1
|
Yang J, Hawthorne L, Stack S, Blagg B, Ali A, Zorlutuna P. Engineered Age-Mimetic Breast Cancer Models Reveal Differential Drug Responses in Young and Aged Microenvironments. Adv Healthc Mater 2025; 14:e2404461. [PMID: 39821643 PMCID: PMC11960616 DOI: 10.1002/adhm.202404461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Indexed: 01/19/2025]
Abstract
Aging is one of the most significant risk factors for breast cancer. With the growing interest in the alterations of the aging breast tissue microenvironment, it is identified that aging is related to tumorigenesis, invasion, and drug resistance. However, current pre-clinical disease models often neglect the impact of aging and sometimes result in worse clinical outcomes. In this study, aged animal-generated materials are utilized to create and validate a novel age-mimetic breast cancer model that generates an aging microenvironment for cells and alters cells toward a more invasive phenotype found in the aged environment. Furthermore, the age-mimetic models are utilized for 3D breast cancer invasion assessment and high-throughput screening of over 700 drugs in the FDA-approved drug library. 36 potential effective drug targets as well as 34 potential drug targets with different drug responses in different age groups are identified, demonstrating the potential of this age-mimetic breast cancer model for further in-depth breast cancer studies and drug development.
Collapse
Affiliation(s)
- Jun Yang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Lauren Hawthorne
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Sharon Stack
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| | - Brian Blagg
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| | - Aktar Ali
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| | - Pinar Zorlutuna
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| |
Collapse
|
2
|
Gola AM, Bucci-Muñoz M, Rigalli JP, Ceballos MP, Ruiz ML. Role of the RNA binding protein IGF2BP1 in cancer multidrug resistance. Biochem Pharmacol 2024; 230:116555. [PMID: 39332691 DOI: 10.1016/j.bcp.2024.116555] [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: 06/12/2024] [Revised: 09/04/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
The insulin-like growth factor-2 mRNA-binding protein 1 (IGF2BP1), a member of a conserved family of single-stranded RNA-binding proteins (IGF2BP1-3), is expressed in a broad range of fetal tissues, placenta and more than sixteen cancer types but only in a limited number of normal adult tissues. IGF2BP1is required for the transport from nucleus to cytoplasm of certain mRNAs that play essential roles in embryogenesis, carcinogenesis, and multidrug resistance (MDR), by affecting their stability, translation, or localization. The purpose of this review is to gather and present information on MDR mechanisms in cancer and the significance of IGF2BP1 in this context. Within this review, we will provide an overview of IGF2BP1, including its tissue distribution, expression, molecular targets in the context of tumorigenesis and its inhibitors. Our main focus will be on elucidating the interplay between IGF2BP1 and MDR, particularly with regard to chemoresistance mediated by ABC transporters.
Collapse
Affiliation(s)
- Aldana Magalí Gola
- Instituto de Fisiología Experimental (CONICET) - Facultad de Ciencias Bioquímicas y Farmacéuticas (UNR), Rosario, Argentina
| | - María Bucci-Muñoz
- Instituto de Fisiología Experimental (CONICET) - Facultad de Ciencias Bioquímicas y Farmacéuticas (UNR), Rosario, Argentina
| | - Juan Pablo Rigalli
- Department of Clinical Pharmacology and Pharmacoepidemiology, Medical Faculty Heidelberg, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - María Paula Ceballos
- Instituto de Fisiología Experimental (CONICET) - Facultad de Ciencias Bioquímicas y Farmacéuticas (UNR), Rosario, Argentina
| | - María Laura Ruiz
- Instituto de Fisiología Experimental (CONICET) - Facultad de Ciencias Bioquímicas y Farmacéuticas (UNR), Rosario, Argentina.
| |
Collapse
|
3
|
Qi Q, Gu R, Zhu J, Anderson KE, Ma X. Roles of the ABCG2 Transporter in Protoporphyrin IX Distribution and Toxicity. Drug Metab Dispos 2024; 52:1201-1207. [PMID: 38351044 PMCID: PMC11495668 DOI: 10.1124/dmd.123.001582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/08/2024] [Indexed: 10/18/2024] Open
Abstract
ATP-binding cassette transporter subfamily G member 2 (ABCG2) is a membrane-bound transporter responsible for the efflux of various xenobiotics and endobiotics, including protoporphyrin IX (PPIX), an intermediate in the heme biosynthesis pathway. Certain genetic mutations and chemicals impair the conversion of PPIX to heme and/or increase PPIX production, leading to PPIX accumulation and toxicity. In mice, deficiency of ABCG2 protects against PPIX-mediated phototoxicity and hepatotoxicity by modulating PPIX distribution. In addition, in vitro studies revealed that ABCG2 inhibition increases the efficacy of PPIX-based photodynamic therapy by retaining PPIX inside target cells. In this review, we discuss the roles of ABCG2 in modulating the tissue distribution of PPIX, PPIX-mediated toxicity, and PPIX-based photodynamic therapy. SIGNIFICANCE STATEMENT: This review summarized the roles of ABCG2 in modulating PPIX distribution and highlighted the therapeutic potential of ABCG2 inhibitors for the management of PPIX-mediated toxicity.
Collapse
Affiliation(s)
- Qian Qi
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| | - Ruizhi Gu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| | - Junjie Zhu
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| | - Karl E Anderson
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| | - Xiaochao Ma
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (Q.Q., R.G., J.Z., X.M.) and Porphyria Laboratory and Center, Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas (K.E.A.)
| |
Collapse
|
4
|
Yang J, Hawthorne L, Stack S, Blagg B, Ali A, Zorlutuna P. Engineered Age-Mimetic Breast Cancer Models Reveal Differential Drug Responses in Young and Aged Microenvironments. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.06.616903. [PMID: 39416111 PMCID: PMC11482747 DOI: 10.1101/2024.10.06.616903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Aging is one of the most significant risk factors for breast cancer. With the growing interests in the alterations of the aging breast tissue microenvironment, it has been identified that aging is related to tumorigenesis, invasion, and drug resistance. However, current pre-clinical disease models often neglect the impact of aging and sometimes result in worse clinical outcomes. In this study, we utilized aged animal-generated materials to create and validate a novel age-mimetic breast cancer model that generates an aging microenvironment for cells and alters cells towards a phenotype found in the aged environment. Furthermore, we utilized the age-mimetic models for 3D breast cancer invasion assessment and high-throughput screening of over 700 drugs in the FDA-approved drug library. We identified 36 potential effective drug targets and 34 potential drug targets with different drug responses in different age groups, demonstrating the potential of this age-mimetic breast cancer model for further in-depth breast cancer studies and drug development.
Collapse
Affiliation(s)
- Jun Yang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Lauren Hawthorne
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Sharon Stack
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| | - Brian Blagg
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| | - Aktar Ali
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| | - Pinar Zorlutuna
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, 46556, USA
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN, 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, 46556, USA
| |
Collapse
|
5
|
Ferrer F, Tetu P, Dousset L, Lebbe C, Ciccolini J, Combarel D, Meyer N, Paci A, Bouchet S. Tyrosine kinase inhibitors in cancers: Treatment optimization - Part II. Crit Rev Oncol Hematol 2024; 200:104385. [PMID: 38810843 DOI: 10.1016/j.critrevonc.2024.104385] [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: 01/24/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
Real-life populations are more heterogeneous than those included in prospective clinical studies. In cancer patients, comorbidities and co-medications favor the appearance of severe adverse effects which can significantly impact quality of life and treatment effectiveness. Most of tyrosine kinase inhibitors (TKI) have been developed with flat oral dosing exposing patients to the risk of poor adherence due to side effects. Additionally, genetic or physiological factors, differences in diet, and drug-drug interactions can lead to inter-individual variability affecting treatment outcomes and increasing the risk of adverse events. Knowledge of the different factors of variability allows individualized patient management. This review examines the effects of adherence, food intake, and pharmaceutical form on the pharmacokinetics of oral TKI, as well as evaluating pharmacokinetics considerations improving TKI management. Concentration-effectiveness and concentration-toxicity data are presented for the selected TKI, and a simple therapeutic drug monitoring schema is outlined to help individualize dosing of oral TKI.
Collapse
Affiliation(s)
- Florent Ferrer
- Department of Pharmacology, Clermont-Ferrand University Hospital, Clermont-Ferrand, France; SMARTc Unit, CRCM Inserm U1068, Aix Marseille Univ and APHM, Marseille, France; Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France
| | - Pauline Tetu
- Department of Dermatology, APHP Dermatology, Paris 7 Diderot University, INSERM U976, Hôpital Saint-Louis, Paris, France; Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France
| | - Léa Dousset
- Dermatology Department, Bordeaux University Hospital, Bordeaux, France; Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France
| | - Céleste Lebbe
- Department of Dermatology, APHP Dermatology, Paris 7 Diderot University, INSERM U976, Hôpital Saint-Louis, Paris, France; Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France
| | - Joseph Ciccolini
- SMARTc Unit, CRCM Inserm U1068, Aix Marseille Univ and APHM, Marseille, France; Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France
| | - David Combarel
- Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France; Service de Pharmacocinétique, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, Châtenay-Malabry 92 296, France
| | - Nicolas Meyer
- Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France; Université Paul Sabatier-Toulouse III, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche, Toulouse 1037-CRCT, France
| | - Angelo Paci
- Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France; Service de Pharmacocinétique, Faculté de Pharmacie, Université Paris-Saclay, Châtenay-Malabry, Châtenay-Malabry 92 296, France
| | - Stéphane Bouchet
- Service de Pharmacologie, Département de Biologie et Pathologie médicales, Gustave Roussy, Villejuif 94805, France; Département de Pharmacologie, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France.
| |
Collapse
|
6
|
Wang Y, Tu MJ, Yu AM. Efflux ABC transporters in drug disposition and their posttranscriptional gene regulation by microRNAs. Front Pharmacol 2024; 15:1423416. [PMID: 39114355 PMCID: PMC11303158 DOI: 10.3389/fphar.2024.1423416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
ATP-binding cassette (ABC) transporters are transmembrane proteins expressed commonly in metabolic and excretory organs to control xenobiotic or endobiotic disposition and maintain their homeostasis. Changes in ABC transporter expression may directly affect the pharmacokinetics of relevant drugs involving absorption, distribution, metabolism, and excretion (ADME) processes. Indeed, overexpression of efflux ABC transporters in cancer cells or bacteria limits drug exposure and causes therapeutic failure that is known as multidrug resistance (MDR). With the discovery of functional noncoding microRNAs (miRNAs) produced from the genome, many miRNAs have been revealed to govern posttranscriptional gene regulation of ABC transporters, which shall improve our understanding of complex mechanism behind the overexpression of ABC transporters linked to MDR. In this article, we first overview the expression and localization of important ABC transporters in human tissues and their clinical importance regarding ADME as well as MDR. Further, we summarize miRNA-controlled posttranscriptional gene regulation of ABC transporters and effects on ADME and MDR. Additionally, we discuss the development and utilization of novel bioengineered miRNA agents to modulate ABC transporter gene expression and subsequent influence on cellular drug accumulation and chemosensitivity. Findings on posttranscriptional gene regulation of ABC transporters shall not only improve our understanding of mechanisms behind variable ADME but also provide insight into developing new means towards rational and more effective pharmacotherapies.
Collapse
Affiliation(s)
| | | | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California at Davis, Sacramento, CA, United States
| |
Collapse
|
7
|
Loer HLH, Kovar C, Rüdesheim S, Marok FZ, Fuhr LM, Selzer D, Schwab M, Lehr T. Physiologically based pharmacokinetic modeling of imatinib and N-desmethyl imatinib for drug-drug interaction predictions. CPT Pharmacometrics Syst Pharmacol 2024; 13:926-940. [PMID: 38482980 PMCID: PMC11179706 DOI: 10.1002/psp4.13127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/20/2024] [Accepted: 03/05/2024] [Indexed: 06/17/2024] Open
Abstract
The first-generation tyrosine kinase inhibitor imatinib has revolutionized the development of targeted cancer therapy and remains among the frontline treatments, for example, against chronic myeloid leukemia. As a substrate of cytochrome P450 (CYP) 2C8, CYP3A4, and various transporters, imatinib is highly susceptible to drug-drug interactions (DDIs) when co-administered with corresponding perpetrator drugs. Additionally, imatinib and its main metabolite N-desmethyl imatinib (NDMI) act as inhibitors of CYP2C8, CYP2D6, and CYP3A4 affecting their own metabolism as well as the exposure of co-medications. This work presents the development of a parent-metabolite whole-body physiologically based pharmacokinetic (PBPK) model for imatinib and NDMI used for the investigation and prediction of different DDI scenarios centered around imatinib as both a victim and perpetrator drug. Model development was performed in PK-Sim® using a total of 60 plasma concentration-time profiles of imatinib and NDMI in healthy subjects and cancer patients. Metabolism of both compounds was integrated via CYP2C8 and CYP3A4, with imatinib additionally transported via P-glycoprotein. The subsequently developed DDI network demonstrated good predictive performance. DDIs involving imatinib and NDMI were simulated with perpetrator drugs rifampicin, ketoconazole, and gemfibrozil as well as victim drugs simvastatin and metoprolol. Overall, 12/12 predicted DDI area under the curve determined between first and last plasma concentration measurements (AUClast) ratios and 12/12 predicted DDI maximum plasma concentration (Cmax) ratios were within twofold of the respective observed ratios. Potential applications of the final model include model-informed drug development or the support of model-informed precision dosing.
Collapse
Affiliation(s)
| | - Christina Kovar
- Clinical PharmacySaarland UniversitySaarbrückenGermany
- Dr. Margarete Fischer‐Bosch‐Institute of Clinical PharmacologyStuttgartGermany
| | - Simeon Rüdesheim
- Clinical PharmacySaarland UniversitySaarbrückenGermany
- Dr. Margarete Fischer‐Bosch‐Institute of Clinical PharmacologyStuttgartGermany
| | | | | | | | - Matthias Schwab
- Dr. Margarete Fischer‐Bosch‐Institute of Clinical PharmacologyStuttgartGermany
- Departments of Clinical Pharmacology, and Pharmacy and BiochemistryUniversity of TübingenTübingenGermany
- Cluster of Excellence iFIT (EXC2180), Image‐Guided and Functionally Instructed Tumor TherapiesUniversity of TübingenTübingenGermany
| | - Thorsten Lehr
- Clinical PharmacySaarland UniversitySaarbrückenGermany
| |
Collapse
|
8
|
Balhara A, Yin M, Unadkat JD. Successful Prediction of Fetal Exposure to Dual BCRP/P-gp Drug Substrates Using the Efflux Ratio-Relative Expression Factor Approach and PBPK M&S. Clin Pharmacol Ther 2024; 115:1044-1053. [PMID: 38124355 DOI: 10.1002/cpt.3157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023]
Abstract
To inform fetal drug safety, it is important to determine or predict fetal drug exposure throughout pregnancy. The former is not possible in the first or second trimester. In contrast, at the time of birth, fetal drug exposure, relative to maternal exposure, can be estimated as Kp,uu (unbound fetal umbilical venous (UV) plasma area under the curve (AUC)/unbound maternal plasma (MP) AUC), provided the observed UV/MP values, spanning the dosing interval, are available from multiple maternal-fetal dyads. However, this fetal Kp,uu cannot be extrapolated to other drugs. To overcome the above limitations, we have used an efflux ratio-relative expression factor (ER-REF) approach to successfully predict the fetal Kp,uu of P-gp substrates. Because many drugs taken by pregnant people are also BCRP substrates, here, we extend this approach to drugs that are effluxed by both placental BCRP and P-gp or P-gp alone. To verify our predictions, we chose drugs for which UV/MP data were available at term: glyburide and imatinib (both BCRP and P-gp substrates) and nelfinavir (only P-gp substrate). First, the ER of the drugs was determined using Transwells and MDCKII cells expressing either BCRP or P-gp. Then, the ER was scaled using the proteomics-informed REF value to predict the fetal Kp,uu of the drug at term. The ER-REF predicted fetal Kp,uu of glyburide (0.43), imatinib (0.42), and nelfinavir (0.40) fell within two-fold of the corresponding in vivo fetal Kp,uu (0.44, 0.37, and 0.46, respectively). These data confirm that the ER-REF approach can successfully predict fetal drug exposure to BCRP/P-gp and P-gp substrates, at term.
Collapse
Affiliation(s)
- Ankit Balhara
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Mengyue Yin
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
9
|
Chandratre S, Olsen J, Howley R, Chen B. Targeting ABCG2 transporter to enhance 5-aminolevulinic acid for tumor visualization and photodynamic therapy. Biochem Pharmacol 2023; 217:115851. [PMID: 37858868 PMCID: PMC10842008 DOI: 10.1016/j.bcp.2023.115851] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023]
Abstract
5-Aminolevulinic acid (ALA) has been approved by the U. S. FDA for fluorescence-guided resection of high-grade glioma and photodynamic therapy (PDT) of superficial skin precancerous and cancerous lesions. As a prodrug, ALA administered orally or topically is metabolized in the heme biosynthesis pathway to produce protoporphyrin IX (PpIX), the active drug with red fluorescence and photosensitizing property. Preferential accumulation of PpIX in tumors after ALA administration enables the use of ALA for PpIX-mediated tumor fluorescence diagnosis and PDT, functioning as a photo-theranostic agent. Extensive research is currently underway to further enhance ALA-mediated PpIX tumor disposition for better tumor visualization and treatment. Particularly, the discovery of PpIX as a specific substrate of ATP binding cassette subfamily G member 2 (ABCG2) opens the door to therapeutic enhancement with ABCG2 inhibitors. Studies with human tumor cell lines and human tumor samples have demonstrated ABCG2 as an important biological determinant of reduced ALA-PpIX tumor accumulation, inhibition of which greatly enhances ALA-PpIX fluorescence and PDT response. These studies strongly support targeting ABCG2 as an effective therapeutic enhancement approach. In this review, we would like to summarize current research of ABCG2 as a drug efflux transporter in multidrug resistance, highlight previous works on targeting ABCG2 for therapeutic enhancement of ALA, and provide future perspectives on how to translate this ABCG2-targeted therapeutic enhancement strategy from bench to bedside.
Collapse
Affiliation(s)
- Sharayu Chandratre
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph's University, Philadelphia, PA, USA
| | - Jordyn Olsen
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph's University, Philadelphia, PA, USA
| | - Richard Howley
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph's University, Philadelphia, PA, USA
| | - Bin Chen
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph's University, Philadelphia, PA, USA; Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Verhagen NE, Koenderink JB, Blijlevens NMA, Janssen JJWM, Russel FGM. Transporter-Mediated Cellular Distribution of Tyrosine Kinase Inhibitors as a Potential Resistance Mechanism in Chronic Myeloid Leukemia. Pharmaceutics 2023; 15:2535. [PMID: 38004514 PMCID: PMC10675650 DOI: 10.3390/pharmaceutics15112535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023] Open
Abstract
Chronic myeloid leukemia (CML) is a hematologic neoplasm characterized by the expression of the BCR::ABL1 oncoprotein, a constitutively active tyrosine kinase, resulting in uncontrolled growth and proliferation of cells in the myeloid lineage. Targeted therapy using tyrosine kinase inhibitors (TKIs) such as imatinib, nilotinib, dasatinib, bosutinib, ponatinib and asciminib has drastically improved the life expectancy of CML patients. However, treatment resistance occurs in 10-20% of CML patients, which is a multifactorial problem that is only partially clarified by the presence of TKI inactivating BCR::ABL1 mutations. It may also be a consequence of a reduction in cytosolic TKI concentrations in the target cells due to transporter-mediated cellular distribution. This review focuses on drug-transporting proteins in stem cells and progenitor cells involved in the distribution of TKIs approved for the treatment of CML. Special attention will be given to ATP-binding cassette transporters expressed in lysosomes, which may facilitate the extracytosolic sequestration of these compounds.
Collapse
Affiliation(s)
- Noor E. Verhagen
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.E.V.); (J.B.K.)
| | - Jan B. Koenderink
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.E.V.); (J.B.K.)
| | - Nicole M. A. Blijlevens
- Department of Haematology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.M.A.B.); (J.J.W.M.J.)
| | - Jeroen J. W. M. Janssen
- Department of Haematology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.M.A.B.); (J.J.W.M.J.)
| | - Frans G. M. Russel
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.E.V.); (J.B.K.)
| |
Collapse
|
11
|
Pathak S, Zajac KK, Annaji M, Govindarajulu M, Nadar RM, Bowen D, Babu RJ, Dhanasekaran M. Clinical outcomes of chemotherapy in cancer patients with different ethnicities. Cancer Rep (Hoboken) 2023; 6 Suppl 1:e1830. [PMID: 37150853 PMCID: PMC10440845 DOI: 10.1002/cnr2.1830] [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: 12/31/2022] [Revised: 04/18/2023] [Accepted: 04/21/2023] [Indexed: 05/09/2023] Open
Abstract
BACKGROUND Choosing the most effective chemotherapeutic agent with safest side effect profile is a common challenge in cancer treatment. Although there are standardized chemotherapy protocols in place, protocol changes made after extensive clinical trials demonstrate significant improvement in the efficacy and tolerability of certain drugs. The pharmacokinetics, pharmacodynamics, and tolerance of anti-cancer medications are all highly individualized. A driving force behind these differences lies within a person's genetic makeup. RECENT FINDINGS Pharmacogenomics, the study of how an individual's genes impact the processing and action of a drug, can optimize drug responsiveness and reduce toxicities by creating a customized medication regimen. However, these differences are rarely considered in the initial determination of standardized chemotherapeutic protocols and treatment algorithms. Because pharmacoethnicity is influenced by both genetic and nongenetic variables, clinical data highlighting disparities in the frequency of polymorphisms between different ethnicities is steadily growing. Recent data suggests that ethnic variations in the expression of allelic variants may result in different pharmacokinetic properties of the anti-cancer medication. In this article, the clinical outcomes of various chemotherapy classes in patients of different ethnicities were reviewed. CONCLUSION Genetic and nongenetic variables contribute to the interindividual variability in response to chemotherapeutic drugs. Considering pharmacoethnicity in the initial determination of standard chemotherapeutic protocols and treatment algorithms can lead to better clinical outcomes of patients of different ethnicities.
Collapse
Affiliation(s)
- Suhrud Pathak
- Department of Drug Discovery and Development, Harrison College of PharmacyAuburn UniversityAuburnAlabamaUSA
| | - Kelsee K. Zajac
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical SciencesUniversity of ToledoToledoOhioUSA
| | - Manjusha Annaji
- Department of Drug Discovery and Development, Harrison College of PharmacyAuburn UniversityAuburnAlabamaUSA
| | - Manoj Govindarajulu
- Department of Drug Discovery and Development, Harrison College of PharmacyAuburn UniversityAuburnAlabamaUSA
| | - Rishi M. Nadar
- Department of Drug Discovery and Development, Harrison College of PharmacyAuburn UniversityAuburnAlabamaUSA
| | - Dylan Bowen
- Department of Drug Discovery and Development, Harrison College of PharmacyAuburn UniversityAuburnAlabamaUSA
| | - R. Jayachandra Babu
- Department of Drug Discovery and Development, Harrison College of PharmacyAuburn UniversityAuburnAlabamaUSA
| | | |
Collapse
|
12
|
Impact of ABCG2 and ABCB1 Polymorphisms on Imatinib Plasmatic Exposure: An Original Work and Meta-Analysis. Int J Mol Sci 2023; 24:ijms24043303. [PMID: 36834713 PMCID: PMC9963452 DOI: 10.3390/ijms24043303] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Adequate imatinib plasma levels are necessary to guarantee an efficacious and safe treatment in gastrointestinal stromal tumor (GIST) and chronic myeloid leukemia (CML) patients. Imatinib is a substrate of the drug transporters ATP-binding cassette subfamily B member 1 (ABCB1) and ATP-binding cassette subfamily G member 2 (ABCG2) that can affect its plasma concentration. In the present study, the association between three genetic polymorphisms in ABCB1 (rs1045642, rs2032582, rs1128503) and one in ABCG2 (rs2231142) and the imatinib plasma trough concentration (Ctrough) was investigated in 33 GIST patients enrolled in a prospective clinical trial. The results of the study were meta-analyzed with those of other seven studies (including a total of 649 patients) selected from the literature through a systematic review process. The ABCG2 c.421C>A genotype demonstrated, in our cohort of patients, a borderline association with imatinib plasma trough levels that became significant in the meta-analysis. Specifically, homozygous carriers of the ABCG2 c.421 A allele showed higher imatinib plasma Ctrough with respect to the CC/CA carriers (Ctrough, 1463.2 ng/mL AA, vs. 1196.6 ng/mL CC + AC, p = 0.04) in 293 patients eligible for the evaluation of this polymorphism in the meta-analysis. The results remained significant under the additive model. No significant association could be described between ABCB1 polymorphisms and imatinib Ctrough, neither in our cohort nor in the meta-analysis. In conclusion, our results and the available literature studies sustain an association between ABCG2 c.421C>A and imatinib plasma Ctrough in GIST and CML patients.
Collapse
|
13
|
Huang J, Ecker GF. A Structure-Based View on ABC-Transporter Linked to Multidrug Resistance. Molecules 2023; 28:495. [PMID: 36677553 PMCID: PMC9862083 DOI: 10.3390/molecules28020495] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/29/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Abstract
The discovery of the first ATP-binding cassette (ABC) transporter, whose overexpression in cancer cells is responsible for exporting anticancer drugs out of tumor cells, initiated enormous efforts to overcome tumor cell multidrug resistance (MDR) by inhibition of ABC-transporter. Because of its many physiological functions, diverse studies have been conducted on the mechanism, function and regulation of this important group of transmembrane transport proteins. In this review, we will focus on the structural aspects of this transporter superfamily. Since the resolution revolution of electron microscope, experimentally solved structures increased rapidly. A summary of the structures available and an overview of recent structure-based studies are provided. More specifically, the artificial intelligence (AI)-based predictions from AlphaFold-2 will be discussed.
Collapse
Affiliation(s)
| | - Gerhard F. Ecker
- Department of Pharmaceutical Sciences, University of Vienna, 1140 Vienna, Austria
| |
Collapse
|
14
|
Marie S, Frost KL, Hau RK, Martinez-Guerrero L, Izu JM, Myers CM, Wright SH, Cherrington NJ. Predicting disruptions to drug pharmacokinetics and the risk of adverse drug reactions in non-alcoholic steatohepatitis patients. Acta Pharm Sin B 2023; 13:1-28. [PMID: 36815037 PMCID: PMC9939324 DOI: 10.1016/j.apsb.2022.08.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 12/18/2022] Open
Abstract
The liver plays a central role in the pharmacokinetics of drugs through drug metabolizing enzymes and transporters. Non-alcoholic steatohepatitis (NASH) causes disease-specific alterations to the absorption, distribution, metabolism, and excretion (ADME) processes, including a decrease in protein expression of basolateral uptake transporters, an increase in efflux transporters, and modifications to enzyme activity. This can result in increased drug exposure and adverse drug reactions (ADRs). Our goal was to predict drugs that pose increased risks for ADRs in NASH patients. Bibliographic research identified 71 drugs with reported ADRs in patients with liver disease, mainly non-alcoholic fatty liver disease (NAFLD), 54 of which are known substrates of transporters and/or metabolizing enzymes. Since NASH is the progressive form of NAFLD but is most frequently undiagnosed, we identified other drugs at risk based on NASH-specific alterations to ADME processes. Here, we present another list of 71 drugs at risk of pharmacokinetic disruption in NASH, based on their transport and/or metabolism processes. It encompasses drugs from various pharmacological classes for which ADRs may occur when used in NASH patients, especially when eliminated through multiple pathways altered by the disease. Therefore, these results may inform clinicians regarding the selection of drugs for use in NASH patients.
Collapse
Affiliation(s)
- Solène Marie
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - Kayla L. Frost
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - Raymond K. Hau
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - Lucy Martinez-Guerrero
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - Jailyn M. Izu
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - Cassandra M. Myers
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA
| | - Stephen H. Wright
- College of Medicine, Department of Physiology, University of Arizona, Tucson, AZ 85724, USA
| | - Nathan J. Cherrington
- College of Pharmacy, Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, USA,Corresponding author. Tel.: +1 520 6260219; fax: +1 520 6266944.
| |
Collapse
|
15
|
Lee J, Kim J, Kang J, Lee HJ. COVID-19 drugs: potential interaction with ATP-binding cassette transporters P-glycoprotein and breast cancer resistance protein. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2023; 53:1-22. [PMID: 36320434 PMCID: PMC9607806 DOI: 10.1007/s40005-022-00596-6] [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: 01/27/2022] [Accepted: 08/30/2022] [Indexed: 01/08/2023]
Abstract
Background The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2, has resulted in acute respiratory distress, fatal systemic manifestations (extrapulmonary as well as pulmonary), and premature mortality among many patients. Therapy for COVID-19 has focused on the treatment of symptoms and of acute inflammation (cytokine storm) and the prevention of viral infection. Although the mechanism of COVID-19 is not fully understood, potential clinical targets have been identified for pharmacological, immunological, and vaccinal approaches. Area covered Pharmacological approaches including drug repositioning have been a priority for initial COVID-19 therapy due to the time-consuming nature of the vaccine development process. COVID-19 drugs have been shown to manage the antiviral infection cycle (cell entry and replication of proteins and genomic RNA) and anti-inflammation. In this review, we evaluated the interaction of current COVID-19 drugs with two ATP-binding cassette transporters [P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP)] and potential drug-drug interactions (DDIs) among COVID-19 drugs, especially those associated with P-gp and BCRP efflux transporters. Expert opinion Overall, understanding the pharmacodynamic/pharmacokinetic DDIs of COVID-19 drugs can be useful for pharmacological therapy in COVID-19 patients.
Collapse
Affiliation(s)
- Jaeok Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760 Republic of Korea
| | - Jihye Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760 Republic of Korea
| | - Jiyeon Kang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760 Republic of Korea
| | - Hwa Jeong Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760 Republic of Korea
| |
Collapse
|
16
|
Saghafian Larijani R, Shabani Ravari N, Goodarzi N, Akhlaghpour S, Saghafian Larijani S, Rouini MR, Dinarvand R. Current status of transarterial chemoembolization (TACE) agents in hepatocellular carcinoma treatment. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
17
|
Taskar KS, Yang X, Neuhoff S, Patel M, Yoshida K, Paine MF, Brouwer KL, Chu X, Sugiyama Y, Cook J, Polli JW, Hanna I, Lai Y, Zamek-Gliszczynski M. Clinical Relevance of Hepatic and Renal P-gp/BCRP Inhibition of Drugs: An International Transporter Consortium Perspective. Clin Pharmacol Ther 2022; 112:573-592. [PMID: 35612761 PMCID: PMC9436425 DOI: 10.1002/cpt.2670] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 05/16/2022] [Indexed: 12/11/2022]
Abstract
The role of P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) in drug-drug interactions (DDIs) and limiting drug absorption as well as restricting the brain penetration of drugs with certain physicochemical properties is well known. P-gp/BCRP inhibition by drugs in the gut has been reported to increase the systemic exposure to substrate drugs. A previous International Transporter Consortium (ITC) perspective discussed the feasibility of P-gp/BCRP inhibition at the blood-brain barrier and its implications. This ITC perspective elaborates and discusses specifically the hepatic and renal P-gp/BCRP (referred as systemic) inhibition of drugs and whether there is any consequence for substrate drug disposition. This perspective summarizes the clinical evidence-based recommendations regarding systemic P-gp and BCRP inhibition of drugs with a focus on biliary and active renal excretion pathways. Approaches to assess the clinical relevance of systemic P-gp and BCRP inhibition in the liver and kidneys included (i) curation of DDIs involving intravenously administered substrates or inhibitors; (ii) in vitro-to-in vivo extrapolation of P-gp-mediated DDIs at the systemic level; and (iii) curation of drugs with information available about the contribution of biliary excretion and related DDIs. Based on the totality of evidence reported to date, this perspective supports limited clinical DDI risk upon P-gp or BCRP inhibition in the liver or kidneys.
Collapse
Affiliation(s)
- Kunal S. Taskar
- Drug Metabolism and Pharmacokinetics, IVIVT, GlaxoSmithKline, Stevenage, UK
| | - Xinning Yang
- Office of Clinical Pharmacology, Center of Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD
| | - Sibylle Neuhoff
- Certara UK Ltd, Simcyp Division, 1 Concourse Way, Level 2-Acero, Sheffield, S1 2BJ, UK
| | - Mitesh Patel
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Kenta Yoshida
- Clinical Pharmacology, Genentech Early Research and Development, South San Francisco, CA 94080, USA
| | - Mary F. Paine
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA
| | - Kim L.R. Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Xiaoyan Chu
- Department of ADME and Discovery Toxicology, Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ 07033 USA
| | - Yuichi Sugiyama
- Laboratory of Quantitative System PK/Pharmacodynamics, School of Pharmacy, Kioicho campus, Josai International University, Tokyo 102-0093, Japan
| | - Jack Cook
- Clinical Pharmacology, Global Product Development, Pfizer Inc., Groton, Connecticut, USA
| | - Joseph W. Polli
- Global Medical Sciences, ViiV Healthcare, Research Triangle Park NC USA
| | - Imad Hanna
- Pharmacokinetic Sciences-Oncology, Novartis Institute for Biomedical Research, East Hanover, NJ
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc. Foster City, CA USA
| | | |
Collapse
|
18
|
Peng J, Ladumor MK, Unadkat JD. Estimation of Fetal-to-Maternal Unbound Steady-State Plasma Concentration Ratio of P-Glycoprotein and/or Breast Cancer Resistance Protein Substrate Drugs Using a Maternal-Fetal Physiologically Based Pharmacokinetic Model. Drug Metab Dispos 2022; 50:613-623. [PMID: 35149540 PMCID: PMC9073947 DOI: 10.1124/dmd.121.000733] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/18/2022] [Indexed: 11/22/2022] Open
Abstract
Pregnant women are frequently prescribed drugs to treat chronic diseases such as human immunodeficiency virus infection, but little is known about the benefits and risks of these drugs to the fetus that are driven by fetal drug exposure. The latter can be estimated by fetal-to-maternal unbound plasma concentration at steady state (Kp,uu,fetal). For drugs that are substrates of placental efflux transporters [i.e., P-glycoprotein (P-gp) or breast cancer resistance protein (BCRP)], Kp,uu,fetal is expected to be <1. Here, we estimated the in vivo Kp,uu,fetal of selective P-gp and BCRP substrate drugs by maternal-fetal physiologically based pharmacokinetic (m-f-PBPK) modeling of umbilical vein (UV) plasma and maternal plasma (MP) concentrations obtained simultaneously at term from multiple maternal-fetal dyads. To do so, three drugs were selected: nelfinavir (P-gp substrate), efavirenz (BCRP substrate), and imatinib (P-gp/BCRP substrate). An m-f-PBPK model for each drug was developed and validated for the nonpregnant population and pregnant women using the Simcyp simulator (v20). Then, after incorporating placental passive diffusion clearance, the in vivo Kp,uu,fetal of the drug was estimated by adjusting the placental efflux clearance until the predicted UV/MP values best matched the observed data (Kp,uu,fetal) of nelfinavir = 0.41, efavirenz = 0.39, and imatinib = 0.35. Furthermore, Kp,uu,fetal of nelfinavir and efavirenz at gestational weeks (GWs) 25 and 15 were predicted to be 0.34 and 0.23 (GW25) and 0.33 and 0.27 (GW15). These Kp,uu,fetal values can be used to adjust dosing regimens of these drugs to optimize maternal-fetal drug therapy throughout pregnancy, to assess fetal benefits and risks of these dosing regimens, and to determine if these estimated in vivo Kp,uu,fetal values can be predicted from in vitro studies. SIGNIFICANCE STATEMENT: The in vivo fetal-to-maternal unbound steady-state plasma concentration ratio (Kp,uu,fetal) of nelfinavir [P-glycoprotein (P-gp) substrate], efavirenz [breast cancer resistance protein (BCRP) substrate], and imatinib (P-gp and BCRP substrate) was successfully estimated using maternal-fetal physiologically based pharmacokinetic (m-f-PBPK) modeling. These Kp,uu,fetal values can be used to adjust dosing regimens of these drugs to optimize maternal-fetal drug therapy throughout pregnancy, to assess fetal benefits and risks of these dosing regimens, and to determine if these estimated in vivo Kp,uu,fetal values can be predicted from in vitro studies.
Collapse
Affiliation(s)
- Jinfu Peng
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (J.P., M.K.L., J.D.U.) and Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China (J.P.)
| | - Mayur K Ladumor
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (J.P., M.K.L., J.D.U.) and Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China (J.P.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (J.P., M.K.L., J.D.U.) and Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China (J.P.)
| |
Collapse
|
19
|
Zheng Q, Zhang B, Li C, Zhang X. Overcome Drug Resistance in Cholangiocarcinoma: New Insight Into Mechanisms and Refining the Preclinical Experiment Models. Front Oncol 2022; 12:850732. [PMID: 35372014 PMCID: PMC8970309 DOI: 10.3389/fonc.2022.850732] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/14/2022] [Indexed: 11/19/2022] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive tumor characterized by a poor prognosis. Therapeutic options are limited in patients with advanced stage of CCA, as a result of the intrinsic or acquired resistance to currently available chemotherapeutic agents, and the lack of new drugs entering into clinical application. The challenge in translating basic research to the clinical setting, caused by preclinical models not being able to recapitulate the tumor characteristics of the patient, seems to be an important reason for the lack of effective and specific therapies for CCA. So, there seems to be two ways to improve patient outcomes. The first one is developing the combination therapies based on a better understanding of the mechanisms contributing to the resistance to currently available chemotherapeutic agents. The second one is developing novel preclinical experimental models that better recapitulate the genetic and histopathological features of the primary tumor, facilitating the screening of new drugs for CCA patients. In this review, we discussed the evidence implicating the mechanisms underlying treatment resistance to currently investigated drugs, and the development of preclinical experiment models for CCA.
Collapse
Affiliation(s)
- Qingfan Zheng
- Department of Hepatobiliary and Pancreas Surgery, the Second Hospital of Jilin University, Changchun, China
| | - Bin Zhang
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xuewen Zhang
- Department of Hepatobiliary and Pancreas Surgery, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
20
|
Boichuk S, Dunaev P, Mustafin I, Mani S, Syuzov K, Valeeva E, Bikinieva F, Galembikova A. Infigratinib (BGJ 398), a Pan-FGFR Inhibitor, Targets P-Glycoprotein and Increases Chemotherapeutic-Induced Mortality of Multidrug-Resistant Tumor Cells. Biomedicines 2022; 10:biomedicines10030601. [PMID: 35327403 PMCID: PMC8945560 DOI: 10.3390/biomedicines10030601] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 02/01/2023] Open
Abstract
The microtubule-targeting agents (MTAs) are well-known chemotherapeutic agents commonly used for therapy of a broad spectrum of human malignancies, exhibiting epithelial origin, including breast, lung, and prostate cancer. Despite the impressive response rates shortly after initiation of MTA-based therapy, the vast majority of human malignancies develop resistance to MTAs due to the different mechanisms. Here, we report that infigratinib (BGJ 398), a potent FGFR1-4 inhibitor, restores sensitivity of a broad spectrum of ABCB1-overexpressing cancer cells to certain chemotherapeutic agents, including paclitaxel (PTX) and doxorubicin (Dox). This was evidenced for the triple-negative breast cancer (TNBC), and gastrointestinal stromal tumor (GIST) cell lines, as well. Indeed, when MDR-overexpressing cancer cells were treated with a combination of BGJ 398 and PTX (or Dox), we observed a significant increase of apoptosis which was evidenced by an increased expression of cleaved forms of PARP, caspase-3, and increased numbers of Annexin V-positive cells, as well. Moreover, BGJ 398 used in combination with PTX significantly decreased the viability and proliferation of the resistant cancer cells. As expected, no apoptosis was found in ABCB1-overexpressing cancer cells treated with PTX, Dox, or BGJ 398 alone. Inhibition of FGFR-signaling by BGJ 398 was evidenced by the decreased expression of phosphorylated (i.e., activated) forms of FGFR and FRS-2, a well-known adaptor protein of FGFR signaling, and downstream signaling molecules (e.g., STAT-1, -3, and S6). In contrast, expression of MDR-related ABC-transporters did not change after BGJ 398 treatment, thereby suggesting an impaired function of MDR-related ABC-transporters. By using the fluorescent-labeled chemotherapeutic agent PTX-Alexa488 (Flutax-2) and doxorubicin, exhibiting an intrinsic fluorescence, we found that BGJ 398 substantially impairs their efflux from MDR-overexpressing TNBC cells. Moreover, the efflux of Calcein AM, a well-known substrate for ABCB1, was also significantly impaired in BGJ 398-treated cancer cells, thereby suggesting the ABCB1 as a novel molecular target for BGJ 398. Of note, PD 173074, a potent FGFR1 and VEGFR2 inhibitor failed to retain chemotherapeutic agents inside ABCB1-overexpressing cells. This was consistent with the inability of PD 173074 to sensitize Tx-R cancer cells to PTX and Dox. Collectively, we show here for the first time that BGJ 398 reverses the sensitivity of MDR-overexpressing cancer cells to certain chemotherapeutic agents due to inhibition of their efflux from cancer cells via ABCB1-mediated mechanism.
Collapse
Affiliation(s)
- Sergei Boichuk
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (P.D.); (S.M.); (K.S.); (F.B.); (A.G.)
- Сentral Research Laboratory, Kazan State Medical University, 420012 Kazan, Russia;
- Department of Radiotherapy and Radiology, Faculty of Surgery, Russian Medical Academy of Continuous Professional Education, 125993 Moscow, Russia
- Correspondence: ; Tel.: +7-917-397-80-93; Fax: +7-843-236-06-52
| | - Pavel Dunaev
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (P.D.); (S.M.); (K.S.); (F.B.); (A.G.)
| | - Ilshat Mustafin
- Department of Biochemistry, Kazan State Medical University, 420012 Kazan, Russia;
| | - Shinjit Mani
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (P.D.); (S.M.); (K.S.); (F.B.); (A.G.)
| | - Kirill Syuzov
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (P.D.); (S.M.); (K.S.); (F.B.); (A.G.)
| | - Elena Valeeva
- Сentral Research Laboratory, Kazan State Medical University, 420012 Kazan, Russia;
| | - Firuza Bikinieva
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (P.D.); (S.M.); (K.S.); (F.B.); (A.G.)
| | - Aigul Galembikova
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (P.D.); (S.M.); (K.S.); (F.B.); (A.G.)
| |
Collapse
|
21
|
Transport and metabolism of tyrosine kinase inhibitors associated with chronic myeloid leukemia therapy: a review. Mol Cell Biochem 2022; 477:1261-1279. [DOI: 10.1007/s11010-022-04376-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 01/27/2022] [Indexed: 12/14/2022]
|
22
|
A Phenylfurocoumarin Derivative Reverses ABCG2-Mediated Multidrug Resistance In Vitro and In Vivo. Int J Mol Sci 2021; 22:ijms222212502. [PMID: 34830383 PMCID: PMC8618058 DOI: 10.3390/ijms222212502] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 11/17/2022] Open
Abstract
The ATP-binding cassette subfamily G member 2 (ABCG2) transporter is involved in the development of multidrug resistance in cancer patients. Many inhibitors of ABCG2 have been reported to enhance the chemosensitivity of cancer cells. However, none of these inhibitors are being used clinically. The aim of this study was to identify novel ABCG2 inhibitors by high-throughput screening of a chemical library. Among the 5812 compounds in the library, 23 compounds were selected in the first screening, using a fluorescent plate reader-based pheophorbide a (PhA) efflux assay. Thereafter, to validate these compounds, a flow cytometry-based PhA efflux assay was performed and 16 compounds were identified as potential inhibitors. A cytotoxic assay was then performed to assess the effect these 16 compounds had on ABCG2-mediated chemosensitivity. We found that the phenylfurocoumarin derivative (R)-9-(3,4-dimethoxyphenyl)-4-((3,3-dimethyloxiran-2-yl)methoxy)-7H-furo [3,2-g]chromen-7-one (PFC) significantly decreased the IC50 of SN-38 in HCT-116/BCRP colon cancer cells. In addition, PFC stimulated ABCG2-mediated ATP hydrolysis, suggesting that this compound interacts with the substrate-binding site of ABCG2. Furthermore, PFC reversed the resistance to irinotecan without causing toxicity in the ABCG2-overexpressing HCT-116/BCRP cell xenograft mouse model. In conclusion, PFC is a novel inhibitor of ABCG2 and has promise as a therapeutic to overcome ABCG2-mediated MDR, to improve the efficiency of cancer chemotherapy.
Collapse
|
23
|
Wang JQ, Wu ZX, Yang Y, Teng QX, Li YD, Lei ZN, Jani KA, Kaushal N, Chen ZS. ATP-binding cassette (ABC) transporters in cancer: A review of recent updates. J Evid Based Med 2021; 14:232-256. [PMID: 34388310 DOI: 10.1111/jebm.12434] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023]
Abstract
The ATP-binding cassette (ABC) transporter superfamily is one of the largest membrane protein families existing in wide spectrum of organisms from prokaryotes to human. ABC transporters are also known as efflux pumps because they mediate the cross-membrane transportation of various endo- and xenobiotic molecules energized by ATP hydrolysis. Therefore, ABC transporters have been considered closely to multidrug resistance (MDR) in cancer, where the efflux of structurally distinct chemotherapeutic drugs causes reduced itherapeutic efficacy. Besides, ABC transporters also play other critical biological roles in cancer such as signal transduction. During the past decades, extensive efforts have been made in understanding the structure-function relationship, transportation profile of ABC transporters, as well as the possibility to overcome MDR via targeting these transporters. In this review, we discuss the most recent knowledge regarding ABC transporters and cancer drug resistance in order to provide insights for the development of more effective therapies.
Collapse
Affiliation(s)
- Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Yi-Dong Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
- School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Khushboo A Jani
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Neeraj Kaushal
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| |
Collapse
|
24
|
Meriggi F. Controversial link between proton pump inhibitors and anticancer agents: review of the literature. TUMORI JOURNAL 2021; 108:204-212. [PMID: 34159850 DOI: 10.1177/03008916211025091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Drug-drug interactions represent a topic of great interest, not only due to the risk of unexpected adverse events but also due to the possibility of altering the effectiveness of a specific treatment. Inappropriate or concomitant use of drugs can often lead to changes in the bioavailability of various compounds, resulting in pharmacokinetic alterations. A recent example is the concomitant administration of proton pump inhibitors (PPIs) and anticancer agents. PPIs are overused beyond their classic indications, resulting in a high risk of interactions with other drugs, such as anticancer agents, both PO and intravenous. However, the real clinical impact of concomitant acid suppression therapy and anticancer therapies remains controversial and is not yet fully understood. Certainly, the gut microbiota plays a key role in regulating the response of the immune system, and PPIs can significantly alter the gut microbiome, resulting in gut dysbiosis. Indeed, while the link sometimes appears to lead to negative outcomes, as in the case of immunotherapy, oral capecitabine, or tyrosine kinase inhibitors, in other cases, it seems to enhance the effectiveness of intravenous chemotherapy. In this review, I analyse the possible drug interactions between PPIs and the main classes of anticancer drugs.
Collapse
Affiliation(s)
- Fausto Meriggi
- Oncology Department, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| |
Collapse
|
25
|
Ibrahim MAA, Badr EAA, Abdelrahman AHM, Almansour NM, Shawky AM, Mekhemer GAH, Alrumaihi F, Moustafa MF, Atia MAM. Prospective Drug Candidates as Human Multidrug Transporter ABCG2 Inhibitors: an In Silico Drug Discovery Study. Cell Biochem Biophys 2021; 79:189-200. [PMID: 33954893 DOI: 10.1007/s12013-021-00985-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2021] [Indexed: 12/22/2022]
Abstract
Breast cancer resistance protein (ABCG2) is a human ATP-binding cassette (ABC) that plays a paramount role in multidrug resistance (MDR) in cancer therapy. The discovery of ABCG2 inhibitors could assist in designing unprecedented therapeutic strategies for cancer treatment. There is as yet no approved drug targeting ABCG2, although a large number of drug candidates have been clinically investigated. In this work, binding affinities of 181 drug candidates in clinical-trial or investigational stages as ABCG2 inhibitors were inspected using in silico techniques. Based on available experimental data, the performance of AutoDock4.2.6 software was first validated to predict the inhibitor-ABCG2 binding mode and affinity. Combined molecular docking calculations and molecular dynamics (MD) simulations, followed by molecular mechanics-generalized Born surface area (MM-GBSA) binding energy calculations, were then performed to filter out the studied drug candidates. From the estimated docking scores and MM-GBSA binding energies, six auspicious drug candidates-namely, pibrentasvir, venetoclax, ledipasvir, avatrombopag, cobicistat, and revefenacin-exhibited auspicious binding energies with value < -70.0 kcal/mol. Interestingly, pibrentasvir, venetoclax, and ledipasvir were observed to show even higher binding affinities with the ABCG2 transporter with binding energies of < -80.0 kcal/mol over long MD simulations of 100 ns. The stabilities of these three promising candidates in complex with ABCG2 transporter were demonstrated by their energetics and structural analyses throughout the 100 ns MD simulations. The current study throws new light on pibrentasvir, venetoclax, and ledipasvir as curative options for multidrug resistant cancers by inhibiting ABCG2 transporter.
Collapse
Affiliation(s)
- Mahmoud A A Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt.
| | - Esraa A A Badr
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt
| | - Alaa H M Abdelrahman
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt
| | - Nahlah Makki Almansour
- Department of Biology, College of Science, University of Hafr Al Batin, Hafr Al Batin, Saudi Arabia
| | - Ahmed M Shawky
- Science and Technology Unit (STU), Umm Al-Qura University, Makkah, Saudi Arabia
| | - Gamal A H Mekhemer
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Mahmoud F Moustafa
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
- Department of Botany & Microbiology, Faculty of Science, South Valley University, Qena, Egypt
| | - Mohamed A M Atia
- Molecular Genetics and Genome Mapping Laboratory, Genome Mapping Department, Agricultural Genetic Engineering Research Institute (AGERI), Agricultural Research Center (ARC), Giza, Egypt.
| |
Collapse
|
26
|
Ma XY, Wei L, Lei Z, Chen Y, Ding Z, Chen ZS. Recent progress on targeting leukemia stem cells. Drug Discov Today 2021; 26:1904-1913. [PMID: 34029689 DOI: 10.1016/j.drudis.2021.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/14/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022]
Abstract
Leukemia is a type of malignant clonal disease of hematopoietic stem cells (HSCs). A small population of leukemic stem cells (LSCs) are responsible for the initiation, drug resistance, and relapse of leukemia. LSCs have the ability to form tumors after xenotransplantation in immunodeficient mice and appear to be common in most human leukemias. Therefore, the eradication of LSCs is an approach with the potential to improve survival or even to cure leukemia. Using recent research in the field of LSCs, we summarize the targeted therapy approaches for the removal of LSCs through surface markers including immune checkpoint molecules, pathways influencing LSC survival, or the survival microenvironment of LSCs. In addition, we introduce the survival microenvironment and survival regulation of LSCs.
Collapse
Affiliation(s)
- Xiang-Yu Ma
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China
| | - Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China.
| | - Zining Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Yanglu Chen
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Zhiyong Ding
- Mills Institute for Personalized Cancer Care, Fynn Biotechnologies Ltd., Gangxing 3rd Rd, High-Tech and Innovation Zone, Jinan, Shandong 250101, PR China
| | - Zhe-Sheng Chen
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China.
| |
Collapse
|
27
|
Park JW, Chung H, Kim KA, Kim JM, Park IH, Lee S, Park JY. ABCG2 Single Nucleotide Polymorphism Affects Imatinib Pharmacokinetics in Lower Alpha-1-Acid Glycoprotein Levels in Humans. Front Pharmacol 2021; 12:658039. [PMID: 33995081 PMCID: PMC8116740 DOI: 10.3389/fphar.2021.658039] [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: 01/25/2021] [Accepted: 04/06/2021] [Indexed: 11/21/2022] Open
Abstract
Imatinib is transported extracellularly by ABCB1 and ABCG2 efflux transporters and bound to alpha-1-acid glycoprotein (AGP) in the bloodstream. However, the clinical and pharmacokinetic effects of ABCB1 and ABCG2 on imatinib were inconsistent in the previous literature and have not been confirmed. Therefore, in the present study, we explored the effects of the ABCG2 and ABCB1 genetic polymorphisms on imatinib pharmacokinetics in association with plasma AGP levels in healthy subjects. Twenty-seven healthy individuals were recruited, genotyped for ABCG2 and ABCB1, and given a single oral dose of 400 mg imatinib. Plasma imatinib concentrations were measured and its pharmacokinetics was assessed with respect to ABCG2 (c.421C>A and c.34G>A) and ABCB1 (c.1236C>T, c.2677C>T/A, and c.3435C>T) genotypes, and plasma AGP levels. AGP levels showed a strong positive correlation with imatinib pharmacokinetics. ABCG2 c.421C>A single nucleotide polymorphism showed a statistically significant effect on imatinib pharmacokinetics in low plasma AGP levels groups (<80 mg/dl); subjects with high plasma AGP levels (n = 5, ≥80 mg/dl) were excluded. The results indicate that plasma AGP levels and ABCG2 polymorphisms modulated imatinib pharmacokinetics; however, the effects of the ABCG2 transporter was masked at high plasma AGP levels.
Collapse
Affiliation(s)
- Jin-Woo Park
- Department of Clinical Pharmacology and Toxicology, Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Hyewon Chung
- Department of Clinical Pharmacology and Toxicology, Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Kyoung-Ah Kim
- Department of Clinical Pharmacology and Toxicology, Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Jong-Min Kim
- Department of Clinical Pharmacology and Toxicology, Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - In-Hwan Park
- Department of Clinical Pharmacology and Toxicology, Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Sangjin Lee
- Department of Clinical Pharmacology and Toxicology, Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Ji-Young Park
- Department of Clinical Pharmacology and Toxicology, Anam Hospital, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
28
|
Medically Important Alterations in Transport Function and Trafficking of ABCG2. Int J Mol Sci 2021; 22:ijms22062786. [PMID: 33801813 PMCID: PMC8001156 DOI: 10.3390/ijms22062786] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
Several polymorphisms and mutations in the human ABCG2 multidrug transporter result in reduced plasma membrane expression and/or diminished transport function. Since ABCG2 plays a pivotal role in uric acid clearance, its malfunction may lead to hyperuricemia and gout. On the other hand, ABCG2 residing in various barrier tissues is involved in the innate defense mechanisms of the body; thus, genetic alterations in ABCG2 may modify the absorption, distribution, excretion of potentially toxic endo- and exogenous substances. In turn, this can lead either to altered therapy responses or to drug-related toxic reactions. This paper reviews the various types of mutations and polymorphisms in ABCG2, as well as the ways how altered cellular processing, trafficking, and transport activity of the protein can contribute to phenotypic manifestations. In addition, the various methods used for the identification of the impairments in ABCG2 variants and the different approaches to correct these defects are overviewed.
Collapse
|
29
|
Uchiyama AAT, Silva PAIA, Lopes MSM, Yen CT, Ricardo ED, Mutão T, Pimenta JR, Machado LM, Shimba DS, Peixoto RD. Proton Pump Inhibitors and Oncologic Treatment Efficacy: A Practical Review of the Literature for Oncologists. Curr Oncol 2021; 28:783-799. [PMID: 33546228 PMCID: PMC7985775 DOI: 10.3390/curroncol28010076] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 12/29/2020] [Accepted: 01/07/2021] [Indexed: 12/19/2022] Open
Abstract
Proton pump inhibitors (PPIs) are the most commonly used anti-acid drugs worldwide, including among cancer patients. However, drug-drug interactions between PPIs and other agents may lead to decreased drug absorption with possible reduced therapeutic benefit, or even increased toxicity. Unfortunately, only scarce data exist regarding the safety of concomitant PPI use with anti-cancer agents. We aim at reviewing current evidence on this possible interaction by dividing anti-cancer agents by class. Until further data is available, we encourage healthcare providers to limit unnecessary PPI overuse.
Collapse
Affiliation(s)
- Angel A. T. Uchiyama
- Centro Especializado em Oncologia, Hospital Alemão Oswaldo Cruz, São Paulo 01327-001, Brazil; (A.A.T.U.); (P.A.I.A.S.); (M.S.M.L.); (C.T.Y.); (E.D.R.); (T.M.); (J.R.P.); (L.M.M.); (D.S.S.)
| | - Pedro A. I. A. Silva
- Centro Especializado em Oncologia, Hospital Alemão Oswaldo Cruz, São Paulo 01327-001, Brazil; (A.A.T.U.); (P.A.I.A.S.); (M.S.M.L.); (C.T.Y.); (E.D.R.); (T.M.); (J.R.P.); (L.M.M.); (D.S.S.)
| | - Moisés S. M. Lopes
- Centro Especializado em Oncologia, Hospital Alemão Oswaldo Cruz, São Paulo 01327-001, Brazil; (A.A.T.U.); (P.A.I.A.S.); (M.S.M.L.); (C.T.Y.); (E.D.R.); (T.M.); (J.R.P.); (L.M.M.); (D.S.S.)
| | - Cheng T. Yen
- Centro Especializado em Oncologia, Hospital Alemão Oswaldo Cruz, São Paulo 01327-001, Brazil; (A.A.T.U.); (P.A.I.A.S.); (M.S.M.L.); (C.T.Y.); (E.D.R.); (T.M.); (J.R.P.); (L.M.M.); (D.S.S.)
| | - Eliza D. Ricardo
- Centro Especializado em Oncologia, Hospital Alemão Oswaldo Cruz, São Paulo 01327-001, Brazil; (A.A.T.U.); (P.A.I.A.S.); (M.S.M.L.); (C.T.Y.); (E.D.R.); (T.M.); (J.R.P.); (L.M.M.); (D.S.S.)
| | - Taciana Mutão
- Centro Especializado em Oncologia, Hospital Alemão Oswaldo Cruz, São Paulo 01327-001, Brazil; (A.A.T.U.); (P.A.I.A.S.); (M.S.M.L.); (C.T.Y.); (E.D.R.); (T.M.); (J.R.P.); (L.M.M.); (D.S.S.)
| | - Jefferson R. Pimenta
- Centro Especializado em Oncologia, Hospital Alemão Oswaldo Cruz, São Paulo 01327-001, Brazil; (A.A.T.U.); (P.A.I.A.S.); (M.S.M.L.); (C.T.Y.); (E.D.R.); (T.M.); (J.R.P.); (L.M.M.); (D.S.S.)
| | - Larissa M. Machado
- Centro Especializado em Oncologia, Hospital Alemão Oswaldo Cruz, São Paulo 01327-001, Brazil; (A.A.T.U.); (P.A.I.A.S.); (M.S.M.L.); (C.T.Y.); (E.D.R.); (T.M.); (J.R.P.); (L.M.M.); (D.S.S.)
| | - Denis S. Shimba
- Centro Especializado em Oncologia, Hospital Alemão Oswaldo Cruz, São Paulo 01327-001, Brazil; (A.A.T.U.); (P.A.I.A.S.); (M.S.M.L.); (C.T.Y.); (E.D.R.); (T.M.); (J.R.P.); (L.M.M.); (D.S.S.)
| | - Renata D. Peixoto
- Centro Paulista de Oncologia, Grupo Oncoclínicas, São Paulo 04538-132, Brazil
| |
Collapse
|
30
|
Paris J, Angeli E, Bousquet G. The Pharmacology of Xenobiotics after Intracerebro Spinal Fluid Administration: Implications for the Treatment of Brain Tumors. Int J Mol Sci 2021; 22:1281. [PMID: 33525427 PMCID: PMC7865853 DOI: 10.3390/ijms22031281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 01/16/2023] Open
Abstract
The incidence of brain metastasis has been increasing for 10 years, with poor prognosis, unlike the improvement in survival for extracranial tumor localizations. Since recent advances in molecular biology and the development of specific molecular targets, knowledge of the brain distribution of drugs has become a pharmaceutical challenge. Most anticancer drugs fail to cross the blood-brain barrier. In order to get around this problem and penetrate the brain parenchyma, the use of intrathecal administration has been developed, but the mechanisms governing drug distribution from the cerebrospinal fluid to the brain parenchyma are poorly understood. Thus, in this review we discuss the pharmacokinetics of drugs after intrathecal administration, their penetration of the brain parenchyma and the different systems causing their efflux from the brain to the blood.
Collapse
Affiliation(s)
- Justine Paris
- Institut National de la Santé Et de la Recherche Médicale (INSERM), U942, 9 Rue de Chablis, 93000 Bobigny, France; (J.P.); (E.A.)
| | - Eurydice Angeli
- Institut National de la Santé Et de la Recherche Médicale (INSERM), U942, 9 Rue de Chablis, 93000 Bobigny, France; (J.P.); (E.A.)
- Assistance Publique Hôpitaux de Paris, Avicenne Hospital, Department of Medical Oncology, 93000 Bobigny, France
- Sorbonne Paris Nord University, 99 Avenue Jean Baptiste Clément, 93430 Villetaneuse, France
| | - Guilhem Bousquet
- Institut National de la Santé Et de la Recherche Médicale (INSERM), U942, 9 Rue de Chablis, 93000 Bobigny, France; (J.P.); (E.A.)
- Assistance Publique Hôpitaux de Paris, Avicenne Hospital, Department of Medical Oncology, 93000 Bobigny, France
- Sorbonne Paris Nord University, 99 Avenue Jean Baptiste Clément, 93430 Villetaneuse, France
| |
Collapse
|
31
|
Emami Nejad A, Najafgholian S, Rostami A, Sistani A, Shojaeifar S, Esparvarinha M, Nedaeinia R, Haghjooy Javanmard S, Taherian M, Ahmadlou M, Salehi R, Sadeghi B, Manian M. The role of hypoxia in the tumor microenvironment and development of cancer stem cell: a novel approach to developing treatment. Cancer Cell Int 2021; 21:62. [PMID: 33472628 PMCID: PMC7816485 DOI: 10.1186/s12935-020-01719-5] [Citation(s) in RCA: 361] [Impact Index Per Article: 90.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
Hypoxia is a common feature of solid tumors, and develops because of the rapid growth of the tumor that outstrips the oxygen supply, and impaired blood flow due to the formation of abnormal blood vessels supplying the tumor. It has been reported that tumor hypoxia can: activate angiogenesis, thereby enhancing invasiveness and risk of metastasis; increase survival of tumor, as well as suppress anti-tumor immunity and hamper the therapeutic response. Hypoxia mediates these effects by several potential mechanisms: altering gene expression, the activation of oncogenes, inactivation of suppressor genes, reducing genomic stability and clonal selection. We have reviewed the effects of hypoxia on tumor biology and the possible strategiesto manage the hypoxic tumor microenvironment (TME), highlighting the potential use of cancer stem cells in tumor treatment.
Collapse
Affiliation(s)
- Asieh Emami Nejad
- Department of Biology, Payame Noor University (PNU), P.O.Box 19395-3697, Tehran, Iran
| | - Simin Najafgholian
- Department of Emergency Medicine, School of Medicine , Arak University of Medical Sciences, Arak, Iran
| | - Alireza Rostami
- Department of Surgery, School of Medicine Amiralmomenin Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Alireza Sistani
- Department of Emergency Medicine, School of Medicine Valiasr Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Samaneh Shojaeifar
- Department of Midwifery, Faculty of Nursing and Midwifery , Arak University of Medical Sciences , Arak, Iran
| | - Mojgan Esparvarinha
- Department of Immunology, School of Medicine , Tabriz University of Medical Sciences , Tabriz, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease , Isfahan University of Medical Sciences , Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences , Isfahan, Iran
| | - Marjan Taherian
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Ahmadlou
- Sciences Medical of University Arak, Hospital Amiralmomenin, Center Development Research Clinical, Arak, Iran
| | - Rasoul Salehi
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease , Isfahan University of Medical Sciences , Isfahan, Iran.,Department of Genetics and Molecular Biology, School of Medicine , Isfahan University of Medical Sciences , Isfahan, Iran
| | - Bahman Sadeghi
- Department of Health and Community Medicine, School of Medicine, Arak University of Medical Sciences, Arak, 3848176341, Iran.
| | - Mostafa Manian
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran. .,Department of Medical Laboratory Science, Faculty of Medical Science Kermanshah Branch, Islamic Azad University, Imam Khomeini Campus, Farhikhtegan Bld., Shahid J'afari St., Kermanshah, 3848176341, Iran.
| |
Collapse
|
32
|
Zhang JY, Luo Q, Xu JR, Bai J, Mu LM, Yan Y, Duan JL, Cui YN, Su ZB, Xie Y, Lu WL. Regulating Stem Cell-Related Genes Induces the Plastic Differentiation of Cancer Stem Cells to Treat Breast Cancer. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:396-408. [PMID: 32913889 PMCID: PMC7452009 DOI: 10.1016/j.omto.2020.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/27/2020] [Indexed: 11/02/2022]
Abstract
Relapse of cancer is associated with multidirectional differentiation and unrestricted proliferative replication potential of cancer stem cells. Herein, we propose the plastic differentiation strategy for irreversible differentiation of cancer stem cells; further, salinomycin and its newly constructed functional liposomes are used to implement this strategy. Whole gene, cancer stem cell-related RNA, and protein expression analyses reveal that salinomycin induces the cancer stem cells into normal cells, dormant cells, and mature cancer cells. Besides, the results indicate that the gatekeeper is related to the inhibition of the protein kinase C (PKC) α signaling pathway. The differentiated normal or dormant cells are incorporated into normal tissue, whereas the rest are killed by chemotherapy. The findings would offer the evidence for plastic differentiation of cancer stem cells and propose a novel strategy for cancer therapy.
Collapse
Affiliation(s)
- Jing-Ying Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Qian Luo
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jia-Rui Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jing Bai
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Li-Min Mu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yan Yan
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jia-Lun Duan
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yi-Nuo Cui
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhan-Bo Su
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ying Xie
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wan-Liang Lu
- State Key Laboratory of Natural and Biomimetic Drugs, Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, and School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
33
|
Wu ZX, Yang Y, Wang G, Wang JQ, Teng QX, Sun L, Lei ZN, Lin L, Chen ZS, Zou C. Dual TTK/CLK2 inhibitor, CC-671, selectively antagonizes ABCG2-mediated multidrug resistance in lung cancer cells. Cancer Sci 2020; 111:2872-2882. [PMID: 32478948 PMCID: PMC7419038 DOI: 10.1111/cas.14505] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/13/2022] Open
Abstract
One pivotal factor that leads to multidrug resistance (MDR) is the overexpression of ABCG2. Therefore, tremendous effort has been devoted to the search of effective reversal agents to overcome ABCG2‐mediated MDR. CC‐671 is a potent and selective inhibitor of both TTK (human protein kinase monopolar spindle 1 [hMps1]) and CDC like kinase 2 (CLK2). It represents a new class of cancer therapeutic drugs. In this study, we show that CC‐671 is an effective ABCG2 reversal agent that enhances the efficacy of chemotherapeutic drugs in ABCG2‐overexpressing lung cancer cells. Mechanistic studies show that the reversal effect of CC‐671 is primarily attributed to the inhibition of the drug efflux activity of ABCG2, which leads to an increased intracellular level of chemotherapeutic drugs. In addition, CC‐671 does not alter the protein expression or subcellular localization of ABCG2. The computational molecule docking analysis suggests CC‐671 has high binding affinity to the drug‐binding site of ABCG2. In conclusion, we reveal the interaction between CC‐671 and ABCG2, providing a rationale for the potential combined use of CC‐671 with ABCG2 substrate to overcome MDR.
Collapse
Affiliation(s)
- Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Guangsuo Wang
- Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Lingling Sun
- Cancer Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Lizhu Lin
- Cancer Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Chang Zou
- Shenzhen People's Hospital, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen, China.,Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| |
Collapse
|
34
|
Orlando BJ, Liao M. ABCG2 transports anticancer drugs via a closed-to-open switch. Nat Commun 2020; 11:2264. [PMID: 32385283 PMCID: PMC7210939 DOI: 10.1038/s41467-020-16155-2] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/16/2020] [Indexed: 12/18/2022] Open
Abstract
ABCG2 is an ABC transporter that extrudes a variety of compounds from cells, and presents an obstacle in treating chemotherapy-resistant cancers. Despite recent structural insights, no anticancer drug bound to ABCG2 has been resolved, and the mechanisms of multidrug transport remain obscure. Such a gap of knowledge limits the development of novel compounds that block or evade this critical molecular pump. Here we present single-particle cryo-EM studies of ABCG2 in the apo state, and bound to the three structurally distinct chemotherapeutics. Without the binding of conformation-selective antibody fragments or inhibitors, the resting ABCG2 adopts a closed conformation. Our cryo-EM, biochemical, and functional analyses reveal the binding mode of three chemotherapeutic compounds, demonstrate how these molecules open the closed conformation of the transporter, and establish that imatinib is particularly effective in stabilizing the inward facing conformation of ABCG2. Together these studies reveal the previously unrecognized conformational cycle of ABCG2. ABCG2 is a human ABC transporter that actively extrudes a wide variety of compounds from cells but the mechanisms of multidrug transport remain obscure. Here authors present cryo-EM structures of ABCG2 in the apo state, and bound to the three structurally distinct chemotherapeutics and demonstrate how these molecules open the closed conformation of the transporter.
Collapse
Affiliation(s)
- Benjamin J Orlando
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Maofu Liao
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
35
|
Westerdijk K, Desar IME, Steeghs N, van der Graaf WTA, van Erp NP. Imatinib, sunitinib and pazopanib: From flat-fixed dosing towards a pharmacokinetically guided personalized dose. Br J Clin Pharmacol 2020; 86:258-273. [PMID: 31782166 PMCID: PMC7015742 DOI: 10.1111/bcp.14185] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/21/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are anti‐cancer drugs that target tyrosine kinases, enzymes that are involved in multiple cellular processes. Currently, multiple oral TKIs have been introduced in the treatment of solid tumours, all administered in a fixed dose, although large interpatient pharmacokinetic (PK) variability is described. For imatinib, sunitinib and pazopanib exposure‐treatment outcome (efficacy and toxicity) relationships have been established and therapeutic windows have been defined, therefore dose optimization based on the measured blood concentration, called therapeutic drug monitoring (TDM), can be valuable in increasing efficacy and reducing the toxicity of these drugs. In this review, an overview of the current knowledge on TDM guided individualized dosing of imatinib, sunitinib and pazopanib for the treatment of solid tumours is presented. We summarize preclinical and clinical data that have defined thresholds for efficacy and toxicity. Furthermore, PK models and factors that influence the PK of these drugs which partly explain the interpatient PK variability are summarized. Finally, pharmacological interventions that have been performed to optimize plasma concentrations are described. Based on current literature, we advise which methods should be used to optimize exposure to imatinib, sunitinib and pazopanib.
Collapse
Affiliation(s)
- Kim Westerdijk
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ingrid M E Desar
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek hospital, Amsterdam, the Netherlands
| | - Winette T A van der Graaf
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Medical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek hospital, Amsterdam, the Netherlands
| | - Nielka P van Erp
- Department of Clinical Pharmacy, Radboud University Medical Center, Nijmegen, the Netherlands
| | | |
Collapse
|
36
|
Muselli F, Peyron JF, Mary D. Druggable Biochemical Pathways and Potential Therapeutic Alternatives to Target Leukemic Stem Cells and Eliminate the Residual Disease in Chronic Myeloid Leukemia. Int J Mol Sci 2019; 20:E5616. [PMID: 31717629 PMCID: PMC6888542 DOI: 10.3390/ijms20225616] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/14/2022] Open
Abstract
Chronic Myeloid Leukemia (CML) is a disease arising in stem cells expressing the BCR-ABL oncogenic tyrosine kinase that transforms one Hematopoietic stem/progenitor Cell into a Leukemic Stem Cell (LSC) at the origin of differentiated and proliferating leukemic cells in the bone marrow (BM). CML-LSCs are recognized as being responsible for resistances and relapses that occur despite the advent of BCR-ABL-targeting therapies with Tyrosine Kinase Inhibitors (TKIs). LSCs share a lot of functional properties with Hematopoietic Stem Cells (HSCs) although some phenotypical and functional differences have been described during the last two decades. Subverted mechanisms affecting epigenetic processes, apoptosis, autophagy and more recently metabolism and immunology in the bone marrow microenvironment (BMM) have been reported. The aim of this review is to bring together the modifications and molecular mechanisms that are known to account for TKI resistance in primary CML-LSCs and to focus on the potential solutions that can circumvent these resistances, in particular those that have been, or will be tested in clinical trials.
Collapse
MESH Headings
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Neoplasm, Residual/drug therapy
- Neoplasm, Residual/metabolism
- Neoplasm, Residual/pathology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Protein Kinase Inhibitors/therapeutic use
- Tumor Microenvironment/drug effects
Collapse
Affiliation(s)
| | | | - Didier Mary
- Université Côte d’Azur, Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Centre Méditerranéen de Médecine Moléculaire, CEDEX 3, 06204 Nice, France; (F.M.); (J.-F.P.)
| |
Collapse
|
37
|
Kato Y, Kondo S, Itakura T, Tokunaga M, Hatayama S, Katayama K, Sugimoto Y. SNAIL- and SLUG-induced side population phenotype of HCT116 human colorectal cancer cells and its regulation by BET inhibitors. Biochem Biophys Res Commun 2019; 521:152-157. [PMID: 31653342 DOI: 10.1016/j.bbrc.2019.10.094] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 01/18/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is associated with cancer malignancies such as invasion, metastasis, and drug resistance. In this study, HCT116 human colorectal cancer cells were transduced with SLUG or SNAIL retroviruses, and EMT cells with mesenchymal morphology were established. The EMT cells showed a high invasive activity and resistance to several anticancer agents such as methotrexate, SN-38, and cisplatin. Furthermore, they contained about 1-10% side population (SP) cells that were not stained by Hoechst 33342. This SP phenotype was not stable; the isolated SP cells generated both SP and non-SP cells, suggesting a potential for differentiation. Gene expression analysis of SP cells suggested the alteration of genes that are involved in epigenetic changes. Therefore, we examined the effect of 74 epigenetic inhibitors, and found that two inhibitors, namely I-BET151 and bromosporine, targeting the bromodomain and extra-terminal motif (BET) proteins, decreased the ratio of SP cells to <50% compared with the control, without affecting the immediate efflux of Hoechst 33342 by transporters. In addition, compared with the parental cells, the EMT cells showed a higher sensitivity to I-BET151 and bromosporine. This study suggests that EMT development and SP phenotype can be independent events but both are regulated by BET inhibitors in SLUG- or SNAIL-transducted HCT116 cells.
Collapse
Affiliation(s)
- Yu Kato
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan.
| | - Shingo Kondo
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Taira Itakura
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Miku Tokunaga
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Shiori Hatayama
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Kazuhiro Katayama
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Yoshikazu Sugimoto
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| |
Collapse
|
38
|
Park TE, Mustafaoglu N, Herland A, Hasselkus R, Mannix R, FitzGerald EA, Prantil-Baun R, Watters A, Henry O, Benz M, Sanchez H, McCrea HJ, Goumnerova LC, Song HW, Palecek SP, Shusta E, Ingber DE. Hypoxia-enhanced Blood-Brain Barrier Chip recapitulates human barrier function and shuttling of drugs and antibodies. Nat Commun 2019; 10:2621. [PMID: 31197168 DOI: 10.1101/482463v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/16/2019] [Indexed: 05/21/2023] Open
Abstract
The high selectivity of the human blood-brain barrier (BBB) restricts delivery of many pharmaceuticals and therapeutic antibodies to the central nervous system. Here, we describe an in vitro microfluidic organ-on-a-chip BBB model lined by induced pluripotent stem cell-derived human brain microvascular endothelium interfaced with primary human brain astrocytes and pericytes that recapitulates the high level of barrier function of the in vivo human BBB for at least one week in culture. The endothelium expresses high levels of tight junction proteins and functional efflux pumps, and it displays selective transcytosis of peptides and antibodies previously observed in vivo. Increased barrier functionality was accomplished using a developmentally-inspired induction protocol that includes a period of differentiation under hypoxic conditions. This enhanced BBB Chip may therefore represent a new in vitro tool for development and validation of delivery systems that transport drugs and therapeutic antibodies across the human BBB.
Collapse
Affiliation(s)
- Tae-Eun Park
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
- Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Nur Mustafaoglu
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Anna Herland
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- Swedish Medical Nanoscience Center, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Ryan Hasselkus
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Robert Mannix
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Edward A FitzGerald
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Alexander Watters
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Olivier Henry
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Maximilian Benz
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Henry Sanchez
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Heather J McCrea
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | | | - Hannah W Song
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Eric Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
39
|
Park TE, Mustafaoglu N, Herland A, Hasselkus R, Mannix R, FitzGerald EA, Prantil-Baun R, Watters A, Henry O, Benz M, Sanchez H, McCrea HJ, Goumnerova LC, Song HW, Palecek SP, Shusta E, Ingber DE. Hypoxia-enhanced Blood-Brain Barrier Chip recapitulates human barrier function and shuttling of drugs and antibodies. Nat Commun 2019; 10:2621. [PMID: 31197168 PMCID: PMC6565686 DOI: 10.1038/s41467-019-10588-0] [Citation(s) in RCA: 361] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/16/2019] [Indexed: 12/20/2022] Open
Abstract
The high selectivity of the human blood-brain barrier (BBB) restricts delivery of many pharmaceuticals and therapeutic antibodies to the central nervous system. Here, we describe an in vitro microfluidic organ-on-a-chip BBB model lined by induced pluripotent stem cell-derived human brain microvascular endothelium interfaced with primary human brain astrocytes and pericytes that recapitulates the high level of barrier function of the in vivo human BBB for at least one week in culture. The endothelium expresses high levels of tight junction proteins and functional efflux pumps, and it displays selective transcytosis of peptides and antibodies previously observed in vivo. Increased barrier functionality was accomplished using a developmentally-inspired induction protocol that includes a period of differentiation under hypoxic conditions. This enhanced BBB Chip may therefore represent a new in vitro tool for development and validation of delivery systems that transport drugs and therapeutic antibodies across the human BBB.
Collapse
Affiliation(s)
- Tae-Eun Park
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
- Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan, 44919, Republic of Korea
| | - Nur Mustafaoglu
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Anna Herland
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden
- Swedish Medical Nanoscience Center, Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Ryan Hasselkus
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Robert Mannix
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Edward A FitzGerald
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Alexander Watters
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Olivier Henry
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Maximilian Benz
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Henry Sanchez
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Heather J McCrea
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | | | - Hannah W Song
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Eric Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
40
|
Clerbaux LA, Paini A, Lumen A, Osman-Ponchet H, Worth AP, Fardel O. Membrane transporter data to support kinetically-informed chemical risk assessment using non-animal methods: Scientific and regulatory perspectives. ENVIRONMENT INTERNATIONAL 2019; 126:659-671. [PMID: 30856453 PMCID: PMC6441651 DOI: 10.1016/j.envint.2019.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/10/2019] [Accepted: 03/01/2019] [Indexed: 06/01/2023]
Abstract
Humans are continuously exposed to low levels of thousands of industrial chemicals, most of which are poorly characterised in terms of their potential toxicity. The new paradigm in chemical risk assessment (CRA) aims to rely on animal-free testing, with kinetics being a key determinant of toxicity when moving from traditional animal studies to integrated in vitro-in silico approaches. In a kinetically informed CRA, membrane transporters, which have been intensively studied during drug development, are an essential piece of information. However, how existing knowledge on transporters gained in the drug field can be applied to CRA is not yet fully understood. This review outlines the opportunities, challenges and existing tools for investigating chemical-transporter interactions in kinetically informed CRA without animal studies. Various environmental chemicals acting as substrates, inhibitors or modulators of transporter activity or expression have been shown to impact TK, just as drugs do. However, because pollutant concentrations are often lower in humans than drugs and because exposure levels and internal chemical doses are not usually known in contrast to drugs, new approaches are required to translate transporter data and reasoning from the drug sector to CRA. Here, the generation of in vitro chemical-transporter interaction data and the development of transporter databases and classification systems trained on chemical datasets (and not only drugs) are proposed. Furtheremore, improving the use of human biomonitoring data to evaluate the in vitro-in silico transporter-related predicted values and developing means to assess uncertainties could also lead to increase confidence of scientists and regulators in animal-free CRA. Finally, a systematic characterisation of the transportome (quantitative monitoring of transporter abundance, activity and maintenance over time) would reinforce confidence in the use of experimental transporter/barrier systems as well as in established cell-based toxicological assays currently used for CRA.
Collapse
Affiliation(s)
| | - Alicia Paini
- European Commission, Joint Research Centre, Ispra, Italy.
| | - Annie Lumen
- National Center for Toxicological Research, US Food and Drug Administration (FDA), Jefferson, AR, USA
| | | | - Andrew P Worth
- European Commission, Joint Research Centre, Ispra, Italy
| | - Olivier Fardel
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environment et travail), UMR_S 1085, F-35000 Rennes, France
| |
Collapse
|
41
|
Verboom MC, Kloth JSL, Swen JJ, Sleijfer S, Reyners AKL, Steeghs N, Mathijssen RHJ, Gelderblom H, Guchelaar HJ. Genetic polymorphisms in ABCG2 and CYP1A2 are associated with imatinib dose reduction in patients treated for gastrointestinal stromal tumors. THE PHARMACOGENOMICS JOURNAL 2019; 19:473-479. [PMID: 30713339 DOI: 10.1038/s41397-019-0079-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/24/2018] [Accepted: 12/20/2018] [Indexed: 12/23/2022]
Abstract
Imatinib has a mild toxicity profile, although severe adverse events may develop. In this pharmacogenetic pathway analysis the need for dose reduction and cessation of therapy was tested for an association with single nucleotide polymorphisms (SNPs) in genes related to imatinib pharmacology. Retrospective data from 315 patients with a gastrointestinal stromal tumor who received imatinib 400 mg o.d. was associated with 36 SNPs. SNPs that showed a trend in univariate testing were tested in a multivariate model with clinical factors and correction for multiple testing was performed. Dose reduction was associated with carriership of the A-allele in rs2231137 in ABCG2 (OR 7.35, p = 0.0002) and two C-alleles in rs762551 in CYP1A2 (OR 7.12, p = 0.001). Results remained significant after correction for multiple testing. Therapy cessation did not show an association with any of the tested SNPs. These results may help identifying patients at increased risk for toxicity who could benefit from intensified follow-up.
Collapse
Affiliation(s)
- Michiel C Verboom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Jacqueline S L Kloth
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Anna K L Reyners
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology and Clinical Pharmacology, Antoni van Leeuwenhoek - Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
42
|
Enantioselective Drug Recognition by Drug Transporters. Molecules 2018; 23:molecules23123062. [PMID: 30467304 PMCID: PMC6321737 DOI: 10.3390/molecules23123062] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/21/2018] [Accepted: 11/22/2018] [Indexed: 01/16/2023] Open
Abstract
Drug transporters mediate the absorption, tissue distribution, and excretion of drugs. The cDNAs of P-glycoprotein, multidrug resistance proteins (MRPs/ABCC), breast cancer resistance protein (BCRP/ABCG2), peptide transporters (PEPTs/SLC15), proton-coupled folate transporters (PCFT/SLC46A1), organic anion transporting polypeptides (OATPs/SLCO), organic anion transporters (OATs/SLC22), organic cation transporters (OCTs/SLC22), and multidrug and toxin extrusions (MATEs/SLC47) have been isolated, and their functions have been elucidated. Enantioselectivity has been demonstrated in the pharmacokinetics and efficacy of drugs, and is important for elucidating the relationship with recognition of drugs by drug transporters from a chiral aspect. Enantioselectivity in the transport of drugs by drug transporters and the inhibitory effects of drugs on drug transporters has been summarized in this review.
Collapse
|
43
|
Lu L, Kok CH, Saunders VA, Wang J, McLean JA, Hughes TP, White DL. Modelling ponatinib resistance in tyrosine kinase inhibitor-naïve and dasatinib resistant BCR-ABL1+ cell lines. Oncotarget 2018; 9:34735-34747. [PMID: 30410673 PMCID: PMC6205183 DOI: 10.18632/oncotarget.26187] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/15/2018] [Indexed: 11/25/2022] Open
Abstract
TKI resistance remains a major impediment to successful treatment of CML. In this study, we investigated the emerging modes of ponatinib resistance in TKI-naïve and dasatinib resistant BCR-ABL1+ cell lines. To investigate potential resistance mechanisms, ponatinib resistance was generated in BCR-ABL1+ cell-lines by long-term exposure to increasing concentrations of ponatinib. Two cell lines with prior dasatinib resistance demonstrated BCR-ABL1 kinase domain (KD) mutation(s) upon exposure to ponatinib. In one of these cell lines the T315I mutation had emerged during dasatinib exposure. When further cultured with ponatinib, the T315I mutation level and BCR-ABL1 mRNA expression level were increased. In the other cell line, compound mutations G250E/E255K developed with ponatinib exposure. In contrast, the ponatinib resistant cell lines that had no prior exposure to other TKIs (TKI-naïve) did not develop BCR-ABL1 KD mutations. Rather, both of these cell lines demonstrated Bcr-Abl-independent resistance via Axl overexpression. Axl, a receptor tyrosine kinase, has previously been associated with imatinib and nilotinib resistance. Ponatinib sensitivity was restored following Axl inhibition or shRNA-mediated-knockdown of Axl, suggesting that Axl was the primary driver of resistance and a potential target for therapy in this setting.
Collapse
Affiliation(s)
- Liu Lu
- South Australian Health and Medical Research Institute (SAHMRI), Cancer Theme, Adelaide, SA, Australia.,School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Chung Hoow Kok
- South Australian Health and Medical Research Institute (SAHMRI), Cancer Theme, Adelaide, SA, Australia.,School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Verity Ann Saunders
- South Australian Health and Medical Research Institute (SAHMRI), Cancer Theme, Adelaide, SA, Australia
| | - Jueqiong Wang
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Jennifer Anne McLean
- South Australian Health and Medical Research Institute (SAHMRI), Cancer Theme, Adelaide, SA, Australia
| | - Timothy Peter Hughes
- South Australian Health and Medical Research Institute (SAHMRI), Cancer Theme, Adelaide, SA, Australia.,School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Department of Haematology, SA Pathology, Adelaide, SA, Australia
| | - Deborah Lee White
- South Australian Health and Medical Research Institute (SAHMRI), Cancer Theme, Adelaide, SA, Australia.,School of Medicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,Discipline of Paediatrics, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia.,School of Biological Sciences, Faculty of Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
44
|
Abstract
Transporters play important roles in absorption, distribution, metabolism, and elimination (ADME) processes, as well as drug pharmacokinetics (PK) and pharmacodynamics (PD). They are also important in maintaining the homeostasis of endogenous compounds and nutrients in the body. Increasing evidences also suggest that they are important in mediating drug-drug interactions (DDIs). While the significance of transporters in drug pharmacodynamics and DDIs are beyond the scope of this overview, the basic concepts of transporters, their contributions in membrane permeation processes, and their roles in influencing drug ADME pathway and PK will be discussed. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Yan Zhang
- Drug Metabolism Pharmacokinetics & Clinical Pharmacology, Incyte Corporation, Wilmington, Delaware
| |
Collapse
|
45
|
Chen KG, Mallon BS, Park K, Robey PG, McKay RDG, Gottesman MM, Zheng W. Pluripotent Stem Cell Platforms for Drug Discovery. Trends Mol Med 2018; 24:805-820. [PMID: 30006147 DOI: 10.1016/j.molmed.2018.06.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/13/2018] [Accepted: 06/20/2018] [Indexed: 12/30/2022]
Abstract
Use of human pluripotent stem cells (hPSCs) and their differentiated derivatives have led to recent proof-of-principle drug discoveries, defining a pathway to the implementation of hPSC-based drug discovery (hPDD). Current hPDD strategies, however, have inevitable conceptual biases and technological limitations, including the dimensionality of cell-culture methods, cell maturity and functionality, experimental variability, and data reproducibility. In this review, we dissect representative hPDD systems via analysis of hPSC-based 2D-monolayers, 3D culture, and organoids. We discuss mechanisms of drug discovery and drug repurposing, and roles of membrane drug transporters in tissue maturation and hPDD using the example of drugs that target various mutations of CFTR, the cystic fibrosis transmembrane conductance regulator gene, in patients with cystic fibrosis.
Collapse
Affiliation(s)
- Kevin G Chen
- NIH Stem Cell Characterization Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Barbara S Mallon
- NIH Stem Cell Characterization Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kyeyoon Park
- NIH Stem Cell Characterization Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Pamela G Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ronald D G McKay
- The Lieber Institute for Brain Development, Baltimore, MD 21205, USA
| | - Michael M Gottesman
- The Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| |
Collapse
|
46
|
Nath A, Wang J, Stephanie Huang R. Pharmacogenetics and Pharmacogenomics of Targeted Therapeutics in Chronic Myeloid Leukemia. Mol Diagn Ther 2018; 21:621-631. [PMID: 28698977 DOI: 10.1007/s40291-017-0292-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The advent of targeted therapeutics has greatly improved outcomes of chronic myeloid leukemia (CML) patients. Despite increased efficacy and better clinical responses over cytotoxic chemotherapies, many patients receiving targeted drugs exhibit a poor initial response, develop drug resistance, or undergo relapse after initial success. This inter-individual variation in response has heightened the interest in studying pharmacogenetics and pharmacogenomics (PGx) of cancer drugs. In this review, we discuss the influence of various germline and somatic factors on targeted drug response in CML. Specifically, we examine the role of genetic variants in drug metabolism genes, i.e. CYP3A family genes, and drug transporters, i.e. ABC and SLC family genes. Additionally, we focus on acquired somatic variations in BCR-ABL1, and the potential role played by additional downstream signaling pathways, in conferring resistance to targeted drugs in CML. This review highlights the importance of PGx of targeted therapeutics and its potential application to improving treatment decisions and patient outcomes.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cytochrome P-450 CYP3A/genetics
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Fusion Proteins, bcr-abl/genetics
- Glucuronosyltransferase/genetics
- Humans
- Inactivation, Metabolic/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Organic Cation Transporter 1/genetics
- Pharmacogenetics
- Protein Kinase Inhibitors/therapeutic use
Collapse
Affiliation(s)
- Aritro Nath
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Jacqueline Wang
- Biological Sciences Collegiate Division, The University of Chicago, Chicago, IL, USA
| | - R Stephanie Huang
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
47
|
Stefan SM, Wiese M. Small-molecule inhibitors of multidrug resistance-associated protein 1 and related processes: A historic approach and recent advances. Med Res Rev 2018; 39:176-264. [DOI: 10.1002/med.21510] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/05/2018] [Accepted: 04/28/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Sven Marcel Stefan
- Pharmaceutical Institute; Rheinische Friedrich-Wilhelms-University; Bonn Germany
| | - Michael Wiese
- Pharmaceutical Institute; Rheinische Friedrich-Wilhelms-University; Bonn Germany
| |
Collapse
|
48
|
Bircsak KM, Moscovitz JE, Wen X, Archer F, Yuen PYS, Mohammed M, Memon N, Weinberger BI, Saba LM, Vetrano AM, Aleksunes LM. Interindividual Regulation of the Breast Cancer Resistance Protein/ ABCG2 Transporter in Term Human Placentas. Drug Metab Dispos 2018; 46:619-627. [PMID: 29386232 PMCID: PMC5896368 DOI: 10.1124/dmd.117.079228] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 01/25/2018] [Indexed: 01/16/2023] Open
Abstract
The breast cancer resistance protein (BCRP/ABCG2) is a maternally-facing efflux transporter that regulates the placental disposition of chemicals. Transcription factors and gene variants are important regulatory factors that influence transporter expression. In this study, we sought to identify the genetic and transcriptional mechanisms underlying the interindividual expression of BCRP mRNA and protein across 137 term placentas from uncomplicated pregnancies. Placental expression of BCRP and regulatory transcription factor mRNAs was measured using multiplex-branched DNA analysis. BCRP expression and ABCG2 genotypes were determined using Western blot and Fluidigm Biomark genetic analysis, respectively. Placentas were obtained from a racially and ethnically diverse population, including Caucasian (33%), African American (14%), Asian (14%), Hispanic (15%), and mixed (16%) backgrounds, as well as unknown origins (7%). Between placentas, BCRP mRNA and protein varied up to 47-fold and 14-fold, respectively. In particular, BCRP mRNA correlated significantly with known transcription factor mRNAs, including nuclear factor erythroid 2-related factor 2 and aryl hydrocarbon receptor. Somewhat surprisingly, single-nucleotide polymorphisms (SNPs) in the ABCG2 noncoding regions were not associated with variation in placental BCRP mRNA or protein. Instead, the coding region polymorphism (C421A/Q141K) corresponded with 40%-50% lower BCRP protein in 421C/A and 421A/A placentas compared with wild types (421C/C). Although BCRP protein and mRNA expression weakly correlated (r = 0.25, P = 0.040), this relationship was absent in individuals expressing the C421A variant allele. Study results contribute to our understanding of the interindividual regulation of BCRP expression in term placentas and may help to identify infants at risk for increased fetal exposure to chemicals due to low expression of this efflux protein.
Collapse
Affiliation(s)
- Kristin M Bircsak
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (K.M.B., J.E.M., X.W., L.M.A.), Environmental and Occupational Health Sciences Institute (L.M.A.), and Lipid Center (L.M.A.), Rutgers, The State University of New Jersey, Piscataway, New Jersey; Department of Pediatrics, Rutgers University Robert Wood Johnson Medical School, New Brunswick, New Jersey (F.A., P.Y.S.Y., M.M., N.M., A.M.V.); Hofstra Northwell School of Medicine, Cohen Children's Medical Center of New York, New Hyde Park, New York (B.I.W.); and Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (L.M.S.)
| | - Jamie E Moscovitz
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (K.M.B., J.E.M., X.W., L.M.A.), Environmental and Occupational Health Sciences Institute (L.M.A.), and Lipid Center (L.M.A.), Rutgers, The State University of New Jersey, Piscataway, New Jersey; Department of Pediatrics, Rutgers University Robert Wood Johnson Medical School, New Brunswick, New Jersey (F.A., P.Y.S.Y., M.M., N.M., A.M.V.); Hofstra Northwell School of Medicine, Cohen Children's Medical Center of New York, New Hyde Park, New York (B.I.W.); and Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (L.M.S.)
| | - Xia Wen
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (K.M.B., J.E.M., X.W., L.M.A.), Environmental and Occupational Health Sciences Institute (L.M.A.), and Lipid Center (L.M.A.), Rutgers, The State University of New Jersey, Piscataway, New Jersey; Department of Pediatrics, Rutgers University Robert Wood Johnson Medical School, New Brunswick, New Jersey (F.A., P.Y.S.Y., M.M., N.M., A.M.V.); Hofstra Northwell School of Medicine, Cohen Children's Medical Center of New York, New Hyde Park, New York (B.I.W.); and Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (L.M.S.)
| | - Faith Archer
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (K.M.B., J.E.M., X.W., L.M.A.), Environmental and Occupational Health Sciences Institute (L.M.A.), and Lipid Center (L.M.A.), Rutgers, The State University of New Jersey, Piscataway, New Jersey; Department of Pediatrics, Rutgers University Robert Wood Johnson Medical School, New Brunswick, New Jersey (F.A., P.Y.S.Y., M.M., N.M., A.M.V.); Hofstra Northwell School of Medicine, Cohen Children's Medical Center of New York, New Hyde Park, New York (B.I.W.); and Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (L.M.S.)
| | - Poi Yu Sofia Yuen
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (K.M.B., J.E.M., X.W., L.M.A.), Environmental and Occupational Health Sciences Institute (L.M.A.), and Lipid Center (L.M.A.), Rutgers, The State University of New Jersey, Piscataway, New Jersey; Department of Pediatrics, Rutgers University Robert Wood Johnson Medical School, New Brunswick, New Jersey (F.A., P.Y.S.Y., M.M., N.M., A.M.V.); Hofstra Northwell School of Medicine, Cohen Children's Medical Center of New York, New Hyde Park, New York (B.I.W.); and Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (L.M.S.)
| | - Moiz Mohammed
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (K.M.B., J.E.M., X.W., L.M.A.), Environmental and Occupational Health Sciences Institute (L.M.A.), and Lipid Center (L.M.A.), Rutgers, The State University of New Jersey, Piscataway, New Jersey; Department of Pediatrics, Rutgers University Robert Wood Johnson Medical School, New Brunswick, New Jersey (F.A., P.Y.S.Y., M.M., N.M., A.M.V.); Hofstra Northwell School of Medicine, Cohen Children's Medical Center of New York, New Hyde Park, New York (B.I.W.); and Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (L.M.S.)
| | - Naureen Memon
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (K.M.B., J.E.M., X.W., L.M.A.), Environmental and Occupational Health Sciences Institute (L.M.A.), and Lipid Center (L.M.A.), Rutgers, The State University of New Jersey, Piscataway, New Jersey; Department of Pediatrics, Rutgers University Robert Wood Johnson Medical School, New Brunswick, New Jersey (F.A., P.Y.S.Y., M.M., N.M., A.M.V.); Hofstra Northwell School of Medicine, Cohen Children's Medical Center of New York, New Hyde Park, New York (B.I.W.); and Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (L.M.S.)
| | - Barry I Weinberger
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (K.M.B., J.E.M., X.W., L.M.A.), Environmental and Occupational Health Sciences Institute (L.M.A.), and Lipid Center (L.M.A.), Rutgers, The State University of New Jersey, Piscataway, New Jersey; Department of Pediatrics, Rutgers University Robert Wood Johnson Medical School, New Brunswick, New Jersey (F.A., P.Y.S.Y., M.M., N.M., A.M.V.); Hofstra Northwell School of Medicine, Cohen Children's Medical Center of New York, New Hyde Park, New York (B.I.W.); and Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (L.M.S.)
| | - Laura M Saba
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (K.M.B., J.E.M., X.W., L.M.A.), Environmental and Occupational Health Sciences Institute (L.M.A.), and Lipid Center (L.M.A.), Rutgers, The State University of New Jersey, Piscataway, New Jersey; Department of Pediatrics, Rutgers University Robert Wood Johnson Medical School, New Brunswick, New Jersey (F.A., P.Y.S.Y., M.M., N.M., A.M.V.); Hofstra Northwell School of Medicine, Cohen Children's Medical Center of New York, New Hyde Park, New York (B.I.W.); and Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (L.M.S.)
| | - Anna M Vetrano
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (K.M.B., J.E.M., X.W., L.M.A.), Environmental and Occupational Health Sciences Institute (L.M.A.), and Lipid Center (L.M.A.), Rutgers, The State University of New Jersey, Piscataway, New Jersey; Department of Pediatrics, Rutgers University Robert Wood Johnson Medical School, New Brunswick, New Jersey (F.A., P.Y.S.Y., M.M., N.M., A.M.V.); Hofstra Northwell School of Medicine, Cohen Children's Medical Center of New York, New Hyde Park, New York (B.I.W.); and Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (L.M.S.)
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (K.M.B., J.E.M., X.W., L.M.A.), Environmental and Occupational Health Sciences Institute (L.M.A.), and Lipid Center (L.M.A.), Rutgers, The State University of New Jersey, Piscataway, New Jersey; Department of Pediatrics, Rutgers University Robert Wood Johnson Medical School, New Brunswick, New Jersey (F.A., P.Y.S.Y., M.M., N.M., A.M.V.); Hofstra Northwell School of Medicine, Cohen Children's Medical Center of New York, New Hyde Park, New York (B.I.W.); and Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado (L.M.S.)
| |
Collapse
|
49
|
Characterization of a Cancer Stem Cell-Like Side Population Derived from Human Pancreatic Adenocarcinoma Cells. TUMORI JOURNAL 2018. [DOI: 10.1177/548.6520] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aims and background To identify and partially characterize the side population cells derived from three human pancreatic adenocarcinoma cell lines. Methods Side population cells were sorted from the human pancreatic adenocarcinoma cell lines SW1990, Capan-2, and BxPC-3 using flow cytometry and then analyzed for cell proliferation, clone formation, differentiation, chemoresistance, invasive potential, and tumorigenicity in a mouse model. Results Human pancreatic carcinoma cell lines SW1990, Capan-2, and BxPC-3 contain 2.7% ±0.35%, 3.6%± 1.2%, and 2.8%± 0.8% side population cells, respectively. We further investigated cancer stem cell characteristics with the moderately differentiated human pancreatic adenocarcinoma cell line SW1990. Flow cytometry analysis revealed that side population cells could differentiate into side population and non-side population cells and could exhibit differentiation potential. Using a clone formation assay, side population cells were shown to have a higher proliferation than non-side population cells. Compared to non-side population cells, side population cells were also more resistant to gemcitabine, a commonly used anti-cancer agent against pancreatic carcinoma, and were more invasive. Importantly, the CD133 level in side population cells was significantly higher than that in non-side population cells. The enhanced tumorigenecity was further confirmed in a male diabetic/severe combined immunodeficiency mouse model. As few as 3 × 103 side population cells were sufficient to induce tumor formation in the mouse model, compared to 107 non-side population or unsorted cells. Conclusions Side population cells isolated from human pancreatic adenocarcinoma cell lines harbor cancer stem cell-like properties that may be related to the invasive potential and therapeutic-resistance of pancreatic adenocarcinoma. Free full text available at www.tumorionline.it
Collapse
|
50
|
Vidal RS, Quarti J, Rodrigues MF, Rumjanek FD, Rumjanek VM. Metabolic Reprogramming During Multidrug Resistance in Leukemias. Front Oncol 2018; 8:90. [PMID: 29675398 PMCID: PMC5895924 DOI: 10.3389/fonc.2018.00090] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/15/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer outcome has improved since introduction of target therapy. However, treatment success is still impaired by the same drug resistance mechanism of classical chemotherapy, known as multidrug resistance (MDR) phenotype. This phenotype promotes resistance to drugs with different structures and mechanism of action. Recent reports have shown that resistance acquisition is coupled to metabolic reprogramming. High-gene expression, increase of active transport, and conservation of redox status are one of the few examples that increase energy and substrate demands. It is not clear if the role of this metabolic shift in the MDR phenotype is related to its maintenance or to its induction. Apart from the nature of this relation, the metabolism may represent a new target to avoid or to block the mechanism that has been impairing treatment success. In this mini-review, we discuss the relation between metabolism and MDR resistance focusing on the multiple non-metabolic functions that enzymes of the glycolytic pathway are known to display, with emphasis with the diverse activities of glyceraldehyde-3-phosphate dehydrogenase.
Collapse
Affiliation(s)
- Raphael Silveira Vidal
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julia Quarti
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto de Nutrição Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Franklin D Rumjanek
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivian M Rumjanek
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|