1
|
Dvoriashyna M, Bentley-Ford M, Yu J, Chatterjee S, Pardue MT, Kane MA, Repetto R, Ethier CR. All- trans retinoic acid and fluid transport in myopigenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636685. [PMID: 39975307 PMCID: PMC11839105 DOI: 10.1101/2025.02.05.636685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Myopia, or near-sightedness, is rapidly growing in prevalence, with significant long-term implications for ocular health. There is thus great impetus to better understand molecular signaling pathways leading to myopia. We and others have reported that all-trans retinoic acid (atRA) is involved in myopigenic signaling, yet the understanding of how atRA is transported and exerts a myopigenic influence is poor. Here we measured the concentrations of atRA in the serum in wild-type C57BL/6 mice under control conditions and after atRA feeding, previously shown to induce myopia. We also developed a mathematical model that describes fluid fluxes and the advective-diffusive transport of atRA in choroid and sclera, including atRA synthesis in the choriocapillaris, atRA degradation by scleral cells, and binding of atRA to the carrier protein serum albumin. This model, developed for both mice and humans, showed that atRA produced in the choriocapillaris was able to permeate well into the sclera in both mice and humans at biologically-relevant concentrations, and that atRA feeding greatly increased tissue levels of atRA across both the choroid and sclera. We were also able to identify which parameters most influence atRA concentration in ocular tissues, guiding future experimental work. Our findings support atRA's role in myopigenic signaling.
Collapse
Affiliation(s)
- Mariia Dvoriashyna
- School of Mathematics and Maxwell Institute for Mathematical Sciences, University of Edinburgh, Edinburgh, UK
| | - Melissa Bentley-Ford
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, GA, USA
| | - Jianshi Yu
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD, 21201, USA
| | - Saptarshi Chatterjee
- Wallace H. Coulter Dept. of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA
| | - Machelle T. Pardue
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, USA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, GA, USA
- Wallace H. Coulter Dept. of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA
| | - Maureen A. Kane
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, MD, 21201, USA
| | - Rodolfo Repetto
- Department of Civil, Chemical and Environmental Engineering, University of Genoa, Genoa, Italy
| | - C. Ross Ethier
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, GA, USA
- Wallace H. Coulter Dept. of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA
| |
Collapse
|
2
|
Isoherranen N, Wen YW. The interplay between retinoic acid binding proteins and retinoic acid degrading enzymes in modulating retinoic acid concentrations. Curr Top Dev Biol 2024; 161:167-200. [PMID: 39870433 DOI: 10.1016/bs.ctdb.2024.09.001] [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] [Indexed: 01/29/2025]
Abstract
The active metabolite of vitamin A, all-trans-retinoic acid (atRA), is critical for maintenance of many cellular processes. Although the enzymes that can synthesize and clear atRA in mammals have been identified, their tissue and cell-type specific roles are still not fully established. Based on the plasma protein binding, tissue distribution and lipophilicity of atRA, atRA partitions extensively to lipid membranes and other neutral lipids in cells. As a consequence, free atRA concentrations in cells are expected to be exceedingly low. As such mechanisms must exist that allow sufficiently high atRA concentrations to occur for binding to retinoic acid receptor (RARs) and for RAR mediated signaling. Kinetic simulations suggest that cellular retinoic acid binding proteins (CRABPs) provide a cytosolic reservoir for atRA to allow high enough cytosolic concentrations that enable RAR signaling. Yet, the different CRABP family members CRABP1 and CRABP2 may serve different functions in this context. CRABP1 may reside in the cytosol as a member of a cytosolic signalosome and CRABP2 may bind atRA in the cytosol and localize to the nucleus. Both CRABPs appear to interact with the atRA-degrading cytochrome P450 (CYP) family 26 enzymes in the endoplasmic reticulum. These interactions, together with the expression levels of the CRABPs and CYP26s, likely modulate cellular atRA concentration gradients and tissue atRA concentrations in a tightly coordinated manner. This review provides a summary of the current knowledge of atRA distribution, metabolism and protein binding and how these characteristics may alter tissue atRA concentrations.
Collapse
Affiliation(s)
- Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington.
| | - Yue Winnie Wen
- Department of Pharmaceutics, School of Pharmacy, University of Washington
| |
Collapse
|
3
|
Pasha M, Zamir A, Rasool MF, Saeed H, Ahmad T, Alqahtani NS, Alqahtani LS, Alqahtani F. A Comprehensive Physiologically Based Pharmacokinetic Model for Predicting Vildagliptin Pharmacokinetics: Insights into Dosing in Renal Impairment. Pharmaceuticals (Basel) 2024; 17:924. [PMID: 39065773 PMCID: PMC11280059 DOI: 10.3390/ph17070924] [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: 05/28/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Physiologically based pharmacokinetic (PBPK) modeling is of great importance in the field of medicine. This study aims to construct a PBPK model, which can provide reliable drug pharmacokinetic (PK) predictions in both healthy and chronic kidney disease (CKD) subjects. To do so, firstly a review of the literature was thoroughly conducted and the PK information of vildagliptin was collected. PBPK modeling software, PK-Sim®, was then used to build and assess the IV, oral, and drug-specific models. Next, the average fold error, visual predictive checks, and predicted/observed ratios were used for the assessment of the robustness of the model for all the essential PK parameters. This evaluation demonstrated that all PK parameters were within an acceptable limit of error, i.e., 2 fold. Also to display the influence of CKD on the total and unbound AUC (the area under the plasma concentration-time curve) and to make modifications in dose, the analysis results of the model on this aspect were further examined. This PBPK model has successfully depicted the variations of PK of vildagliptin in healthy subjects and patients with CKD, which can be useful for medical practitioners in dosage optimization in renal disease patients.
Collapse
Affiliation(s)
- Mahnoor Pasha
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan; (M.P.); (A.Z.)
| | - Ammara Zamir
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan; (M.P.); (A.Z.)
| | - Muhammad Fawad Rasool
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60800, Pakistan; (M.P.); (A.Z.)
| | - Hamid Saeed
- Section of Pharmaceutics, University College of Pharmacy, Allama Iqbal Campus, University of the Punjab, Lahore 54000, Pakistan;
| | - Tanveer Ahmad
- Institute for Advanced Biosciences (IAB), CNRS UMR5309, INSERM U1209, Grenoble Alpes University, 38700 La Tronche, France;
| | - Nawaf Shalih Alqahtani
- King Abdulaziz Medical City, Riyadh Region Ministry of National Guard, Health Affairs, Riyadh 11426, Saudi Arabia;
| | - Lamya Saif Alqahtani
- Department of Cardiology, Prince Sultan Cardiac Center, Riyadh 11625, Saudi Arabia;
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
4
|
Farruggia G, Anconelli L, Galassi L, Voltattorni M, Rossi M, Lodeserto P, Blasi P, Orienti I. Nano-fenretinide demonstrates remarkable activity in acute promyeloid leukemia cells. Sci Rep 2024; 14:13737. [PMID: 38877119 PMCID: PMC11178801 DOI: 10.1038/s41598-024-64629-w] [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: 12/15/2023] [Accepted: 03/26/2024] [Indexed: 06/16/2024] Open
Abstract
Acute promyelocytic leukemia (APL) is characterized by rearrangements of the retinoic acid receptor, RARα, which makes all-trans retinoic acid (ATRA) highly effective in the treatment of this disease, inducing promyelocytes differentiation. Current therapy, based on ATRA in combination with arsenic trioxide, with or without chemotherapy, provides high rates of event-free survival and overall survival. However, a decline in the drug activity, due to increased ATRA metabolism and RARα mutations, is often observed over long-term treatments. Furthermore, dedifferentiation can occur providing relapse of the disease. In this study we evaluated fenretinide, a semisynthetic ATRA derivative, encapsulated in nanomicelles (nano-fenretinide) as an alternative treatment to ATRA in APL. Nano-fenretinide was prepared by fenretinide encapsulation in a self-assembling phospholipid mixture. Physico-chemical characterization was carried out by dinamic light scattering and spectrophotometry. The biological activity was evaluated by MTT assay, flow cytometry and confocal laser-scanning fluorescence microscopy. Nano-fenretinide induced apoptosis in acute promyelocytic leukemia cells (HL60) by an early increase of reactive oxygen species and a mitochondrial potential decrease. The fenretinide concentration that induced 90-100% decrease in cell viability was about 2.0 µM at 24 h, a concentration easily achievable in vivo when nano-fenretinide is administered by oral or intravenous route, as demonstrated in previous studies. Nano-fenretinide was effective, albeit at slightly higher concentrations, also in doxorubicin-resistant HL60 cells, while a comparison with TK6 lymphoblasts indicated a lack of toxicity on normal cells. The results indicate that nano-fenretinide can be considered an alternative therapy to ATRA in acute promyelocytic leukemia when decreased efficacy, resistance or recurrence of disease emerge after protracted treatments with ATRA.
Collapse
Affiliation(s)
- Giovanna Farruggia
- Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 19/2, 40127, Bologna, Italy
- Center for Applied Biomedical Research (CRBA), University of Bologna, 40126, Bologna, Italy
- National Institute of Biostructures and Biosystems, Via Delle Medaglie d'Oro 305, 00136, Rome, Italy
| | - Lorenzo Anconelli
- Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 19/2, 40127, Bologna, Italy
| | - Lucrezia Galassi
- Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 19/2, 40127, Bologna, Italy
- Center for Applied Biomedical Research (CRBA), University of Bologna, 40126, Bologna, Italy
| | - Manuela Voltattorni
- Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 19/2, 40127, Bologna, Italy
| | - Martina Rossi
- Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 19/2, 40127, Bologna, Italy
- Center for Applied Biomedical Research (CRBA), University of Bologna, 40126, Bologna, Italy
| | - Pietro Lodeserto
- Section of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Paolo Blasi
- Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 19/2, 40127, Bologna, Italy.
- Center for Applied Biomedical Research (CRBA), University of Bologna, 40126, Bologna, Italy.
| | - Isabella Orienti
- Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 19/2, 40127, Bologna, Italy.
| |
Collapse
|
5
|
Han J, Jang KL. All-trans retinoic acid downregulates HBx levels via E6-associated protein-mediated proteasomal degradation to suppress hepatitis B virus replication. PLoS One 2024; 19:e0305350. [PMID: 38861553 PMCID: PMC11166335 DOI: 10.1371/journal.pone.0305350] [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: 02/07/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
All-trans retinoic acid (ATRA), recognized as the principal and most biologically potent metabolite of vitamin A, has been identified for its inhibitory effects on hepatitis B virus (HBV) replication. Nevertheless, the underlying mechanism remains elusive. The present study reveals that ATRA induces E6-associated protein (E6AP)-mediated proteasomal degradation of HBx to suppress HBV replication in human hepatoma cells in a p53-dependent pathway. For this effect, ATRA induced promoter hypomethylation of E6AP in the presence of HBx, which resulted in the upregulation of E6AP levels in HepG2 but not in Hep3B cells, emphasizing the p53-dependent nature of this effect. As a consequence, ATRA augmented the interaction between E6AP and HBx, resulting in substantial ubiquitination of HBx and consequent reduction in HBx protein levels in both the HBx overexpression system and the in vitro HBV replication model. Additionally, the knockdown of E6AP under ATRA treatment reduced the interaction between HBx and E6AP and decreased the ubiquitin-dependent proteasomal degradation of HBx, which prompted a recovery of HBV replication in the presence of ATRA, as confirmed by increased levels of intracellular HBV proteins and secreted HBV levels. This study not only contributes to the understanding of the complex interactions between ATRA, p53, E6AP, and HBx but also provides an academic basis for the clinical employment of ATRA in the treatment of HBV infection.
Collapse
Affiliation(s)
- Jiwoo Han
- Department of Integrated Biological Science, The Graduate School, Pusan National University, Busan, Republic of Korea
| | - Kyung Lib Jang
- Department of Integrated Biological Science, The Graduate School, Pusan National University, Busan, Republic of Korea
- Department of Microbiology, College of Natural Science, Pusan National University, Busan, Republic of Korea
- Microbiological Resource Research Institute, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
6
|
Nagai Y, Ambinder AJ. The Promise of Retinoids in the Treatment of Cancer: Neither Burnt Out Nor Fading Away. Cancers (Basel) 2023; 15:3535. [PMID: 37509198 PMCID: PMC10377082 DOI: 10.3390/cancers15143535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Since the introduction of all-trans retinoic acid (ATRA), acute promyelocytic leukemia (APL) has become a highly curable malignancy, especially in combination with arsenic trioxide (ATO). ATRA's success has deepened our understanding of the role of the RARα pathway in normal hematopoiesis and leukemogenesis, and it has influenced a generation of cancer drug development. Retinoids have also demonstrated some efficacy in a handful of other disease entities, including as a maintenance therapy for neuroblastoma and in the treatment of cutaneous T-cell lymphomas; nevertheless, the promise of retinoids as a differentiating therapy in acute myeloid leukemia (AML) more broadly, and as a cancer preventative, have largely gone unfulfilled. Recent research into the mechanisms of ATRA resistance and the biomarkers of RARα pathway dysregulation in AML have reinvigorated efforts to successfully deploy retinoid therapy in a broader subset of myeloid malignancies. Recent studies have demonstrated that the bone marrow environment is highly protected from exogenous ATRA via local homeostasis controlled by stromal cells expressing CYP26, a key enzyme responsible for ATRA inactivation. Synthetic CYP26-resistant retinoids such as tamibarotene bypass this stromal protection and have shown superior anti-leukemic effects. Furthermore, recent super-enhancer (SE) analysis has identified a novel AML subgroup characterized by high expression of RARα through strong SE levels in the gene locus and increased sensitivity to tamibarotene. Combined with a hypomethylating agent, synthetic retinoids have shown synergistic anti-leukemic effects in non-APL AML preclinical models and are now being studied in phase II and III clinical trials.
Collapse
Affiliation(s)
- Yuya Nagai
- Department of Hematology, Kobe City Medical Center General Hospital, Kobe 650-0047, Hyogo, Japan
| | - Alexander J Ambinder
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
7
|
McBride DA, Kerr MD, Johnson WT, Nguyen A, Zoccheddu M, Yao M, Prideaux EB, Dorn NC, Wang W, Svensson MN, Bottini N, Shah NJ. Immunomodulatory Microparticles Epigenetically Modulate T Cells and Systemically Ameliorate Autoimmune Arthritis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2202720. [PMID: 36890657 PMCID: PMC10104670 DOI: 10.1002/advs.202202720] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 01/09/2023] [Indexed: 05/10/2023]
Abstract
Disease modifying antirheumatic drugs (DMARDs) have improved the prognosis of autoimmune inflammatory arthritides but a large fraction of patients display partial or nonresponsiveness to front-line DMARDs. Here, an immunoregulatory approach based on sustained joint-localized release of all-trans retinoic acid (ATRA), which modulates local immune activation and enhances disease-protective T cells and leads to systemic disease control is reported. ATRA imprints a unique chromatin landscape in T cells, which is associated with an enhancement in the differentiation of naïve T cells into anti-inflammatory regulatory T cells (Treg ) and suppression of Treg destabilization. Sustained release poly-(lactic-co-glycolic) acid (PLGA)-based biodegradable microparticles encapsulating ATRA (PLGA-ATRA MP) are retained in arthritic mouse joints after intra-articular (IA) injection. IA PLGA-ATRA MP enhance migratory Treg which in turn reduce inflammation and modify disease in injected and uninjected joints, a phenotype that is also reproduced by IA injection of Treg . PLGA-ATRA MP reduce proteoglycan loss and bone erosions in the SKG and collagen-induced arthritis mouse models of autoimmune arthritis. Strikingly, systemic disease modulation by PLGA-ATRA MP is not associated with generalized immune suppression. PLGA-ATRA MP have the potential to be developed as a disease modifying agent for autoimmune arthritis.
Collapse
Affiliation(s)
- David A. McBride
- Department of NanoengineeringUniversity of CaliforniaLa JollaSan DiegoCA92093USA
- Chemical Engineering ProgramUniversity of CaliforniaLa JollaSan DiegoCA92093USA
| | - Matthew D. Kerr
- Department of NanoengineeringUniversity of CaliforniaLa JollaSan DiegoCA92093USA
- Chemical Engineering ProgramUniversity of CaliforniaLa JollaSan DiegoCA92093USA
| | - Wade T. Johnson
- Department of NanoengineeringUniversity of CaliforniaLa JollaSan DiegoCA92093USA
| | - Anders Nguyen
- Department of Rheumatology and Inflammation ResearchSahlgrenska AcademyInstitute of MedicineUniversity of GothenburgGothenburg41346Sweden
| | - Martina Zoccheddu
- Department of MedicineDivision of RheumatologyAllergy and ImmunologyUniversity of CaliforniaLa JollaSan DiegoCA92093USA
| | - Mina Yao
- Department of Chemistry and BiochemistryUniversity of CaliforniaLa JollaSan DiegoCA92093USA
| | - Edward B. Prideaux
- Department of Chemistry and BiochemistryUniversity of CaliforniaLa JollaSan DiegoCA92093USA
| | - Nicholas C. Dorn
- Department of NanoengineeringUniversity of CaliforniaLa JollaSan DiegoCA92093USA
- Chemical Engineering ProgramUniversity of CaliforniaLa JollaSan DiegoCA92093USA
| | - Wei Wang
- Department of Chemistry and BiochemistryUniversity of CaliforniaLa JollaSan DiegoCA92093USA
- Department of Cellular and Molecular MedicineUniversity of CaliforniaLa JollaSan DiegoCA92093USA
| | - Mattias N.D. Svensson
- Department of Rheumatology and Inflammation ResearchSahlgrenska AcademyInstitute of MedicineUniversity of GothenburgGothenburg41346Sweden
| | - Nunzio Bottini
- Department of MedicineDivision of RheumatologyAllergy and ImmunologyUniversity of CaliforniaLa JollaSan DiegoCA92093USA
| | - Nisarg J. Shah
- Department of NanoengineeringUniversity of CaliforniaLa JollaSan DiegoCA92093USA
- Chemical Engineering ProgramUniversity of CaliforniaLa JollaSan DiegoCA92093USA
| |
Collapse
|
8
|
Chen J, Yuan Z, Tu Y, Hu W, Xie C, Ye L. Experimental and computational models to investigate intestinal drug permeability and metabolism. Xenobiotica 2023; 53:25-45. [PMID: 36779684 DOI: 10.1080/00498254.2023.2180454] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Oral administration is the preferred route for drug administration that leads to better therapy compliance. The intestine plays a key role in the absorption and metabolism of oral drugs, therefore, new intestinal models are being continuously proposed, which contribute to the study of intestinal physiology, drug screening, drug side effects, and drug-drug interactions.Advances in pharmaceutical processes have produced more drug formulations, causing challenges for intestinal models. To adapt to the rapid evolution of pharmaceuticals, more intestinal models have been created. However, because of the complexity of the intestine, few models can take all aspects of the intestine into account, and some functions must be sacrificed to investigate other areas. Therefore, investigators need to choose appropriate models according to the experimental stage and other requirements to obtain the desired results.To help researchers achieve this goal, this review summarised the advantages and disadvantages of current commonly used intestinal models and discusses possible future directions, providing a better understanding of intestinal models.
Collapse
Affiliation(s)
- Jinyuan Chen
- Institute of Scientific Research, Southern Medical University, Guangzhou, P.R. China.,TCM-Integrated Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Ziyun Yuan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Yifan Tu
- Boehringer-Ingelheim, Connecticut, P.R. USA
| | - Wanyu Hu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Cong Xie
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Ling Ye
- TCM-Integrated Hospital, Southern Medical University, Guangzhou, P.R. China
| |
Collapse
|
9
|
Maślanka T. Effect of IL-27, Teriflunomide and Retinoic Acid and Their Combinations on CD4 + T Regulatory T Cells-An In Vitro Study. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238471. [PMID: 36500570 PMCID: PMC9739213 DOI: 10.3390/molecules27238471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/11/2022]
Abstract
The principal goal of the study was to verify the concept of pharmacological induction of Foxp3+CD25+CD4+ T regulatory (Treg) cells which will additionally be characterized by a highly suppressive phenotype, i.e., by extensive CD25 and CD39 expression and IL-10 and TGF-β production. Stimulated and unstimulated murine lymphocytes were exposed to IL-27, teriflunomide (TER), and all trans retinoic acid (ATRA) alone and to their combinations. The study demonstrated that: (a) IL-27 alone induced CD39 expression on Treg cells and the generation of Tr1 cells; (b) TER alone induced Foxp3-expressing CD4+ T cells and up-regulated density of CD25 on these cells; TER also induced the ability of Treg cells to TGF-β production; (c) ATRA alone induced CD39 expression on Treg cells. The experiments revealed a strong superadditive effect between IL-27 and ATRA with respect to increasing CD39 expression on Treg cells. Moreover, IL-27 and ATRA in combination, but not alone, induced the ability of Treg cells to IL-10 production. However, the combination of IL-27, TER, and ATRA did not induce the generation of Treg cell subset with all described above features. This was due to the fact that TER abolished all listed above desired effects induced by IL-27 alone, ATRA alone, and their combination. IL-27 alone, ATRA alone, and their combination affected TER-induced effects to a lesser extent. Therefore, it can be concluded that in the aspect of pharmacological induction of Treg cells with a highly suppressive phenotype, the triple combination treatment with TER, IL-27, and ATRA does not provide any benefits over TER alone or dual combination including IL-27 and ATRA.
Collapse
Affiliation(s)
- Tomasz Maślanka
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego Street 13, 10-719 Olsztyn, Poland
| |
Collapse
|
10
|
Nuclear Proteomics of Induced Leukemia Cell Differentiation. Cells 2022; 11:cells11203221. [PMID: 36291090 PMCID: PMC9600443 DOI: 10.3390/cells11203221] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022] Open
Abstract
Studies of induced granulocytic differentiation help to reveal molecular mechanisms of cell maturation. The nuclear proteome represents a rich source of regulatory molecules, including transcription factors (TFs). It is important to have an understanding of molecular perturbations at the early stages of the differentiation processes. By applying the proteomic quantitative profiling using isobaric labeling, we found that the contents of 214, 319, 376, 426, and 391 proteins were altered at 3, 6, 9, 12, and 72 h, respectively, compared to 0 h in the HL-60 cell nuclear fraction under all-trans-retinoid acid (ATRA) treatment. From 1860 identified nuclear proteins, 231 proteins were annotated as proteins with transcription factor (TF) activity. Six TFs (RREB1, SRCAP, CCDC124, TRIM24, BRD7, and BUD31) were downregulated and three TFs EWSR1, ENO1, and FUS were upregulated at early time points (3–12 h) after ATRA treatment. Bioinformatic annotation indicates involvement of the HL-60 nuclear proteome in DNA damage recognition in the RUNX1-triggered pathway, and in the p53-regulation pathway. By applying scheduled multiple reaction monitoring using stable isotopically labeled peptide standards (MRM/SIS), we found a persistent increase in the content of the following proteins: PRAM1, CEPBP, RBPJ, and HIC1 in the HL-60 cell nuclear fraction during ATRA-induced granulocytic differentiation. In the case of STAT1, CASP3, PARP1, and PRKDC proteins, a transient increase in their content was observed at early time points (3–12 h) after the ATRA treatment. Obtained data on nuclear proteome composition and dynamics during granulocytic differentiation could be beneficial for the development of new treatment approaches for leukemias with the mutated p53 gene.
Collapse
|
11
|
Yadav AS, Stevison F, Kosaka M, Wong S, Kenny JR, Amory JK, Isoherranen N. Isotretinoin and its Metabolites Alter mRNA of Multiple Enzyme and Transporter Genes In Vitro, but Downregulation of Organic Anion Transporting Polypeptide Does Not Translate to the Clinic. Drug Metab Dispos 2022; 50:1042-1052. [PMID: 35545255 PMCID: PMC11022860 DOI: 10.1124/dmd.122.000882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/23/2022] [Indexed: 11/22/2022] Open
Abstract
Isotretinoin [13-cis-retinoic acid (13cisRA)] is widely used for the treatment of neuroblastoma and acne. It acts via regulating gene transcription through binding to retinoic acid receptors. Yet, the potential for isotretinoin to cause transcriptionally mediated drug-drug interactions (DDIs) has not been fully explored. We hypothesized that isotretinoin and its active metabolites all-trans-retinoic acid (atRA) and 4-oxo-13cisRA would alter the transcription of enzymes and transporters in the human liver via binding to nuclear receptors. The goal of this study was to define the DDI potential of isotretinoin and its metabolites resulting from transcriptional regulation of cytochrome P450 and transporter mRNAs. In human hepatocytes (n = 3), 13cisRA, atRA, and 4-oxo-13cisRA decreased OATP1B1, CYP1A2, CYP2C9, and CYP2D6 mRNA and increased CYP2B6 and CYP3A4 mRNA in a concentration-dependent manner. The EC50 values for OATP1B1 mRNA downregulation ranged from 2 to 110 nM, with maximum effect (Emax ) ranging from 0.17- to 0.54-fold. Based on the EC50 and Emax values and the known circulating concentrations of 13cisRA and its metabolites after isotretinoin dosing, a 55% decrease in OATP1B1 activity was predicted in vivo. In vivo DDI potential was evaluated clinically in participants dosed with isotretinoin for up to 32 weeks using coproporphyrin-I (CP-I) as an OATP1B1 biomarker. CP-I steady-state serum concentrations were unaltered following 2, 8, or 16 weeks of isotretinoin treatment. These data show that isotretinoin and its metabolites alter transcription of multiple enzymes and transporters in vitro, but translation of these changes to in vivo drug-drug interactions requires clinical evaluation for each enzyme. SIGNIFICANCE STATEMENT: Isotretinoin and its metabolites alter the mRNA expression of multiple cytochrome P450s (CYPs) and transporters in human hepatocytes, suggesting that isotretinoin may cause clinically significant drug-drug interactions (DDIs). Despite the observed changes in organic anion transporting polypeptide 1B1 (OATP1B1) mRNA in human hepatocytes, no clinical DDI was observed when measuring a biomarker, coproporphyrin-I. Further work is needed to determine whether these findings can be extrapolated to a lack of a DDI with CYP1A2, CYP2B6, and CYP2C9 substrates.
Collapse
Affiliation(s)
- Aprajita S Yadav
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (A.S.Y., F.S., N.I.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (M.K., S.W., J.R.K.); and Department of Medicine, University of Washington, Seattle, Washington (J.K.A.)
| | - Faith Stevison
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (A.S.Y., F.S., N.I.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (M.K., S.W., J.R.K.); and Department of Medicine, University of Washington, Seattle, Washington (J.K.A.)
| | - Mika Kosaka
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (A.S.Y., F.S., N.I.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (M.K., S.W., J.R.K.); and Department of Medicine, University of Washington, Seattle, Washington (J.K.A.)
| | - Susan Wong
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (A.S.Y., F.S., N.I.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (M.K., S.W., J.R.K.); and Department of Medicine, University of Washington, Seattle, Washington (J.K.A.)
| | - Jane R Kenny
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (A.S.Y., F.S., N.I.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (M.K., S.W., J.R.K.); and Department of Medicine, University of Washington, Seattle, Washington (J.K.A.)
| | - John K Amory
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (A.S.Y., F.S., N.I.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (M.K., S.W., J.R.K.); and Department of Medicine, University of Washington, Seattle, Washington (J.K.A.)
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (A.S.Y., F.S., N.I.); Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (M.K., S.W., J.R.K.); and Department of Medicine, University of Washington, Seattle, Washington (J.K.A.)
| |
Collapse
|
12
|
Assessment of cytochrome P450 3A4-mediated drug–drug interactions for ipatasertib using a fit-for-purpose physiologically based pharmacokinetic model. Cancer Chemother Pharmacol 2022; 89:707-720. [PMID: 35428895 PMCID: PMC9054915 DOI: 10.1007/s00280-022-04434-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 04/02/2022] [Indexed: 12/04/2022]
Abstract
Purpose Ipatasertib, a potent and highly selective small-molecule inhibitor of AKT, is currently under investigation for treatment of cancer. Ipatasertib is a substrate and a time-dependent inhibitor of CYP3A4. It exhibits non-linear pharmacokinetics at subclinical doses in the clinical dose escalation study. To assess the DDI risk of ipatasertib at the intended clinical dose of 400 mg with CYP3A4 inhibitors, inducers, and substrates, a fit-for-purpose physiologically based pharmacokinetic (PBPK) model of ipatasertib was developed. Methods The PBPK model was constructed in Simcyp using in silico, in vitro, and clinical data and was optimized and verified using clinical data. Results The PBPK model described non-linear pharmacokinetics of ipatasertib and captured the magnitude of the observed clinical DDIs. Following repeated doses of 400 mg ipatasertib once daily (QD), the PBPK model predicted a 3.3-fold increase of ipatasertib exposure with itraconazole; a 2–2.5-fold increase with moderate CYP3A4 inhibitors, erythromycin and diltiazem; and no change with a weak CYP3A4 inhibitor, fluvoxamine. Additionally, in the presence of strong or moderate CYP3A4 inducers, rifampicin and efavirenz, ipatasertib exposures were predicted to decrease by 86% and 74%, respectively. As a perpetrator, the model predicted that ipatasertib (400 mg) caused a 1.7-fold increase in midazolam exposure. Conclusion This study demonstrates the value of using a fit-for-purpose PBPK model to assess the clinical DDIs for ipatasertib and to provide dosing strategies for the concurrent use of other CYP3A4 perpetrators or victims. Supplementary Information The online version contains supplementary material available at 10.1007/s00280-022-04434-2.
Collapse
|
13
|
Czuba LC, Fay EE, LaFrance J, Smith CK, Shum S, Moreni SL, Mao J, Isoherranen N, Hebert MF. Plasma Retinoid Concentrations Are Altered in Pregnant Women. Nutrients 2022; 14:1365. [PMID: 35405978 PMCID: PMC9002937 DOI: 10.3390/nu14071365] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/18/2022] [Indexed: 11/22/2022] Open
Abstract
Vitamin A is vital to maternal-fetal health and pregnancy outcomes. However, little is known about pregnancy associated changes in maternal vitamin A homeostasis and concentrations of circulating retinol metabolites. The goal of this study was to characterize retinoid concentrations in healthy women (n = 23) during two stages of pregnancy (25-28 weeks gestation and 28-32 weeks gestation) as compared to ≥3 months postpartum. It was hypothesized that plasma retinol, retinol binding protein 4 (RBP4), transthyretin and albumin concentrations would decline during pregnancy and return to baseline by 3 months postpartum. At 25-28 weeks gestation, plasma retinol (-27%), 4-oxo-13-cis-retinoic acid (-34%), and albumin (-22%) concentrations were significantly lower, and all-trans-retinoic acid (+48%) concentrations were significantly higher compared to ≥3 months postpartum in healthy women. In addition, at 28-32 weeks gestation, plasma retinol (-41%), retinol binding protein 4 (RBP4; -17%), transthyretin (TTR; -21%), albumin (-26%), 13-cis-retinoic acid (-23%) and 4-oxo-13-cis-retinoic acid (-48%) concentrations were significantly lower, whereas plasma all-trans-retinoic acid concentrations (+30%) were significantly higher than ≥3 months postpartum. Collectively, the data demonstrates that in healthy pregnancies, retinol plasma concentrations are lower, but all-trans-retinoic acid concentrations are higher than postpartum.
Collapse
Affiliation(s)
- Lindsay C. Czuba
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA; (L.C.C.); (J.L.); (S.S.); (N.I.)
| | - Emily E. Fay
- Department of Obstetrics and Gynecology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (E.E.F.); (S.L.M.); (J.M.)
| | - Jeffrey LaFrance
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA; (L.C.C.); (J.L.); (S.S.); (N.I.)
| | - Chase K. Smith
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA 98195, USA;
| | - Sara Shum
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA; (L.C.C.); (J.L.); (S.S.); (N.I.)
| | - Sue L. Moreni
- Department of Obstetrics and Gynecology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (E.E.F.); (S.L.M.); (J.M.)
| | - Jennie Mao
- Department of Obstetrics and Gynecology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (E.E.F.); (S.L.M.); (J.M.)
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA; (L.C.C.); (J.L.); (S.S.); (N.I.)
| | - Mary F. Hebert
- Department of Obstetrics and Gynecology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (E.E.F.); (S.L.M.); (J.M.)
- Department of Pharmacy, School of Pharmacy, University of Washington, Seattle, WA 98195, USA;
| |
Collapse
|
14
|
Baybutt RC, Standard JT, Dim D, Quinn T, Hamdan H, Lin D, Kunz K, Bomstein ZS, Estorge BJ, Herndon B, Zia H, Mansour A, Lankachandra M, Molteni A. Cod Liver Oil, but Not Retinoic Acid, Treatment Restores Bone Thickness in a Vitamin A-Deficient Rat. Nutrients 2022; 14:486. [PMID: 35276845 PMCID: PMC8838835 DOI: 10.3390/nu14030486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/19/2022] [Accepted: 01/19/2022] [Indexed: 02/01/2023] Open
Abstract
Vitamin A plays a prominent role for maintaining optimal bone status, but its impact upon the bone in response to vitamin A deficiency is not well defined. The purpose of this study was to evaluate how replenishing vitamin A by either whole food cod liver oil (COD) or the active metabolite of vitamin A, retinoic acid (RA), altered bone thickness of vitamin A-deficient (VAD) rats. Weanling rats were administered a control diet (CTRL) or VAD diet for 9 weeks. This was followed by four weeks of treatment in which the VAD group was divided into the following 4 subgroups: (1) VAD (9 weeks)-VAD (4 weeks); (2) VAD-CTRL; (3) VAD-COD; and (4) VAD-RA. Compared to controls, VAD rats had thicker bones which showed marked dysplasia. VAD-rats treated with COD produced a thinner bone that was not significantly different from that of untreated rats. In contrast, RA did not significantly change the thicker bone, and also had significantly greater periosteal and endosteal osteoblast numbers compared to VAD-COD. Active osteoclasts were not detected in VAD rats, nor during the treatment period. These findings suggest that the abnormal bone thickness in VAD rats appears to be more effectively restored to bone thickness of untreated control rats when treated with COD.
Collapse
Affiliation(s)
- Richard C. Baybutt
- Department of Applied Health Science, Wheaton College, Wheaton, IL 60187, USA; (J.T.S.); (K.K.)
- Department of Nutrition Science, East Carolina University, Greenville, NC 27834, USA; (Z.S.B.); (B.J.E.)
| | - Joseph T. Standard
- Department of Applied Health Science, Wheaton College, Wheaton, IL 60187, USA; (J.T.S.); (K.K.)
| | - Daniel Dim
- Department of Pathology and Pharmacology, UMKC School of Medicine, Kansas City, MO 64110, USA; (D.D.); (T.Q.); (H.H.); (B.H.); (H.Z.); (A.M.); (A.M.)
| | - Tim Quinn
- Department of Pathology and Pharmacology, UMKC School of Medicine, Kansas City, MO 64110, USA; (D.D.); (T.Q.); (H.H.); (B.H.); (H.Z.); (A.M.); (A.M.)
| | - Hana Hamdan
- Department of Pathology and Pharmacology, UMKC School of Medicine, Kansas City, MO 64110, USA; (D.D.); (T.Q.); (H.H.); (B.H.); (H.Z.); (A.M.); (A.M.)
| | - Dingbo Lin
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Kyle Kunz
- Department of Applied Health Science, Wheaton College, Wheaton, IL 60187, USA; (J.T.S.); (K.K.)
| | - Zachary S. Bomstein
- Department of Nutrition Science, East Carolina University, Greenville, NC 27834, USA; (Z.S.B.); (B.J.E.)
| | - Benjamin J. Estorge
- Department of Nutrition Science, East Carolina University, Greenville, NC 27834, USA; (Z.S.B.); (B.J.E.)
| | - Betty Herndon
- Department of Pathology and Pharmacology, UMKC School of Medicine, Kansas City, MO 64110, USA; (D.D.); (T.Q.); (H.H.); (B.H.); (H.Z.); (A.M.); (A.M.)
| | - Hamid Zia
- Department of Pathology and Pharmacology, UMKC School of Medicine, Kansas City, MO 64110, USA; (D.D.); (T.Q.); (H.H.); (B.H.); (H.Z.); (A.M.); (A.M.)
| | - Ahmad Mansour
- Department of Pathology and Pharmacology, UMKC School of Medicine, Kansas City, MO 64110, USA; (D.D.); (T.Q.); (H.H.); (B.H.); (H.Z.); (A.M.); (A.M.)
| | - Manesha Lankachandra
- Englewood Orthopedic Associates, 410 South Van Brunt Street, Englewood, NJ 07631, USA;
| | - Agostino Molteni
- Department of Pathology and Pharmacology, UMKC School of Medicine, Kansas City, MO 64110, USA; (D.D.); (T.Q.); (H.H.); (B.H.); (H.Z.); (A.M.); (A.M.)
| |
Collapse
|
15
|
Bos AV, Erkelens MN, Koenders STA, van der Stelt M, van Egmond M, Mebius RE. Clickable Vitamins as a New Tool to Track Vitamin A and Retinoic Acid in Immune Cells. Front Immunol 2021; 12:671283. [PMID: 34305901 PMCID: PMC8298001 DOI: 10.3389/fimmu.2021.671283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/02/2021] [Indexed: 01/24/2023] Open
Abstract
The vitamin A derivative, retinoid acid (RA) is key player in guiding adaptive mucosal immune responses. However, data on the uptake and metabolism of vitamin A within human immune cells has remained largely elusive because retinoids are small, lipophilic molecules which are difficult to detect. To overcome this problem and to be able to study the effect of vitamin A metabolism in human immune cell subsets, we have synthesized novel bio-orthogonal retinoid-based probes (clickable probes), which are structurally and functionally indistinguishable from vitamin A. The probes contain a functional group (an alkyne) to conjugate to a fluorogenic dye to monitor retinoid molecules in real-time in immune cells. We demonstrate, by using flow cytometry and microscopy, that multiple immune cells have the capacity to internalize retinoids to varying degrees, including human monocyte-derived dendritic cells (DCs) and naïve B lymphocytes. We observed that naïve B cells lack the enzymatic machinery to produce RA, but use exogenous retinoic acid to enhance CD38 expression. Furthermore, we showed that human DCs metabolize retinal into retinoic acid, which in co-culture with naïve B cells led to of the induction of CD38 expression. These data demonstrate that in humans, DCs can serve as an exogenous source of RA for naïve B cells. Taken together, through the use of clickable vitamins our data provide valuable insight in the mechanism of vitamin A metabolism and its importance for human adaptive immunity.
Collapse
Affiliation(s)
- Amelie V Bos
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands
| | - Martje N Erkelens
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands
| | - Sebastiaan T A Koenders
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands.,Department of Surgery, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands
| | - Reina E Mebius
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, location VUmc, Amsterdam, Netherlands
| |
Collapse
|
16
|
Ambinder AJ, Norsworthy K, Hernandez D, Palau L, Paun B, Duffield A, Chandraratna R, Sanders M, Varadhan R, Jones RJ, Douglas Smith B, Ghiaur G. A Phase 1 Study of IRX195183, a RARα-Selective CYP26 Resistant Retinoid, in Patients With Relapsed or Refractory AML. Front Oncol 2020; 10:587062. [PMID: 33194741 PMCID: PMC7645224 DOI: 10.3389/fonc.2020.587062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/05/2020] [Indexed: 12/31/2022] Open
Abstract
Subsets of non-acute promyelocytic leukemia (APL) acute myelogenous leukemia (AML) exhibit aberrant retinoid signaling and demonstrate sensitivity to retinoids in vitro. We present the results of a phase 1 dose-escalation study that evaluated the safety, pharmacodynamics, and efficacy of IRX195183, a novel retinoic acid receptor α agonist, in patients with relapsed or refractory myelodysplastic syndrome (MDS) or AML. In this single center, single arm study, eleven patients with relapsed or refractory MDS/AML were enrolled and treated. Oral IRX195183 was administered at two dose levels: 50 mg daily or 75 mg daily for a total of two 28-day cycles. Patients with stable disease or better were allowed to continue on the drug for four additional 28-day cycles. Common adverse events included hypertriglyceridemia, fatigue, dyspnea, and edema. Three patients at the first dose level developed asymptomatic Grade 3 hypertriglyceridemia. The maximally tolerated dose was not reached. Four of the eleven patients had (36%) stable disease or better. One had a morphological complete remission with incomplete hematologic recovery while on the study drug. Two patients had evidence of in vivo leukemic blast maturation, as reflected by increased CD38 expression. In a pharmacodynamics study, plasma samples from four patients treated at the lowest dose level demonstrated the capacity to differentiate leukemic cells from the NB4 cell line in vitro. These results suggest that IRX195183 is safe, achieves biologically meaningful plasma concentrations and may be efficacious in a subset of patients with MDS/AML. Clinical Trial Registration: clinicaltrials.gov, identifier NCT02749708.
Collapse
Affiliation(s)
- Alexander J. Ambinder
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kelly Norsworthy
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniela Hernandez
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Laura Palau
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Bogdan Paun
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Amy Duffield
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | | | | | - Ravi Varadhan
- Division of Biostatistics and Bioinformatics, Johns Hopkins/Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, United States
| | - Richard J. Jones
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - B. Douglas Smith
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Gabriel Ghiaur
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
17
|
Kocher HM, Basu B, Froeling FEM, Sarker D, Slater S, Carlin D, deSouza NM, De Paepe KN, Goulart MR, Hughes C, Imrali A, Roberts R, Pawula M, Houghton R, Lawrence C, Yogeswaran Y, Mousa K, Coetzee C, Sasieni P, Prendergast A, Propper DJ. Phase I clinical trial repurposing all-trans retinoic acid as a stromal targeting agent for pancreatic cancer. Nat Commun 2020; 11:4841. [PMID: 32973176 PMCID: PMC7518421 DOI: 10.1038/s41467-020-18636-w] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/02/2020] [Indexed: 12/20/2022] Open
Abstract
Pre-clinical models have shown that targeting pancreatic stellate cells with all-trans-retinoic-acid (ATRA) reprograms pancreatic stroma to suppress pancreatic ductal adenocarcinoma (PDAC) growth. Here, in a phase Ib, dose escalation and expansion, trial for patients with advanced, unresectable PDAC (n = 27), ATRA is re-purposed as a stromal-targeting agent in combination with gemcitabine-nab-paclitaxel chemotherapy using a two-step adaptive continual re-assessment method trial design. The maximum tolerated dose (MTD) and recommended phase 2 dose (RP2D, primary outcome) is the FDA/EMEA approved dose of gemcitabine-nab-paclitaxel along-with ATRA (45 mg/m2 orally, days 1-15/cycle). Dose limiting toxicity (DLT) is grade 4 thrombocytopenia (n = 2). Secondary outcomes show no detriment to ATRA pharmacokinetics.. Median overall survival for RP2D treated evaluable population, is 11.7 months (95%CI 8.6-15.7 m, n = 15, locally advanced (2) and metastatic (13)). Exploratory pharmacodynamics studies including changes in diffusion-weighted (DW)-MRI measured apparent diffusion coefficient after one cycle, and, modulation of cycle-specific serum pentraxin 3 levels over various cycles indicate stromal modulation. Baseline stromal-specific retinoid transport protein (FABP5, CRABP2) expression may be predicitve of response. Re-purposing ATRA as a stromal-targeting agent with gemcitabine-nab-paclitaxel is safe and tolerable. This combination will be evaluated in a phase II randomized controlled trial for locally advanced PDAC. Clinical trial numbers: EudraCT: 2015-002662-23; NCT03307148. Trial acronym: STARPAC.
Collapse
Affiliation(s)
- Hemant M Kocher
- Centre for Tumour Biology, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University London, London, EC1M 6BQ, UK.
- Centre for Experimental Cancer Medicine, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University of London, London, EC1M 6BQ, UK.
- Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, Whitechapel, London, E1 1FR, UK.
- Barts Pancreas Tissue Bank, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University London, London, EC1M 6BQ, UK.
| | - Bristi Basu
- Department of Oncology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust-Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Fieke E M Froeling
- Department of Surgery and Cancer, Imperial College London-Hammersmith Hospital, London, W12 0HS, UK
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY, 11724, USA
| | - Debashis Sarker
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital Campus, London, SE1 9RT, UK
| | - Sarah Slater
- Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, Whitechapel, London, E1 1FR, UK
| | - Dominic Carlin
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Nandita M deSouza
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Katja N De Paepe
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, SW7 3RP, UK
| | - Michelle R Goulart
- Centre for Tumour Biology, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University London, London, EC1M 6BQ, UK
| | - Christine Hughes
- Centre for Tumour Biology, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University London, London, EC1M 6BQ, UK
| | - Ahmet Imrali
- Barts Pancreas Tissue Bank, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University London, London, EC1M 6BQ, UK
| | - Rhiannon Roberts
- Barts Pancreas Tissue Bank, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University London, London, EC1M 6BQ, UK
| | - Maria Pawula
- PK/Bioanalytics Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Richard Houghton
- PK/Bioanalytics Core Facility, Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Cheryl Lawrence
- Centre for Experimental Cancer Medicine, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Yathushan Yogeswaran
- Centre for Experimental Cancer Medicine, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Kelly Mousa
- Centre for Experimental Cancer Medicine, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Carike Coetzee
- Centre for Experimental Cancer Medicine, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Peter Sasieni
- Cancer Prevention Trials Unit, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, EC1M 6BQ, UK
- School of Cancer & Pharmaceutical Sciences, and King's Clinical Trials Unit, King's College London, London, SE1 9RT, UK
| | - Aaron Prendergast
- Centre for Experimental Cancer Medicine, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University of London, London, EC1M 6BQ, UK
| | - David J Propper
- Centre for Experimental Cancer Medicine, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University of London, London, EC1M 6BQ, UK
- Barts and the London HPB Centre, The Royal London Hospital, Barts Health NHS Trust, Whitechapel, London, E1 1FR, UK
- Centre for Cancer and Inflammation, Barts Cancer Institute-A CRUK Centre of Excellence, Queen Mary University London, London, EC1M 6BQ, UK
| |
Collapse
|
18
|
Novotná E, Morell A, Büküm N, Hofman J, Danielisová P, Wsól V. Interactions of antileukemic drugs with daunorubicin reductases: could reductases affect the clinical efficacy of daunorubicin chemoregimens? Arch Toxicol 2020; 94:3059-3068. [PMID: 32588086 DOI: 10.1007/s00204-020-02818-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/18/2020] [Indexed: 11/27/2022]
Abstract
Although novel anticancer drugs are being developed intensively, anthracyclines remain the gold standard in the treatment of acute myeloid leukaemia (AML). The reductive conversion of daunorubicin (Dau) to less active daunorubicinol (Dau-ol) is an important mechanism that contributes to the development of pharmacokinetic anthracycline resistance. Dau is a key component in many AML regimes, in which it is combined with many drugs, including all-trans-retinoic acid (ATRA), cytarabine, cladribine and prednisolone. In the present study, we investigated the influence of these anticancer drugs on the reductive Dau metabolism mediated by the aldo-keto reductases AKR1A1, 1B10, 1C3, and 7A2 and carbonyl reductase 1 (CBR1). In incubation experiments with recombinant enzymes, cladribine and cytarabine did not significantly inhibit the activity of the tested enzymes. Prednisolone inhibited AKR1C3 with an IC50 of 41.73 µM, while ATRA decreased the activity of AKR1B10 (IC50 = 78.33 µM) and AKR1C3 (IC50 = 1.17 µM). Subsequent studies showed that AKR1C3 inhibition mediated by ATRA exhibited tight binding (Kiapp = 0.54 µM). Further, the combination of 1 µM ATRA with different concentrations of Dau demonstrated synergistic effects in HCT116 and KG1a human cells expressing AKR1C3. Our results suggest that ATRA-mediated inhibition of AKR1C3 can contribute to the mechanisms that are hidden beyond the beneficial clinical outcome of the ATRA-Dau combination.
Collapse
Affiliation(s)
- Eva Novotná
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovskeho 1203, Hradec Kralove, 50005, Czech Republic
| | - Anselm Morell
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovskeho 1203, Hradec Kralove, 50005, Czech Republic
| | - Neslihan Büküm
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovskeho 1203, Hradec Kralove, 50005, Czech Republic
| | - Jakub Hofman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Akademika Heyrovskeho 1203, Hradec Kralove, 50005, Czech Republic
| | - Petra Danielisová
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovskeho 1203, Hradec Kralove, 50005, Czech Republic
| | - Vladimír Wsól
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovskeho 1203, Hradec Kralove, 50005, Czech Republic.
| |
Collapse
|
19
|
Marques Borges GS, Oliveira Ferencs MD, Mello Gomide Loures CD, Abdel-Salam MAL, Gontijo Evangelista FC, Sales CC, Reis da Silva PH, de Oliveira RB, Malachias Â, Yoshida MI, de Souza-Fagundes EM, Paula Sabino AD, Fernandes C, Miranda Ferreira LA. Novel self-nanoemulsifying drug-delivery system enhances antileukemic properties of all-trans retinoic acid. Nanomedicine (Lond) 2020; 15:1471-1486. [DOI: 10.2217/nnm-2020-0061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: All- trans retinoic acid (ATRA) shows erratic oral bioavailability when administered orally against leukemia, which can be solved through its incorporation in self-nanoemulsifying drug-delivery systems (SEDDS). The SEDDS developed contained a hydrophobic ion pair between benzathine (BZT) and ATRA and was enriched with tocotrienols by the input of a palm oil tocotrienol rich fraction (TRF) in its composition. Results: SEDDS-TRF-ATRA-BZT allowed the formation of emulsions with nanometric size that retained ATRA within their core after dispersion. Pharmacokinetic parameters after oral administration of SEDDS-TRF-ATRA-BZT in mice were improved compared with what was seen for an ATRA solution. Moreover, SEDDS-TRF-ATRA-BZT had improved activity against HL-60 cells compared with SEDDS without TRF. Conclusion: SEDDS-TRF-ATRA-BZT is a promising therapeutic choice over ATRA conventional medicine.
Collapse
Affiliation(s)
- Gabriel Silva Marques Borges
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Micael de Oliveira Ferencs
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cristina de Mello Gomide Loures
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mostafa AL Abdel-Salam
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Camila Campos Sales
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pedro Henrique Reis da Silva
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Renata Barbosa de Oliveira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ângelo Malachias
- Departamento de Física, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria Irene Yoshida
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Elaine Maria de Souza-Fagundes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adriano de Paula Sabino
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Christian Fernandes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Antônio Miranda Ferreira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
20
|
Czuba LC, Zhong G, Yabut KC, Isoherranen N. Analysis of vitamin A and retinoids in biological matrices. Methods Enzymol 2020; 637:309-340. [PMID: 32359651 DOI: 10.1016/bs.mie.2020.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Vitamin A signaling pathways are predominantly driven by the cellular concentrations of all-trans-retinoic acid (atRA), as the main mechanism of retinoid signaling is via activation of retinoic acid receptors. atRA concentrations are in turn controlled by the storage of vitamin A and enzymatic processes that synthesize and clear atRA. This has resulted in the need for robust and highly specific analytical methods to accurately quantify retinoids in diverse biological matrices. Tissue-specific differences in both the quantity of retinoids and background matrix interferences can confound the quantification of retinoids, and the bioanalysis requires high performance instrumentation, such as liquid chromatography mass-spectrometry (LC-MS). Successful bioanalysis of retinoids is further complicated by the innate structural instability of retinoids and their relatively high lipophilicity. Further, in vitro experiments with retinoids require attention to experimental design and interpretation to account for the instability of retinoids due to isomerization and degradation, sequential metabolism to numerous structurally similar metabolites, and substrate depletion during experiments. In addition, in vitro biological activity is often confounded by residual presence of retinoids in common biological reagents such as cell culture media. This chapter identifies common biological and analytical complexities in retinoid bioanalysis in diverse biological matrices, and in the use of retinoids in cell culture and metabolic incubations. In addition, this chapter highlights best practices for the successful detection and quantification of the vitamin A metabolome in a wide range of biological matrices.
Collapse
Affiliation(s)
- Lindsay C Czuba
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Guo Zhong
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - King C Yabut
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States.
| |
Collapse
|
21
|
Ventrella D, Forni M, Bacci ML, Annaert P. Non-clinical Models to Determine Drug Passage into Human Breast Milk. Curr Pharm Des 2020; 25:534-548. [PMID: 30894104 DOI: 10.2174/1381612825666190320165904] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Successful practice of clinical perinatal pharmacology requires a thorough understanding of the pronounced physiological changes during lactation and how these changes affect various drug disposition processes. In addition, pharmacokinetic processes unique to lactation have remained understudied. Hence, determination of drug disposition mechanisms in lactating women and their babies remains a domain with important knowledge gaps. Indeed, lack of data regarding infant risk during breastfeeding far too often results in discontinuation of breastfeeding and subsequent loss of all the associated benefits to the breastfed infant. In the absence of age-specific toxicity data, human lactation data alone are considered insufficient to rapidly generate the required evidence regarding risks associated with medication use during lactation. METHODS Systematic review of literature to summarize state-of-the art non-clinical approaches that have been developed to explore the mechanisms underlying drug milk excretion. RESULTS Several studies have reported methods to predict (to some extent) milk drug excretion rates based on physicochemical properties of the compounds. In vitro studies with primary mammary epithelial cells appear excellent approaches to determine transepithelial drug transport rates across the mammary epithelium. Several of these in vitro tools have been characterized in terms of transporter expression and activity as compared to the mammary gland tissue. In addition, with the advent of physiology-based pharmacokinetic (PBPK) modelling, these in vitro transport data may prove instrumental in predicting drug milk concentration time profiles prior to the availability of data from clinical lactation studies. In vivo studies in lactating animals have proven their utility in elucidating the mechanisms underlying drug milk excretion. CONCLUSION By combining various non-clinical tools (physicochemistry-based, in vitro and PBPK, in vivo animal) for drug milk excretion, valuable and unique information regarding drug milk concentrations during lactation can be obtained. The recently approved IMI project ConcePTION will address several of the challenges outlined in this review.
Collapse
Affiliation(s)
- Domenico Ventrella
- University of Bologna, Department of Veterinary Medical Science, 40064 Ozzano Emilia Bologna, Italy
| | - Monica Forni
- University of Bologna, Department of Veterinary Medical Science, 40064 Ozzano Emilia Bologna, Italy
| | - Maria Laura Bacci
- University of Bologna, Department of Veterinary Medical Science, 40064 Ozzano Emilia Bologna, Italy
| | - Pieter Annaert
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Herestraat 49-box 921, 3000 Leuven, Belgium
| |
Collapse
|
22
|
Mosquera N, Rodriguez-Trillo A, Blanco FJ, Mera-Varela A, Gonzalez A, Conde C. All-Trans Retinoic Acid Inhibits Migration and Invasiveness of Rheumatoid Fibroblast-Like Synoviocytes. J Pharmacol Exp Ther 2020; 372:185-192. [PMID: 31801802 DOI: 10.1124/jpet.119.261370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/26/2019] [Indexed: 12/23/2022] Open
Abstract
Fibroblast-like synoviocytes (FLSs) are pivotal in inflammation and joint damage of rheumatoid arthritis (RA). They acquire an active and aggressive phenotype, displaying increased migration and invasiveness and contributing to perpetuate synovial inflammation and destruction of cartilage and bone. The main current therapies of RA are focused against inflammatory factors and immune cells; however, a significant percentage of patients do not successfully respond. Combined treatments with drugs that control inflammation and that reverse the pathogenic phenotype of FLS could improve the prognosis of these patients. An unexplored area includes the retinoic acid, the main biologic retinoid, which is a candidate drug for many diseases but has reached clinical use only for a few. Here, we explored the effect of all-trans retinoic acid (ATRA) on the aggressive phenotype of FLS from patients with RA. RA FLSs were treated with ATRA, tumor necrosis factor (TNF), or TNF+ATRA, and cell migration and invasion were analyzed. In addition, a microarray analysis of expression, followed by gene-set analysis and quantitative polymerase chain reaction validation, was performed. We showed that ATRA induced a notable decrease in FLS migration and invasion that was accompanied by complex changes in gene expression. At supraphysiological doses, many of these effects were overridden or reverted by the concomitant presence of TNF. In conclusion, these results have demonstrated the therapeutic potential of retinoic acid on RA FLS provided TNF could be counterbalanced, either with high ATRA doses or with TNF inhibitors. SIGNIFICANCE STATEMENT: All-trans retinoic acid (ATRA) reduced the rheumatoid arthritis (RA) fibroblast-like synoviocyte migration and invasiveness and down-regulated gene expression of cell motility and migration genes. At supraphysiological doses, some of these effects were reverted by tumor necrosis factor. Therefore, ATRA could be an RA drug candidate that would require high doses or combined treatment with anti-inflammatory drugs.
Collapse
Affiliation(s)
- Nerea Mosquera
- Laboratorio de Reumatología Experimental y Observacional, y Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Travesía da Choupana s/n, Santiago de Compostela, Spain (N.M., A.R.-T., A.G., C.C.); Servicio de Reumatología, Instituto de Investigacion Biomedica de A Coruña (INIBIC)-Hospital Universitario A Coruña, A Xubias 84, A Coruña, Spain (F.J.B.); and Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Travesía da Choupana s/n, Santiago de Compostela, Spain (A.M.-V.)
| | - Angela Rodriguez-Trillo
- Laboratorio de Reumatología Experimental y Observacional, y Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Travesía da Choupana s/n, Santiago de Compostela, Spain (N.M., A.R.-T., A.G., C.C.); Servicio de Reumatología, Instituto de Investigacion Biomedica de A Coruña (INIBIC)-Hospital Universitario A Coruña, A Xubias 84, A Coruña, Spain (F.J.B.); and Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Travesía da Choupana s/n, Santiago de Compostela, Spain (A.M.-V.)
| | - Francisco J Blanco
- Laboratorio de Reumatología Experimental y Observacional, y Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Travesía da Choupana s/n, Santiago de Compostela, Spain (N.M., A.R.-T., A.G., C.C.); Servicio de Reumatología, Instituto de Investigacion Biomedica de A Coruña (INIBIC)-Hospital Universitario A Coruña, A Xubias 84, A Coruña, Spain (F.J.B.); and Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Travesía da Choupana s/n, Santiago de Compostela, Spain (A.M.-V.)
| | - Antonio Mera-Varela
- Laboratorio de Reumatología Experimental y Observacional, y Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Travesía da Choupana s/n, Santiago de Compostela, Spain (N.M., A.R.-T., A.G., C.C.); Servicio de Reumatología, Instituto de Investigacion Biomedica de A Coruña (INIBIC)-Hospital Universitario A Coruña, A Xubias 84, A Coruña, Spain (F.J.B.); and Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Travesía da Choupana s/n, Santiago de Compostela, Spain (A.M.-V.)
| | - Antonio Gonzalez
- Laboratorio de Reumatología Experimental y Observacional, y Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Travesía da Choupana s/n, Santiago de Compostela, Spain (N.M., A.R.-T., A.G., C.C.); Servicio de Reumatología, Instituto de Investigacion Biomedica de A Coruña (INIBIC)-Hospital Universitario A Coruña, A Xubias 84, A Coruña, Spain (F.J.B.); and Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Travesía da Choupana s/n, Santiago de Compostela, Spain (A.M.-V.)
| | - Carmen Conde
- Laboratorio de Reumatología Experimental y Observacional, y Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Travesía da Choupana s/n, Santiago de Compostela, Spain (N.M., A.R.-T., A.G., C.C.); Servicio de Reumatología, Instituto de Investigacion Biomedica de A Coruña (INIBIC)-Hospital Universitario A Coruña, A Xubias 84, A Coruña, Spain (F.J.B.); and Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Travesía da Choupana s/n, Santiago de Compostela, Spain (A.M.-V.)
| |
Collapse
|
23
|
Population Life-course exposure to health effects model (PLETHEM): An R package for PBPK modeling. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.comtox.2019.100115] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
24
|
Isoherranen N, Zhong G. Biochemical and physiological importance of the CYP26 retinoic acid hydroxylases. Pharmacol Ther 2019; 204:107400. [PMID: 31419517 PMCID: PMC6881548 DOI: 10.1016/j.pharmthera.2019.107400] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022]
Abstract
The Cytochrome P450 (CYP) family 26 enzymes contribute to retinoic acid (RA) metabolism and homeostasis in humans, mammals and other chordates. The three CYP26 family enzymes, CYP26A1, CYP26B1 and CYP26C1 have all been shown to metabolize all-trans-retinoic acid (atRA) it's 9-cisRA and 13-cisRA isomers and primary metabolites 4-OH-RA and 4-oxo-RA with high efficiency. While no crystal structures of CYP26 enzymes are available, the binding of various ligands has been extensively explored via homology modeling. All three CYP26 enzymes are inducible by treatment with atRA in various prenatal and postnatal tissues and cell types. However, current literature shows that in addition to regulation by atRA, CYP26 enzyme expression is also regulated by other endogenous processes and inflammatory cytokines. In humans and in animal models the expression patterns of CYP26 enzymes have been shown to be tissue and cell type specific, and the expression of the CYP26 enzymes is believed to regulate the formation of critical atRA concentration gradients in various tissue types. Yet, very little data exists on direct disease associations of altered CYP26 expression or activity. Nevertheless, data is emerging describing a variety of human genetic variations in the CYP26 enzymes that are associated with different pathologies. Interestingly, some of these genetic variants result in increased activity of the CYP26 enzymes potentially leading to complex gene-environment interactions due to variability in dietary intake of retinoids. This review highlights the current knowledge of structure-function of CYP26 enzymes and focuses on their role in human retinoid metabolism in different tissues.
Collapse
Affiliation(s)
- Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA.
| | - Guo Zhong
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| |
Collapse
|
25
|
Buechler MB, Kim KW, Onufer EJ, Williams JW, Little CC, Dominguez CX, Li Q, Sandoval W, Cooper JE, Harris CA, Junttila MR, Randolph GJ, Turley SJ. A Stromal Niche Defined by Expression of the Transcription Factor WT1 Mediates Programming and Homeostasis of Cavity-Resident Macrophages. Immunity 2019; 51:119-130.e5. [PMID: 31231034 DOI: 10.1016/j.immuni.2019.05.010] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 02/20/2019] [Accepted: 05/20/2019] [Indexed: 12/21/2022]
Abstract
Tissue-resident macrophages require specific milieus for the maintenance of defining gene-expression programs. Expression of the transcription factor GATA6 is required for the homeostasis, function and localization of peritoneal cavity-resident macrophages. Gata6 expression is maintained in a non-cell autonomous manner and is elicited by the vitamin A metabolite, retinoic acid. Here, we found that the GATA6 transcriptional program is a common feature of macrophages residing in all visceral body cavities. Retinoic acid-dependent and -independent hallmark genes of GATA6+ macrophages were induced by mesothelial and fibroblastic stromal cells that express the transcription factor Wilms' Tumor 1 (WT1), which drives the expression of two rate-limiting enzymes in retinol metabolism. Depletion of Wt1+ stromal cells reduced the frequency of GATA6+ macrophages in the peritoneal, pleural and pericardial cavities. Thus, Wt1+ mesothelial and fibroblastic stromal cells constitute essential niche components supporting the tissue-specifying transcriptional landscape and homeostasis of cavity-resident macrophages.
Collapse
Affiliation(s)
- Matthew B Buechler
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Ki-Wook Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Emily J Onufer
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jesse W Williams
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christine C Little
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Claudia X Dominguez
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Qingling Li
- Microchemistry and Proteomics, Genentech, South San Francisco, CA 94080, USA
| | - Wendy Sandoval
- Microchemistry and Proteomics, Genentech, South San Francisco, CA 94080, USA
| | - Jonathan E Cooper
- Translational Oncology, Genentech, South San Francisco, CA 94080, USA
| | - Charles A Harris
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Shannon J Turley
- Department of Cancer Immunology, Genentech, South San Francisco, CA 94080, USA.
| |
Collapse
|
26
|
Zhong G, Hogarth C, Snyder JM, Palau L, Topping T, Huang W, Czuba LC, LaFrance J, Ghiaur G, Isoherranen N. The retinoic acid hydroxylase Cyp26a1 has minor effects on postnatal vitamin A homeostasis, but is required for exogenous atRA clearance. J Biol Chem 2019; 294:11166-11179. [PMID: 31167781 DOI: 10.1074/jbc.ra119.009023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/01/2019] [Indexed: 12/20/2022] Open
Abstract
The all-trans-retinoic acid (atRA) hydroxylase Cyp26a1 is essential for embryonic development and may play a key role in regulating atRA clearance also in adults. We hypothesized that loss of Cyp26a1 activity via inducible knockout in juvenile or adult mice would result in decreased atRA clearance and increased tissue atRA concentrations and atRA-related adverse effects. To test these hypotheses, Cyp26a1 was knocked out in juvenile and adult male and female Cyp26a1 floxed mice using standard Cre-Lox technology and tamoxifen injections. Biochemical and histological methods were used to study the effects of global Cyp26a1 knockout. The Cyp26a1 knockout did not result in consistent histopathological changes in any major organs. Cyp26a1 -/- mice gained weight normally and exhibited no adverse phenotypes for up to 1 year after loss of Cyp26a1 expression. Similarly, atRA concentrations were not increased in the liver, testes, spleen, or serum of these mice, and the Cyp26a1 knockout did not cause compensatory induction of lecithin:retinol acetyltransferase (Lrat) or retinol dehydrogenase 11 (Rdh11) mRNA or a decrease in aldehyde dehydrogenase 1a1 (Aldh1a1) mRNA in the liver compared with tamoxifen-treated controls. However, the Cyp26a1 -/- mice showed increased bone marrow cellularity and decreased frequency of erythroid progenitor cells in the bone marrow consistent with a retinoid-induced myeloid skewing of hematopoiesis. In addition, the Cyp26a1 knockout decreased clearance of exogenous atRA by 70% and increased atRA half-life 6-fold. These findings demonstrate that despite lacking a major impact on endogenous atRA signaling, Cyp26a1 critically contributes as a barrier for exogenous atRA exposure.
Collapse
Affiliation(s)
- Guo Zhong
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195
| | - Cathryn Hogarth
- School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| | - Jessica M Snyder
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, Washington 98195
| | - Laura Palau
- School of Medicine, The Johns Hopkins University, Baltimore, Maryland 21231
| | - Traci Topping
- School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| | - Weize Huang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195
| | - Lindsay C Czuba
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195
| | - Jeffrey LaFrance
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195
| | - Gabriel Ghiaur
- School of Medicine, The Johns Hopkins University, Baltimore, Maryland 21231
| | - Nina Isoherranen
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington 98195
| |
Collapse
|
27
|
Jiang L, Dong R, Ying M, He Q, Cao J, Yang B. Immune cells in the tumour: new routes of retinoids for chemoprevention and chemotherapeutics. Br J Pharmacol 2018; 175:4285-4294. [PMID: 30298911 DOI: 10.1111/bph.14511] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/18/2018] [Accepted: 09/24/2018] [Indexed: 01/27/2023] Open
Abstract
Retinoids, vitamin A and its natural and synthetic analogues have various functions, including being involved in cell proliferation and differentiation and participating in the formation of vertebrate morphology. In addition, they may activate certain tumour suppressor genes that then act as tumour inhibitors. In the past decades, retinoids have been regarded as promising chemotherapeutic and chemopreventive agents; however, their mechanisms are still not fully understood. Immune cells that participate in or are associated with the immune response play vital roles in the initiation and development of many cancers. Interestingly, recent studies have demonstrated that retinoids can also exert various effects on immune cells including macrophages, T cells and dendritic cells in tumour tissues to execute anti-tumour actions, providing new insights into chemoprevention and chemotherapeutics. In this review, we focus on the effects of retinoids on immune cells in the tumour, which may provide new approaches for antineoplastic strategies.
Collapse
Affiliation(s)
- Li Jiang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Rong Dong
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Meidan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ji Cao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
28
|
All-Trans Retinoic Acid Enhances Matrix Metalloproteinase 2 Expression and Secretion in Human Myeloid Leukemia THP-1 Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5971080. [PMID: 30225259 PMCID: PMC6129365 DOI: 10.1155/2018/5971080] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/13/2018] [Indexed: 12/15/2022]
Abstract
All-trans retinoic acid (ATRA) is an effective drug for the induction therapy of acute promyelocytic leukemia. However, the treatment is associated with adverse events such as retinoic acid syndrome (RAS) in some patients, whose histologic characteristics included organ infiltration by leukemic cells. Matrix metalloproteinase 2 (MMP-2) is often upregulated in tumor cells and plays a role in tumor cell migration and invasion by degrading the extracellular matrix. In this study, we examined the possible modulatory effects of ATRA on MMP-2 expression and secretion in human myeloid leukemia cell line THP-1. The cells were treated with various concentrations of ATRA, and MMP-2 expression and secretion were examined. MMP-2 expression and secretion started to increase with ATRA concentration as low as 0.1 nM and gradually increased thereafter. Agonists of retinoic acid receptor (RAR) or retinoid X receptor (RXR) alone could enhance MMP-2 secretion, and RAR or RXR antagonists alone could reverse ATRA-induced MMP-2 secretion. ATRA increased intracellular calcium ion levels, and a calcium-channel blocker inhibited ATRA-induced MMP-2 secretion. Dexamethasone suppressed ATRA-induced MMP-2 secretion. Our results suggest that ATRA enhances MMP-2 expression and secretion in human myeloid leukemia THP-1 cells in a calcium ion dependent manner through RAR/RXR signaling pathways, and this enhanced expression and secretion may be associated with the possible mechanisms of RAS.
Collapse
|
29
|
Rubinow KB. An intracrine view of sex steroids, immunity, and metabolic regulation. Mol Metab 2018; 15:92-103. [PMID: 29551633 PMCID: PMC6066741 DOI: 10.1016/j.molmet.2018.03.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 02/26/2018] [Accepted: 03/01/2018] [Indexed: 12/21/2022] Open
Abstract
Background Over the past two decades, parallel recognition has grown of the importance of both sex steroids and immune activity in metabolic regulation. More recently, these discrete areas have been integrated in studies examining the metabolic effects of sex steroid immunomodulation. Implicit in these studies has been a traditional, endocrine model of sex steroid delivery from the gonads to target cells, including immune cells. Thus, research to date has focused on the metabolic effects of sex steroid receptor signaling in immune cells. This endocrine model, however, overlooks the extensive capacity of immune cells to generate and metabolize sex steroids, enabling the production of sex steroids for intracrine signaling – that is, sex steroid production for signaling within the cell of origin. Intracrine function allows highly cell-autonomous regulation of sex steroid exposure, and sex steroid secretion by immune cells could confer paracrine signaling effects in neighboring cells within metabolic tissues. In this review, immune cell intracrinology will denote sex steroid production within immune cells for either intracrine or paracrine signaling. This intracrine capacity of immune cells has been well established, and prior work has supported its importance in autoimmune disorders, trauma, and cancer. The potential relevance of immune cell intracrine function to the regulation of energy balance, body weight, body composition, and insulin sensitivity has yet to be explored. Scope of review The following review will detail findings to date regarding the steroidogenic and steroid metabolizing capacity of immune cells, the regulation of immune cell intracrine function, and the biological effects of immune-derived sex steroids, including the clinical relevance of immune cell intracrinology in fields other than metabolism. These findings will serve as the basis for a proposed model of immune cell intracrinology constituting a new frontier in metabolism research. Major conclusions The development of highly sensitive mass spectrometric methods for sex steroid measurement and quantitation of metabolic flux now allows unprecedented ability to interrogate sex steroid production, metabolism and secretion by immune cells. Immune cell intracrinology could reveal key mechanisms underlying immune cell-mediated metabolic regulation. Sex steroids exert immunomodulatory effects that may influence metabolic health. Immune cells can synthesize, modify, and metabolize sex steroids. Immune cell-derived sex steroids may play intracrine, autocrine, paracrine, and possibly even endocrine roles. Immune cell steroidogenesis is a largely unexplored area of metabolism research.
Collapse
Affiliation(s)
- Katya B Rubinow
- Diabetes Institute, Department of Medicine, University of Washington, School of Medicine, 850 Republican St., Box 358055, Seattle, WA 98109, USA.
| |
Collapse
|
30
|
Jing J, Petroff R, Shum S, Crouthamel B, Topletz AR, Grant KS, Burbacher TM, Isoherranen N. Toxicokinetics and Physiologically Based Pharmacokinetic Modeling of the Shellfish Toxin Domoic Acid in Nonhuman Primates. Drug Metab Dispos 2018; 46:155-165. [PMID: 29150543 PMCID: PMC5776359 DOI: 10.1124/dmd.117.078485] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/13/2017] [Indexed: 12/16/2022] Open
Abstract
Domoic acid (DA), a neurotoxin, is produced by marine algae and has caused toxications worldwide in animals and humans. However, the toxicokinetics of DA have not been fully evaluated, and information is missing on the disposition of DA following oral exposures at doses that are considered safe for human consumption. In this study, toxicokinetics of DA were investigated in cynomolgus monkeys following single doses of 5 µg/kg DA intravenously, 0.075 mg/kg DA orally, and 0.15 mg/kg DA orally. After intravenous dosing, DA had a systemic clearance of 124 ± 71 (ml/h)/kg, volume of distribution at steady state of 131 ± 71 ml/kg and elimination half-life of 1.2 ± 1.1 hours. However, following oral dosing, the average terminal half-life of DA was 11.3 ± 2.4 hours, indicating that DA disposition follows flip-flop kinetics with slow, rate-limiting absorption. The absorption of DA was low after oral dosing with absolute bioavailability of 6% ± 4%. The renal clearance of DA was variable [21-152 (ml/h)/kg] with 42% ± 11% of the intravenous DA dose recovered in urine. A physiologically based pharmacokinetic model was developed for DA in monkeys and humans that replicated the flip-flop kinetics observed after oral administration and allowed simulation of urinary excretion and brain and kidney distribution of DA following intravenous and oral dosing. This study is the first to characterize DA disposition at exposure levels close to the current estimated tolerable daily intake and to mechanistically model DA disposition in a model species, providing important information of the toxicokinetics of DA for human safety assessment.
Collapse
Affiliation(s)
- Jing Jing
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Rebekah Petroff
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Sara Shum
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Brenda Crouthamel
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Ariel R Topletz
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Kimberly S Grant
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Thomas M Burbacher
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| | - Nina Isoherranen
- Department of Pharmaceutics (J.J., S.S., A.R.T., N.I.), Department of and Environmental and Occupational Health Sciences (R.P., B.C., K.S.G., T.M.B.), Center on Human Development and Disability (K.S.G., T.M.B.), and Infant Primate Research Laboratory, Washington National Primate Research Center, University of Washington, Seattle, Washington (K.S.G., T.M.B.)
| |
Collapse
|
31
|
Prediction of drug–drug interaction potential using physiologically based pharmacokinetic modeling. Arch Pharm Res 2017; 40:1356-1379. [DOI: 10.1007/s12272-017-0976-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 10/19/2017] [Indexed: 12/22/2022]
|