1
|
Pu W, Shen X, Fan X, Zheng Y, Liu X, Li J, Zhou JK, He J, Wei R, Gong Y, Zheng Q, Luo Y, Guo Y, Ai M, Ming Y, Ye Z, Zhao Y, Wang C, Peng Y. Structure-Guided Optimization and Preclinical Evaluation of 6- O-Benzylguanine-Based Pin1 Inhibitor for Hepatocellular Carcinoma Treatment. J Med Chem 2025. [PMID: 39868498 DOI: 10.1021/acs.jmedchem.4c02144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related deaths globally, and the need for effective systemic therapies for HCC is urgent. Our previous work reveals that Pin1 is a potential anti-HCC target, which regulates miRNA biogenesis and identifies API-1 as a novel Pin1 inhibitor to suppresses HCC. However, a great demand in HCC therapy as well as the limited chemical stability and pharmacokinetic feature of API-1 motivated us to find improved Pin1 inhibitors. Herein, we designed and synthesized diverse 6-O-benzylguanine derivatives and discovered API-32 as a novel Pin1 inhibitor with better stability and pharmacokinetic property over API-1. API-32 directly interacted with the Pin1 PPIase domain to inhibit Pin1 activity. API-32 significantly suppressed the cell proliferation and migration of HCC cells by blocking Pin1's downstream signal. Moreover, API-32 exhibited an enhanced inhibitory function against the HCC tumor in mice models without obvious toxicity, making it a promising drug candidate for HCC treatment.
Collapse
Affiliation(s)
- Wenchen Pu
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Xianyan Shen
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Xin Fan
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610041, China
| | - Yuanyuan Zheng
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Xuesha Liu
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Jiao Li
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Jian-Kang Zhou
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, China
| | - Juan He
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Rong Wei
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yanqiu Gong
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Qingquan Zheng
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yao Luo
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yingli Guo
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Min Ai
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yue Ming
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Zixia Ye
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
| | - Yun Zhao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610041, China
| | - Chun Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Yong Peng
- Center for Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610064, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China
| |
Collapse
|
2
|
Zhu S, Jin G, He X, Li Y, Xu F, Guo H. Mechano-assisted strategies to improve cancer chemotherapy. Life Sci 2024; 359:123178. [PMID: 39471901 DOI: 10.1016/j.lfs.2024.123178] [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: 05/23/2024] [Revised: 07/25/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024]
Abstract
Chemotherapy remains a cornerstone in cancer treatment; however, its effectiveness is frequently undermined by the development of drug resistance. Recent studies underscores the pivotal role of the tumor mechanical microenvironment (TMME) and the emerging field of mechanical nanomedicine in tackling chemo-resistance. This review offers an in-depth analysis of mechano-assisted strategies aimed at mitigating chemo-resistance through the modification of the TMME and the refinement of mechanical nanomedicine delivery systems. We explore the potential of targeting abnormal tumor mechanical properties as a promising avenue for enhancing the efficacy of cancer chemotherapy, which offers novel directions for advancing future cancer therapies, especially from the mechanomedicine perspective.
Collapse
Affiliation(s)
- Shanshan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Guorui Jin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Xiaocong He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuan Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| | - Hui Guo
- Department of Medical Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, PR China.
| |
Collapse
|
3
|
Liu H, Xing F, Yu P, Shakya S, Peng K, Liu M, Xiang Z, Ritz U. Integrated design and application of stimuli-responsive metal-organic frameworks in biomedicine: current status and future perspectives. J Mater Chem B 2024; 12:8235-8266. [PMID: 39058314 DOI: 10.1039/d4tb00768a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
In recent years, metal-organic frameworks (MOFs) have garnered widespread attention due to their distinctive attributes, such as high surface area, tunable properties, biodegradability, extremely low density, high loading capacity, diverse chemical functionalities, thermal stability, well-defined pore sizes, and molecular dimensions. Increasingly, biomedical researchers have turned their focus towards their multifaceted development. Among these, stimuli-responsive MOFs, with their unique advantages, have captured greater interest from researchers. This review will delve into the merits and drawbacks of both endogenous and exogenous stimuli-responsive MOFs, along with their application directions. Furthermore, it will outline the characteristics of different synthesis routes of MOFs, exploring various design schemes and modification strategies and their impacts on the properties of MOF products, as well as how to control them. Additionally, we will survey different types of stimuli-responsive MOFs, discussing the significance of various MOF products reported in biomedical applications. We will categorically summarize different strategies such as anticancer therapy, antibacterial treatment, tissue repair, and biomedical imaging, as well as insights into the development of novel MOFs nanomaterials in the future. Finally, this review will conclude by summarizing the challenges in the development of stimuli-responsive MOFs in the field of biomedicine and providing prospects for future research endeavors.
Collapse
Affiliation(s)
- Hao Liu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, 610041 Chengdu, China.
| | - Fei Xing
- Department of Pediatric Surgery, West China Hospital, Sichuan University, 610041 Chengdu, China
| | - Peiyun Yu
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Sujan Shakya
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, 610041 Chengdu, China.
| | - Kun Peng
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiang Xi, China
| | - Ming Liu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, 610041 Chengdu, China.
| | - Zhou Xiang
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, 610041 Chengdu, China.
- Department of Orthopedics, Sanya People's Hospital, 572000 Sanya, Hainan, China
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
4
|
Bozuyuk U, Wrede P, Yildiz E, Sitti M. Roadmap for Clinical Translation of Mobile Microrobotics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311462. [PMID: 38380776 DOI: 10.1002/adma.202311462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/24/2024] [Indexed: 02/22/2024]
Abstract
Medical microrobotics is an emerging field to revolutionize clinical applications in diagnostics and therapeutics of various diseases. On the other hand, the mobile microrobotics field has important obstacles to pass before clinical translation. This article focuses on these challenges and provides a roadmap of medical microrobots to enable their clinical use. From the concept of a "magic bullet" to the physicochemical interactions of microrobots in complex biological environments in medical applications, there are several translational steps to consider. Clinical translation of mobile microrobots is only possible with a close collaboration between clinical experts and microrobotics researchers to address the technical challenges in microfabrication, safety, and imaging. The clinical application potential can be materialized by designing microrobots that can solve the current main challenges, such as actuation limitations, material stability, and imaging constraints. The strengths and weaknesses of the current progress in the microrobotics field are discussed and a roadmap for their clinical applications in the near future is outlined.
Collapse
Affiliation(s)
- Ugur Bozuyuk
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Paul Wrede
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Institute for Biomedical Engineering, ETH Zurich, Zurich, 8093, Switzerland
| | - Erdost Yildiz
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Metin Sitti
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- School of Medicine and College of Engineering, Koc University, Istanbul, 34450, Turkey
| |
Collapse
|
5
|
Ferdous J, Bhuia MS, Chowdhury R, Rakib AI, Aktar MA, Al Hasan MS, Melo Coutinho HD, Islam MT. Pharmacological Activities of Plant-Derived Fraxin with Molecular Mechanisms: A Comprehensive Review. Chem Biodivers 2024; 21:e202301615. [PMID: 38506600 DOI: 10.1002/cbdv.202301615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 03/21/2024]
Abstract
Fruits and vegetables serve not only as sources of nutrition but also as medicinal agents for the treatment of diverse diseases and maladies. These dietary components are significant resources of phytochemicals that demonstrate therapeutic properties against many illnesses. Fraxin is a naturally occurring coumarin glycoside mainly present in various species of Fraxinus genera, having a multitude of therapeutic uses against various diseases and disorders. This study focuses to investigate the pharmacological activities, botanical sources, and biopharmaceutical profile of the phytochemical fraxin based on different preclinical and non-clinical studies to show the scientific evidence and to evaluate the underlying molecular mechanisms of the therapeutic effects against various ailments. For this, data was searched and collected (as of February 15, 2024) in a variety of credible electronic databases, including PubMed/Medline, Scopus, Springer Link, ScienceDirect, Wiley Online, Web of Science, and Google Scholar. The findings demonstrated favorable outcomes in relation to a range of diseases or medical conditions, including inflammation, neurodegenerative disorders such as cerebral ischemia-reperfusion (I/R) and depression, viral infection, as well as diabetic nephropathy. The phytochemical also showed protective effects such as osteoprotective, renoprotective, pulmoprotective, hepatoprotective, and gastroprotective effects due to its antioxidant capacity. Fraxin has a great capability to diminish oxidative stress-related damage in different organs by stimulating the antioxidant enzymes, downregulating nuclear factor kappa B and NLRP3, and triggering the Nrf2/ARE signaling pathways. Fraxin exhibited poor oral bioavailability because of reduced absorption and a wide distribution into tissues of different organs. However, extensive research is required to decipher the biopharmaceutical profiles, and clinical studies are necessary to establish the efficacy of the natural compound as a reliable therapeutic agent.
Collapse
Affiliation(s)
- Jannatul Ferdous
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- BioLuster Research Center, Gopalganj, 8100, Dhaka, Bangladesh
| | - Md Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- BioLuster Research Center, Gopalganj, 8100, Dhaka, Bangladesh
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Asraful Islam Rakib
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Mst Asma Aktar
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Md Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- BioLuster Research Center, Gopalganj, 8100, Dhaka, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna, 9208, Bangladesh
| |
Collapse
|
6
|
Sztukowski KE, Yaufman Z, Cook MR, Aarnes TK, Husbands BD. Vincristine-induced adverse events related to body weight in dogs treated for lymphoma. J Vet Intern Med 2024; 38:1686-1692. [PMID: 38563346 PMCID: PMC11099714 DOI: 10.1111/jvim.17063] [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: 08/08/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Traditional dosing of chemotherapy drugs based on body surface area may overdose small dogs, leading to an increased frequency of adverse events (AEs). HYPOTHESIS/OBJECTIVES Evaluate the frequency of hematologic and gastrointestinal AEs in dogs with newly diagnosed lymphoma treated with vincristine weighing ≤15 kg in comparison to dogs weighing >15 kg. We hypothesized that dogs weighing ≤15 kg would experience a higher frequency of AEs. ANIMALS One hundred and thirty-eight dogs with newly diagnosed lymphoma were treated with vincristine. METHODS A multicenter retrospective study reviewing hematologic data and medical record information. Complete blood counts were performed no more than 24 hours before vincristine administration and then between 4 and 8 days post-administration. Data were evaluated using logistic regression or ordinal logistic regression. RESULTS Thirty-eight dogs weighing ≤15 kg and 100 dogs weighing >15 kg were included. The median vincristine dose for both groups was 0.6 mg/m2. Seventeen (12.3%) instances of neutropenia occurred with no significant difference in overall frequency or grade between groups. Thirty initially asymptomatic substage A dogs (29.4%) experienced gastrointestinal AEs. Because of the widespread use of gastrointestinal supportive care medications, statistical comparison between groups could not be performed. Seven instances of hospitalization occurred (5.0%) and the risk of hospitalization did not differ significantly between groups (P = .37). CONCLUSIONS AND CLINICAL IMPORTANCE Vincristine dosed at ≤0.6 mg/m2 does not increase the risk of hematologic AEs in dogs weighing ≤15 kg.
Collapse
Affiliation(s)
- Keira E. Sztukowski
- Department of Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
| | - Zachary Yaufman
- Department of Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
- Present address:
Department of Small Animal Clinical SciencesVirginia‐Maryland College of Veterinary MedicineRoanokeVirginiaUSA
| | - Matthew R. Cook
- Nashville Veterinary SpecialistsNashvilleTennesseeUSA
- Present address:
Metropolitan Veterinary HospitalHighland HeightsOhioUSA
| | - Turi K. Aarnes
- Department of Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
| | - Brian D. Husbands
- Department of Clinical SciencesThe Ohio State University College of Veterinary MedicineColumbusOhioUSA
| |
Collapse
|
7
|
Zhou D, Byers LA, Sable B, Smit MAD, Sadraei NH, Dutta S, Upreti VV. Clinical Pharmacology Profile of AMG 119, the First Chimeric Antigen Receptor T (CAR-T) Cell Therapy Targeting Delta-Like Ligand 3 (DLL3), in Patients with Relapsed/Refractory Small Cell Lung Cancer (SCLC). J Clin Pharmacol 2024; 64:362-370. [PMID: 37694295 DOI: 10.1002/jcph.2346] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/06/2023] [Indexed: 09/12/2023]
Abstract
With the promise of a potentially single-dose curative regimen, CAR-T cell therapies have brought a paradigm shift in the treatment and management of hematological malignancies with 6 approved products in the USA. However, there are no approved CAR-T cell therapies for solid tumors. Herein, we report the clinical pharmacology profile of AMG 119, the first CAR-T cell therapy targeting delta-like ligand 3 (DLL3), in patients with relapsed/refractory (R/R) small cell lung cancer (SCLC). AMG 119 demonstrated robust cellular expansion with long-lasting cell persistence and a favorable exposure-response relationship. AMG 119 has been demonstrated to be clinically safe and well tolerated at the doses tested, with no dose-limiting toxicities (DLTs) reported. This is the first publication of the clinical pharmacology profile of a CAR-T cell therapy in SCLC, with encouraging cellular kinetics data supporting the potential for CAR-T cell therapy in solid tumor space.
Collapse
Affiliation(s)
- Di Zhou
- Clinical Pharmacology, Modeling & Simulation, Amgen Inc, South San Francisco, CA, USA
| | - Lauren A Byers
- Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Beate Sable
- Clinical Biomarker, Amgen Inc, Thousand Oaks, CA, USA
| | | | | | - Sandeep Dutta
- Clinical Pharmacology, Modeling & Simulation, Amgen Inc, Thousand Oaks, CA, USA
| | - Vijay V Upreti
- Clinical Pharmacology, Modeling & Simulation, Amgen Inc, South San Francisco, CA, USA
| |
Collapse
|
8
|
Tangsiri M, Hheidari A, Liaghat M, Razlansari M, Ebrahimi N, Akbari A, Varnosfaderani SMN, Maleki-Sheikhabadi F, Norouzi A, Bakhtiyari M, Zalpoor H, Nabi-Afjadi M, Rahdar A. Promising applications of nanotechnology in inhibiting chemo-resistance in solid tumors by targeting epithelial-mesenchymal transition (EMT). Biomed Pharmacother 2024; 170:115973. [PMID: 38064969 DOI: 10.1016/j.biopha.2023.115973] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/25/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024] Open
Abstract
The resistance of cancer cells to chemotherapy, also known as chemo-resistance, poses a significant obstacle to cancer treatment and can ultimately result in patient mortality. Epithelial-mesenchymal transition (EMT) is one of the many factors and processes responsible for chemo-resistance. Studies have shown that targeting EMT can help overcome chemo-resistance, and nanotechnology and nanomedicine have emerged as promising approaches to achieve this goal. This article discusses the potential of nanotechnology in inhibiting EMT and proposes a viable strategy to combat chemo-resistance in various solid tumors, including breast cancer, lung cancer, pancreatic cancer, glioblastoma, ovarian cancer, gastric cancer, and hepatocellular carcinoma. While nanotechnology has shown promising results in targeting EMT, further research is necessary to explore its full potential in overcoming chemo-resistance and discovering more effective methods in the future.
Collapse
Affiliation(s)
- Mona Tangsiri
- Department of Medical Entomology and Vector Control, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Hheidari
- Department of Mechanical Engineering, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mahsa Liaghat
- Department of Medical Laboratory sciences, Faculty of Medical Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran; Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mahtab Razlansari
- Faculty of Mathematics and Natural Sciences, Tübingen University, Tübingen 72076, Germany
| | - Narges Ebrahimi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Abdullatif Akbari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran; Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Fahimeh Maleki-Sheikhabadi
- Department of Hematology and Blood Banking, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Norouzi
- Dental Research Center, Faculty of Dentistry, Mazandaran University of Medical Sciences, Sari, Iran
| | - Maryam Bakhtiyari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran; Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran; Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran.
| |
Collapse
|
9
|
Kumar P, Yadav N, Chaudhary B, Umakanthan S, Chattu VK, Kazmi I, Al-Abbasi FA, Alzarea SI, Afzal O, Altamimi ASA, Gupta G, Gupta MM. Lipid Nanocapsule: A Novel Approach to Drug Delivery System Formulation Development. Curr Pharm Biotechnol 2024; 25:268-284. [PMID: 37231750 DOI: 10.2174/1389201024666230523114350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/08/2022] [Accepted: 01/05/2023] [Indexed: 05/27/2023]
Abstract
Nanocapsules are polymeric nanoparticles encased in a polymeric coating composed of a predominantly non-ionic surfactant, macromolecules, phospholipids, and an oil core. Lipophilic drugs have been entrapped using various nanocarriers, including lipid cores, likely lipid nanocapsules, solid lipid nanoparticles, and others. A phase inversion temperature approach is used to create lipid nanocapsules. The PEG (polyethyleneglycol) is primarily utilised to produce nanocapsules and is a critical parameter influencing capsule residence time. With their broad drug-loading features, lipid nanocapsules have a distinct advantage in drug delivery systems, such as the capacity to encapsulate hydrophilic or lipophilic pharmaceuticals. Lipid nanocapsules, as detailed in this review, are surface modified, contain target-specific patterns, and have stable physical and chemical properties. Furthermore, lipid nanocapsules have target-specific delivery and are commonly employed as a marker in the diagnosis of numerous illnesses. This review focuses on nanocapsule synthesis, characterisation, and application, which will help understand the unique features of nanocapsules and their application in drug delivery systems.
Collapse
Affiliation(s)
- Parveen Kumar
- Shri Ram College of Pharmacy, Karnal, Haryana, India
| | - Nishant Yadav
- B. S. Anangpuria Institute of Pharmacy, Faridabad, Haryana, India
| | - Benu Chaudhary
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar, Haryana, India
| | - Srikant Umakanthan
- Department of Paraclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago, WI
| | - Vijay K Chattu
- Department of OS & OT, Temerty Faculty of Medicine, University of Toronto, ON M5G 1V7, Canada
- Center for Transdisciplinary Research, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
- Center for Technology and Innovations, Global Health Research and Innovations Canada Inc. (GHRIC), ON, Toronto, Canada
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University Jeddah 21589, Saudi Arabia
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University Jeddah 21589, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, 11942, Saudi Arabia
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, 11942, Saudi Arabia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura 302017, Mahal Road, Jaipur, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Madan M Gupta
- School of Pharmacy, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago, West Indies
| |
Collapse
|
10
|
Li S, Dai J, Zhu M, Arroyo-Currás N, Li H, Wang Y, Wang Q, Lou X, Kippin TE, Wang S, Plaxco KW, Li H, Xia F. Implantable Hydrogel-Protective DNA Aptamer-Based Sensor Supports Accurate, Continuous Electrochemical Analysis of Drugs at Multiple Sites in Living Rats. ACS NANO 2023; 17:18525-18538. [PMID: 37703911 DOI: 10.1021/acsnano.3c06520] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
The ability to track the levels of specific molecules, such as drugs, metabolites, and biomarkers, in the living body, in real time and for long durations, would improve our understanding of health and our ability to diagnose, treat, and monitor disease. To this end, we are developing electrochemical aptamer-based (EAB) biosensors, a general platform supporting high-frequency, real-time molecular measurements in the living body. Here we report that the use of an agarose hydrogel protective layer for EAB sensors significantly improves their signaling stability when deployed in the complex, highly time-varying environments found in vivo. The improved stability is sufficient that these hydrogel-protected sensors achieved good baseline stability and precision when deployed in situ in the veins, muscles, bladder, or tumors of living rats without the use of the drift correction approaches traditionally required in such placements. Finally, our implantable gel-protective EAB sensors achieved good biocompatibility when deployed in vivo in the living rats without causing any severe inflammation.
Collapse
Affiliation(s)
- Shaoguang Li
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, People's Republic of China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| | - Man Zhu
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, People's Republic of China
| | - Netzahualcóyotl Arroyo-Currás
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Hongxing Li
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, People's Republic of China
| | - Yuanyuan Wang
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, People's Republic of China
| | - Quan Wang
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, People's Republic of China
| | - Xiaoding Lou
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, People's Republic of China
| | - Tod E Kippin
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, California 93106, United States
- The Neuroscience Research Institute, University of California, Santa Barbara, California 93106, United States
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, People's Republic of China
| | - Kevin W Plaxco
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, California 93106, United States
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
- Center for Bioengineering, University of California, Santa Barbara, California 93106, United States
- Interdepartmental Program in Biomolecular Science and Engineering, University of California, Santa Barbara, California 93106, United States
| | - Hui Li
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, People's Republic of China
| | - Fan Xia
- State Key Laboratory of Biogeology Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, People's Republic of China
| |
Collapse
|
11
|
Foudah AI, Alam A, Salkini MA, Ross SA, Kumar P, Aldawsari MF, Alqarni MH, Sweilam SH. Synergistic Combination of Letrozole and Berberine in Ascorbic Acid-Stabilized AuNPs: A Promising Solution for Breast Cancer. Pharmaceuticals (Basel) 2023; 16:1099. [PMID: 37631014 PMCID: PMC10459502 DOI: 10.3390/ph16081099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer is a deadly disease that affects countless women worldwide. The most conventional treatments for breast cancer, such as the administration of anticancer medications such as letrozole (LTZ), pose significant barriers due to the non-selective delivery and low bioavailability of cytotoxic drugs leading to serious adverse effects and multidrug resistance (MDR). Addressing these obstacles requires an innovative approach, and we propose a combined strategy that synergistically incorporates LTZ with berberine (BBR) into stabilised AuNPs coated with ascorbic acid (AA), known as LTZ-BBR@AA-AuNPs. The LTZ-BBR@AA-AuNPs, a novel combined drug delivery system, were carefully designed to maximise the entrapment of both LTZ and BBR. The resulting spherical nanoparticles exhibited remarkable efficiency in trapping these two compounds, with rates of 58% and 54%, respectively. In particular, the average hydrodynamic diameter of these nanoparticles was determined to be 81.23 ± 4.0 nm with a PDI value of only 0.286, indicating excellent uniformity between them. Furthermore, their zeta potential was observed to be -14.5 mV, suggesting high stability even under physiological conditions. The release profiles showed that after being incubated for about 24 h at pH levels ranging from acidic (pH = 5) to basic (pH = 7), the percentage released for both drugs ranged from 56-72%. This sustained and controlled drug release can reduce any negative side effects while improving therapeutic efficacy when administered directly to cancer. MDA-MB-231 cells treated with LTZ-BBR@AA-AuNPs for 48 h exhibited IC50 values of 2.04 ± 0.011 μg/mL, indicating potent cytotoxicity against cells. Furthermore, the nanoparticles demonstrated excellent stability throughout the duration of the treatment.
Collapse
Affiliation(s)
- Ahmed I. Foudah
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia; (A.A.); (M.A.S.); (M.H.A.); (S.H.S.)
| | - Aftab Alam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia; (A.A.); (M.A.S.); (M.H.A.); (S.H.S.)
| | - Mohammad Ayman Salkini
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia; (A.A.); (M.A.S.); (M.H.A.); (S.H.S.)
| | - Samir A. Ross
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA;
- Department of Biomolecular Sciences, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA
| | - Piyush Kumar
- Department of Chemistry, Indian Institute of Technology, NH-44, PO Nagrota, Jagti, Jammu 181221, India;
| | - Mohammed F. Aldawsari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia;
| | - Mohammed H. Alqarni
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia; (A.A.); (M.A.S.); (M.H.A.); (S.H.S.)
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia; (A.A.); (M.A.S.); (M.H.A.); (S.H.S.)
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo 11829, Egypt
| |
Collapse
|
12
|
Nishikubo K, Ohgaki R, Liu X, Okanishi H, Xu M, Endou H, Kanai Y. Combination effects of amino acid transporter LAT1 inhibitor nanvuranlat and cytotoxic anticancer drug gemcitabine on pancreatic and biliary tract cancer cells. Cancer Cell Int 2023; 23:116. [PMID: 37322479 DOI: 10.1186/s12935-023-02957-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/26/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Cytotoxic anticancer drugs widely used in cancer chemotherapy have some limitations, such as the development of side effects and drug resistance. Furthermore, monotherapy is often less effective against heterogeneous cancer tissues. Combination therapies of cytotoxic anticancer drugs with molecularly targeted drugs have been pursued to solve such fundamental problems. Nanvuranlat (JPH203 or KYT-0353), an inhibitor for L-type amino acid transporter 1 (LAT1; SLC7A5), has novel mechanisms of action to suppress the cancer cell proliferation and tumor growth by inhibiting the transport of large neutral amino acids into cancer cells. This study investigated the potential of the combined use of nanvuranlat and cytotoxic anticancer drugs. METHODS The combination effects of cytotoxic anticancer drugs and nanvuranlat on cell growth were examined by a water-soluble tetrazolium salt assay in two-dimensional cultures of pancreatic and biliary tract cancer cell lines. To elucidate the pharmacological mechanisms underlying the combination of gemcitabine and nanvuranlat, we investigated apoptotic cell death and cell cycle by flow cytometry. The phosphorylation levels of amino acid-related signaling pathways were analyzed by Western blot. Furthermore, growth inhibition was examined in cancer cell spheroids. RESULTS All the tested seven types of cytotoxic anticancer drugs combined with nanvuranlat significantly inhibited the cell growth of pancreatic cancer MIA PaCa-2 cells compared to their single treatment. Among them, the combined effects of gemcitabine and nanvuranlat were relatively high and confirmed in multiple pancreatic and biliary tract cell lines in two-dimensional cultures. The growth inhibitory effects were suggested to be additive but not synergistic under the tested conditions. Gemcitabine generally induced cell cycle arrest at the S phase and apoptotic cell death, while nanvuranlat induced cell cycle arrest at the G0/G1 phase and affected amino acid-related mTORC1 and GAAC signaling pathways. In combination, each anticancer drug basically exerted its own pharmacological activities, although gemcitabine more strongly influenced the cell cycle than nanvuranlat. The combination effects of growth inhibition were also verified in cancer cell spheroids. CONCLUSIONS Our study demonstrates the potential of first-in-class LAT1 inhibitor nanvuranlat as a concomitant drug with cytotoxic anticancer drugs, especially gemcitabine, on pancreatic and biliary tract cancers.
Collapse
Affiliation(s)
- Kou Nishikubo
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ryuichi Ohgaki
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan.
| | - Xingming Liu
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroki Okanishi
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Minhui Xu
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | | | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan.
| |
Collapse
|
13
|
Abstract
Cooperativity (homotropic allostery) is the primary mechanism by which evolution steepens the binding curves of biomolecular receptors to produce more responsive input-output behavior in biomolecular systems. Motivated by the ubiquity with which nature employs this effect, over the past 15 years we, together with other groups, have engineered this mechanism into several otherwise noncooperative receptors. These efforts largely aimed to improve the utility of such receptors in artificial biotechnologies, such as synthetic biology and biosensors, but they have also provided the first quantitative, experimental tests of longstanding ideas about the mechanisms underlying cooperativity. In this article, we review the literature on the design of this effect, paying particular attention to the design strategies involved, the extent to which each can be rationally applied to (and optimized for) new receptors, and what each teaches us about the origins and optimization of this important phenomenon.
Collapse
Affiliation(s)
- Gabriel Ortega
- Precision Medicine and Metabolism Laboratory, CIC bioGUNE, Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Alejandro Chamorro-Garcia
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California, USA;
- Biological Engineering Graduate Program, University of California, Santa Barbara, California, USA
- Dipartimento di Scienze e Tecnologie Chimiche, University of Rome Tor Vergata, Rome, Italy
| | - Francesco Ricci
- Dipartimento di Scienze e Tecnologie Chimiche, University of Rome Tor Vergata, Rome, Italy
| | - Kevin W Plaxco
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California, USA;
- Biological Engineering Graduate Program, University of California, Santa Barbara, California, USA
| |
Collapse
|
14
|
Chamorro-Garcia A, Gerson J, Flatebo C, Fetter L, Downs AM, Emmons N, Ennis HL, Milosavić N, Yang K, Stojanovic M, Ricci F, Kippin TE, Plaxco KW. Real-Time, Seconds-Resolved Measurements of Plasma Methotrexate In Situ in the Living Body. ACS Sens 2023; 8:150-157. [PMID: 36534756 DOI: 10.1021/acssensors.2c01894] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dose-limiting toxicity and significant patient-to-patient pharmacokinetic variability often render it difficult to achieve the safe and effective dosing of drugs. This is further compounded by the slow, cumbersome nature of the analytical methods used to monitor patient-specific pharmacokinetics, which inevitably rely on blood draws followed by post-facto laboratory analysis. Motivated by the pressing need for improved "therapeutic drug monitoring", we are developing electrochemical aptamer-based (EAB) sensors, a minimally invasive biosensor architecture that can provide real-time, seconds-resolved measurements of drug levels in situ in the living body. A key advantage of EAB sensors is that they are generalizable to the detection of a wide range of therapeutic agents because they are independent of the chemical or enzymatic reactivity of their targets. Three of the four therapeutic drug classes that have, to date, been shown measurable using in vivo EAB sensors, however, bind to nucleic acids as part of their mode of action, leaving open questions regarding the extent to which the approach can be generalized to therapeutics that do not. Here, we demonstrate real-time, in vivo measurements of plasma methotrexate, an antimetabolite (a mode of action not reliant on DNA binding) chemotherapeutic, following human-relevant dosing in a live rat animal model. By providing hundreds of drug concentration values, the resulting seconds-resolved measurements succeed in defining key pharmacokinetic parameters, including the drug's elimination rate, peak plasma concentration, and exposure (area under the curve), with unprecedented 5 to 10% precision. With this level of precision, we easily identify significant (>2-fold) differences in drug exposure occurring between even healthy rats given the same mass-adjusted methotrexate dose. By providing a real-time, seconds-resolved window into methotrexate pharmacokinetics, such measurements can be used to precisely "individualize" the dosing of this significantly toxic yet vitally important chemotherapeutic.
Collapse
Affiliation(s)
- Alejandro Chamorro-Garcia
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States.,Dipartimento di Scienze e Tecnologie Chimiche, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Julian Gerson
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Charlotte Flatebo
- Institute for Collaborative Biotechnologies, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Lisa Fetter
- Biomolecular Science and Engineering Program, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Alex M Downs
- Department of Mechanical Engineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Nicole Emmons
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Herbert L Ennis
- Center for Innovative Diagnostic and Therapeutic Approaches, Department of Medicine, Columbia University New York, New York, New York 10032, United States
| | - Nenad Milosavić
- Center for Innovative Diagnostic and Therapeutic Approaches, Department of Medicine, Columbia University New York, New York, New York 10032, United States
| | - Kyungae Yang
- Center for Innovative Diagnostic and Therapeutic Approaches, Department of Medicine, Columbia University New York, New York, New York 10032, United States
| | - Milan Stojanovic
- Center for Innovative Diagnostic and Therapeutic Approaches, Department of Medicine, Columbia University New York, New York, New York 10032, United States.,Department of Biomedical Engineering and Systems Biology, Columbia University New York, New York, New York 10032, United States
| | - Francesco Ricci
- Dipartimento di Scienze e Tecnologie Chimiche, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Tod E Kippin
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Kevin W Plaxco
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States.,Biomolecular Science and Engineering Program, University of California Santa Barbara, Santa Barbara, California 93106, United States.,Department of Mechanical Engineering, University of California Santa Barbara, Santa Barbara, California 93106, United States.,Biological Engineering Graduate Program, University of California Santa Barbara, Santa Barbara, California 93106, United States
| |
Collapse
|
15
|
Fatima M, Karwasra R, Almalki WH, Sahebkar A, Kesharwani P. Galactose engineered nanocarriers: Hopes and hypes in cancer therapy. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
16
|
Khan MW, Zou C, Hassan S, Din FU, Abdoul Razak MY, Nawaz A, Alam Zeb, Wahab A, Bangash SA. Cisplatin and oleanolic acid Co-loaded pH-sensitive CaCO 3 nanoparticles for synergistic chemotherapy. RSC Adv 2022; 12:14808-14818. [PMID: 35702211 PMCID: PMC9109477 DOI: 10.1039/d2ra00742h] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/01/2022] [Indexed: 01/06/2023] Open
Abstract
Despite being one of the most potent anticancer agents, cisplatin (CDDP) clinical usage is limited owing to the acquired resistance and severe adverse effects including nephrotoxicity. The current work has offered a unique approach by designing a pH-sensitive calcium carbonate drug delivery system for CDDP and oleanolic acid (OA) co-delivery, with an enhanced tumor efficacy and reduced unwanted effects. Micro emulsion method was employed to generate calcium carbonate cores (CDDP encapsulated) followed by lipid coating along with the OA loading resulting in the generation of lipid-coated cisplatin/oleanolic acid calcium carbonate nanoparticles (CDDP/OA-LCC NPs). In vitro biological assays confirmed the synergistic apoptotic effect of CDDP and OA against HepG2 cells. It was further verified in vivo through the tumor-bearing nude mice model where NPs exhibited enhanced satisfactory antitumor efficacy in contrast to free drug solutions. In vivo pharmacokinetic study demonstrated that a remarkable long circulation time with a constant therapeutic concentration for both drugs could be achieved via this drug delivery system. In addition, the in vivo imaging study revealed that DiR-loaded NPs were concentrated more in tumors for a longer period of time as compared to other peritoneal tissues in tumor bearing mice, demonstrating the site specificity of the delivery system. On the other hand, hematoxylin and eosin (H&E) staining of Kunming mice kidney tissue sections revealed that OA greatly reduced CDDP induced nephrotoxicity in the formulation. Overall, these results confirmed that our pH-sensitive dual loaded drug delivery system offers a handy direction for effective and safer combination chemotherapy.
Collapse
Affiliation(s)
- Muhammad Waseem Khan
- Institute of Pharmaceutical Sciences, Khyber Medical University Peshawar Pakistan +92-3459146065
| | - Chenming Zou
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology Wuhan Hubei 430030 China
| | - Said Hassan
- Institute of Biotechnology and Microbiology, Bacha Khan University Charsadda Pakistan
| | - Fakhar Ud Din
- Department of Pharmacy, Quaid-I-Azam University Islamabad 45320 Pakistan
| | - Mahaman Yacoubou Abdoul Razak
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 China
| | - Asif Nawaz
- Faculty of Pharmacy, Gomal University Dera Ismail Khan Pakistan
| | - Alam Zeb
- Riphah Institute of Pharmaceutical Sciences, Riphah International University Islamabad Pakistan
| | - Abdul Wahab
- Department of Pharmacy, Kohat University of Science and Technology Kohat Pakistan
| | - Sudhair Abbas Bangash
- Faculty of Life Science, Department of Pharmacy, Sarhad University of Science and Information Technology Peshawar Pakistan
| |
Collapse
|
17
|
Xiao J, Lu Y, Lu D, Chen W, Hu W, Zhao Y, Chen S. Co‐delivery of paclitaxel and
CXCL1 shRNA
via cationic polymeric micelles for synergistic therapy against ovarian cancer. POLYM INT 2022. [DOI: 10.1002/pi.6406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Jingjing Xiao
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Yingying Lu
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Deng Lu
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Wulian Chen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science Fudan University Shanghai 200433 PR China
| | - Weiguo Hu
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Yuqing Zhao
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| | - Shouzhen Chen
- Obstetrics and gynecology hospital, Shanghai Medical college Fudan University Shen Yang road, No 128 Shanghai 200090 PR China
| |
Collapse
|
18
|
Winkler S, Winkler I, Figaschewski M, Tiede T, Nordheim A, Kohlbacher O. De novo identification of maximally deregulated subnetworks based on multi-omics data with DeRegNet. BMC Bioinformatics 2022; 23:139. [PMID: 35439941 PMCID: PMC9020058 DOI: 10.1186/s12859-022-04670-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 03/29/2022] [Indexed: 12/14/2022] Open
Abstract
Background With a growing amount of (multi-)omics data being available, the extraction of knowledge from these datasets is still a difficult problem. Classical enrichment-style analyses require predefined pathways or gene sets that are tested for significant deregulation to assess whether the pathway is functionally involved in the biological process under study. De novo identification of these pathways can reduce the bias inherent in predefined pathways or gene sets. At the same time, the definition and efficient identification of these pathways de novo from large biological networks is a challenging problem. Results We present a novel algorithm, DeRegNet, for the identification of maximally deregulated subnetworks on directed graphs based on deregulation scores derived from (multi-)omics data. DeRegNet can be interpreted as maximum likelihood estimation given a certain probabilistic model for de-novo subgraph identification. We use fractional integer programming to solve the resulting combinatorial optimization problem. We can show that the approach outperforms related algorithms on simulated data with known ground truths. On a publicly available liver cancer dataset we can show that DeRegNet can identify biologically meaningful subgraphs suitable for patient stratification. DeRegNet can also be used to find explicitly multi-omics subgraphs which we demonstrate by presenting subgraphs with consistent methylation-transcription patterns. DeRegNet is freely available as open-source software. Conclusion The proposed algorithmic framework and its available implementation can serve as a valuable heuristic hypothesis generation tool contextualizing omics data within biomolecular networks.
Collapse
Affiliation(s)
- Sebastian Winkler
- Applied Bioinformatics, Department of Computer Science, University of Tuebingen, Tübingen, Germany. .,International Max Planck Research School (IMPRS) "From Molecules to Organism", Tübingen, Germany.
| | - Ivana Winkler
- International Max Planck Research School (IMPRS) "From Molecules to Organism", Tübingen, Germany.,Interfaculty Institute for Cell Biology (IFIZ), University of Tuebingen, Tübingen, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mirjam Figaschewski
- Applied Bioinformatics, Department of Computer Science, University of Tuebingen, Tübingen, Germany
| | - Thorsten Tiede
- Applied Bioinformatics, Department of Computer Science, University of Tuebingen, Tübingen, Germany
| | - Alfred Nordheim
- Interfaculty Institute for Cell Biology (IFIZ), University of Tuebingen, Tübingen, Germany.,Leibniz Institute on Aging (FLI), Jena, Germany
| | - Oliver Kohlbacher
- Applied Bioinformatics, Department of Computer Science, University of Tuebingen, Tübingen, Germany.,Institute for Bioinformatics and Medical Informatics, University of Tuebingen, Tübingen, Germany.,Translational Bioinformatics, University Hospital Tuebingen, Tübingen, Germany
| |
Collapse
|
19
|
Degu A, Terefe EM, Some ES, Tegegne GT. Treatment Outcomes and Its Associated Factors Among Adult Patients with Selected Solid Malignancies at Kenyatta National Hospital: A Hospital-Based Prospective Cohort Study. Cancer Manag Res 2022; 14:1525-1540. [PMID: 35498512 PMCID: PMC9042075 DOI: 10.2147/cmar.s361485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/07/2022] [Indexed: 11/27/2022] Open
Abstract
Introduction The treatment outcome of cancer is poor in the African setting due to inadequate treatment and diagnostic facilities. However, there is a paucity of data on solid cancers in Kenya. Hence, this study aimed to investigate the treatment outcomes and its determinant factors among adult patients diagnosed with selected solid malignancies at Kenyatta National Hospital (KNH). Materials and Methods A prospective cohort study was employed at the Oncology Department of KNH from 1st July 2020 to 31st December 2021. All new patients with a confirmed diagnosis of lymphoma, prostate cancer and breast cancer were studied. A consecutive sample of 99 breast cancer, 50 lymphomas, and 82 prostate cancer patients was included in the study. Semi-structured questionnaires consisting of socio-demographics, clinical characteristics, and quality of life were employed to collect the data. All enrolled patients were followed prospectively for 12 months. Treatment outcomes were reported as mortality, cancer-specific survival and health-related quality of life. The data were entered and analyzed using the SPSS 20.0 statistical software. Survival outcomes and its predictors were evaluated using the Kaplan–Meier analysis and Cox regression analyses, respectively. Results The study showed that the mortality rate among breast and prostate cancer patients was 3% and 4.9%, respectively. In contrast, the mortality rate was 10% among lymphoma patients. Most of the patients had partial remission and a good overall global health-related quality of life. Older age above 60 years, co-morbidity, distant metastasis and advanced stages of disease were significant predictors of mortality. Conclusion Although the mortality was not high at 12 months, only a few patients had complete remission. For many patients, the disease was progressing, despite 12-month mortality was not high. Therefore, longer follow-up will be required to report cancer mortality accurately. In addition, most of the patients had a good overall global health-related quality of life.
Collapse
Affiliation(s)
- Amsalu Degu
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, Nairobi, Kenya
- Correspondence: Amsalu Degu, United States International University-Africa, School of Pharmacy and Health Sciences, Nairobi, Kenya, Tel +254745063687, Email
| | - Ermias Mergia Terefe
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, Nairobi, Kenya
| | - Eliab Seroney Some
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, Nairobi, Kenya
| | - Gobezie T Tegegne
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
20
|
The Value of Pharmacogenetics to Reduce Drug-Related Toxicity in Cancer Patients. Mol Diagn Ther 2022; 26:137-151. [PMID: 35113367 PMCID: PMC8975257 DOI: 10.1007/s40291-021-00575-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2021] [Indexed: 10/19/2022]
Abstract
Many anticancer drugs cause adverse drug reactions (ADRs) that negatively impact safety and reduce quality of life. The typical narrow therapeutic range and exposure-response relationships described for anticancer drugs make precision dosing critical to ensure safe and effective drug exposure. Germline mutations in pharmacogenes contribute to inter-patient variability in pharmacokinetics and pharmacodynamics of anticancer drugs. Patients carrying reduced-activity or loss-of-function alleles are at increased risk for ADRs. Pretreatment genotyping offers a proactive approach to identify these high-risk patients, administer an individualized dose, and minimize the risk of ADRs. In the field of oncology, the most well-studied gene-drug pairs for which pharmacogenetic dosing recommendations have been published to improve safety are DPYD-fluoropyrimidines, TPMT/NUDT15-thiopurines, and UGT1A1-irinotecan. Despite the presence of these guidelines, the scientific evidence showing the benefits of pharmacogenetic testing (e.g., improved safety and cost-effectiveness) and the development of efficient multi-gene genotyping panels, routine pretreatment testing for these gene-drug pairs has not been implemented widely in the clinic. Important considerations required for widespread clinical implementation include pharmacogenetic education of physicians, availability or allocation of institutional resources to build an efficient clinical infrastructure, international standardization of guidelines, uniform adoption of guidelines by regulatory agencies leading to genotyping requirements in drug labels, and development of cohesive reimbursement policies for pretreatment genotyping. Without clinical implementation, the potential of pharmacogenetics to improve patient safety remains unfulfilled.
Collapse
|
21
|
Yan M, Fan X, Si H, Wang X, Wang Z, Wang Z, Lv X, Yin H, Jia Y, Jiang L, Xia Y, Liu Y. Association between gene polymorphism and adverse effects in cancer patients receiving docetaxel treatment: a meta-analysis. Cancer Chemother Pharmacol 2022; 89:173-181. [PMID: 34988655 DOI: 10.1007/s00280-021-04374-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 11/10/2021] [Indexed: 11/02/2022]
Abstract
PURPOSE Large interindividual variability in the pharmacokinetic properties of docetaxel has been reported, with the clearance of docetaxel varying nearly six fold, in which pharmacogenetics of docetaxel may play an essential role in addition to physiological factors. The association between the gene polymorphism and risk of adverse clinical effects in docetaxel treated patients has been examined in several studies, but their conclusions are, to some extent, controversial. To clarify the role of gene polymorphism in the clinical outcomes of docetaxel treatment, a meta-analysis was performed in the present study. METHODS Pooled odds ratios (ORs) and corresponding 95% confidence intervals (CIs) were employed to evaluate the impact of gene polymorphisms of CYP3A4, CYP3A5 and ABCB1. Four studies with 485 subjects were included in this study. Fixed or random-effects model was chosen according to heterogeneity to conduct the meta-analysis. Publication bias was evaluated by fail-safe numbers. RESULTS Significant association was identified between the ABCB1 C3435T (rs1045642) polymorphism and risk of short-term recurrent hematological toxicity (TT vs. CC + TC OR = 2.91, 95% CI 1.30-6.52, P = 0.009; TT vs. CC OR = 4.23, 95% CI 1.69-10.57 P = 0.002). The association of the ABCB1 G2677T/A (rs2032582) polymorphism with risk of fluid retention was statistically significant (T(A)/T(A) vs. GG + GT(A) OR = 2.08, 95% CI 1.16-3.73, P = 0.01). No statistically significant association between the CYP3A5 A6986G (rs776746) polymorphism and adverse effects was observed in this study. Due to the limitations of included literature, we did not conduct meta-analysis on CYP3A4 gene polymorphism and adverse effects. CONCLUSION An association between the ABCB1 C3435T (rs1045642), ABCB1 G2677T/A (rs2032582) polymorphism and risk of adverse effects of docetaxel was found by our meta-analysis. Namely, the TT homozygotes of the ABCB1 C3435T polymorphism may be associated with the risk of hematological toxicity. ABCB1 G2677T T(A)/T(A) genotype may be associated with the fluid retention. TRAIL REGISTRATION PROSPERO 2020 CRD42020203132.
Collapse
Affiliation(s)
- Mingrui Yan
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Liaodongwan New District, Panjin, 124221, China
| | - Xiaoyu Fan
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Liaodongwan New District, Panjin, 124221, China
| | - Hongyanhua Si
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Liaodongwan New District, Panjin, 124221, China
| | - Xiaoyu Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Liaodongwan New District, Panjin, 124221, China
| | - Zhe Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Liaodongwan New District, Panjin, 124221, China
| | - Zhen Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Liaodongwan New District, Panjin, 124221, China
| | - Xin Lv
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Liaodongwan New District, Panjin, 124221, China
| | - Hang Yin
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Liaodongwan New District, Panjin, 124221, China
| | - Yanyan Jia
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Liaodongwan New District, Panjin, 124221, China
| | - Lili Jiang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Liaodongwan New District, Panjin, 124221, China
| | - Yangliu Xia
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Liaodongwan New District, Panjin, 124221, China.
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Liaodongwan New District, Panjin, 124221, China.
| |
Collapse
|
22
|
Wu Q, Zhou Y, Fan X, Ma H, Gu W, Sun F. Evaluation of nine formulas for estimating the body surface area of children with hematological malignancies. Front Pediatr 2022; 10:989049. [PMID: 36160791 PMCID: PMC9489993 DOI: 10.3389/fped.2022.989049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Body surface area (BSA) is an important parameter in clinical practice for children. To find out the most accurate BSA formula for Chinese children, nine formulas were compared. METHODS This single-center study comprised children who were diagnosed with acute lymphoblastic leukemia and treated with anticancer agents in a specialized children's hospital in China from January 2017 to December 2020. The BSA values were calculated using the formulas from Boyd, Banerjee and Bhattacharya, Costeff, Fujimoto and Watanabe, Haycock, Gehan and George, Mosteller, Stevenson and a Pediatrics textbook. The arithmetic mean of formulas was calculated as the "gold standard" for comparison. RESULTS A total of 666 children (389 males and 277 females) were included. All nine formulas showed a strong positive correlation with the "gold standard." Underestimation was observed with the Banerjee and Bhattacharya, Fujimoto and Watanabe formulas. The Gehan and George formula showed overestimation. Values estimated from the Haycock and Mosteller formulas were the closest to the mean BSA. CONCLUSION The Haycock and Mosteller formulas are the most recommended formulas for Chinese children with hematological malignancies.
Collapse
Affiliation(s)
- Qing Wu
- Department of Pharmacy, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Chongqing, China
| | - Yan Zhou
- Department of Pharmacy, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Chongqing, China
| | - Xin Fan
- Department of Pharmacy, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, China International Science and Technology Cooperation Base of Child development and Critical Disorders, Chongqing, China
| | - Huan Ma
- Department of Pharmacy, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Wenrui Gu
- Department of Pharmacy, First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Fengjun Sun
- Department of Pharmacy, First Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
23
|
Poetto AS, Posocco B, Gagno S, Orleni M, Zanchetta M, Iacuzzi V, Canil G, Buzzo M, Montico M, Guardascione M, Basile D, Pelizzari G, Alberti M, Gerratana L, Puglisi F, Toffoli G. A new dried blood spot LC-MS/MS method for therapeutic drug monitoring of palbociclib, ribociclib, and letrozole in patients with cancer. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1185:122985. [PMID: 34700133 DOI: 10.1016/j.jchromb.2021.122985] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/06/2021] [Accepted: 10/07/2021] [Indexed: 11/30/2022]
Abstract
Therapeutic drug monitoring (TDM) is strongly suggested to define the proper drug dosage to overcome inter- and intra-patient variability in drug exposure, which is typically observed with oral anticancer agents, such as palbociclib (PALBO), ribociclib (RIBO) and letrozole (LETRO), all approved for the treatment of HR+, HER2- locally advanced or metastatic breast cancer (BC). Optimal TDM implementation requires a blood sampling organization that can be hampered by limited availability of health and laboratory personnel. Dried Blood Spot (DBS) sampling is proposed to overcome such limitations. The aim of this work was the development of a new LC-MS/MS method to analyze DBS samples containing PALBO, RIBO, and LETRO. Analytes extraction from DBS was performed by adding a methanolic solution containing the corresponding internal standards. LC-MS/MS analysis was performed using a LC Nexera (Shimadzu) system coupled with an API 4000 QTrap (SCIEX) mass spectrometer. The chromatographic separation was performed on a Luna Omega Polar C18 column (Phenomenex). The method was applied to 38 clinical samples collected by finger prick. The influence of hematocrit and spot size, sample homogeneity, stability, and correlation between finger prick and venous DBS measurement were assessed. The analytical validation was performed according to EMA and FDA guidelines. The analytical range of the method was 1 to 250 ng/mL for PALBO, 40 to 10000 ng/mL for RIBO, and 2 to 500 ng/mL for LETRO, where linearity was assessed, obtaining mean coefficients of determination (R2) of 0.9979 for PALBO, 0.9980 for RIBO, and 0.9987 for LETRO). The LC-MS/MS method runtime was 6.6 min. Incurred sample reanalysis demonstrated reproducibility, as the percentage difference between the two quantifications was lower than 20% for 100% of PALBO, 81.8% of RIBO, and 90.9% of LETRO paired samples. Intra- and inter-day precision (CV (%)) was lower than 11.4% and intra- and inter-day accuracy was between 90.0 and 106.5%. DBS sample stability at room temperature was confirmed for 2.5 months. A positive correlation was observed between DBS and plasma concentrations for the 3 drugs, Lin's concordance correlation coefficients obtained by DBS normalization applying a selected strategy were 0.958 for PALBO, 0.957 for RIBO, and 0.963 for LETRO. In conclusion, a fast, easy, and reproducible DBS LC-MS/MS method for the simultaneous quantification of palbociclib; ribociclib and letrozole was developed to be used in clinical practice.
Collapse
Affiliation(s)
- Ariana Soledad Poetto
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy; Doctoral School in Pharmacological Sciences, University of Padua, Lgo Meneghetti 2, Padova 35131, Italy
| | - Bianca Posocco
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Sara Gagno
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Marco Orleni
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy; Doctoral School in Pharmacological Sciences, University of Padua, Lgo Meneghetti 2, Padova 35131, Italy
| | - Martina Zanchetta
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy; Department of Chemical and Pharmaceutical Sciences, University of Trieste, via Valerio 8/3, Trieste 34127, Italy
| | - Valentina Iacuzzi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Giovanni Canil
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Mauro Buzzo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Marcella Montico
- Clinical Trial Office, Centro di Riferimento Oncologico di Aviano (CRO), IRCSS, Aviano 33081, Italy
| | - Michela Guardascione
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Debora Basile
- Department of Medicine (DAME), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Giacomo Pelizzari
- Department of Medicine (DAME), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Martina Alberti
- Department of Medicine (DAME), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Lorenzo Gerratana
- Department of Medicine (DAME), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Fabio Puglisi
- Department of Medicine (DAME), University of Udine, Udine 33100, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano 33081, Italy.
| |
Collapse
|
24
|
Vloemans D, Van Hileghem L, Verbist W, Thomas D, Dal Dosso F, Lammertyn J. Precise sample metering method by coordinated burst action of hydrophobic burst valves applied to dried blood spot collection. LAB ON A CHIP 2021; 21:4445-4454. [PMID: 34651158 DOI: 10.1039/d1lc00422k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Dried blood spot (DBS) sampling by finger-pricking has recently gained a lot of interest as an alternative sample collection method. The reduced invasiveness, requirement of lower sample volumes and suitability for long-term storage at room temperature make DBS ideal for use in home settings or low-resource environments. However, traditional protocols often suffer from biased analysis data due to variable and not exactly known blood volumes present in the samples. In this work, a novel device has been developed to split-off precisely metered volumes from a blood drop and load them on pre-cut filter paper. Hereto, hydrophobic burst valves (HBV) were developed to temporarily retain a fluid flow, configurable to burst at pressures within a range of 175-600 Pa. By combining HBVs with different burst pressures, a volume metering system was developed to allow parallel metering of multiple pre-defined sample volumes. The system was shown to be accurate and consistent for blood volumes between 5-15 μL and for hematocrit levels spanning the range of 25-70%. Finally, a point-of-care DBS sampling device was developed combining the self-powered microfluidic SIMPLE technology. To evaluate the system's practical applicability, a validation study in the context of therapeutic drug monitoring of biologicals was performed using adalimumab-spiked blood samples. Microfluidic DBS samples showed good performance compared to the traditional DBS method with improved recovery rates (86% over 62%). This innovative metering system, allowing for parallelization and integration with complex liquid manipulations, will greatly impact the field of robust sampling, sample preparation, storage and analysis at the point-of-care.
Collapse
Affiliation(s)
- Dries Vloemans
- KU Leuven, Department of Biosystems - Biosensors Group, Willem de Croylaan 42, box 2428, 3001 Leuven, Belgium.
| | - Lorenz Van Hileghem
- KU Leuven, Department of Biosystems - Biosensors Group, Willem de Croylaan 42, box 2428, 3001 Leuven, Belgium.
| | - Wannes Verbist
- KU Leuven, Department of Biosystems - Biosensors Group, Willem de Croylaan 42, box 2428, 3001 Leuven, Belgium.
| | - Debby Thomas
- KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Herestraat 49, box 424, 3000 Leuven, Belgium
| | - Francesco Dal Dosso
- KU Leuven, Department of Biosystems - Biosensors Group, Willem de Croylaan 42, box 2428, 3001 Leuven, Belgium.
| | - Jeroen Lammertyn
- KU Leuven, Department of Biosystems - Biosensors Group, Willem de Croylaan 42, box 2428, 3001 Leuven, Belgium.
| |
Collapse
|
25
|
Liva S, Chen M, Mortazavi A, Walker A, Wang J, Dittmar K, Hofmeister C, Coss CC, Phelps MA. Population Pharmacokinetic Analysis from First-in-Human Data for HDAC Inhibitor, REC-2282 (AR-42), in Patients with Solid Tumors and Hematologic Malignancies: A Case Study for Evaluating Flat vs. Body Size Normalized Dosing. Eur J Drug Metab Pharmacokinet 2021; 46:807-816. [PMID: 34618345 PMCID: PMC8599380 DOI: 10.1007/s13318-021-00722-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2021] [Indexed: 12/26/2022]
Abstract
Background and Objectives REC-2282 is a novel histone deacetylase inhibitor that has shown antitumor activity in in vitro and in vivo models of malignancy. The aims of this study were to characterize the population pharmacokinetics of REC-2282 (AR-42) from the first-in-human (NCT01129193) and phase I acute myeloid leukemia trials (NCT01798901) and to evaluate potential sources of variability. Additionally, we sought to understand alternate body size descriptors as sources of inter-individual variability (IIV), which was significant for dose-normalized maximum observed concentration and area under the concentration-time curve (AUC). Methods Datasets from two clinical trials were combined, and population pharmacokinetic analysis was performed using NONMEM and R softwares; patient demographics were tested as covariates. Results A successful population pharmacokinetic model was constructed. The pharmacokinetics of REC-2282 were best described by a two-compartment model with one transit compartment for absorption, first-order elimination and a proportional error model. Fat-free mass (FFM) was retained as a single covariate on clearance (CL), though it explained < 3% of the observed variability on CL. Tumor type and formulation were retained as covariates on lag time, and a majority of variability, attributed to absorption, remained unexplained. Computed tomography (CT)-derived lean body weight estimates were lower than estimated lean body weight and fat-free mass measures in most patients. Analysis of dose-normalized AUC vs. body size descriptors suggests flat dosing is most appropriate for REC-2282. Conclusions FFM was identified as a significant covariate on CL; however, it explained only a very small portion of the IIV; major factors contributing significantly to REC-2282 pharmacokinetic variability remain unidentified. Supplementary Information The online version contains supplementary material available at 10.1007/s13318-021-00722-z.
Collapse
Affiliation(s)
- Sophia Liva
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Min Chen
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA.,Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Alison Walker
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.,Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Jiang Wang
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Kristin Dittmar
- Department of Radiology, Wexner Medical Center, Columbus, OH, USA
| | - Craig Hofmeister
- Division of Hematology, Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Christopher C Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA. .,Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| | - Mitch A Phelps
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA. .,Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
26
|
High-throughput screening and genome-wide analyses of 44 anticancer drugs in the 1000 Genomes cell lines reveals an association of the NQO1 gene with the response of multiple anticancer drugs. PLoS Genet 2021; 17:e1009732. [PMID: 34437536 PMCID: PMC8439493 DOI: 10.1371/journal.pgen.1009732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 09/14/2021] [Accepted: 07/22/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer patients exhibit a broad range of inter-individual variability in response and toxicity to widely used anticancer drugs, and genetic variation is a major contributor to this variability. To identify new genes that influence the response of 44 FDA-approved anticancer drug treatments widely used to treat various types of cancer, we conducted high-throughput screening and genome-wide association mapping using 680 lymphoblastoid cell lines from the 1000 Genomes Project. The drug treatments considered in this study represent nine drug classes widely used in the treatment of cancer in addition to the paclitaxel + epirubicin combination therapy commonly used for breast cancer patients. Our genome-wide association study (GWAS) found several significant and suggestive associations. We prioritized consistent associations for functional follow-up using gene-expression analyses. The NAD(P)H quinone dehydrogenase 1 (NQO1) gene was found to be associated with the dose-response of arsenic trioxide, erlotinib, trametinib, and a combination treatment of paclitaxel + epirubicin. NQO1 has previously been shown as a biomarker of epirubicin response, but our results reveal novel associations with these additional treatments. Baseline gene expression of NQO1 was positively correlated with response for 43 of the 44 treatments surveyed. By interrogating the functional mechanisms of this association, the results demonstrate differences in both baseline and drug-exposed induction. In the burgeoning field of personalized medicine, genetic variation is recognized as a major contributor to patients’ differential responses to drugs. Lymphoblastoid cell lines (LCLs) are a consistent and convenient representation of cells used for in vitro research. Human genome sequencing with LCLs can identify new genes that influence individuals’ drug responses, including the dose-response relationship, which describes the relationship between physiological response and the amount of exposure to a substance. In this work, we conduct high-throughput screening and genome-wide association mapping using 680 LCLs from the 1000 Genomes Project to identify new genes that influence individual response to 44 widely used anticancer drugs. We found the NQO1 gene to be associated with the dose-response of several drugs, namely arsenic trioxide, erlotinib, trametinib, and the paclitaxel + epirubicin combination, and performed follow-up analyses to better understand its functional role in drug response. Our results indicate NQO1 expression is correlated with increased drug resistance and provide some evidence that SNP rs1800566 influences drug response by altering protein activity for these four treatments. With further research, NQO1 has potential use as a therapeutic target, for example, suppressing NQO1 expression to increase sensitivity to particular drugs.
Collapse
|
27
|
Liao M, Jeziorski KG, Tomaszewska-Kiecana M, Láng I, Jasiówka M, Skarbová V, Centkowski P, Ramlau R, Górnaś M, Lee J, Edwards S, Habeck J, Nash E, Grechko N, Xiao JJ. A phase 1, open-label, drug-drug interaction study of rucaparib with rosuvastatin and oral contraceptives in patients with advanced solid tumors. Cancer Chemother Pharmacol 2021; 88:887-897. [PMID: 34370076 PMCID: PMC8484168 DOI: 10.1007/s00280-021-04338-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/23/2021] [Indexed: 12/14/2022]
Abstract
Purpose This study aimed at evaluating the effect of rucaparib on the pharmacokinetics of rosuvastatin and oral contraceptives in patients with advanced solid tumors and the safety of rucaparib with and without coadministration of rosuvastatin or oral contraceptives. Methods Patients received single doses of oral rosuvastatin 20 mg (Arm A) or oral contraceptives ethinylestradiol 30 µg + levonorgestrel 150 µg (Arm B) on days 1 and 19 and continuous doses of rucaparib 600 mg BID from day 5 to 23. Serial blood samples were collected with and without rucaparib for pharmacokinetic analysis. Results Thirty-six patients (n = 18 each arm) were enrolled and received at least 1 dose of study drug. In the drug–drug interaction analysis (n = 15 each arm), the geometric mean ratio (GMR) of maximum concentration (Cmax) with and without rucaparib was 1.29 for rosuvastatin, 1.09 for ethinylestradiol, and 1.19 for levonorgestrel. GMR of area under the concentration–time curve from time zero to last quantifiable measurement (AUC0–last) was 1.34 for rosuvastatin, 1.43 for ethinylestradiol, and 1.56 for levonorgestrel. There was no increase in frequency of treatment-emergent adverse events (TEAEs) when rucaparib was given with either of the probe drugs. In both arms, most TEAEs were mild in severity and considered unrelated to study treatment. Conclusion Rucaparib 600 mg BID weakly increased the plasma exposure to rosuvastatin or oral contraceptives. Rucaparib safety profile when coadministered with rosuvastatin or oral contraceptives was consistent with that of rucaparib monotherapy. Dose adjustments of rosuvastatin and oral contraceptives are not necessary when coadministered with rucaparib. ClinicalTrials.gov NCT03954366; Date of registration May 17, 2019. Supplementary Information The online version contains supplementary material available at 10.1007/s00280-021-04338-7.
Collapse
Affiliation(s)
- Mingxiang Liao
- Clinical Pharmacology, Clovis Oncology, Inc 5500 Flatrion Pkwy, Boulder, CO, 80301, USA
| | - Krzysztof G Jeziorski
- Department of Gerontology, Public Health and Didactics, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland.,Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland.,BioVirtus Research Site Sp. Z.O.O., BioVirtus Medical Centre, Józefów, Poland
| | | | - István Láng
- Oncology Unit, Istenhegy Private Health Center, Budapest, Hungary
| | - Marek Jasiówka
- Gynecological Oncology Clinic, Centre of Oncology, Maria Skłodowska-Curie Memorial Institute, Krakow, Poland.,Pleiades Medical Centre, Krakow, Poland
| | - Viera Skarbová
- Department of Internal Medicine and Clinical Pharmacology, Summit Clinical Research, Bratislava, Slovakia
| | - Piotr Centkowski
- Department of Oncology and Hematology, Provincial Specialist Hospital, Biala Podlaska, Poland
| | - Rodryg Ramlau
- Department of Oncology, Poznan University of Medical Sciences, Poznań, Poland
| | - Maria Górnaś
- Department of Chemotherapy, ATTIS Centre, Warsaw, Poland
| | - John Lee
- Regulatory Affairs, Clovis Oncology UK, Ltd., Cambridge, UK
| | - Sarah Edwards
- Medical Affairs, Clovis Oncology UK, Ltd., Cambridge, UK
| | - Jenn Habeck
- Biostatistics, Clovis Oncology, Inc., Boulder, CO, USA
| | - Eileen Nash
- Clinical Operations, Clovis Oncology, Inc., Boulder, CO, USA
| | | | - Jim J Xiao
- Clinical Pharmacology, Clovis Oncology, Inc 5500 Flatrion Pkwy, Boulder, CO, 80301, USA.
| |
Collapse
|
28
|
Živojević K, Mladenović M, Djisalov M, Mundzic M, Ruiz-Hernandez E, Gadjanski I, Knežević NŽ. Advanced mesoporous silica nanocarriers in cancer theranostics and gene editing applications. J Control Release 2021; 337:193-211. [PMID: 34293320 DOI: 10.1016/j.jconrel.2021.07.029] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/17/2022]
Abstract
Targeted nanomaterials for cancer theranostics have been the subject of an expanding volume of research studies in recent years. Mesoporous silica nanoparticles (MSNs) are particularly attractive for such applications due to possibilities to synthesize nanoparticles (NPs) of different morphologies, pore diameters and pore arrangements, large surface areas and various options for surface functionalization. Functionalization of MSNs with different organic and inorganic molecules, polymers, surface-attachment of other NPs, loading and entrapping cargo molecules with on-desire release capabilities, lead to seemingly endless prospects for designing advanced nanoconstructs exerting multiple functions, such as simultaneous cancer-targeting, imaging and therapy. Describing composition and multifunctional capabilities of these advanced nanoassemblies for targeted therapy (passive, ligand-functionalized MSNs, stimuli-responsive therapy), including one or more modalities for imaging of tumors, is the subject of this review article, along with an overview of developments within a novel and attractive research trend, comprising the use of MSNs for CRISPR/Cas9 systems delivery and gene editing in cancer. Such advanced nanconstructs exhibit high potential for applications in image-guided therapies and the development of personalized cancer treatment.
Collapse
Affiliation(s)
- Kristina Živojević
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia
| | - Minja Mladenović
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia
| | - Mila Djisalov
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia
| | - Mirjana Mundzic
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia
| | | | - Ivana Gadjanski
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia
| | - Nikola Ž Knežević
- BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, 21000 Novi Sad, Serbia.
| |
Collapse
|
29
|
Ma Y, Zhao X, Chen X, Huang X, Lin Q, Lin Y, Salamone SJ, Zhou X, Wang C, Liang W, Zhao H, Wu K, Yang Y, Zhang L. Therapeutic drug monitoring of docetaxel by pharmacokinetics and pharmacogenetics: A randomized clinical trial of AUC-guided dosing in nonsmall cell lung cancer. Clin Transl Med 2021; 11:e354. [PMID: 33931979 PMCID: PMC8021539 DOI: 10.1002/ctm2.354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/24/2021] [Accepted: 02/23/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Yuxiang Ma
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | | | - Xi Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xinxin Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qingguang Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuehao Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | | | | | | | - Weiting Liang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hongyun Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kui Wu
- BGI-Shenzhen, Shenzhen, China
| | - Yunpeng Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
30
|
Ramos H, Soares MIL, Silva J, Raimundo L, Calheiros J, Gomes C, Reis F, Monteiro FA, Nunes C, Reis S, Bosco B, Piazza S, Domingues L, Chlapek P, Vlcek P, Fabian P, Rajado AT, Carvalho ATP, Veselska R, Inga A, Pinho E Melo TMVD, Saraiva L. A selective p53 activator and anticancer agent to improve colorectal cancer therapy. Cell Rep 2021; 35:108982. [PMID: 33852837 DOI: 10.1016/j.celrep.2021.108982] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 03/08/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Impairment of the p53 pathway is a critical event in cancer. Therefore, reestablishing p53 activity has become one of the most appealing anticancer therapeutic strategies. Here, we disclose the p53-activating anticancer drug (3S)-6,7-bis(hydroxymethyl)-5-methyl-3-phenyl-1H,3H-pyrrolo[1,2-c]thiazole (MANIO). MANIO demonstrates a notable selectivity to the p53 pathway, activating wild-type (WT)p53 and restoring WT-like function to mutant (mut)p53 in human cancer cells. MANIO directly binds to the WT/mutp53 DNA-binding domain, enhancing the protein thermal stability, DNA-binding ability, and transcriptional activity. The high efficacy of MANIO as an anticancer agent toward cancers harboring WT/mutp53 is further demonstrated in patient-derived cells and xenograft mouse models of colorectal cancer (CRC), with no signs of undesirable side effects. MANIO synergizes with conventional chemotherapeutic drugs, and in vitro and in vivo studies predict its adequate drug-likeness and pharmacokinetic properties for a clinical candidate. As a single agent or in combination, MANIO will advance anticancer-targeted therapy, particularly benefiting CRC patients harboring distinct p53 status.
Collapse
Affiliation(s)
- Helena Ramos
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Maria I L Soares
- University of Coimbra, Coimbra Chemistry Centre and Department of Chemistry, 3004-535 Coimbra, Portugal
| | - Joana Silva
- CEB-Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Liliana Raimundo
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Juliana Calheiros
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Célia Gomes
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Flávio Reis
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Filipe A Monteiro
- Departamento de Biomedicina, Unidade de Biologia Experimental, FMUP - Faculdade de Medicina da Universidade do Porto, 4200-319 Porto, Portugal; Pain Research Group, IBMC - Instituto de Biologia Celular e Molecular, 4150-180 Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4150-180 Porto, Portugal
| | - Cláudia Nunes
- LAQV/REQUIMTE, Laboratório de Química Aplicada, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Salette Reis
- LAQV/REQUIMTE, Laboratório de Química Aplicada, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Bartolomeo Bosco
- Department CIBIO, Laboratory of Transcriptional Networks, University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Silvano Piazza
- Department CIBIO, Laboratory of Transcriptional Networks, University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Lucília Domingues
- CEB-Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Petr Chlapek
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Petr Vlcek
- 1st Department of Surgery, St. Anne's University Hospital, Brno, Czech Republic
| | - Pavel Fabian
- Department of Oncological and Experimental Pathology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Ana Teresa Rajado
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - A T P Carvalho
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Renata Veselska
- Laboratory of Tumor Biology, Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Alberto Inga
- Department CIBIO, Laboratory of Transcriptional Networks, University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Teresa M V D Pinho E Melo
- University of Coimbra, Coimbra Chemistry Centre and Department of Chemistry, 3004-535 Coimbra, Portugal.
| | - Lucília Saraiva
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
31
|
Orgel E, Nabais T, Douglas C, Mittelman SD, Neely M. Effect of Body Fat on Population Pharmacokinetics of High-Dose Methotrexate in Pediatric Patients With Acute Lymphoblastic Leukemia. J Clin Pharmacol 2021; 61:755-762. [PMID: 33314168 DOI: 10.1002/jcph.1799] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/07/2020] [Indexed: 11/10/2022]
Abstract
Nearly all international regimens for pediatric acute lymphoblastic leukemia (ALL) incorporate intravenous "high-dose" methotrexate (HDMTX, ≥1 g/m2 ) to penetrate the central nervous system. Dosing is routinely adjusted for body surface area (BSA), but limited data describe the pharmacokinetics of HDMTX, particularly in obese and/or large patients. To understand the impact of body size (BSA) and body fat percentage (BFP) on HDMTX pharmacokinetics, we performed a secondary analysis of 36 children and adolescents 10-21 years old treated for newly diagnosed ALL and who were enrolled in a prospective study examining body composition. All patients received 5 g/m2 of HDMTX infused over 24 hours. Plasma methotrexate concentrations were measured at 24, 42, and 48 hours. At 48 hours, ≥0.4 μmol/L was defined as "delayed elimination," necessitating prolonged supportive care. BFP was measured using dual-energy x-ray absorptiometry. A nonparametric population pharmacokinetic model was constructed with subsequent simulations to explore effects of BSA and BFP extremes. Despite standard BSA-adjusted dosing, we found significant intrapatient variability in mean MTX concentration (38%; range, 1.2%-86%). BSA and BFP were not linearly associated with increased area under the curve (AUC, P = 0.74 and P = 0.12), but both larger size (BSA) and greater obesity (BFP) were associated with an approximately 2-fold higher risk for delayed elimination at 48 hours. HDMTX AUC was not associated with toxicity. MTX pharmacokinetics vary among and even within patients despite BSA-adjusted dosing. Obesity and large size are identified as new risk factors for delayed elimination, requiring further investigation.
Collapse
Affiliation(s)
- Etan Orgel
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, California, USA.,Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Teresa Nabais
- Laboratory of Applied Pharmacokinetics and Bioinformatics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Christopher Douglas
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Steven D Mittelman
- Children's Discovery and Innovation Institute, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Michael Neely
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Laboratory of Applied Pharmacokinetics and Bioinformatics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
| |
Collapse
|
32
|
Marcum BA, Li Z, Turek JJ, Moore GE, Nolte DD, Childress MO. Biodynamic signatures from ex vivo bone marrow aspirates are associated with chemotherapy-induced neutropenia in cancer-bearing dogs. Vet Med Sci 2020; 7:665-673. [PMID: 33369129 PMCID: PMC8136957 DOI: 10.1002/vms3.423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/18/2020] [Accepted: 12/09/2020] [Indexed: 12/27/2022] Open
Abstract
Background Neutropenia is the most common dose‐limiting side effect of cytotoxic chemotherapy in cancer‐bearing dogs. Biodynamic imaging (BDI) is a functional imaging technology that measures dynamic light scattering from living, three‐dimensional tissues to characterize intracellular motion within those tissues. Previous studies have associated BDI biomarkers with tumour sensitivity to chemotherapy agents in dogs with naturally occurring cancer. We hypothesized that BDI, performed ex vivo on bone marrow aspirate samples, would identify dynamic biomarkers associated with the occurrence of specific degrees of neutropenia in tumour‐bearing dogs receiving doxorubicin chemotherapy. Materials and Methods Bone marrow aspirates were collected from 10 dogs with naturally occurring cancers prior to initiation of doxorubicin treatment. BDI was performed on bone marrow samples treated ex vivo with doxorubicin at 0.1, 1, 10 and 100 μM along with 0.1% DMSO as a control. Dogs then were treated with doxorubicin (30 mg/m2, intravenously). Peripheral blood neutrophil counts were obtained on the day of treatment and again 7 days later. Receiver operating characteristic curves identified provisional breakpoints for BDI biomarkers that correlated with specific changes in neutrophil counts between the two time points. Results Provisional breakpoints for several BDI biomarkers were identified, specifying dogs with the largest proportionate change in neutrophils and with neutropenia that was grade 2 or higher following doxorubicin treatment. Conclusions Biodynamic imaging of bone marrow aspirates may identify those dogs at greater risk for neutropenia following doxorubicin chemotherapy. This approach may be useful for pre‐emptively modifying chemotherapy dosing in dogs to avoid unacceptable side effects.
Collapse
Affiliation(s)
- Blake A Marcum
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Zhe Li
- Department of Physics and Astronomy, College of Science, Purdue University, West Lafayette, IN, USA
| | - John J Turek
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - George E Moore
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - David D Nolte
- Department of Physics and Astronomy, College of Science, Purdue University, West Lafayette, IN, USA
| | - Michael O Childress
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
33
|
Salman B, Al-Khabori M. Applications and challenges in therapeutic drug monitoring of cancer treatment: A review. J Oncol Pharm Pract 2020; 27:693-701. [PMID: 33302823 DOI: 10.1177/1078155220979048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Most anticancer agents show wide variability in pharmacokinetics (PK) and have a narrow therapeutic index which makes fixed dosing suboptimal. To achieve the best therapeutic outcomes with these agents, many studies have postulated using PK or therapeutic drug monitoring (TDM)-guided dosing. However, multiple factors contribute to the variability in PKs making the application of TDM in practice challenging. Also, despite the known association with clinical outcomes, standard guidelines on PK-guided dosing are lacking for most agents. Understanding the factors that contribute to PK variability and their impact is essential for dose individualization. The purpose of this review is to discuss the factors that contribute to the PK variability of anticancer agents and the challenges faced in practice when individualizing doses for certain widely used agents. Searching the literature has identified several gaps and efforts are needed to ensure better targeting of cancer therapeutics.
Collapse
Affiliation(s)
- Bushra Salman
- Pharmacy Department, Sultan Qaboos University Hospital, Muscat, Oman
| | - Murtadha Al-Khabori
- Department of Hematology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
34
|
Sebuhyan M, Crichi B, Abdallah NA, Bonnet C, Deville L, Marjanovic Z, Farge D. Drug-drug interaction (DDI) with direct oral anticoagulant (DOAC) in patients with cancer. JOURNAL DE MEDECINE VASCULAIRE 2020; 45:6S31-6S38. [PMID: 33276942 DOI: 10.1016/s2542-4513(20)30517-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cancer-associated thrombosis (CAT) is the second leading cause of death in cancer patients after tumor progression. The treatment of CAT is challenging because of a high risk of VTE recurrence, a high risk of bleeding, common presence of comorbidities, poly-medication, and potential drug-drug interactions (DDI). Since 2018, direct oral anticoagulants (DOACs) represent a promising therapeutic alternative and have been recently included into the 2019 update of the International Initiative on Thrombosis and Cancer (ITAC-CME) clinical practice guidelines for management of CAT. However, pharmacokinetic studies suggest that concomitant treatment with P-gp or CYP3A4 inhibitors will result in an increased exposure to rivaroxaban and apixaban, but the clinical relevance of these studies is unknown. In addition, there is an important inter-individual variability in drug absorption, distribution, metabolism and elimination, even more in cancer patients. Overall, the risk of pharmacokinetic DDI should be estimated based on several individual (patient age, renal and liver function, number of comedications) and diseases-related factors, including inflammation, sarcopenia, and low body weight. In this context, DDI with clinical implications could be expected with anti-neoplastic agents or supportive care treatments, especially with drugs known to be moderate or strong inhibitors/inducers of CYP3A4 and P-gp. Consequently, in the presence of potential DDIs through CYP3A4, and/or P-gp, LMWHs remain the first-line anticoagulant of choice for the long-term treatment of CAT. Multidisciplinary consultation meetings and therapeutic patient education should be emphasized in the complex management of CAT.
Collapse
Affiliation(s)
- M Sebuhyan
- Unité de médecine interne : maladies auto-immunes et pathologie vasculaire (UF04), hôpital Saint-Louis, Assistance publique des Hôpitaux de Paris (APHP), 1 avenue Claude-Vellefaux, 75010 Paris, France.
| | - B Crichi
- Unité de médecine interne : maladies auto-immunes et pathologie vasculaire (UF04), hôpital Saint-Louis, Assistance publique des Hôpitaux de Paris (APHP), 1 avenue Claude-Vellefaux, 75010 Paris, France
| | - N Ait Abdallah
- Unité de médecine interne : maladies auto-immunes et pathologie vasculaire (UF04), hôpital Saint-Louis, Assistance publique des Hôpitaux de Paris (APHP), 1 avenue Claude-Vellefaux, 75010 Paris, France
| | - C Bonnet
- Service d'oncologie médicale, hôpital Saint-Louis, Assistance publique des Hôpitaux de Paris (APHP), 1 avenue Claude-Vellefaux, 75010 Paris, France.
| | - L Deville
- Service de pharmacie, hôpital Saint-Louis, Assistance publique des Hôpitaux de Paris (APHP), 1 avenue Claude-Vellefaux, 75010 Paris, France
| | - Z Marjanovic
- Service d'hématologie clinique et thérapie cellulaire, hôpital Saint-Antoine, Assistance publique des Hôpitaux de Paris (APHP), 184 rue du Faubourg Saint-Antoine, 75012 Paris, France
| | - D Farge
- Unité de médecine interne : maladies auto-immunes et pathologie vasculaire (UF04), hôpital Saint-Louis, Assistance publique des Hôpitaux de Paris (APHP), 1 avenue Claude-Vellefaux, 75010 Paris, France; Université de Paris, IRSL, EA-3518, Recherche clinique appliquée à l'hématologie, F-75010 Paris, France; Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
35
|
Chien JC, Baker SW, Soh HT, Arbabian A. Design and Analysis of a Sample-and-Hold CMOS Electrochemical Sensor for Aptamer-based Therapeutic Drug Monitoring. IEEE JOURNAL OF SOLID-STATE CIRCUITS 2020; 55:2914-2929. [PMID: 33343021 PMCID: PMC7742970 DOI: 10.1109/jssc.2020.3020789] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
In this paper, we present the design and the analysis of an electrochemical circuit for measuring the concentrations of therapeutic drugs using structure-switching aptamers. Aptamers are single-stranded nucleic acids, whose sequence is selected to exhibit high affinity and specificity toward a molecular target, and change its conformation upon binding. This property, when coupled with a redox reporter and electrochemical detection, enables reagent-free biosensing with a sub-minute temporal resolution for in vivo therapeutic drug monitoring. Specifically, we design a chronoamperometry-based electrochemical circuit that measures the direct changes in the electron transfer (ET) kinetics of a methylene blue reporter conjugated at the distal-end of the aptamer. To overcome the high-frequency noise amplification issue when interfacing with a large-size (> 0.25 mm2) implantable electrode, we present a sample-and-hold (S/H) circuit technique in which the desired electrode potentials are held onto noiseless capacitors during the recording of the redox currents. This allows disconnecting the feedback amplifiers to avoid its noise injection while reducing the total power consumption. A prototype circuit implemented in 65-nm CMOS demonstrates a cell-capacitance-insensitive input-referred noise (IRN) current of 15.2 pArms at a 2.5-kHz filtering bandwidth. We tested our system in human whole blood samples and measured the changes in the ET kinetics from the redox-labeled aptamers at different kanamycin concentrations. By employing principal component analysis (PCA) to compensate for the sampling errors, we report a molecular noise floor (at SNR = 1) of 3.1 µM with sub 1-sec acquisition time at 0.22-mW power consumption.
Collapse
Affiliation(s)
- Jun-Chau Chien
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305 USA
| | - Sam W Baker
- Department of Comparative Medicine, Stanford University, Stanford, CA 94305 USA
| | - H Tom Soh
- Department of Radiology and the Department of Electrical Engineering, Stanford University, Stanford, CA 94305 USA
| | - Amin Arbabian
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305 USA
| |
Collapse
|
36
|
Biological evaluation and pharmacokinetic profiling of a coumarin-benzothiazole hybrid as a new scaffold for human gliomas. MEDICINE IN DRUG DISCOVERY 2020. [DOI: 10.1016/j.medidd.2020.100012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
37
|
CDK4/6 Inhibitors in Breast Cancer Treatment: Potential Interactions with Drug, Gene, and Pathophysiological Conditions. Int J Mol Sci 2020; 21:ijms21176350. [PMID: 32883002 PMCID: PMC7504705 DOI: 10.3390/ijms21176350] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/11/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022] Open
Abstract
Palbociclib, ribociclib, and abemaciclib belong to the third generation of cyclin-dependent kinases inhibitors (CDKis), an established therapeutic class for advanced and metastatic breast cancer. Interindividual variability in the therapeutic response of CDKis has been reported and some individuals may experience increased and unexpected toxicity. This narrative review aims at identifying the factors potentially concurring at this variability for driving the most appropriate and tailored use of CDKis in the clinic. Specifically, concomitant medications, pharmacogenetic profile, and pathophysiological conditions could influence absorption, distribution, metabolism, and elimination pharmacokinetics. A personalized therapeutic approach taking into consideration all factors potentially contributing to an altered pharmacokinetic/pharmacodynamic profile could better drive safe and effective clinical use.
Collapse
|
38
|
5-Fluorouracil Response Prediction and Blood Level-Guided Therapy in Oncology: Existing Evidence Fundamentally Supports Instigation. Ther Drug Monit 2020; 42:660-664. [PMID: 32649488 DOI: 10.1097/ftd.0000000000000788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
5-Fluorouracil (5-FU) response prediction and therapeutic drug monitoring (TDM) are required to minimize toxicity while preserving efficacy. Conventional 5-FU dose normalization uses body surface area. It is characterized by up to 100-fold interindividual variability of pharmacokinetic (PK) parameters, and typically >50% of patients have plasma 5-FU concentrations outside the optimal range. This underscores the need for a different dose rationalization paradigm, hence there is a case for 5-FU TDM. An association between 5-FU PK parameters and efficacy/toxicity has been established. It is believed that 5-FU response is enhanced and toxicity is reduced by PK management of its dosing. The area under the concentration-time curve is the most relevant PK parameter associated with 5-FU efficacy/toxicity, and optimal therapeutic windows have been proposed. Currently, there is no universally applied a priori test for predicting 5-FU response and identifying individuals with an elevated risk of toxicity. The following two-step strategy: prediction of response/toxicity and TDM for subsequent doses seems plausible. Approximately 80% of 5-FU is degraded in a three-step sequential metabolic pathway. Dihydropyrimidine dehydrogenase (DPD) is the initial and rate-limiting enzyme. Its deficiency can cause toxicity with standard 5-FU doses. DPD also metabolizes uracil (U) into 5,6-dihydrouracil (UH2). The UH2/U ratio is an index of DPD activity and a credible biomarker of response and toxicity. This article outlines the UH2/U ratio as a parameter for 5-FU response/toxicity prediction and highlights key studies emphasizing the value of 5-FU TDM. Broad application of 5-FU response/toxicity prediction and blood level-guided therapy remains unmet, despite ever-increasing clinical interest. Considered collectively, existing evidence is compelling and fundamentally supports universal instigation of response/toxicity prediction and TDM.
Collapse
|
39
|
Stimuli-responsive nano-assemblies for remotely controlled drug delivery. J Control Release 2020; 322:566-592. [DOI: 10.1016/j.jconrel.2020.03.051] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/19/2020] [Accepted: 03/31/2020] [Indexed: 12/30/2022]
|
40
|
Sakamoto S, Sato K, Takita Y, Izumiya Y, Kumagai N, Sudo K, Hasegawa Y, Yokota H, Akamine Y, Okuda Y, Asano M, Takeda M, Sano M, Miura M, Nakayama K. ABCG2 C421A polymorphisms affect exposure of the epidermal growth factor receptor inhibitor gefitinib. Invest New Drugs 2020; 38:1687-1695. [PMID: 32436059 DOI: 10.1007/s10637-020-00946-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/06/2020] [Indexed: 01/05/2023]
Abstract
ATP-binding castle protein G2 (ABCG2) is thought to inhibit the activities of certain gefitinib transporters, thereby affecting drug pharmacokinetics. The C421A polymorphism affects the function and expression of ABCG2 on the cell membrane. Previous studies have shown that proton-pump inhibitors (PPIs) inhibit gefitinib absorption, as well as the function of ABCG2. We evaluated the plasma concentrations of gefitinib in patients with and without the ABCG2 C421A polymorphism, who were or were not taking PPIs. In total, 61 patients with advanced epidermal-growth-factor-positive non-small-cell lung cancer were enrolled in this study. They were treated with gefitinib at a dose of 250 mg per day. Plasma gefitinib concentration and ABCG2 C421A status were determined after 2 weeks. The patients were divided into CC- and CA/AA genotype groups. We compared the trough and peak gefitinib levels and the area under the curve (AUC) values for 24-h gefitinib concentrations. We also compared these parameters among four groups distinguished according to the presence or absence of the polymorphism and PPI use. The mean trough gefitinib level and AUC value for 24-h gefitinib concentration were significantly lower in the CA/AA group compared to the CC group (mean trough level: 333.2 vs. 454.5 ng/mL, respectively, P = 0.021; AUC: 9949.9 vs. 13,085.4 ng・h/mL, respectively, P = 0.034). Among patients taking PPIs, the mean trough gefitinib level was significantly lower in the CA/AA group than the CC group (220.1 vs. 340.5 ng/mL, respectively, P = 0.033). The CA/AA-type of ABCG2 C421A polymorphism may be associated with lower gefitinib plasma concentrations.
Collapse
Affiliation(s)
- Sho Sakamoto
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Kazuhiro Sato
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Yuri Takita
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Yuka Izumiya
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Naho Kumagai
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Kazuhisa Sudo
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Yukiyasu Hasegawa
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Hayato Yokota
- Department of Pharmacy, Akita University Hospital, Akita, Japan
| | - Yumiko Akamine
- Department of Pharmacy, Akita University Hospital, Akita, Japan
| | - Yuji Okuda
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Mariko Asano
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Masahide Takeda
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Masaaki Sano
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Masatomo Miura
- Department of Pharmacy, Akita University Hospital, Akita, Japan
| | - Katsutoshi Nakayama
- Department of Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan.
| |
Collapse
|
41
|
Biltaji E, Enioutina EY, Yellepeddi V, Rower JE, Sherwin CMT, Ward RM, Lemons RS, Constance JE. Supportive care medications coinciding with chemotherapy among children with hematologic malignancy. Leuk Lymphoma 2020; 61:1920-1931. [PMID: 32264729 PMCID: PMC7725403 DOI: 10.1080/10428194.2020.1749604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pharmacokinetic (PK) conflicts can arise between supportive care medications (SCM) and chemotherapy in children with hematologic malignancy (HM). In this retrospective study, medical records for children (28 days-18 years) diagnosed with HM and receiving an SCM antimicrobial were collected from a hospital network between 1 May 2000 and 31 December 2014. PK drug-gene associations were obtained from a curated pharmacogenomics database. Among 730 patients (median age of 7.5 (IQR 3.7-13.9) years), primarily diagnosed with lymphoid leukemia (52%), lymphoma (28%), or acute myeloid leukemia (16%), chemotherapy was administered in 2846 hospitalizations. SCM accounted for 90.5% (n = 448) of distinct drugs with 93% (n = 679) of children, receiving ≥5 different SCM/hospitalization. Same-day SCM/chemotherapeutic PK gene overlap occurred in 48.3% of hospitalizations and was associated with age (p = 0.026), number of SCM, HM subtype, surgery, and hematopoietic stem cell transplant (p < 0.0001). A high and variable SCM burden among children with HM receiving chemotherapy poses a risk for unanticipated PK conflicts.
Collapse
Affiliation(s)
- Eman Biltaji
- Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Elena Y Enioutina
- Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Venkata Yellepeddi
- Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Joseph E Rower
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Catherine M T Sherwin
- Department of Pediatrics, Wright State University Boonshoft School of Medicine, Dayton Children's Hospital, Dayton, OH, USA
| | - Robert M Ward
- Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Richard S Lemons
- Division of Hematology and Oncology, Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Jonathan E Constance
- Division of Clinical Pharmacology, Department of Pediatrics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
42
|
|
43
|
Gabel F, Aubry AS, Hovhannisyan V, Chavant V, Weinsanto I, Maduna T, Darbon P, Goumon Y. Unveiling the Impact of Morphine on Tamoxifen Metabolism in Mice in vivo. Front Oncol 2020; 10:25. [PMID: 32154159 PMCID: PMC7046683 DOI: 10.3389/fonc.2020.00025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 01/08/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Tamoxifen is used to treat breast cancer and cancer recurrences. After administration, tamoxifen is converted into two more potent antitumor compounds, 4OH-tamoxifen and endoxifen by the CYP3A4/5 and 2D6 enzymes in human. These active compounds are inactivated by the same UDP-glucuronosyltransferase isoforms as those involved in the metabolism of morphine. Importantly, cancer-associated pain can be treated with morphine, and the common metabolic pathway of morphine and tamoxifen suggests potential clinically relevant interactions. Methods: Mouse liver microsomes were used to determine the impact of morphine on 4OH-tamoxifen metabolism in vitro. For in vivo experiments, female mice were first injected with tamoxifen alone and then with tamoxifen and morphine. Blood was collected, and LC-MS/MS was used to quantify tamoxifen, 4OH-tamoxifen, N-desmethyltamoxifen, endoxifen, 4OH-tamoxifen-glucuronide, and endoxifen-glucuronide. Results:In vitro, we found increased Km values for the production of 4OH-tamoxifen-glucuronide in the presence of morphine, suggesting an inhibitory effect on 4OH-tamoxifen glucuronidation. Conversely, in vivo morphine treatment decreased 4OH-tamoxifen levels in the blood while dramatically increasing the formation of inactive metabolites 4OH-tamoxifen-glucuronide and endoxifen-glucuronide. Conclusions: Our findings emphasize the need for caution when extrapolating results from in vitro metabolic assays to in vivo drug metabolism interactions. Importantly, morphine strongly impacts tamoxifen metabolism in mice. It suggests that tamoxifen efficiency could be reduced when both drugs are co-administered in a clinical setting, e.g., to relieve pain in breast cancer patients. Further studies are needed to assess the potential for tamoxifen-morphine metabolic interactions in humans.
Collapse
Affiliation(s)
- Florian Gabel
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, University of Strasbourg, Strasbourg, France
| | - Anne-Sophie Aubry
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, University of Strasbourg, Strasbourg, France
| | - Volodya Hovhannisyan
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, University of Strasbourg, Strasbourg, France
| | - Virginie Chavant
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, University of Strasbourg, Strasbourg, France.,Mass Spectrometry Facilities of the CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Ivan Weinsanto
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, University of Strasbourg, Strasbourg, France
| | - Tando Maduna
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, University of Strasbourg, Strasbourg, France
| | - Pascal Darbon
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, University of Strasbourg, Strasbourg, France
| | - Yannick Goumon
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, University of Strasbourg, Strasbourg, France.,Mass Spectrometry Facilities of the CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| |
Collapse
|
44
|
Nandi S, Kale NR, Takale V, Chate GC, Bhave M, Banerjee SS, Khandare JJ. Cell deformation and acquired drug resistance: elucidating the major influence of drug-nanocarrier delivery systems. J Mater Chem B 2020; 8:1852-1862. [PMID: 32022091 DOI: 10.1039/c9tb02744k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cancer diagnosis and its stage-wise assessment are determined through invasive solid tissue biopsies. Conversely, cancer imaging is enriched through emission tomography and longitudinal high-resolution analysis for the early detection of cancer through altered cell morphology and cell-deformation. Similarly, in post multiple chemo-cycle exposures, the tumor regression and progression thereafter are not well understood. Here, we report chemo-cycles of doxorubicin (Dox) carrying nanoparticles (NPs) to be highly indicative of cell deformation and a progressive indicator of phenotypic expressions of acquired drug resistance (ADR). We designed graphene (G) based nanocarriers by chemically conjugating multiple components: (i) G; (ii) iron oxide (Fe3O4) NPs; and (iii) Dox through a cysteine (Cys) linker (G-Dox and G-Cys-Fe3O4-Dox). Although Dox underwent cell diffusion, the G-based nanocarriers followed a receptor-mediated endocytosis which created a profound impact on the cell membrane integrity. ADR owing to Dox and G-based nanocarriers was analyzed through a cytotoxicity assay, cell morphology deformation parameters and cellular uptake kinetic patterns. Interestingly, after the third chemo-cycle, G-Dox incubated cells showed the greatest decrease in the alteration of the nuclear surface area (NSA) of ∼28%, a ∼40% reduction of the cell surface area (CSA) and a ∼32% increase in the cell roundness (CRd). Our results suggested that the G-based nanocarriers induced the cell deformation process, subsequently resulting in ADR. Although the G-based nanocarriers initiated ADR, G-Dox was most cytotoxic to cancer cells and induced the maximum cell morphology deformation within our scope of study. This outcome implies caution is needed when using G-based nanocarriers and other multi-component nanosystems for Dox delivery as they lead to possible phenotypic expressions of drug resistance in cancer cells.
Collapse
Affiliation(s)
- Semonti Nandi
- MAEER's Maharashtra Institute of Pharmacy, Kothrud, Pune 411038, India
| | - Narendra R Kale
- MAEER's Maharashtra Institute of Pharmacy, Kothrud, Pune 411038, India
| | - Vijay Takale
- MAEER's Maharashtra Institute of Pharmacy, Kothrud, Pune 411038, India
| | - Govind C Chate
- MAEER's Maharashtra Institute of Pharmacy, Kothrud, Pune 411038, India
| | - Madhura Bhave
- MAEER's Maharashtra Institute of Pharmacy, Kothrud, Pune 411038, India
| | - Shashwat S Banerjee
- Maharashtra Institute of Medical Education and Research Medical College, Talegaon Dabhade, Pune 410507, India.
| | | |
Collapse
|
45
|
Ma Y, Lin Q, Yang Y, Liang W, Salamone SJ, Li Y, Lin Y, Zhao H, Zhao Y, Fang W, Huang Y, Zhang L. Clinical pharmacokinetics and drug exposure-toxicity correlation study of docetaxel based chemotherapy in Chinese head and neck cancer patients. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:236. [PMID: 32309383 PMCID: PMC7154422 DOI: 10.21037/atm.2020.01.76] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Area under time-concentration curve (AUC) of docetaxel is related with its toxicity and efficacy. The aim of this study is to investigate the target range of docetaxel AUC in Chinese head and neck cancer (HNC) patients. Methods Eligible HNC patients were enrolled and received at least 2 cycles of docetaxel-based chemotherapy. A simplified pharmacokinetic (PK) strategy (2 monitored samples) was developed to simulate docetaxel AUC using the nonlinear mixed-effect modelling program. Preliminary target range of AUC was pre-set as 2.5–3.7 µg·hr/mL according to pooled analysis from 8 previous studies. Fisher exact test was used to analyze the relationship between AUC with neutropenia and efficacy, and to verify the target range. Results Thirty-nine eligible patients were enrolled. Grade 3-4 and grade 4 neutropenia rate in 1st cycle was 64% and 36%, respectively. AUC simulation by simplified PK strategy was acceptable compared to full sampling method from the analysis of archived 300 patients’ data, with −5.67% of mean prediction error (MPE). Median AUC of all patients was 2.58 µg·hr/mL (range from 1.28 to 9.39). A significant correlation (P=0.007) was detected between AUC and body surface area (BSA)-dosage, but BSA contributed only 18.3% of AUC inter-individual variability. Docetaxel AUC was significantly related with the severity (grade 3–4) of neutropenia (correlation of coefficient was 0.452, P=0.004). Fourteen patients (36%) were within the target AUC range. Patients with AUC above the target experienced more severe neutropenia (grade 3–4 rate 100% vs. 56%, P=0.036; grade 4 rate 86% vs. 25%, P=0.005). No significant difference of response rate was found between patients within the target or not. Conclusions A simplified samples PK strategy was developed for docetaxel AUC simulation. The target range of docetaxel AUC in Chinese HNC patients was suggested at 2.5–3.7 µg·hr/mL for reduced toxicity without compromising efficacy of docetaxel treatment.
Collapse
Affiliation(s)
- Yuxiang Ma
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Qingguang Lin
- Department of ultrasonography, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yunpeng Yang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Weiting Liang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | | | - Yunying Li
- Saladax Biomedical Inc., Bethlehem, PA, USA
| | - Yuehao Lin
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Hongyun Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yuanyuan Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Wenfeng Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yan Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| |
Collapse
|
46
|
Mindt S, Aida S, Merx K, Müller A, Gutting T, Hedtke M, Neumaier M, Hofheinz RD. Therapeutic drug monitoring (TDM) of 5-fluorouracil (5-FU): new preanalytic aspects. Clin Chem Lab Med 2020; 57:1012-1016. [PMID: 30699067 DOI: 10.1515/cclm-2018-1177] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 12/16/2018] [Indexed: 12/31/2022]
Abstract
Background 5-Fluorouracil (5-FU) is frequently used for the treatment of gastrointestinal tumors. The pharmacological effect of 5-FU is influenced by genetic polymorphisms as well as differently dosed regimens. Currently, 5-FU is generally administered as a continuous infusion via an implanted port system using a body surface area (BSA)-based dose calculation. In order to optimize treatment, the area under the curve (AUC) can be estimated to allow for individual dose adjustment. A 5-FU AUC range between 20 and 30 [mg×h×L] is recommended. The aim of the current study was to assess if blood for AUC analysis could also be drawn at the side where the port system had been placed. Methods We collected EDTA blood samples of patients receiving infusional 5-FU simultaneously from different sampling points (right/left cubital vein). 5-FU concentrations were measured in a steady-state equilibrium based on nanoparticle immunoassay (My5-FU; Saladax). Results A total of 39 patients took part in this study. About half of the patients did not reach the target 5-FU concentration window (37% were under- and 16% of the patients were overdosed). Calculated median AUC was 23.3 for the right arm (range 5.8-59.4) and a median of 23.4 for the left arm (range 5.3-61.0). AUC values showed no difference between right compared to left arms (p=0.99). Conclusions In all, these results confirm that a high percentage of patients are not treated with 5-FU doses reaching suggested AUC levels of 20-30. The location of venepuncture, however, had no impact on the results of plasma 5-FU concentration.
Collapse
Affiliation(s)
- Sonani Mindt
- Institute for Clinical Chemistry, Mannheim Medical Faculty of Heidelberg University, University Hospital Mannheim, Mannheim, Germany
| | - Sihem Aida
- Institute for Clinical Chemistry, Mannheim Medical Faculty of Heidelberg University, University Hospital Mannheim, Mannheim, Germany
| | - Kirsten Merx
- Day Treatment Center (TTZ), Interdisciplinary Tumor Center Mannheim (ITM) & III Medical Clinic, Mannheim, Germany
| | - Annette Müller
- Day Treatment Center (TTZ), Interdisciplinary Tumor Center Mannheim (ITM) & III Medical Clinic, Mannheim, Germany
| | - Tobias Gutting
- Day Treatment Center (TTZ), Interdisciplinary Tumor Center Mannheim (ITM) & III Medical Clinic, Mannheim, Germany.,Department of Medicine II, Mannheim Medical Faculty of Heidelberg University, University Hospital Mannheim, Mannheim, Germany
| | - Maren Hedtke
- Institute for Clinical Chemistry, Mannheim Medical Faculty of Heidelberg University, University Hospital Mannheim, Mannheim, Germany
| | - Michael Neumaier
- Institute for Clinical Chemistry, Mannheim Medical Faculty of Heidelberg University, University Hospital Mannheim, Mannheim, Germany
| | - Ralf-Dieter Hofheinz
- Day Treatment Center (TTZ), Interdisciplinary Tumor Center Mannheim (ITM) & III Medical Clinic, Mannheim, Germany
| |
Collapse
|
47
|
|
48
|
Li X, Zhao Z, Yang Y, Liu Z, Wang J, Xu Y, Zhang Y. Novel β-1,3-d-glucan porous microcapsule enveloped folate-functionalized liposomes as a Trojan horse for facilitated oral tumor-targeted co-delivery of chemotherapeutic drugs and quantum dots. J Mater Chem B 2020; 8:2307-2320. [DOI: 10.1039/c9tb02674f] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this study, novel β-1,3-d-glucan porous microcapsule enveloped folate-functionalized liposomes were developed for the potential co-delivery of chemotherapeutic drugs and quantum dots with facilitated drug absorption and antitumor efficacy.
Collapse
Affiliation(s)
- Xiaonan Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
| | - Ziming Zhao
- Department of Pharmaceutics, School of Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
| | - Yihua Yang
- Department of Pharmaceutics, School of Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
| | - Zhaorong Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
| | - Jinglei Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
| | - Yalu Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
| | - Yanzhuo Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy
- Xuzhou Medical University
- Xuzhou
- P. R. China
- Department of Pharmaceutics, School of Pharmacy
| |
Collapse
|
49
|
Reyner E, Lum B, Jing J, Kagedal M, Ware JA, Dickmann LJ. Intrinsic and Extrinsic Pharmacokinetic Variability of Small Molecule Targeted Cancer Therapy. Clin Transl Sci 2019; 13:410-418. [PMID: 31729137 PMCID: PMC7070882 DOI: 10.1111/cts.12726] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 10/30/2019] [Indexed: 12/16/2022] Open
Abstract
Pharmacokinetic (PK) variability in cancer clinical trials may be due to heterogeneous populations and identifying sources of variability is important. Use of healthy subjects in clinical pharmacology studies together with detailed knowledge of the characteristics of patients with cancer can allow for quick identification and quantification of factors affecting PK variability. PK data and sources of variability of 40 marketed molecularly targeted oncology therapeutics were compiled from regulatory approval documents covering an 18‐year period (1999–2017). Variability in PK parameters was compared and contributors to variability were identified. The results show that PK variability was ~ 16% higher for peak plasma concentration (Cmax) and area under the concentration time curve (AUC) in patients with cancer compared with healthy subjects. Several factors were identified as major contributors to variability including hepatic/renal impairment and cytochrome P450 inhibition/induction. Lower PK variability in healthy subjects may represent an opportunity to perform rapid and robust pharmacological and PK assessments to inform subsequent studies in the development of new cancer therapies.
Collapse
Affiliation(s)
- Eric Reyner
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Bert Lum
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Jing Jing
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Matts Kagedal
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Joseph A Ware
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA.,Acerta Pharma LLC, South San Francisco, California, USA
| | - Leslie J Dickmann
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
50
|
Zargar A, Chang S, Kothari A, Snijders AM, Mao JH, Wang J, Hernández AC, Keasling JD, Bivona TG. Overcoming the challenges of cancer drug resistance through bacterial-mediated therapy. Chronic Dis Transl Med 2019; 5:258-266. [PMID: 32055785 PMCID: PMC7004931 DOI: 10.1016/j.cdtm.2019.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Indexed: 12/23/2022] Open
Abstract
Despite tremendous efforts to fight cancer, it remains a major public health problem and a leading cause of death worldwide. With increased knowledge of cancer pathways and improved technological platforms, precision therapeutics that specifically target aberrant cancer pathways have improved patient outcomes. Nevertheless, a primary cause of unsuccessful cancer therapy remains cancer drug resistance. In this review, we summarize the broad classes of resistance to cancer therapy, particularly pharmacokinetics, the tumor microenvironment, and drug resistance mechanisms. Furthermore, we describe how bacterial-mediated cancer therapy, a bygone mode of treatment, has been revitalized by synthetic biology and is uniquely suited to address the primary resistance mechanisms that confound traditional therapies. Through genetic engineering, we discuss how bacteria can be potent anticancer agents given their tumor targeting potential, anti-tumor activity, safety, and coordinated delivery of anti-cancer drugs.
Collapse
Affiliation(s)
- Amin Zargar
- Joint BioEnergy Institute, Lawrence Berkeley National Laboratory, Emeryville, CA 94608, USA
- QB3 Institute, University of California-Berkeley, 174 Stanley Hall, Berkeley, CA 94720, USA
| | - Samantha Chang
- Joint BioEnergy Institute, Lawrence Berkeley National Laboratory, Emeryville, CA 94608, USA
| | - Ankita Kothari
- Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Antoine M. Snijders
- Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Jian-Hua Mao
- Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Jessica Wang
- Joint BioEnergy Institute, Lawrence Berkeley National Laboratory, Emeryville, CA 94608, USA
| | - Amanda C. Hernández
- Joint BioEnergy Institute, Lawrence Berkeley National Laboratory, Emeryville, CA 94608, USA
- Department of Chemical & Biomolecular Engineering, Department of Bioengineering, University of California, Berkeley, CA 94720, USA
| | - Jay D. Keasling
- Joint BioEnergy Institute, Lawrence Berkeley National Laboratory, Emeryville, CA 94608, USA
- QB3 Institute, University of California-Berkeley, 174 Stanley Hall, Berkeley, CA 94720, USA
| | - Trever G. Bivona
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94143, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94143, USA
| |
Collapse
|