1
|
Nakagawa A, Morwood K, Johnson K, Berra L, Boal L, Azar S, Huang M, Heeney MM, Bloch DB, Ichinose F. Screen of the ReFRAME Compound Library for Therapeutic Agents to Prevent Red Blood Cell Sickling Using an Improved High Throughput Sickling Assay. ACS OMEGA 2025; 10:16497-16505. [PMID: 40321528 PMCID: PMC12044578 DOI: 10.1021/acsomega.4c11077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/30/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025]
Abstract
Sickle cell disease (SCD) is an autosomal recessive disorder of blood characterized by a mutation in the β chain of hemoglobin (Hb), leading to the production of sickle Hb (HbS). In SCD, under low oxygen conditions, red blood cells (RBCs) containing HbS form a characteristic "sickle" shape, resulting in chronic hemolytic anemia and acute vaso-occlusive crises. Current therapies for SCD have limitations in efficacy or availability, highlighting the need for new anti-sickling drugs. To facilitate the discovery of new anti-sickling compounds, we previously developed a high throughput sickling assay, which permits rapid screening of thousands of compounds for the ability to inhibit RBC sickling. In this study, we improved the sickling assay by optimizing the assay condition and expanded our screening efforts by evaluating the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) compound library, which contains approximately 2.5 times more compounds than previously screened. We were able to increase the number of blood samples that were adequate for identifying anti-sickling compounds in the improved sickling assay and identified voxelotor and SNS-314 as compounds that successfully prevented sickling. The improved sickling assay will increase access to valuable blood samples from SCD volunteers, providing more opportunities to develop anti-sickling compounds for treating SCD.
Collapse
Affiliation(s)
- Akito Nakagawa
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Kaycie Morwood
- The
Calibr-Skaggs Institute for Innovative Medicines, Scripps Research, San Diego, California 92037, United States
| | - Kristen Johnson
- The
Calibr-Skaggs Institute for Innovative Medicines, Scripps Research, San Diego, California 92037, United States
| | - Lorenzo Berra
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Lauren Boal
- Division
of Pediatric Hematology and Oncology, Massachusetts
General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Sharl Azar
- Comprehensive
Sickle Cell Disease Treatment Center, Massachusetts
General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Mary Huang
- Division
of Pediatric Hematology and Oncology, Massachusetts
General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Matthew M. Heeney
- Dana-Farber/Boston
Children’s Cancer and Blood Disorders Center and Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Donald B. Bloch
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
- Division
of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital and Harvard Medical
School, Boston, Massachusetts 02114, United States
| | - Fumito Ichinose
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
2
|
Cavazzana M, Corsia A, Brusson M, Miccio A, Semeraro M. Treating Sickle Cell Disease: Gene Therapy Approaches. Annu Rev Pharmacol Toxicol 2025; 65:397-413. [PMID: 39259977 DOI: 10.1146/annurev-pharmtox-022124-022000] [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: 09/13/2024]
Abstract
Sickle cell disease (SCD) is a hereditary blood disorder characterized by the presence of abnormal hemoglobin molecules and thus distortion (sickling) of the red blood cells. SCD causes chronic pain and organ damage and shortens life expectancy. Gene therapy emerges as a potentially curative approach for people with SCD who lack a matched sibling donor for hematopoietic stem cell transplantation. Here, we review recent progress in gene therapy for SCD and focus on innovative technologies that target the genetic roots of the disease. We also review the challenges associated with gene therapy, including oncogenic risks, and the need for refined delivery methods. Despite these hurdles, the rapidly evolving landscape of gene therapy for SCD raises hope for a paradigm shift in the treatment of this debilitating disease. As research progresses, a deeper understanding of the molecular mechanisms involved and continuous improvements in gene-editing technologies promise to bring gene therapy for SCD closer to mainstream clinical application, offering a transformative, curative option for patients with this genetic disorder.
Collapse
Affiliation(s)
- Marina Cavazzana
- Imagine Institute, Paris, France
- Centre d'Investigation Clinique en Biothérapie, INSERM UMR1163, Paris, France
- Département de Biothérapie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France;
- Université Paris Cité, Paris, France
| | - Alice Corsia
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
- Université Paris Cité, Paris, France
| | - Megane Brusson
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Annarita Miccio
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Michaela Semeraro
- EA 7323, Université Paris Cité, Pharmacologie et Évaluations Thérapeutiques chez l'Enfant et la Femme Enceinte, Paris, France
- Centre d'Investigation Clinique and Unité de Recherche Clinique, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Paris Cité, Paris, France
| |
Collapse
|
3
|
Klopp C, Zhang X, Campbell MK, Kvaskoff D, Struwe MA, Warren CR, Bajrami B, Scheidig AJ, Jones AK, Clement B. mARC1 Is the Main Contributor to Metabolic Reduction of N-Hydroxyurea. J Med Chem 2024; 67:18090-18097. [PMID: 39397364 PMCID: PMC11513889 DOI: 10.1021/acs.jmedchem.4c01148] [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: 05/17/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024]
Abstract
N-Hydroxyurea has been known since the 1960s as an antiproliferative drug and is used both in oncology and for treatment of hematological disorders such as sickle cell anemia where very high daily doses are administered. It is assumed that the cellular effect of N-hydroxyurea is caused by inhibition of ribonucleotide reductase, while alternative mechanisms, e.g., generation of nitric oxide, have also been proposed. Despite its many therapeutic applications, the metabolism of hydroxyurea is largely unexplored. The major elimination pathway of N-hydroxyurea is the reduction to urea. Since the mitochondrial amidoxime reducing component (mARC) is known for its N-reductive activity, we investigated the reduction of NHU by this enzyme system. This study presents in vitro and in vivo evidence that this reductive biotransformation is specifically mediated by the mARC1. Inactivation by mARC1 is a possible explanation for the high doses of NHU required for treatment.
Collapse
Affiliation(s)
- Cathrin Klopp
- Zoological
Institute − Structural Biology, Kiel
University 24118, Kiel, Germany
- Pharmaceutical
Institute − Medicinal Chemistry, Kiel University 24118, Kiel, Germany
| | - Xiaomei Zhang
- Department
of Cardiometabolic Disease Research, Boehringer
Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Morgan K. Campbell
- Department
of Cardiometabolic Disease Research, Boehringer
Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - David Kvaskoff
- Department
of Drug Discovery Sciences, Discovery Science Technologies (DK, BB), Boehringer Ingelheim Pharma GmbH & Co. 88400, Biberach
an der Riss, Germany
| | - Michel A. Struwe
- Zoological
Institute − Structural Biology, Kiel
University 24118, Kiel, Germany
- Pharmaceutical
Institute − Medicinal Chemistry, Kiel University 24118, Kiel, Germany
| | - Curtis R. Warren
- Department
of Cardiometabolic Disease Research, Boehringer
Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Besnik Bajrami
- Department
of Drug Discovery Sciences, Discovery Science Technologies (DK, BB), Boehringer Ingelheim Pharma GmbH & Co. 88400, Biberach
an der Riss, Germany
| | - Axel J. Scheidig
- Zoological
Institute − Structural Biology, Kiel
University 24118, Kiel, Germany
| | - Amanda K. Jones
- Department
of Cardiometabolic Disease Research, Boehringer
Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Bernd Clement
- Pharmaceutical
Institute − Medicinal Chemistry, Kiel University 24118, Kiel, Germany
| |
Collapse
|
4
|
Gonçalves E, Smaoui S, Brito M, Oliveira JM, Arez AP, Tavares L. Sickle Cell Disease: Current Drug Treatments and Functional Foods with Therapeutic Potential. Curr Issues Mol Biol 2024; 46:5845-5865. [PMID: 38921020 PMCID: PMC11202234 DOI: 10.3390/cimb46060349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Sickle cell anemia (SCA), the most common form of sickle cell disease (SCD), is a genetic blood disorder. Red blood cells break down prematurely, causing anemia and often blocking blood vessels, leading to chronic pain, organ damage, and increased infection risk. SCD arises from a single-nucleotide mutation in the β-globin gene, substituting glutamic acid with valine in the β-globin chain. This review examines treatments evaluated through randomized controlled trials for managing SCD, analyzes the potential of functional foods (dietary components with health benefits) as a complementary strategy, and explores the use of bioactive compounds as functional food ingredients. While randomized trials show promise for certain drugs, functional foods enriched with bioactive compounds also hold therapeutic potential. Further research is needed to confirm clinical efficacy, optimal dosages, and specific effects of these compounds on SCD, potentially offering a cost-effective and accessible approach to managing the disease.
Collapse
Affiliation(s)
- Elisângela Gonçalves
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation Towards Global Health (LA-REAL), Institute of Hygiene and Tropical Medicine, (IHMT), NOVA University of Lisbon (UNL) 1349-008 Lisbon, Portugal; (E.G.); (A.P.A.)
| | - Slim Smaoui
- Laboratory of Microbial and Enzymes Biotechnology and Biomolecules (LBMEB), Centre of Biotechnology of Sfax (CBS), University of Sfax-Tunisia, Road of Sidi Mansour Km 6, P.O. Box 1177, Sfax 3018, Tunisia;
| | - Miguel Brito
- Health Research Centre of Angola (CISA), Caxito, Angola;
- H&TRC—Health & Technology Research Center, Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-092 Lisbon, Portugal
| | - J. M. Oliveira
- School of Design, Management and Production Technologies Northern Aveiro, University of Aveiro, Estrada do Cercal, 449, 3810-193 Oliveira de Azeméis, Portugal;
- EMaRT Group—Emerging Materials, Research, Technology, University of Aveiro, 3810-193 Aveiro, Portugal
- CICECO Aveiro—Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Ana Paula Arez
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation Towards Global Health (LA-REAL), Institute of Hygiene and Tropical Medicine, (IHMT), NOVA University of Lisbon (UNL) 1349-008 Lisbon, Portugal; (E.G.); (A.P.A.)
| | - Loleny Tavares
- School of Design, Management and Production Technologies Northern Aveiro, University of Aveiro, Estrada do Cercal, 449, 3810-193 Oliveira de Azeméis, Portugal;
- EMaRT Group—Emerging Materials, Research, Technology, University of Aveiro, 3810-193 Aveiro, Portugal
- CICECO Aveiro—Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
5
|
Haijer F, Koets-Shajari S, Heegsma J, Serna-Salas S, Blokzijl T, Buist-Homan M, Moshage H, Faber KN. Hydroxyurea attenuates hepatic stellate cell proliferation in vitro and liver fibrogenesis in vivo. FASEB J 2023; 37:e23124. [PMID: 37552464 DOI: 10.1096/fj.202300920r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/24/2023] [Accepted: 07/24/2023] [Indexed: 08/09/2023]
Abstract
Liver fibrosis results from excessive proliferation of, and collagen production by hepatic stellate cells (HSCs) that is caused by chronic liver injury. No drugs are available to cure liver fibrosis. Hydroxyurea is an anti-proliferative drug that is used in benign and malignant disorders. Here, we studied the effect of hydroxyurea on primary HSCs and its anti-fibrotic effect in the CCl4 mouse model of liver fibrosis. Primary rat HSCs were cultured in the absence or presence of hydroxyurea (0.1-1.0 mmol/L). CCl4 or vehicle was administered to C57BL/6/J mice for 4 weeks, with or without hydroxyurea (100 mg/kg/day) co-treatment. We used real-time cell proliferation analysis, Oil Red O (lipid droplet) staining, immunohistochemistry, Acridine Orange staining (apoptosis), Sytox green staining (necrosis), RT-qPCR, ELISA, and Western Blotting for analysis. Hydroxyurea dose-dependently suppressed lipid droplet-loss and mRNA levels of Col1α1 and Acta2 in transdifferentiating HSCs. In fully-activated HSCs, hydroxyurea dose-dependently attenuated PCNA protein levels and BrdU incorporation, but did not reverse Col1α1 and Acta2 mRNA expression. Hydroxyurea did not induce apoptosis or necrosis in HSCs or hepatocytes. Hydroxyurea suppressed accumulation of desmin-positive HSCs and hepatic collagen deposition after CCl4 treatment. CCl4 -induced regenerative hepatocyte proliferation, Col1α1 and Acta2 mRNA expression and α-SMA protein levels were not affected. This study demonstrates that hydroxyurea inhibits HSC proliferation in vitro and attenuates early development of liver fibrosis in vivo, while preserving hepatocyte regeneration after toxic insults by CCl4. Thus, hydroxyurea may have therapeutic value against liver fibrosis.
Collapse
Affiliation(s)
- Floris Haijer
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Shiva Koets-Shajari
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Janette Heegsma
- Department Laboratory Medicine, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sandra Serna-Salas
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tjasso Blokzijl
- Department Laboratory Medicine, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manon Buist-Homan
- Department Laboratory Medicine, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department Laboratory Medicine, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department Laboratory Medicine, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Xu B, Yang Z, Zhang X, Liu Z, Huang Y, Ding X, Chu J, Peng T, Wu D, Jin C, Li W, Cai B, Wang X. 16S rDNA sequencing combined with metabolomics profiling with multi-index scoring method reveals the mechanism of salt-processed Semen Cuscuta in Bushen Antai mixture on kidney yang deficiency syndrome. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1216:123602. [PMID: 36652816 DOI: 10.1016/j.jchromb.2023.123602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/22/2022] [Accepted: 01/08/2023] [Indexed: 01/11/2023]
Abstract
Kidney yang deficiency syndrome (KYDS) is a classic syndrome of traditional Chinese medicine (TCM). The salt-processed product of Semen Cuscuta (YP) is the monarch drug in Bushen Antai Mixture (BAM), can improve the reproductive dysfunction caused by KYDS, and the effect is better than that of raw products of Semen Cuscuta (SP). However, its mechanism is not completely clear yet. In this study, an integrated strategy combining untargeted metabolomics with microbiology was used to explore the mechanism of YP in the BAM improving KYDS. 16S rDNA gene sequencing showed that BAM containing YP (Y-BAM) had a significantly better regulatory effect on Desulfobacterota and Desulfovibrionaceae_unclassified than BAM containing SP (S-BAM). Untargeted metabolomics studies showed that Y-BAM significantly regulated 4 metabolites and 4 metabolic pathways. In addition, multi-index analysis showed that the effect of Y-BAM on arachidonic acid metabolism, tyrosine metabolism, purine metabolism, fructose and mannose metabolism and total metabolism was closer to that of the control group compared to S-BAM. The analysis of serum biochemical indexes showed that Y-BAM had more significant regulating effect on the levels of luteinizing hormone (LH), follicle stimulating hormone (FSH), testosterone (T) and superoxide dismutase (SOD) in serum of KYDS rats compared to S-BAM. Spearman correlation analysis showed that there was a significant correlation between intestinal microorganisms and metabolites and serum biochemical indexes. For example, Desulfovibrionaceae_unclassified was positively correlated with arachidonic acid, and negatively correlated with SOD and LH. This study suggests that YP may enhance the regulation of intestinal flora and endogenous metabolism of KYDS, so that BAM shows a better therapeutic effect on KYDS, which also reasonably explains why BAM uses Semen Cuscuta stir-baked with salt solution.
Collapse
Affiliation(s)
- Baiyang Xu
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Zhitong Yang
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Xue Zhang
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Zilu Liu
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yu Huang
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Ximeng Ding
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jijun Chu
- Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Hefei 230012, China
| | - Tangyi Peng
- The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, China
| | - Deling Wu
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Chuanshan Jin
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; Heritage Base of TCM Processing Technolovgy of NATCM, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Hefei 230012, China
| | - Weidong Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Baochang Cai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaoli Wang
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei 230012, China; Heritage Base of TCM Processing Technolovgy of NATCM, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Traditional Chinese Medicine Decoction Pieces of New Manufacturing Technology, Hefei 230012, China.
| |
Collapse
|
7
|
Coache D, Friciu M, Bernine Marcellin R, Bonnemain L, Viau A, Roullin VG, Forest JM, Leclair G. Stability evaluation of compounded hydroxyurea 100 mg/mL oral liquids using a novel analytical method involving chemical derivatization. PLoS One 2022; 17:e0270206. [PMID: 35749410 PMCID: PMC9231814 DOI: 10.1371/journal.pone.0270206] [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: 03/10/2022] [Accepted: 06/06/2022] [Indexed: 11/21/2022] Open
Abstract
This study assessed the stability of six extemporaneously compounded hydroxyurea oral liquids stored at room temperature. Hydroxyurea oral liquids (100 mg/mL) were prepared using three different mixing methods (mortar, mixer or QuartetRx) from either bulk powder, capsule content, or whole capsules. Two brands of capsules were tested in this study. All formulations were stored at room temperature (25°C / 60% RH) in amber plastic bottles for 90 days and amber plastic syringes for 14 days. Physical stability was assessed visually, while chemical stability was evaluated using a stability-indicating high-performance liquid chromatography method. Chemical derivatization with xanthydrol allowed the retention of hydroxyurea on a reverse-phase column. At least 93.9% and 97.0% of the initial concentration of hydroxyurea remained after 90 days in bottles and 14 days in syringes, respectively. There were no visual changes in formulations over the study period. Changes in pH up to 1.6 units were observed after 90 days of storage and were explained most likely by an ammonium generating degradation pathway. Ammonium was quantified and remained within safe levels in each HU 100 mg/mL oral preparations. Hydroxyurea oral liquids were all stable for 90 days in amber plastic bottles and 14 days in amber plastic syringes.
Collapse
Affiliation(s)
- Daphné Coache
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Mihaela Friciu
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | | | - Lola Bonnemain
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Annie Viau
- Sainte-Justine University Hospital Center, Montréal, Québec, Canada
| | - V Gaëlle Roullin
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Marc Forest
- Sainte-Justine University Hospital Center, Montréal, Québec, Canada
| | - Grégoire Leclair
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
8
|
Epicatechin exerts dual action to shield sickling and hydroxyurea-induced myelosuppression: Implication in sickle cell anemia management. Toxicol Appl Pharmacol 2022; 449:116113. [PMID: 35691369 DOI: 10.1016/j.taap.2022.116113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/04/2022] [Accepted: 06/04/2022] [Indexed: 11/20/2022]
Abstract
Hydroxyurea (HU) is the key drug to treat Sickle cell anemia (SCA). However, its treatment is associated with the liability of myelosuppression. The present study aimed to investigate the potential of epicatechin as a supplementation therapy for the symptomatic management of SCA under HU therapy. A panel of experiments were performed at first to observe epicatechin's effect on sickling and hemolytic behaviour using SCA patient's blood (ex vivo). Thereafter, the effect of HU in the presence or absence of epicatechin was investigated on cytokine inhibition in rat splenocytes (ex vivo) as well as alterations in hematological parameters and kidney function tests in rats (in vivo). Then, any effect of epicatechin on pharmacokinetic modulation of HU in rats was elucidated along with the underlying mechanism using a battery of in vitro and in vivo models. Epicatechin exhibited potent action on anti-sickling, polymerization inhibition, and erythrocyte membrane stability. It did not show any inherent hemolytic activity and reduced TNF-α level during concomitant administration with HU. Based on hematological changes in rats, epicatechin treatment aided to the beneficial effect of HU and prevented the treatment-linked disadvantageous effects of HU like neutropenia. The plasma exposure of HU was significantly augmented in rats upon simultaneous oral administration of epicatechin with HU. Down-regulation of Oatp1b2 and catalase possibly contributed to the pharmacokinetic interaction of HU. Epicatechin is found to be a promising candidate and should be explored at a reduced dose level of HU towards offsetting the dose-dependent myelosuppressive effect of HU under the frame of supplementation therapy in SCA.
Collapse
|
9
|
Hematopoietic Stem Cell Gene-Addition/Editing Therapy in Sickle Cell Disease. Cells 2022; 11:cells11111843. [PMID: 35681538 PMCID: PMC9180595 DOI: 10.3390/cells11111843] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/29/2022] [Accepted: 06/02/2022] [Indexed: 12/17/2022] Open
Abstract
Autologous hematopoietic stem cell (HSC)-targeted gene therapy provides a one-time cure for various genetic diseases including sickle cell disease (SCD) and β-thalassemia. SCD is caused by a point mutation (20A > T) in the β-globin gene. Since SCD is the most common single-gene disorder, curing SCD is a primary goal in HSC gene therapy. β-thalassemia results from either the absence or the reduction of β-globin expression, and it can be cured using similar strategies. In HSC gene-addition therapy, patient CD34+ HSCs are genetically modified by adding a therapeutic β-globin gene with lentiviral transduction, followed by autologous transplantation. Alternatively, novel gene-editing therapies allow for the correction of the mutated β-globin gene, instead of addition. Furthermore, these diseases can be cured by γ-globin induction based on gene addition/editing in HSCs. In this review, we discuss HSC-targeted gene therapy in SCD with gene addition as well as gene editing.
Collapse
|
10
|
Mathematical Modeling of Hydroxyurea Therapy in Individuals with Sickle Cell Disease. Pharmaceutics 2022; 14:pharmaceutics14051065. [PMID: 35631651 PMCID: PMC9144420 DOI: 10.3390/pharmaceutics14051065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 11/19/2022] Open
Abstract
Sickle cell disease (SCD) is a chronic hemolytic anemia affecting millions worldwide with acute and chronic clinical manifestations and early mortality. While hydroxyurea (HU) and other treatment strategies managed to ameliorate disease severity, high inter-individual variability in clinical response and a lack of an ability to predict those variations need to be addressed to maximize the clinical efficacy of HU. We developed pharmacokinetics (PK) and pharmacodynamics (PD) models to study the dosing, efficacy, toxicity, and clinical response of HU treatment in more than eighty children with SCD. The clinical PK parameters were used to model the HU plasma concentration for a 24 h period, and the estimated daily average HU plasma concentration was used as an input to our PD models with approximately 1 to 9 years of data connecting drug exposure with drug response. We modeled the biomarkers mean cell volume and fetal hemoglobin to study treatment efficacy. For myelosuppression, we modeled red blood cells and absolute neutrophil count. Our models provided excellent fits for individuals with known or correctly inferred adherence. Our models can be used to determine the optimal dosing regimens and study the effect of non-adherence on HU-treated individuals.
Collapse
|
11
|
Nakagawa A, Cooper MK, Kost-Alimova M, Berstler J, Yu B, Berra L, Klings ES, Huang MS, Heeney MM, Bloch DB, Zapol WM. High-Throughput Assay to Screen Small Molecules for Their Ability to Prevent Sickling of Red Blood Cells. ACS OMEGA 2022; 7:14009-14016. [PMID: 35559170 PMCID: PMC9089379 DOI: 10.1021/acsomega.2c00541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/06/2022] [Indexed: 06/15/2023]
Abstract
Sickle cell disease (SCD) is an inherited disorder of hemoglobin (Hb); approximately 300,000 babies are born worldwide with SCD each year. In SCD, fibers of polymerized sickle Hb (HbS) form in red blood cells (RBCs), which cause RBCs to develop their characteristic "sickled" shape, resulting in hemolytic anemia and numerous vascular complications including vaso-occlusive crises. The development of novel antisickling compounds will provide new therapeutic options for patients with SCD. We developed a high-throughput "sickling assay" that is based on an automated high-content imaging system to quantify the effects of hypoxia on the shape and size of RBCs from HbSS SCD patients (SS RBCs). We used this assay to screen thousands of compounds for their ability to inhibit sickling. In the assay, voxelotor (an FDA-approved medication used to treat SCD) prevented sickling with a z'-factor > 0.4, suggesting that the assay is capable of identifying compounds that inhibit sickling. We screened the Broad Repurposing Library of 5393 compounds for their ability to prevent sickling in 4% oxygen/96% nitrogen. We identified two compounds, SNS-314 mesylate and voxelotor itself, that successfully prevented sickling. SNS-314 mesylate prevented sickling in the absence of oxygen, while voxelotor did not, suggesting that SNS-314 mesylate acts by a mechanism that is different from that of voxelotor. The sickling assay described in this study will permit the identification of additional, novel antisickling compounds, which will potentially expand the therapeutic options for SCD.
Collapse
Affiliation(s)
- Akito Nakagawa
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Marissa K. Cooper
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Maria Kost-Alimova
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - James Berstler
- Center
for the Development of Therapeutics, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Binglan Yu
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Lorenzo Berra
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Elizabeth S. Klings
- Pulmonary
Center, Boston University School of Medicine, Boston, Massachusetts 02118, United States
| | - Mary S. Huang
- Division
of Pediatric Hematology and Oncology, Massachusetts
General Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Matthew M. Heeney
- Division
of Hematology/Oncology, Boston Children’s
Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Donald B. Bloch
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
- Division
of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital and Harvard Medical
School, Boston, Massachusetts 02114, United States
| | - Warren M. Zapol
- Anesthesia
Center for Critical Care Research, Department of Anesthesia, Critical
Care, and Pain Medicine, Massachusetts General
Hospital and Harvard Medical School, Boston, Massachusetts 02114, United States
| |
Collapse
|
12
|
Gour A, Kotwal P, Dogra A, Kour D, Dhiman S, Kumar A, Digra SK, Kumar A, Singh G, Nandi U. Investigating the Potential Use of Andrographolide as a Coadjuvant in Sickle Cell Anemia Therapy. ACS OMEGA 2022; 7:12765-12771. [PMID: 35474831 PMCID: PMC9026017 DOI: 10.1021/acsomega.1c07339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
Andrographolide is one of the main active principles of Andrographolide paniculata and has been extensively explored for its therapeutic use. Current studies focus on phytotherapeutics-based adjuvant therapy to symptomatically treat sickle cell anemia (SCA) as there is no specific drug/gene therapy available to date. The present study aimed to explore the potential of andrographolide as an adjuvant therapy for SCA in the presence or absence of hydroxyurea (HU), a key drug for SCA treatment. A panel of ex vivo and in vivo experimentations was performed to explore the antisickling activity of andrographolide, followed by evaluating pharmacokinetic and pharmacodynamic (PK/PD) activities in the presence of HU. Andrographolide showed significant antisickling activity using blood from SCA patients (ex vivo) and did not show any deleterious effect to cause hemolysis using rat blood (ex vivo). It displayed a substantial decrease in HU-induced decline in splenic lymphocyte proliferation and cytokine level (TNF-α and IFN-γ) using rat splenocytes (ex vivo). Concomitant oral administration of andrographolide with HU in rats for 15 days exhibited a noticeable improvement in the RBC count and hemoglobin levels comparable to the efficacy of l-glutamine (in vivo). Simultaneous administration of andrographolide with HU caused no marked effect on any pharmacokinetic parameters of HU except the highest plasma concentration of HU and its corresponding time point, which significantly dropped and delayed, respectively (in vivo). No considerable effect of andrographolide was observed on urease and horseradish peroxidase activity (in vitro). Overall, results suggest that andrographolide has several beneficial actions to be an adjuvant therapy to symptomatically manage SCA, but it should be avoided during the prescribed therapy of HU.
Collapse
Affiliation(s)
- Abhishek Gour
- PK-PD
Toxicology Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pankul Kotwal
- PK-PD
Toxicology Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ashish Dogra
- PK-PD
Toxicology Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Dilpreet Kour
- PK-PD
Toxicology Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sumit Dhiman
- PK-PD
Toxicology Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
| | - Amit Kumar
- Natural
Product and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | | | - Ajay Kumar
- PK-PD
Toxicology Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Gurdarshan Singh
- PK-PD
Toxicology Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Utpal Nandi
- PK-PD
Toxicology Division, CSIR-Indian Institute
of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
13
|
Ben Moftah M, Eswayah A. Repurposing of Hydroxyurea Against COVID-19: A Promising Immunomodulatory Role. Assay Drug Dev Technol 2022; 20:55-62. [PMID: 34990284 DOI: 10.1089/adt.2021.090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cytokine release syndrome, a prominent mechanism of morbidity and mortality in patients with coronavirus disease 2019 (COVID-19), can cause multiple bodily reactions, including excessive release of proinflammatory mediators, with tumor necrosis factor-α (TNF-α) being the most prevalent cytokine combined with persistently elevated D-dimer levels that are indicative of potential thrombotic events, low levels of endogenous nitric oxide (NO) generation, and progressive decrease in hemoglobin production. In our argument, the conceptual repurposing of hydroxyurea (HU) for managing COVID-19 can provide a promising therapeutic option originating from a rich history of investigational antiviral activity. HU as a proposed supportive therapeutic agent for treating COVID-19 can exemplify a successful remedial choice through its anti-inflammatory activity along with an intrinsic propensity to control the circulatory levels of key cytokines including TNF-α. HU has the ability to undergo in vivo NO conversion acting as NO donor together with being a prominent inducer of fetal hemoglobin (HbF) production. The combination of the mentioned two properties allows HU to possess evident capability of protecting against thrombotic events by controlling D-dimer levels. The implication of our hypothetical argument sheds light on the curative potential of HU, which can be strategically harnessed against COVID-19.
Collapse
Affiliation(s)
- Moayed Ben Moftah
- Department of Medicinal and Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tripoli, Tripoli, Libya
| | - Asma Eswayah
- Department of Medicinal and Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tripoli, Tripoli, Libya
| |
Collapse
|
14
|
Gour A, Dogra A, Kour D, Singh G, Kumar A, Nandi U. Effect of Concomitant Hydroxyurea Therapy with Rutin and Gallic Acid: Integration of Pharmacokinetic and Pharmacodynamic Approaches. ACS OMEGA 2021; 6:14542-14550. [PMID: 34124477 PMCID: PMC8190911 DOI: 10.1021/acsomega.1c01518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/13/2021] [Indexed: 05/28/2023]
Abstract
Hydroxyurea (HU) is the first-ever approved drug by USFDA for sickle cell anemia (SCA). However, its treatment is associated with severe side effects like myelosuppression. Current studies are focused on the supplementation therapy for symptomatic management of SCA. In the present study, we aimed to explore rutin's and gallic acid's potential individually, for concomitant therapy with HU using pharmacokinetic and pharmacodynamic approaches since there is no such precedent till date. In vivo pharmacokinetic studies of HU in rats showed that rutin could be safely co-administered with HU, while gallic acid significantly raised the plasma concentration of HU. Both the phytochemicals did not have any marked inhibitory effect on urease but have considerable effects on horseradish peroxidase enzyme. The experimental phytoconstituents displayed a very low propensity to cause in vitro hemolysis. Gallic acid markedly enhanced the HU-induced decrease in lymphocyte proliferation. A substantial improvement by rutin or gallic acid was observed in HU-induced reduction of the main hematological parameters in rats. Combined treatment of HU with rutin and gallic acid reduced serum levels of both IL-6 and IL-17A. Overall, both rutin and gallic acid are found to have promising phytotherapy potential with HU. Further exploration needs to be done on both candidates for use as phytotherapeutics for SCA.
Collapse
Affiliation(s)
- Abhishek Gour
- PK-PD,
Toxicology and Formulation Division, CSIR-Indian
Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Ashish Dogra
- PK-PD,
Toxicology and Formulation Division, CSIR-Indian
Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Dilpreet Kour
- PK-PD,
Toxicology and Formulation Division, CSIR-Indian
Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Gurdarshan Singh
- PK-PD,
Toxicology and Formulation Division, CSIR-Indian
Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Ajay Kumar
- PK-PD,
Toxicology and Formulation Division, CSIR-Indian
Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| | - Utpal Nandi
- PK-PD,
Toxicology and Formulation Division, CSIR-Indian
Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201 002, India
| |
Collapse
|
15
|
Karamperis K, Tsoumpeli MT, Kounelis F, Koromina M, Mitropoulou C, Moutinho C, Patrinos GP. Genome-based therapeutic interventions for β-type hemoglobinopathies. Hum Genomics 2021; 15:32. [PMID: 34090531 PMCID: PMC8178887 DOI: 10.1186/s40246-021-00329-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/28/2021] [Indexed: 12/18/2022] Open
Abstract
For decades, various strategies have been proposed to solve the enigma of hemoglobinopathies, especially severe cases. However, most of them seem to be lagging in terms of effectiveness and safety. So far, the most prevalent and promising treatment options for patients with β-types hemoglobinopathies, among others, predominantly include drug treatment and gene therapy. Despite the significant improvements of such interventions to the patient's quality of life, a variable response has been demonstrated among different groups of patients and populations. This is essentially due to the complexity of the disease and other genetic factors. In recent years, a more in-depth understanding of the molecular basis of the β-type hemoglobinopathies has led to significant upgrades to the current technologies, as well as the addition of new ones attempting to elucidate these barriers. Therefore, the purpose of this article is to shed light on pharmacogenomics, gene addition, and genome editing technologies, and consequently, their potential use as direct and indirect genome-based interventions, in different strategies, referring to drug and gene therapy. Furthermore, all the latest progress, updates, and scientific achievements for patients with β-type hemoglobinopathies will be described in detail.
Collapse
Affiliation(s)
- Kariofyllis Karamperis
- Department of Pharmacy, School of Health Sciences, Laboratory of Pharmacogenomics and Individualized Therapy, University of Patras, Patras, Greece
- The Golden Helix Foundation, London, UK
| | - Maria T Tsoumpeli
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, UK
| | - Fotios Kounelis
- Department of Computing, Group of Large-Scale Data & Systems, Imperial College London, London, UK
| | - Maria Koromina
- Department of Pharmacy, School of Health Sciences, Laboratory of Pharmacogenomics and Individualized Therapy, University of Patras, Patras, Greece
| | | | - Catia Moutinho
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Darlinghurst, Sydney, Australia
| | - George P Patrinos
- Department of Pharmacy, School of Health Sciences, Laboratory of Pharmacogenomics and Individualized Therapy, University of Patras, Patras, Greece.
- College of Medicine and Health Sciences, Department of Pathology, United Arab Emirates University, Al-Ain, United Arab Emirates.
- Zayed Center of Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.
| |
Collapse
|
16
|
Drysdale CM, Nassehi T, Gamer J, Yapundich M, Tisdale JF, Uchida N. Hematopoietic-Stem-Cell-Targeted Gene-Addition and Gene-Editing Strategies for β-hemoglobinopathies. Cell Stem Cell 2021; 28:191-208. [PMID: 33545079 DOI: 10.1016/j.stem.2021.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sickle cell disease (SCD) is caused by a well-defined point mutation in the β-globin gene and therefore is an optimal target for hematopoietic stem cell (HSC) gene-addition/editing therapy. In HSC gene-addition therapy, a therapeutic β-globin gene is integrated into patient HSCs via lentiviral transduction, resulting in long-term phenotypic correction. State-of-the-art gene-editing technology has made it possible to repair the β-globin mutation in patient HSCs or target genetic loci associated with reactivation of endogenous γ-globin expression. With both approaches showing signs of therapeutic efficacy in patients, we discuss current genetic treatments, challenges, and technical advances in this field.
Collapse
Affiliation(s)
- Claire M Drysdale
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Tina Nassehi
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Jackson Gamer
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Morgan Yapundich
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Naoya Uchida
- Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD 20892, USA; Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan.
| |
Collapse
|
17
|
Yahouédéhou SCMA, Neres JSDS, da Guarda CC, Carvalho SP, Santiago RP, Figueiredo CVB, Fiuza LM, Ndidi US, de Oliveira RM, Fonseca CA, Nascimento VML, Rocha LC, Adanho CSA, da Rocha TSC, Adorno EV, Goncalves MS. Sickle Cell Anemia: Variants in the CYP2D6, CAT, and SLC14A1 Genes Are Associated With Improved Hydroxyurea Response. Front Pharmacol 2020; 11:553064. [PMID: 33013391 PMCID: PMC7510454 DOI: 10.3389/fphar.2020.553064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/18/2020] [Indexed: 11/13/2022] Open
Abstract
Differences in hydroxyurea response in sickle cell anemia may arise due to a series of factors with genetic factors appearing to be predominant. This study aims to investigate the effects of single nucleotide polymorphisms in genes encoding drug-metabolizing enzymes and solute carriers on hydroxyurea response, in patients with sickle cell anemia. For that purpose, a total number of 90 patients with sickle cell anemia were recruited, 45 were undergoing hydroxyurea treatment, while 45 were not under the treatment. Association analyses were performed between CYP3A4 (rs2740574), CYP2D6 (rs3892097), CAT (rs7943316 and rs1001179), and SLC14A1 (rs2298720) variants and laboratory parameters. According to our findings, patients with hydroxyurea treatment demonstrated higher HbF levels and a significant improvement in hemolytic, hepatic, inflammatory, and lipid parameters in comparison to those without the treatment. We also found significant associations between the CYP2D6 (rs3892097), CAT (rs7943316 and rs1001179), and SLC14A1 (rs2298720) variants and an improvement of the therapeutic effects, specifically the hemolytic, hepatic, inflammatory, lipid, and renal parameters. In conclusion, our results highlight the importance of the investigated variants, and their strong association with hydroxyurea efficacy in patients with sickle cell anemia, which may be considered in the future as genetic markers.
Collapse
Affiliation(s)
- Sètondji Cocou Modeste Alexandre Yahouédéhou
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | | | - Caroline Conceição da Guarda
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - Suellen Pinheiro Carvalho
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - Rayra Pereira Santiago
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - Camylla Vilas Boas Figueiredo
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - Luciana Magalhães Fiuza
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | | | - Rodrigo Mota de Oliveira
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - Cleverson Alves Fonseca
- Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | | | | | | | | | - Elisângela Vitória Adorno
- Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| | - Marilda Souza Goncalves
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Brazil.,Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Brazil
| |
Collapse
|
18
|
Yahouédéhou SCMA, da Guarda CC, Figueiredo CVB, Santiago RP, Carvalho SP, Fiuza LM, Ndidi US, Oliveira RM, Carvalho MOS, Nascimento VML, Rocha LC, Lyra IM, Adorno EV, Goncalves MS. Hydroxyurea alters hematological, biochemical and inflammatory biomarkers in Brazilian children with SCA: Investigating associations with βS haplotype and α-thalassemia. PLoS One 2019; 14:e0218040. [PMID: 31306416 PMCID: PMC6629070 DOI: 10.1371/journal.pone.0218040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 05/24/2019] [Indexed: 12/13/2022] Open
Abstract
This study investigated the effects of hydroxyurea (HU) on hematological, biochemical and inflammatory parameters in children with sickle cell anemia (SCA) in association with βS haplotype and α-thalassemia. We included 22 children with SCA who were followed for an average of 14.5 months. Laboratory parameters were assessed by electronic methods, and molecular analysis was investigated by PCR-RFLP and allele-specific PCR. Results showed significant increases in hemoglobin, HbF, hematocrit, MCV, MCH, glucose, HDL-C and albumin levels, as well as significant decreases in MCHC and AST levels, WBC, neutrophils, eosinophils, lymphocytes and reticulocytes, in children during HU therapy. HbF levels were positively correlated with hemoglobin, hematocrit, MCV and total protein, yet negatively correlated with MCHC, RDW, AAT and AST during HU therapy (p<0.05). Children who carried the Central African Republic haplotype, in response to HU therapy, presented significant increases in hemoglobin, hematocrit, triglycerides and uric acid levels, as well as significant decreases in MCHC, AST and direct bilirubin levels, WBC, neutrophils, eosinophils, lymphocytes and reticulocytes. Those with the Benin haplotype presented increases in HbF and albumin levels, and a reduction in platelet counts (p<0.05). Children with α-thalassemia presented decreased ALT during HU use, while those without this deletion presented increases in hemoglobin, hematocrit, MCV, MCH, HDL-C and albumin, as well as decreases in MCHC, neutrophils, lymphocytes, reticulocytes and AST (p<0.05). Hence, regardless of its use in association with βS haplotypes or α-thalassemia, HU seems to be linked to alterations in hemolytic, inflammatory, hepatic, lipid and glycemic profiles.
Collapse
Affiliation(s)
- Sètondji Cocou Modeste Alexandre Yahouédéhou
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Bahia, Brasil
- Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Bahia, Brasil
| | - Caroline Conceição da Guarda
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Bahia, Brasil
- Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Bahia, Brasil
| | - Camylla Vilas Boas Figueiredo
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Bahia, Brasil
- Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Bahia, Brasil
| | - Rayra Pereira Santiago
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Bahia, Brasil
- Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Bahia, Brasil
| | - Suellen Pinheiro Carvalho
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Bahia, Brasil
- Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Bahia, Brasil
| | - Luciana Magalhães Fiuza
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Bahia, Brasil
- Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Bahia, Brasil
| | | | - Rodrigo Mota Oliveira
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Bahia, Brasil
- Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Bahia, Brasil
| | | | | | | | - Isa Menezes Lyra
- Fundação de Hematologia e Hemoterapia da Bahia, Salvador, Bahia, Brasil
| | - Elisângela Vitória Adorno
- Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Bahia, Brasil
| | - Marilda Souza Goncalves
- Laboratório de Investigação em Genética e Hematologia Translacional, Instituto Gonçalo Moniz, Salvador, Bahia, Brasil
- Laboratório de Pesquisa em Anemia, Departamento de Análises Clínicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Bahia, Brasil
| |
Collapse
|
19
|
|