1
|
Varisli L, Zoumpourlis P, Spandidos DA, Zoumpourlis V, Vlahopoulos S. ALDH1A1 in breast cancer: A prospective target to overcome therapy resistance (Review). Oncol Lett 2025; 29:213. [PMID: 40093866 PMCID: PMC11905208 DOI: 10.3892/ol.2025.14959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
The expression of cytosolic aldehyde dehydrogenases (ALDHs), which mediate the last step in the pathway of the synthesis of all-trans retinoic acid, is dysregulated in various types of human cancer, and has been associated with the development of cancer stem cells (CSCs) in solid tumors and hematological malignancies. CSCs are considered a minor fraction of cancer cells with the capacity to initiate neoplastic tumors. ALDH1A1 serves a crucial role in the emergence of the CSC phenotype, induces the malignant behavior of cancer cells and promotes treatment resistance. Notably, ALDH1A1-induced therapy resistance is not exclusive to just one group of drugs, but affects diverse types of drugs that use different mechanisms to kill cells. This diversity of drug resistance-inducing effects is associated with the stemness-supporting functions of ALDH1A1. The inhibition of ALDH1A1 activity using chemicals or the depletion of ALDH1A1 via genetic approaches, such as the use of small interfering RNA, can overcome diverse pathways of therapy resistance. In the context of breast cancer, it is critical that only a fraction of malignant cells are expected to manifest stem-like features, which include increased expression of ALDH1A1. From the angle of disease prognosis, the extent of the association of ALDH1A1 with increased malignant behavior and drug resistance remains to be determined through the application of cutting-edge methods that detect the expression of tracked biomarkers within tumors.
Collapse
Affiliation(s)
- Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey
| | - Panagiotis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
2
|
Liang D, Li L, Ai Y, Li Z, Hedrich WD, Sakamuru S, Lynch C, Yu W, Watts-Ouattara I, Heyward S, Xia M, MacKerell AD, Wang H, Xue F. Potent and Selective Human Constitutive Androstane Receptor Activator DL5055 Facilitates Cyclophosphamide-Based Chemotherapies. J Med Chem 2025; 68:7044-7061. [PMID: 40145447 DOI: 10.1021/acs.jmedchem.4c02064] [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: 03/28/2025]
Abstract
Enhancement of the metabolic conversion of cyclophosphamide (CPA) increases its therapeutic effects. Activation of the human constitutive androstane receptor (hCAR) induces CYP2B6, a key enzyme responsible for CPA bioactivation. Based on our previous hCAR activator DL5016, we designed and synthesized a series of new hCAR activators. Compared to DL5016, three new compounds 6i, 6k (DL5055), and 7e, showed significantly improved activating potency for hCAR. Particularly, DL5055 activates hCAR with an EC50 of 0.35 μM and EMAX of 4.3, and does not activate hPXR and other related nuclear receptors. It induced the expression of CYP2B6 and caused the translocation of hCAR from the cytoplasm to the nucleus in human primary hepatocytes. DL5055 also induces the expression of Cyp2b10 (the mouse analog of human CYP2B6) in hCAR-transgenic mice. In addition, it significantly enhances the efficacy of CPA-based chemotherapy regimen, CHOP, in a coculture system and a mouse xenograft model in vivo.
Collapse
Affiliation(s)
- Dongdong Liang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Yong Ai
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Zhihui Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - William D Hedrich
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Srilatha Sakamuru
- 9800 Medical Center Drive, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Caitlin Lynch
- 9800 Medical Center Drive, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Wenbo Yu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Ismael Watts-Ouattara
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Scott Heyward
- BioIVT, 1450 S Rolling Rd, Halethorpe, Maryland 21227, United States
| | - Menghang Xia
- 9800 Medical Center Drive, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Alexander D MacKerell
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, Maryland 21201, United States
| |
Collapse
|
3
|
Li T, Feng Y, Liu Y, Wang H. The role of organic anion transport peptides in cyclophosphamide-induced hepatotoxicity in high-fat diet mice. Life Sci 2024; 359:123239. [PMID: 39566716 DOI: 10.1016/j.lfs.2024.123239] [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: 07/25/2024] [Revised: 10/28/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
Clinically, patients with lipid metabolism disorders caused by factors such as high-fat diet (HFD) developed severer liver damage and lipid metabolism disorders after treatment with cyclophosphamide (CTX). This can lead to elevated levels of inflammatory cytokines, which in turn lead to changes in levels of various liver and kidney transporters, to increase drug accumulation, which may be a way to exacerbate liver injury. The role of organic anion transport peptides (OATPs), an important uptake transporter, in the transport process of CTX and in the aggravation of liver injury induced by CTX in HFD mice is unclear. The aim of this study was to characterize the hepatotoxicity and lipid metabolism disorders of HFD mice exposed to CTX and to investigate the possible mechanism from the perspective of drug in vivo process and transporter regulation. It has been verified that CTX induced severer liver injury in HFD mice compared with the control group, accompanied with upregulated Interleukin-1β (IL-1β) expression and down-regulated OATPs expression in liver and renal, and increased blood CTX concentration. This suggested that the down-regulation of OATPs involved in IL-1β may play an important role in HFD-CTX-induced liver injury, and then experiments in Hep G2 cells was used to validate the hypothesis. Pharmacokinetic and primary hepatocyte uptake experiments confirmed that OATPs may be an important factor involved in the in vivo process of CTX. In summary, this study demonstrated that HFD mice exhibited severer liver toxicity after exposure to CTX, which may be caused by the disorder of lipid levels and the up-regulation of inflammatory factors, and then the downregulation of liver and renal OATPs to increase the accumulation of CTX in vivo. These findings suggest that IL-1β and OATPs may be involved in the interactive regulation of CTX accumulation and endogenous lipid disturbance, and play very important role in the aggravation of liver injury induced by CTX in HFD mice.
Collapse
Affiliation(s)
- Tianyi Li
- School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Yuhao Feng
- School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Yan Liu
- Department of Pharmacy, Weifang People's Hospital, Weifang, Shandong, China
| | - Haina Wang
- School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
4
|
Tian Z, Yao W. Chemotherapeutic drugs for soft tissue sarcomas: a review. Front Pharmacol 2023; 14:1199292. [PMID: 37637411 PMCID: PMC10450752 DOI: 10.3389/fphar.2023.1199292] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023] Open
Abstract
Despite the low incidence of soft tissue sarcomas (STSs), hundreds of thousands of new STS cases are diagnosed annually worldwide, and approximately half of them eventually progress to advanced stages. Currently, chemotherapy is the first-line treatment for advanced STSs. There are difficulties in selecting appropriate drugs for multiline chemotherapy, or for combination treatment of different STS histological subtypes. In this study, we first comprehensively reviewed the efficacy of various chemotherapeutic drugs in the treatment of STSs, and then described the current status of sensitive drugs for different STS subtypes. anthracyclines are the most important systemic treatment for advanced STSs. Ifosfamide, trabectedin, gemcitabine, taxanes, dacarbazine, and eribulin exhibit certain activities in STSs. Vinca alkaloid agents (vindesine, vinblastine, vinorelbine, vincristine) have important therapeutic effects in specific STS subtypes, such as rhabdomyosarcoma and Ewing sarcoma family tumors, whereas their activity in other subtypes is weak. Other chemotherapeutic drugs (methotrexate, cisplatin, etoposide, pemetrexed) have weak efficacy in STSs and are rarely used. It is necessary to select specific second- or above-line chemotherapeutic drugs depending on the histological subtype. This review aims to provide a reference for the selection of chemotherapeutic drugs for multi-line therapy for patients with advanced STSs who have an increasingly long survival.
Collapse
Affiliation(s)
| | - Weitao Yao
- Department of Orthopedics, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
5
|
Ferreira FCS, Clementino M, Rodrigues FAP, Veras HN, Martins DS, Queiroga ML, Lima MA, Silva DO, de Freitas TM, Ribeiro SA, Mota MRL, da Silva JA, Lima AAM, Havt A. [8] and [10]-Gingerol reduces urothelial damage in ifosfamide-induced hemorrhagic cystitis via JAK/STAT/FOXO signaling pathway via IL-10. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1773-1786. [PMID: 36843129 DOI: 10.1007/s00210-023-02436-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/19/2023] [Indexed: 02/28/2023]
Abstract
Acrolein is the main toxic metabolite of ifosfamide (IFO) that causes urothelial damage by oxidative stress and inflammation. Here, we investigate the molecular mechanism of action of gingerols, Zingiber officinale bioactive molecules, as an alternative treatment for ifosfamide-induced hemorrhagic cystitis. Female Swiss mice were randomly divided into 5 groups: control; IFO; IFO + Mesna; and IFO + [8]- or [10]-gingerol. Mesna (80 mg/kg, i.p.) was given 5 min before, 4 and 8 h after IFO (400mg/kg, i.p.). Gingerols (25 mg/kg, p.o.) were given 1 h before and 4 and 8 h after IFO. Animals were euthanized 12 h after IFO injection. Bladders were submitted to macroscopic and histological evaluation. Oxidative stress and inflammation were assessed by malondialdehyde (MDA) or myeloperoxidase assays, respectively. mRNA gene expression was performed to evaluate mesna and gingerols mechanisms of action. Mesna was able to protect bladder tissue by activating NF-κB and NrF2 pathways. However, we demonstrated that gingerols acted as an antioxidant and anti-inflammatory agent stimulating the expression of IL-10, which intracellularly activates JAK/STAT/FOXO signaling pathway.
Collapse
Affiliation(s)
- Francisco C S Ferreira
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Marco Clementino
- Institute of Biomedicine for Brazilian Semiarid, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Herlice N Veras
- Institute of Biomedicine for Brazilian Semiarid, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Dainesy S Martins
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Marcus L Queiroga
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Mikael A Lima
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Dayara O Silva
- Institute of Biomedicine for Brazilian Semiarid, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Thiago M de Freitas
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Samilly A Ribeiro
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Mario R L Mota
- Department of Dental Clinic, Division of Oral Pathology, Faculty of Pharmacy, Dentistry and Nursing, Federal University of Ceará, Fortaleza, CE, Brazil
| | - James A da Silva
- Department of Pharmacy, Federal University of Sergipe, Lagarto, SE, Brazil
| | - Aldo A M Lima
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil
- Institute of Biomedicine for Brazilian Semiarid, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Alexandre Havt
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil.
- Institute of Biomedicine for Brazilian Semiarid, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
6
|
Dancik GM, Varisli L, Vlahopoulos SA. The Molecular Context of Oxidant Stress Response in Cancer Establishes ALDH1A1 as a Critical Target: What This Means for Acute Myeloid Leukemia. Int J Mol Sci 2023; 24:ijms24119372. [PMID: 37298333 DOI: 10.3390/ijms24119372] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
The protein family of aldehyde dehydrogenases (ALDH) encompasses nineteen members. The ALDH1 subfamily consists of enzymes with similar activity, having the capacity to neutralize lipid peroxidation products and to generate retinoic acid; however, only ALDH1A1 emerges as a significant risk factor in acute myeloid leukemia. Not only is the gene ALDH1A1 on average significantly overexpressed in the poor prognosis group at the RNA level, but its protein product, ALDH1A1 protects acute myeloid leukemia cells from lipid peroxidation byproducts. This capacity to protect cells can be ascribed to the stability of the enzyme under conditions of oxidant stress. The capacity to protect cells is evident both in vitro, as well as in mouse xenografts of those cells, shielding cells effectively from a number of potent antineoplastic agents. However, the role of ALDH1A1 in acute myeloid leukemia has been unclear in the past due to evidence that normal cells often have higher aldehyde dehydrogenase activity than leukemic cells. This being true, ALDH1A1 RNA expression is significantly associated with poor prognosis. It is hence imperative that ALDH1A1 is methodically targeted, particularly for the acute myeloid leukemia patients of the poor prognosis risk group that overexpress ALDH1A1 RNA.
Collapse
Affiliation(s)
- Garrett M Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA
| | - Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey
| | - Spiros A Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527 Athens, Greece
| |
Collapse
|
7
|
Piperi C, Saurty-Seerunghen MS, Levidou G, Sepsa A, Trigka EA, Klonou A, Markouli M, Strepkos D, Spyropoulou A, Kanakoglou DS, Lakiotaki E, Karatrasoglou EA, Boviatsis E, El-Habr EA, Korkolopoulou P. Glioma Cells Expressing High Levels of ALDH5A1 Exhibit Enhanced Migration Transcriptional Signature in Patient Tumors. Neurotherapeutics 2023; 20:881-895. [PMID: 36976494 PMCID: PMC10275844 DOI: 10.1007/s13311-023-01354-8] [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] [Accepted: 02/10/2023] [Indexed: 03/29/2023] Open
Abstract
Accumulating data shows that altered metabolic activity contributes to glioma development. Recently, modulation of SSADH (succinic semialdehyde dehydrogenase) expression, implicated in the catabolism of GABA neurotransmitter, was shown to impact glioma cell properties, such as proliferation, self-renewal and tumorigenicity. The purpose of this study was to investigate the clinical significance of SSADH expression in human gliomas. Using public single-cell RNA-sequencing data from glioma surgical resections, we initially grouped cancer cells according to ALDH5A1 (Aldehyde dehydrogenase 5 family member A1) expression, which encodes SSADH. Gene ontology enrichment analysis of genes differentially expressed between cancer cells expressing high or low levels of ALDH5A1, highlighted enrichment in genes implicated in cell morphogenesis and motility. In glioblastoma cell lines, ALDH5A1 knockdown inhibited cell proliferation, induced apoptosis and reduced their migratory potential. This was accompanied by a reduction in the mRNA levels of the adherens junction molecule ADAM-15 and deregulation in the expression of EMT biomarkers, with increased CDH1 and decreased vimentin mRNA levels. Evaluation of SSADH expression in a cohort of 95 gliomas using immunohistochemistry showed that SSADH expression was significantly elevated in cancer tissues compared to normal brain tissues, without any significant correlation with clinicopathological characteristics. In summary, our data show that SSADH is upregulated in glioma tissues irrespective of the histological grade and its expression sustains glioma cell motility.
Collapse
Affiliation(s)
- Christina Piperi
- Department of Biological Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Mirca S Saurty-Seerunghen
- CNRS UMR8246, Inserm U1130, Sorbonne Université, Neuroscience Paris Seine-IBPS Laboratory, Paris, France
| | - Georgia Levidou
- Department of Pathology, Medical School, Klinikum Nuremberg, Paracelsus University, Nuremberg, Germany
| | - Athanasia Sepsa
- Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, Athens, Greece
| | - Eleni-Andriana Trigka
- Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, Athens, Greece
| | - Alexia Klonou
- Department of Biological Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Mariam Markouli
- Department of Biological Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Strepkos
- Department of Biological Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia Spyropoulou
- Department of Biological Chemistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios S Kanakoglou
- Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, Athens, Greece.
| | - Eleftheria Lakiotaki
- Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, Athens, Greece
| | - Eleni A Karatrasoglou
- Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, Athens, Greece
| | - Efstathios Boviatsis
- Department of Neurosurgery, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Elias A El-Habr
- CNRS UMR8246, Inserm U1130, Sorbonne Université, Neuroscience Paris Seine-IBPS Laboratory, Paris, France
| | - Penelope Korkolopoulou
- Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 M. Asias Street, Athens, Greece.
| |
Collapse
|
8
|
Identifying polymorphic cis-regulatory variants as risk markers for lung carcinogenesis and chemotherapy responses in tobacco smokers from eastern India. Sci Rep 2023; 13:4019. [PMID: 36899086 PMCID: PMC10006236 DOI: 10.1038/s41598-023-30962-9] [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: 07/09/2022] [Accepted: 03/03/2023] [Indexed: 03/12/2023] Open
Abstract
Aberrant expression of xenobiotic metabolism and DNA repair genes is critical to lung cancer pathogenesis. This study aims to identify the cis-regulatory variants of the genes modulating lung cancer risk among tobacco smokers and altering their chemotherapy responses. From a list of 2984 SNVs, prioritization and functional annotation revealed 22 cis-eQTLs of 14 genes within the gene expression-correlated DNase I hypersensitive sites using lung tissue-specific ENCODE, GTEx, Roadmap Epigenomics, and TCGA datasets. The 22 cis-regulatory variants predictably alter the binding of 44 transcription factors (TFs) expressed in lung tissue. Interestingly, 6 reported lung cancer-associated variants were found in linkage disequilibrium (LD) with 5 prioritized cis-eQTLs from our study. A case-control study with 3 promoter cis-eQTLs (p < 0.01) on 101 lung cancer patients and 401 healthy controls from eastern India with confirmed smoking history revealed an association of rs3764821 (ALDH3B1) (OR = 2.53, 95% CI = 1.57-4.07, p = 0.00014) and rs3748523 (RAD52) (OR = 1.69, 95% CI = 1.17-2.47, p = 0.006) with lung cancer risk. The effect of different chemotherapy regimens on the overall survival of lung cancer patients to the associated variants showed that the risk alleles of both variants significantly decreased (p < 0.05) patient survival.
Collapse
|
9
|
Gökçe F, Kaestli A, Lohasz C, de Geus M, Kaltenbach H, Renggli K, Bornhauser B, Hierlemann A, Modena M. Microphysiological Drug-Testing Platform for Identifying Responses to Prodrug Treatment in Primary Leukemia. Adv Healthc Mater 2023; 12:e2202506. [PMID: 36651229 PMCID: PMC11469234 DOI: 10.1002/adhm.202202506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/20/2022] [Indexed: 01/19/2023]
Abstract
Despite increasing survival rates of pediatric leukemia patients over the past decades, the outcome of some leukemia subtypes has remained dismal. Drug sensitivity and resistance testing on patient-derived leukemia samples provide important information to tailor treatments for high-risk patients. However, currently used well-based drug screening platforms have limitations in predicting the effects of prodrugs, a class of therapeutics that require metabolic activation to become effective. To address this issue, a microphysiological drug-testing platform is developed that enables co-culturing of patient-derived leukemia cells, human bone marrow mesenchymal stromal cells, and human liver microtissues within the same microfluidic platform. This platform also enables to control the physical interaction between the diverse cell types. Herein, it is made possible to recapitulate hepatic prodrug activation of ifosfamide in their platform, which is very difficult in traditional well-based assays. By testing the susceptibility of primary patient-derived leukemia samples to the prodrug ifosfamide, sample-specific sensitivities to ifosfamide in primary leukemia samples are identified. The microfluidic platform is found to enable the recapitulation of physiologically relevant conditions and the testing of prodrugs including short-lived and unstable metabolites. The platform holds great potential for clinical translation and precision chemotherapy selection.
Collapse
Affiliation(s)
- Furkan Gökçe
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| | - Alicia Kaestli
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| | - Christian Lohasz
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| | - Martina de Geus
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| | | | - Kasper Renggli
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| | - Beat Bornhauser
- Children's Research CenterUniversity Children's Hospital ZurichZurichZH, 8008Switzerland
| | - Andreas Hierlemann
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| | - Mario Modena
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| |
Collapse
|
10
|
CITCO as an Adjuvant Facilitates CHOP-Based Lymphoma Treatment in hCAR-Transgenic Mice. Cells 2020; 9:cells9112520. [PMID: 33233444 PMCID: PMC7700167 DOI: 10.3390/cells9112520] [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: 10/16/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 11/17/2022] Open
Abstract
Non-Hodgkin's lymphoma (NHL) is a malignant cancer originating in the lymphatic system with a 25-30% mortality rate. CHOP, consisting of cyclophosphamide (CPA), doxorubicin, vincristine, and prednisone, is a first-generation chemotherapy extensively used to treat NHL. However, poor survival rates among patients in advanced stages of NHL shows a need to improve this standard of care treatment. CPA, an integral component of CHOP, is a prodrug that requires CYP2B6-mediated bioactivation to 4-hydroxy-CPA (4-OH-CPA). The expression of CYP2B6 is transcriptionally regulated by the constitutive androstane receptor (CAR, NRi13). We have previously demonstrated that the induction of hepatic CYP2B6 by CITCO, a selective human CAR (hCAR) agonist, results in CHOP's enhanced antineoplastic effects in vitro. Here, we investigate the in vivo potential of CITCO as an adjuvant of CPA-based NHL treatment in a hCAR-transgenic mouse line. Our results demonstrate that the addition of CITCO to the CHOP regimen leads to significant suppression of the growth of EL-4 xenografts in hCAR-transgenic mice accompanied by reduced expression of cyclin-D1, ki67, Pcna, and increased caspase 3 fragmentation in tumor tissues. CITCO robustly induced the expression of cyp2b10 (murine ortholog of CYP2B6) through hCAR activation and increased plasma concentrations of 4-OH-CPA. Comparing to intraperitoneal injection, oral gavage of CITCO results in optimal hepatic cyp2b10 induction. Our in vivo studies have collectively uncovered CITCO as an effective facilitator for CPA-based NHL treatment with a pharmacokinetic profile favoring oral administration, promoting CITCO as a promising adjuvant candidate for CPA-based regimens.
Collapse
|
11
|
Lohasz C, Bonanini F, Hoelting L, Renggli K, Frey O, Hierlemann A. Predicting Metabolism-Related Drug-Drug Interactions Using a Microphysiological Multitissue System. ACTA ACUST UNITED AC 2020; 4:e2000079. [PMID: 33073544 DOI: 10.1002/adbi.202000079] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 09/30/2020] [Indexed: 12/20/2022]
Abstract
Drug-drug interactions (DDIs) occur when the pharmacological activity of one drug is altered by a second drug. As multimorbidity and polypharmacotherapy are becoming more common due to the increasing age of the population, the risk of DDIs is massively increasing. Therefore, in vitro testing methods are needed to capture such multiorgan events. Here, a scalable, gravity-driven microfluidic system featuring 3D microtissues (MTs) that represent different organs for the prediction of drug-drug interactions is used. Human liver microtissues (hLiMTs) are combined with tumor microtissues (TuMTs) and treated with drug combinations that are known to cause DDIs in vivo. The testing system is able to capture and quantify DDIs upon co-administration of the anticancer prodrugs cyclophosphamide or ifosfamide with the antiretroviral drug ritonavir. Dosage of ritonavir inhibits hepatic metabolization of the two prodrugs to different extents and decreases their efficacy in acting on TuMTs. The flexible MT compartment design of the system, the use of polystyrene as chip material, and the assembly of several chips in stackable plates offer the potential to significantly advance preclinical substance testing. The possibility of testing a broad variety of drug combinations to identify possible DDIs will improve the drug development process and increase patient safety.
Collapse
Affiliation(s)
- Christian Lohasz
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland
| | - Flavio Bonanini
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland
| | | | - Kasper Renggli
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland
| | | | - Andreas Hierlemann
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland
| |
Collapse
|
12
|
Özdemir S, Kucukler S, Çomaklı S, Kandemir FM. The protective effect of Morin against ifosfamide-induced acute liver injury in rats associated with the inhibition of DNA damage and apoptosis. Drug Chem Toxicol 2020; 45:1308-1317. [PMID: 32957801 DOI: 10.1080/01480545.2020.1822390] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Morin is a flavonoid and broadly found in white berry and cranberry branch. Ifosfamide (IFOS) is known as an anticancer and cytotoxic drug especially on the liver. This study aimed to explore the potential protective effects of Morin against IFOS-induced liver toxicity in rats. The model group of rats received a single injection of IFOS (500 mg/kg; i.p.) at day 2, whereas the protective groups of rats were given two different doses of Morin (100 and 200 mg/kg; given by gavage) at days 1 and 2. All animals were then culled 24 h post-IFOS injection. We observed that IFOS caused liver injury, oxidative stress, inflammation, DNA damage, and apoptosis. However, Morin decreased the levels of aspartate aminotransferase (AST), alkaline phosphatase (ALP), alanine aminotransferase (ALT) (p < 0.05). While Morin contributed to the recovery of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione (GSH) levels, Morin decreased the levels of malondialdehyde (MDA) induced by IFOS in the liver (p < 0.05). Besides, the levels of nuclear factor kappa B (NF-κB), tumor necrosis factor-α (TNF-α), and P53 measured by ELISA test were reduced via Morin administration (p < 0.05). Lastly, the mRNA transcript levels of Bax, Apaf-1, Bcl-2, Bcl-xL, and inducible nitric oxide synthase (iNOS) determined by RT-PCR were down-regulated in the Morin groups (p < 0.05). These results indicate that Morin plays a protective role by reducing oxidative stress, inflammation, and apoptosis in the IFOS-induced liver injury in rats.
Collapse
Affiliation(s)
- Selçuk Özdemir
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Sefa Kucukler
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Selim Çomaklı
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Fatih Mehmet Kandemir
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| |
Collapse
|
13
|
Quantification of 3-Hydroxypropyl Mercapturic Acid in the Urine of Patients with Breast Cancer to Monitor Cyclophosphamide Toxicity. Ther Drug Monit 2020; 42:548-553. [DOI: 10.1097/ftd.0000000000000737] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Steinbrecht S, Kiebist J, König R, Thiessen M, Schmidtke KU, Kammerer S, Küpper JH, Scheibner K. Synthesis of cyclophosphamide metabolites by a peroxygenase from Marasmius rotula for toxicological studies on human cancer cells. AMB Express 2020; 10:128. [PMID: 32683510 PMCID: PMC7368878 DOI: 10.1186/s13568-020-01064-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 07/13/2020] [Indexed: 02/08/2023] Open
Abstract
Cyclophosphamide (CPA) represents a widely used anti-cancer prodrug that is converted by liver cytochrome P450 (CYP) enzymes into the primary metabolite 4-hydroxycyclophosphamide (4-OH-CPA), followed by non-enzymatic generation of the bioactive metabolites phosphoramide mustard and acrolein. The use of human drug metabolites as authentic standards to evaluate their toxicity is essential for drug development. However, the chemical synthesis of 4-OH-CPA is complex and leads to only low yields and undesired side products. In past years, fungal unspecific peroxygenases (UPOs) have raised to powerful biocatalysts. They can exert the identical selective oxyfunctionalization of organic compounds and drugs as known for CYP enzymes with hydrogen peroxide being used as sole cosubstrate. Herein, we report the efficient enzymatic hydroxylation of CPA using the unspecific peroxygenase from Marasmius rotula (MroUPO) in a simple reaction design. Depending on the conditions used the primary liver metabolite 4-OH-CPA, its tautomer aldophosphamide (APA) and the overoxidized product 4-ketocyclophosphamide (4-keto-CPA) could be obtained. Using a kinetically controlled approach 4-OH-CPA was isolated with a yield of 32% (purity > 97.6%). Two human cancer cell lines (HepG2 and MCF-7) were treated with purified 4-OH-CPA produced by MroUPO (4-OH-CPAUPO). 4-OH-CPAUPO–induced cytotoxicity as measured by a luminescent cell viability assay and its genotoxicity as measured by γH2AX foci formation was not significantly different to the commercially available standard. The high yield of 4-OH-CPAUPO and its biological activity demonstrate that UPOs can be efficiently used to produce CYP-specific drug metabolites for pharmacological assessment.
Collapse
|
15
|
A molecular electron density theory study of the [3 + 2] cycloaddition reaction of 1,4-diphosphorinium-3-olates with methyl acrylate and methyl methacrylate. Theor Chem Acc 2020. [DOI: 10.1007/s00214-020-02637-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
16
|
El-Haj BM, Ahmed SB. Metabolic-Hydroxy and Carboxy Functionalization of Alkyl Moieties in Drug Molecules: Prediction of Structure Influence and Pharmacologic Activity. Molecules 2020; 25:E1937. [PMID: 32331223 PMCID: PMC7222001 DOI: 10.3390/molecules25081937] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/01/2020] [Accepted: 04/07/2020] [Indexed: 12/11/2022] Open
Abstract
Alkyl moieties-open chain or cyclic, linear, or branched-are common in drug molecules. The hydrophobicity of alkyl moieties in drug molecules is modified by metabolic hydroxy functionalization via free-radical intermediates to give primary, secondary, or tertiary alcohols depending on the class of the substrate carbon. The hydroxymethyl groups resulting from the functionalization of methyl groups are mostly oxidized further to carboxyl groups to give carboxy metabolites. As observed from the surveyed cases in this review, hydroxy functionalization leads to loss, attenuation, or retention of pharmacologic activity with respect to the parent drug. On the other hand, carboxy functionalization leads to a loss of activity with the exception of only a few cases in which activity is retained. The exceptions are those groups in which the carboxy functionalization occurs at a position distant from a well-defined primary pharmacophore. Some hydroxy metabolites, which are equiactive with their parent drugs, have been developed into ester prodrugs while carboxy metabolites, which are equiactive to their parent drugs, have been developed into drugs as per se. In this review, we present and discuss the above state of affairs for a variety of drug classes, using selected drug members to show the effect on pharmacologic activity as well as dependence of the metabolic change on drug molecular structure. The review provides a basis for informed predictions of (i) structural features required for metabolic hydroxy and carboxy functionalization of alkyl moieties in existing or planned small drug molecules, and (ii) pharmacologic activity of the metabolites resulting from hydroxy and/or carboxy functionalization of alkyl moieties.
Collapse
Affiliation(s)
- Babiker M. El-Haj
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, University of Science and Technology of Fujairah, Fufairah 00971, UAE
| | - Samrein B.M. Ahmed
- College of Medicine, Sharjah Institute for Medical Research, University of Sharjah, Sharjah 00971, UAE;
| |
Collapse
|
17
|
Pamuła-Piłat J, Tęcza K, Kalinowska-Herok M, Grzybowska E. Genetic 3'UTR variations and clinical factors significantly contribute to survival prediction and clinical response in breast cancer patients. Sci Rep 2020; 10:5736. [PMID: 32235849 PMCID: PMC7109149 DOI: 10.1038/s41598-020-62662-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 03/13/2020] [Indexed: 11/09/2022] Open
Abstract
The study describes a relationship between the 3′UTR variants, clinicopathological parameters and response to chemotherapy. We analyzed 33 germline polymorphisms in 3′UTRs of ADME genes in 305 breast cancer women treated with FAC regime. Clinical endpoints of this study were: overall survival (OS), progression-free survival (PFS), recurrence-free survival (RFS) and overall response defined as treatment failure-free survival (TFFS). The shortened OS was connected with the presence of NR1/2 rs3732359 AA, SLC22A16 rs7756222 CC, as well as SLC22A16 rs9487402 allele G and clinical factors belonging to TNM classification: tumor size >1 cm, nodal involvement and presence of metastases. PFS was related to two polymorphisms PGR rs1824125 GG, PGR rs12224560 CC and SLC22A16 rs7756222 CC as well as preexisting metastases. The RFS was shortened due to the DPYD rs291593 CC, AKR1C3 rs3209896 AG and negative expression of PGR. The presence of ALDH5A1 rs1054899 allele A, lack of pre-chemotherapy surgery and negative status of PGR correlated with worse treatment response. The germline variants commonly present in the population are important factors determining the response to treatment. We observed the effect of the accumulation of genetic and clinical factors on poor survival prognosis and overall treatment response.
Collapse
Affiliation(s)
- Jolanta Pamuła-Piłat
- Department of Genetic and Molecular Diagnostics of Cancer, Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland.,Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Karolina Tęcza
- Department of Genetic and Molecular Diagnostics of Cancer, Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland.,Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Magdalena Kalinowska-Herok
- Department of Genetic and Molecular Diagnostics of Cancer, Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland.,Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Ewa Grzybowska
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland.
| |
Collapse
|
18
|
Dobrek Ł, Arent Z, Nalik-Iwaniak K, Fic K, Kopańska M. Osteopontin and Fatty Acid Binding Protein in Ifosfamide-treated Rats. Open Med (Wars) 2019; 14:561-571. [PMID: 31410367 PMCID: PMC6689207 DOI: 10.1515/med-2019-0063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/07/2019] [Indexed: 01/04/2023] Open
Abstract
Introduction Ifosfamide (IF) is a cytostatic that exhibits adverse nephrotoxic properties. Clinically, IF-induced nephrotoxicity takes various forms, depending on applied dose and length of treatment. Objectives The aim of the study was to evaluate the two proteins: osteopontin (OP) and fatty acid binding protein (FABP), as markers of kidney function in rats treated with ifosfamide. Material and Methods Rats receiving a single IF dose (250 mg/kg b.w.; group 1) or treated with five consecutive IF doses administrated on following days (50mg/kg b.w.; group 3), compared with control groups 2 and 4, respectively, were studied. Kidney function was assessed using classical (urea, creatinine) and novel (FABP, OP) laboratory parameters and by histopathology. Results Single IF dose administration resulted in significant total proteinuria with urinary concentrations and 24-hour excretions of both FABP and OP comparable to the appropriate control. In rats treated with five consecutive IF doses, the urinary concentrations and 24-hour excretion of both FABP and OP were significantly higher compared to the appropriate control. The development of cystitis was revealed in groups 1 and 3, which was not accompanied by significant histopathological kidney damage. Conclusions Both OP and FABP may be useful laboratory markers of tubulopathy in the early stage of chronic nephrotoxicity of ifosfamide.
Collapse
Affiliation(s)
- Łukasz Dobrek
- Independent Researcher cooperating with Experimental and Innovative Medicine Centre, University Centre of Veterinary Medicine UJ-UR, University of Agriculture in Krakow, Krakow Poland
| | - Zbigniew Arent
- Independent Researcher cooperating with Experimental and Innovative Medicine Centre, University Centre of Veterinary Medicine UJ-UR, University of Agriculture in Krakow, Krakow Poland
| | - Klaudia Nalik-Iwaniak
- Independent Researcher cooperating with Experimental and Innovative Medicine Centre, University Centre of Veterinary Medicine UJ-UR, University of Agriculture in Krakow, Krakow Poland
| | - Kinga Fic
- Independent Researcher cooperating with Experimental and Innovative Medicine Centre, University Centre of Veterinary Medicine UJ-UR, University of Agriculture in Krakow, Krakow Poland
| | - Marta Kopańska
- Department of Human Physiology, Faculty of Medicine, University of Rzeszow, Rzeszow Poland
| |
Collapse
|
19
|
Liang D, Li L, Lynch C, Mackowiak B, Hedrich WD, Ai Y, Yin Y, Heyward S, Xia M, Wang H, Xue F. Human constitutive androstane receptor agonist DL5016: A novel sensitizer for cyclophosphamide-based chemotherapies. Eur J Med Chem 2019; 179:84-99. [PMID: 31247375 DOI: 10.1016/j.ejmech.2019.06.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/29/2022]
Abstract
The DNA alkylating prodrug cyclophosphamide (CPA), alone or in combination with other agents, is one of the most commonly used anti-cancer agents. As a prodrug, CPA is activated by cytochrome P450 2B6 (CYP2B6), which is transcriptionally regulated by the human constitutive androstane receptor (hCAR). Therefore, hCAR agonists represent novel sensitizers for CPA-based therapies. Among known hCAR agonists, compound 6-(4-chlorophenyl)imidazo-[2,1-b]thiazole-5-carbaldehyde-O-(3,4-dichlorobenzyl)oxime (CITCO) is the most potent and broadly utilized in biological studies. Through structural modification of CITCO, we have developed a novel compound DL5016 (32), which has an EC50 value of 0.66 μM and EMAX value of 4.9 when activating hCAR. DL5016 robustly induced the expression of hCAR target gene CYP2B6, at both the mRNA and protein levels, and caused translocation of hCAR from the cytoplasm to the nucleus in human primary hepatocytes. The effects of DL5016 were highlighted by dramatically enhancing the efficacy of CPA-based cytotoxicity to non-Hodgkin lymphoma cells.
Collapse
Affiliation(s)
- Dongdong Liang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States
| | - Caitlin Lynch
- 9800 Medical Center Drive, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Bryan Mackowiak
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States
| | - William D Hedrich
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States
| | - Yong Ai
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States
| | - Yue Yin
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States
| | - Scott Heyward
- BioIVT, 1450 S Rolling Rd, Halethorpe, MD, 21227, United States
| | - Menghang Xia
- 9800 Medical Center Drive, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, United States
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States.
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, United States.
| |
Collapse
|
20
|
Reis-Mendes A, Carvalho F, Remião F, Sousa E, Bastos MDL, Costa VM. The Main Metabolites of Fluorouracil + Adriamycin + Cyclophosphamide (FAC) Are Not Major Contributors to FAC Toxicity in H9c2 Cardiac Differentiated Cells. Biomolecules 2019; 9:98. [PMID: 30862114 PMCID: PMC6468772 DOI: 10.3390/biom9030098] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/21/2019] [Accepted: 03/01/2019] [Indexed: 02/07/2023] Open
Abstract
In the clinical practice, the combination of 5-fluorouracil (5-FU) + Adriamycin (also known as doxorubicin, DOX) + cyclophosphamide (CYA) (known as FAC) is used to treat breast cancer. The FAC therapy, however, carries some serious risks, namely potential cardiotoxic effects, although the mechanisms are still unclear. In the present study, the role of the main metabolites regarding FAC-induced cardiotoxicity was assessed at clinical relevant concentrations. Seven-day differentiated H9c2 cells were exposed for 48 h to the main metabolites of FAC, namely the metabolite of 5-FU, α-fluoro-β-alanine (FBAL, 50 or 100 μM), of DOX, doxorubicinol (DOXOL, 0.2 or 1 μM), and of CYA, acrolein (ACRO, 1 or 10 μM), as well as to their combination. The parent drugs (5-FU 50 μM, DOX 1 μM, and CYA 50 μM) were also tested isolated or in combination with the metabolites. Putative cytotoxicity was evaluated through phase contrast microscopy, Hoechst staining, membrane mitochondrial potential, and by two cytotoxicity assays: the reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) and the neutral red (NR) lysosomal incorporation. The metabolite DOXOL was more toxic than FBAL and ACRO in the MTT and NR assays. When in combination, neither FBAL nor ACRO increased DOXOL-induced cytotoxicity. No nuclear condensation was observed for any of the tested combinations; however, a significant mitochondrial potential depolarization after FBAL 100 μM + DOXOL 1 μM + ACRO 10 μM or FBAL 100 μM + DOXOL 1 μM exposure was seen at 48 h. When tested alone DOX 1 μM was more cytotoxic than all the parent drugs and metabolites in both the cytotoxicity assays performed. These results demonstrated that DOXOL was the most toxic of all the metabolites tested; nonetheless, the metabolites do not seem to be the major contributors to FAC-induced cardiotoxicity in this cardiac model.
Collapse
Affiliation(s)
- Ana Reis-Mendes
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Fernando Remião
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemistry, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
- CIIMAR⁻Interdisciplinary Centre of Marine and Environmental Research, 4450-208 Porto, Portugal.
| | - Maria de Lourdes Bastos
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Vera Marisa Costa
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
21
|
Hallooman D, Ríos-Gutiérrez M, Rhyman L, Alswaidan IA, Domingo LR, Ramasami P. DFT exploration of [3 + 2] cycloaddition reaction of 1 H-phosphorinium-3-olate and 1-methylphosphorinium-3-olate with methyl methacrylate. RSC Adv 2018; 8:27406-27416. [PMID: 35539990 PMCID: PMC9083896 DOI: 10.1039/c8ra04703k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 07/17/2018] [Indexed: 11/22/2022] Open
Abstract
A Molecular Electron Density Theory (MEDT) study of the regio- and stereoselectivity of the [3 + 2] cycloaddition (32CA) reaction of 1H-phosphorinium-3-olate and 1-methylphosphorinium-3-olate with methyl methacrylate was carried out using the B3LYP/6-31G(d) method. In order to test the method dependence for the most favorable reaction path leading to the 1H-substituted 6-exo cycloadduct (CA) various functionals using higher basis sets were taken into consideration in the gas phase. An analysis of the energetic parameters indicates that the reaction path leading to 6-exo CA are kinetically as well as thermodynamically favored in the gas phase, THF and ethanol. The calculated energetic parameters of the 32CA reaction of these phosphorus derivatives were compared with those of methyl acrylate and their nitrogen analogues. Investigation of the global electron density transfer at the TSs indicates that these 32CA reactions have non-polar character, while electron localisation function topological analysis of the C-C bond formation along the most favorable reaction path indicates that these 32CA reactions take place through a non-concerted two-stage one-step mechanism, via highly asynchronous TSs.
Collapse
Affiliation(s)
- Dhanashree Hallooman
- Computational Chemistry Group, Department of Chemistry, Faculty of Science, University of Mauritius Réduit 80837 Mauritius
| | - Mar Ríos-Gutiérrez
- Department of Organic Chemistry, University of Valencia Dr. Moliner 50, E-46100 Burjassot Valencia Spain
| | - Lydia Rhyman
- Computational Chemistry Group, Department of Chemistry, Faculty of Science, University of Mauritius Réduit 80837 Mauritius
- Department of Applied Chemistry, University of Johannesburg Doornfontein Johannesburg 2028 South Africa
| | - Ibrahim A Alswaidan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University Riyadh 11451 Saudi Arabia
| | - Luis R Domingo
- Department of Organic Chemistry, University of Valencia Dr. Moliner 50, E-46100 Burjassot Valencia Spain
| | - Ponnadurai Ramasami
- Computational Chemistry Group, Department of Chemistry, Faculty of Science, University of Mauritius Réduit 80837 Mauritius
- Department of Applied Chemistry, University of Johannesburg Doornfontein Johannesburg 2028 South Africa
| |
Collapse
|
22
|
Skarbek C, Delahousse J, Pioche-Durieu C, Baconnais S, Deroussent A, Renevret P, Rivard M, Desmaele D, Martens T, Le Cam E, Couvreur P, Paci A. Poly-isoprenylated ifosfamide analogs: Preactivated antitumor agents as free formulation or nanoassemblies. Int J Pharm 2017; 532:748-756. [PMID: 28546071 DOI: 10.1016/j.ijpharm.2017.05.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/19/2017] [Accepted: 05/20/2017] [Indexed: 01/28/2023]
Abstract
Oxazaphosphorines including cyclophosphamide, trofosfamide and ifosfamide (IFO) belong to the alkylating agent class and are indicated in the treatment of numerous cancers. However, IFO is subject to limiting side-effects in high-dose protocols. To circumvent IFO drawbacks in clinical practices, preactivated IFO analogs were designed to by-pass the toxic metabolic pathway. Among these IFO analogs, some of them showed the ability to self-assemble due to the use of a poly-isoprenyloxy chain as preactivating moiety. We present here, the in vitro activity of the nanoassembly formulations of preactivated IFO derivatives with a C-4 geranyloxy, farnesyloxy and squalenoxy substituent on a large panel of tumor cell lines. The chemical and colloidal stabilities of the geranyloxy-IFO (G-IFO), farnesyloxy-IFO (F-IFO) and squalenoxy-IFO (SQ-IFO) NAs were further evaluated in comparison to their free formulation. Finally, pharmacokinetic parameters and maximal tolerated dose of the most potent preactivated IFO analog (G-IFO) were determined and compared to IFO, paving the way to in vivo studies.
Collapse
Affiliation(s)
- Charles Skarbek
- Vectorologie des anticancéreux et des acides nucléiques, UMR 8203, CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Julia Delahousse
- Vectorologie des anticancéreux et des acides nucléiques, UMR 8203, CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France; Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, 94805 Villejuif, France
| | - Catherine Pioche-Durieu
- Signalisations, Noyaux et Innovations en Cancérologie, UMR 8126, CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Sonia Baconnais
- Signalisations, Noyaux et Innovations en Cancérologie, UMR 8126, CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Alain Deroussent
- Vectorologie des anticancéreux et des acides nucléiques, UMR 8203, CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Patrice Renevret
- Institut de Chimie et des Matériaux Paris Est Créteil (ICMPE), UMR 7182, CNRS, Université Paris Est (UPEC), 94320 Thiais, France
| | - Michael Rivard
- Institut de Chimie et des Matériaux Paris Est Créteil (ICMPE), UMR 7182, CNRS, Université Paris Est (UPEC), 94320 Thiais, France
| | - Didier Desmaele
- Institut Galien Paris-Sud, UMR 8612, CNRS, Université Paris-Saclay, Faculté de Pharmacie, 92296, Châtenay-Malabry, France
| | - Thierry Martens
- Institut de Chimie et des Matériaux Paris Est Créteil (ICMPE), UMR 7182, CNRS, Université Paris Est (UPEC), 94320 Thiais, France
| | - Eric Le Cam
- Signalisations, Noyaux et Innovations en Cancérologie, UMR 8126, CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France
| | - Patrick Couvreur
- Institut Galien Paris-Sud, UMR 8612, CNRS, Université Paris-Saclay, Faculté de Pharmacie, 92296, Châtenay-Malabry, France
| | - Angelo Paci
- Vectorologie des anticancéreux et des acides nucléiques, UMR 8203, CNRS, Université Paris-Sud, Gustave Roussy, Université Paris-Saclay, 94805 Villejuif, France; Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, 94805 Villejuif, France; Département de Pharmacocinétique & Pharmacie Clinique, Université Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 92296 Châtenay-Malabry, France.
| |
Collapse
|
23
|
Hallooman D, Ríos-Gutiérrez M, Rhyman L, Alswaidan IA, Fun HK, Domingo LR, Ramasami P. [3+2] Cycloaddition reaction of 1H-phosphorinium-3-olate and 1-methylphosphorinium-3-olate with methyl acrylate: A DFT study. COMPUT THEOR CHEM 2016. [DOI: 10.1016/j.comptc.2016.04.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
24
|
Tóth A, Brózik A, Szakács G, Sarkadi B, Hegedüs T. A novel mathematical model describing adaptive cellular drug metabolism and toxicity in the chemoimmune system. PLoS One 2015; 10:e0115533. [PMID: 25699998 PMCID: PMC4338831 DOI: 10.1371/journal.pone.0115533] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 11/25/2014] [Indexed: 02/01/2023] Open
Abstract
Cells cope with the threat of xenobiotic stress by activating a complex molecular network that recognizes and eliminates chemically diverse toxic compounds. This "chemoimmune system" consists of cellular Phase I and Phase II metabolic enzymes, Phase 0 and Phase III ATP Binding Cassette (ABC) membrane transporters, and nuclear receptors regulating these components. In order to provide a systems biology characterization of the chemoimmune network, we designed a reaction kinetic model based on differential equations describing Phase 0-III participants and regulatory elements, and characterized cellular fitness to evaluate toxicity. In spite of the simplifications, the model recapitulates changes associated with acquired drug resistance and allows toxicity predictions under variable protein expression and xenobiotic exposure conditions. Our simulations suggest that multidrug ABC transporters at Phase 0 significantly facilitate the defense function of successive network members by lowering intracellular drug concentrations. The model was extended with a novel toxicity framework which opened the possibility of performing in silico cytotoxicity assays. The alterations of the in silico cytotoxicity curves show good agreement with in vitro cell killing experiments. The behavior of the simplified kinetic model suggests that it can serve as a basis for more complex models to efficiently predict xenobiotic and drug metabolism for human medical applications.
Collapse
Affiliation(s)
- Attila Tóth
- MTA-SE Molecular Biophysics Research Group, Hungarian Academy of Sciences, Budapest, 1094, Hungary
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, 1094, Hungary
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, 1113, Hungary
| | - Anna Brózik
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, 1113, Hungary
| | - Gergely Szakács
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, 1113, Hungary
| | - Balázs Sarkadi
- MTA-SE Molecular Biophysics Research Group, Hungarian Academy of Sciences, Budapest, 1094, Hungary
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, 1094, Hungary
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, 1113, Hungary
| | - Tamás Hegedüs
- MTA-SE Molecular Biophysics Research Group, Hungarian Academy of Sciences, Budapest, 1094, Hungary
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, 1094, Hungary
- * E-mail:
| |
Collapse
|
25
|
Vredenburg G, den Braver-Sewradj S, van Vugt-Lussenburg BMA, Vermeulen NPE, Commandeur JNM, Vos JC. Activation of the anticancer drugs cyclophosphamide and ifosfamide by cytochrome P450 BM3 mutants. Toxicol Lett 2014; 232:182-92. [PMID: 25448283 DOI: 10.1016/j.toxlet.2014.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 10/27/2014] [Accepted: 11/03/2014] [Indexed: 11/17/2022]
Abstract
Cyclophosphamide (CPA) and ifosfamide (IFA) are widely used anticancer agents that require metabolic activation by cytochrome P450 (CYP) enzymes. While 4-hydroxylation yields DNA-alkylating and cytotoxic metabolites, N-dechloroethylation results in the generation of neuro- and nephrotoxic byproducts. Gene-directed enzyme prodrug therapies (GDEPT) have been suggested to facilitate local CPA and IFA bioactivation by expressing CYP enzymes within the tumor cells, thereby increasing efficacy. We screened bacterial CYP BM3 mutants, previously engineered to metabolize drug-like compounds, for their ability to catalyze 4-hydroxylation of CPA and IFA. Two CYP BM3 mutants showed very rapid initial bioactivation of CPA and IFA, followed by a slower phase of product formation. N-dechloroethylation by these mutants was very low (IFA) to undetectable (CPA). Using purified CYP BM3 as an extracellular bioactivation tool, cytotoxicity of CPA and IFA metabolism was confirmed in U2OS cells. This novel application of CYP BM3 possibly provides a clean and catalytically efficient alternative to liver microsomes or S9 for the study of CYP-mediated drug toxicity. To our knowledge, the observed rate of CPA and IFA 4-hydroxylation by these CYP BM3 mutants is the fastest reported to date, and might be of potential interest for CPA and IFA GDEPT.
Collapse
Affiliation(s)
- Galvin Vredenburg
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences (CPS), VU University Amsterdam, De Boelelaan 1083, Amsterdam 1081 HV, The Netherlands
| | - Shalenie den Braver-Sewradj
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences (CPS), VU University Amsterdam, De Boelelaan 1083, Amsterdam 1081 HV, The Netherlands
| | | | - Nico P E Vermeulen
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences (CPS), VU University Amsterdam, De Boelelaan 1083, Amsterdam 1081 HV, The Netherlands
| | - Jan N M Commandeur
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences (CPS), VU University Amsterdam, De Boelelaan 1083, Amsterdam 1081 HV, The Netherlands
| | - J Chris Vos
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences (CPS), VU University Amsterdam, De Boelelaan 1083, Amsterdam 1081 HV, The Netherlands.
| |
Collapse
|
26
|
Ali SA, Danda SK, Basha SAA, Rasheed A, Ahmed O, Ahmed MM. Comparision of uroprotective activity of reduced glutathione with mesna in ifosfamide induced hemorrhagic cystitis in rats. Indian J Pharmacol 2014; 46:105-8. [PMID: 24550594 PMCID: PMC3912792 DOI: 10.4103/0253-7613.125188] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/12/2013] [Accepted: 12/01/2013] [Indexed: 11/17/2022] Open
Abstract
Background: Ifosfamide (IFO) is widely used DNA-alkylating agents in cancer chemotherapy for management of solid tumors and hematological malignancies. However, hemorrhagic cystitis limits the use of IFO. Objectives: To compare the efficiency of reduced glutathione with 2-Mesna in reducing Ifosfamide (IFO) induced hemorrhagic cystitis (HC) in wistar rats. Materials and Methods: Ifosfamide and 2-Mesna were dissolved in sterile water for injection and administered to wistar rats of albino strains. The rats were randomly assigned to one of the four groups of 6 rats each: Group I: Vehicle control; Group II: 120 mg/kg of IFO alone by intraperitoneal injection (i.p); Group III: 40 mg/kg Mesna i.p., at the same time and at 4 and 8 h after IFO administration; Group IV: 500 mg/kg of glutathione i.p., 30 min prior to IFO as above. The animals were observed for 5 days. On 6th day, rats were sacrificed by dissecting the intrajugular vein. The bladders were macroscopically and histopathologically evaluated. Results: Control animals had normal bladders with assigned scores of ‘0’ for the three parameters of edema, hemorrhage and histopathological changes. All the animals receiving IFO (group II) had evidence of HC as evidenced by alterations of edema and hemorrhages. These alterations were almost abolished (P < 0.001) by the glutathione (group III) or Mesna (group IV) in IFO-treated animals. Conclusion: Glutathione could be as useful as Mesna in the preventive management of IFO-induced HC.
Collapse
Affiliation(s)
- Syed Amir Ali
- Deccan School of Pharmacy, Hyderabad, Andhra Pradesh, India
| | | | | | - Asif Rasheed
- Deccan School of Pharmacy, Hyderabad, Andhra Pradesh, India
| | - Osman Ahmed
- Deccan School of Pharmacy, Hyderabad, Andhra Pradesh, India
| | | |
Collapse
|
27
|
Rathod V, Jain S, Nandekar P, Sangamwar AT. Human pregnane X receptor: a novel target for anticancer drug development. Drug Discov Today 2014; 19:63-70. [DOI: 10.1016/j.drudis.2013.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 05/02/2013] [Accepted: 08/15/2013] [Indexed: 02/07/2023]
|
28
|
The constitutive androstane receptor is a novel therapeutic target facilitating cyclophosphamide-based treatment of hematopoietic malignancies. Blood 2012; 121:329-38. [PMID: 23160467 DOI: 10.1182/blood-2012-06-436691] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cyclophosphamide (CPA) is one of the most widely used chemotherapeutic prodrugs that undergoes hepatic bioactivation mediated predominantly by cytochrome P450 (CYP) 2B6. Given that the CYP2B6 gene is primarily regulated by the constitutive androstane receptor (CAR, NR1I3), we hypothesize that selective activation of CAR can enhance systemic exposure of the pharmacologically active 4-hydroxycyclophosamide (4-OH-CPA), with improved efficacy of CPA-based chemotherapy. In this study, we have developed a unique human primary hepatocyte (HPH)-leukemia cell coculture model; the chemotherapeutic effects of CPA on leukemia cells can be directly investigated in vitro in a cellular environment where hepatic metabolism was well maintained. Our results demonstrated that activation of CAR preferentially induces the expression of CYP2B6 over CYP3A4 in HPHs, although endogenous expression of these enzymes in leukemia cells remains negligible. Importantly, coadministration of CPA with a human CAR activator led to significantly enhanced cytotoxicity in leukemia cells by inducing the apoptosis pathways, without concomitant increase in the off-target hepatotoxicity. Associated with the enhanced antitumor activity, a time and concentration-dependent increase in 4-OH-CPA formation was observed in the coculture system. Together, our findings offer proof of concept that CAR as a novel molecular target can facilitate CPA-based chemotherapy by selectively promoting its bioactivation.
Collapse
|