1
|
Patel ZV, Prajjwal P, Bethineedi LD, Patel DJ, Khullar K, Patel H, Khatri K, Marsool MDM, Gadam S, Aleti S, Amir O. Newer Modalities and Updates in the Management of Sickle Cell Disease: A Systematic Review. J Blood Med 2024; 15:435-447. [PMID: 39286637 PMCID: PMC11404495 DOI: 10.2147/jbm.s477507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024] Open
Abstract
Sickle cell disease (SCD), the most common autosomal recessive genetic disorder, affects the hemoglobin (Hb) chains in human red blood cells. It is caused by mutations in the β-globin genes, leading to the production of hemoglobin S, which results in the formation of sickle-shaped red blood cells (RBCs). These abnormal cells cause hemolysis, endothelial damage, and small vessel occlusion, leading to both acute and long-term complications. According to the World Health Organization's 2008 estimates, SCD affects approximately 2.28 per 1000 individuals globally. Despite this high prevalence, therapeutic advancements have been slow. For many years, the only FDA-approved medications for managing SCD complications were hydroxyurea and deferiprone. However, recent years have seen the approval of several new therapies, including L-glutamine (2017), voxelotor and crizanlizumab (2019), as well as exagamglogene autotemcel (Casgevy) and lovotibeglogene autotemcel (Lyfgenia) (2023). These treatments have proven effective in managing both the acute and chronic effects of SCD, including hemolytic anemia, chronic pain, stroke, vaso-occlusive crises, and multiple organ damage syndromes. This review explores the mechanisms of action, practical considerations, and side effects of these emerging therapies, drawing from a comprehensive search of databases such as PubMed, Medline, and Cochrane.
Collapse
Affiliation(s)
| | | | | | - Divyakshi J Patel
- Internal Medicine, Smt. NHL Municipal Medical College, Ahmedabad, India
| | - Kaarvi Khullar
- Internal Medicine, Maharashtra University of Health Sciences, Government Medical College, Gondia, India
| | - Hinal Patel
- Internal Medicine, GMERS Medical College and Hospital Sola, Ahmedabad, India
| | - Kanishka Khatri
- Internal Medicine, Bharati Vidyapeeth University Medical College, Pune, India
| | | | - Srikanth Gadam
- Internal Medicine, NYC Health+ Hospitals, New York, NYC, USA
| | - Soumya Aleti
- Internal Medicine, Berkshire Medical Center, Pittsfield, MA, USA
| | - Omniat Amir
- Internal Medicine, Al Manhal Academy of Science, Sudan
| |
Collapse
|
2
|
Youssry I, Ayad N. Sickle cell disease: combination new therapies vs. CRISPR-Cas9 potential and challenges - review article. Ann Hematol 2024; 103:2613-2619. [PMID: 37867187 DOI: 10.1007/s00277-023-05510-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/12/2023] [Indexed: 10/24/2023]
Abstract
In 2022, sickle cell disease (SCD) continues to affect the lives of millions of people, being one of the most frequently inherited blood disorders worldwide. Recently, several new therapies have been FDA approved for the treatment of SCD. The complexity of the pathophysiology of sickling has given opportunity to the evolution of several modalities of therapies. Nonetheless, the potential for complementary targeting of HbS polymerization, vasocclusion, and other inflammatory pathways remains controversial. None of these drugs can be considered a single curative line of treatment. With the advancement of CRISPR/Cas9 technology, autologous transplant of gene-edited hematopoietic stem cells could possibly provide a cure for most patients with SCD. The advantage of this approach over the conventional stem cell transplantation is that it decreases the need for immuno-suppressive drugs and the risk of graft-versus-host disease. In addition, recent technological advances can reduce the off-target effects, but long-term monitoring is needed to ensure the reliability of these methods in the clinical setting. This review explores the efficacy and safety of combination therapies and contrasting this alternative with the challenges that exist with sickle cell gene therapy using CRISPR.
Collapse
Affiliation(s)
- Ilham Youssry
- Pediatric Hematology and BMT Unit, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nardeen Ayad
- Pediatric Hematology and BMT Unit, Faculty of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
3
|
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
|
4
|
Bhatt S, Argueta DA, Gupta K, Kundu S. Red Blood Cells as Therapeutic Target to Treat Sickle Cell Disease. Antioxid Redox Signal 2024; 40:1025-1049. [PMID: 37975291 DOI: 10.1089/ars.2023.0348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Significance: Sickle cell disease (SCD) is the most common inherited diathesis affecting mostly underserved populations globally. SCD is characterized by chronic pain and fatigue, severe acute painful crises requiring hospitalization and opioids, strokes, multiorgan damage, and a shortened life span. Symptoms may appear shortly after birth, and, in less developed countries, most children with SCD die before attaining age 5. Hematopoietic stem cell transplant and gene therapy offer a curative therapeutic approach, but, due to many challenges, are limited in their availability and effectiveness for a majority of persons with SCD. A critical unmet need is to develop safe and effective novel targeted therapies. A wide array of drugs currently undergoing clinical investigation hold promise for an expanded pharmacological armamentarium against SCD. Recent Advances: Hydroxyurea, the most widely used intervention for SCD management, has improved the survival in the Western world and more recently, voxelotor (R-state-stabilizer), l-glutamine, and crizanlizumab (anti-P-selectin antibody) have been approved by the Food and Drug Administration (FDA) for use in SCD. The recent FDA approval emphasizes the need to revisit the advances in understanding the core pathophysiology of SCD to accelerate novel evidence-based strategies to treat SCD. The biomechanical breakdown of erythrocytesis, the core pathophysiology of SCD, is associated with intrinsic factors, including the composition of hemoglobin, membrane integrity, cellular volume, hydration, andoxidative stress. Critical Issues and Future Directions: In this context, this review focuses on advances in emerging nongenetic interventions directed toward the therapeutic targets intrinsic to sickle red blood cells (RBCs), which can prevent impaired rheology of RBCs to impede disease progression and reduce the sequelae of comorbidities, including pain, vasculopathy, and organ damage. In addition, given the intricate pathophysiology of the disease, it is unlikely that a single pharmacotherapeutic intervention will comprehensively ameliorate the multifaceted complications associated with SCD. However, the availability of multiple drug options affords the opportunity for individualized therapeutic regimens tailored to specific SCD-related complications. Furthermore, it opens avenues for combination drug therapy, capitalizing on distinct mechanisms of action and profiles of adverse effects.
Collapse
Affiliation(s)
- Shruti Bhatt
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
| | - Donovan A Argueta
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, California, USA
| | - Kalpna Gupta
- Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Irvine, California, USA
| | - Suman Kundu
- Department of Biochemistry, University of Delhi South Campus, New Delhi, India
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, KK Birla Goa Campus, Goa, India
| |
Collapse
|
5
|
Dimitrievska M, Bansal D, Vitale M, Strouboulis J, Miccio A, Nicolaides KH, El Hoss S, Shangaris P, Jacków-Malinowska J. Revolutionising healing: Gene Editing's breakthrough against sickle cell disease. Blood Rev 2024; 65:101185. [PMID: 38493007 DOI: 10.1016/j.blre.2024.101185] [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: 10/25/2023] [Revised: 03/01/2024] [Accepted: 03/01/2024] [Indexed: 03/18/2024]
Abstract
Recent advancements in gene editing illuminate new potential therapeutic approaches for Sickle Cell Disease (SCD), a debilitating monogenic disorder caused by a point mutation in the β-globin gene. Despite the availability of several FDA-approved medications for symptomatic relief, allogeneic hematopoietic stem cell transplantation (HSCT) remains the sole curative option, underscoring a persistent need for novel treatments. This review delves into the growing field of gene editing, particularly the extensive research focused on curing haemoglobinopathies like SCD. We examine the use of techniques such as CRISPR-Cas9 and homology-directed repair, base editing, and prime editing to either correct the pathogenic variant into a non-pathogenic or wild-type one or augment fetal haemoglobin (HbF) production. The article elucidates ways to optimize these tools for efficacious gene editing with minimal off-target effects and offers insights into their effective delivery into cells. Furthermore, we explore clinical trials involving alternative SCD treatment strategies, such as LentiGlobin therapy and autologous HSCT, distilling the current findings. This review consolidates vital information for the clinical translation of gene editing for SCD, providing strategic insights for investigators eager to further the development of gene editing for SCD.
Collapse
Affiliation(s)
- Marija Dimitrievska
- St John's Institute of Dermatology, King's College London, London SE1 9RT, UK
| | - Dravie Bansal
- St John's Institute of Dermatology, King's College London, London SE1 9RT, UK
| | - Marta Vitale
- St John's Institute of Dermatology, King's College London, London SE1 9RT, UK
| | - John Strouboulis
- Red Cell Hematology Lab, Comprehensive Cancer Center, School of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom
| | - Annarita Miccio
- Laboratory of Chromatin and Gene Regulation During Development, Imagine Institute, INSERM UMR1163, Paris 75015, France
| | - Kypros H Nicolaides
- Women and Children's Health, School of Life Course & Population Sciences, Kings College London, London, United Kingdom; Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom
| | - Sara El Hoss
- Red Cell Hematology Lab, Comprehensive Cancer Center, School of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom.
| | - Panicos Shangaris
- Women and Children's Health, School of Life Course & Population Sciences, Kings College London, London, United Kingdom; Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom.
| | | |
Collapse
|
6
|
Kuriri FA. Hope on the Horizon: New and Future Therapies for Sickle Cell Disease. J Clin Med 2023; 12:5692. [PMID: 37685759 PMCID: PMC10488840 DOI: 10.3390/jcm12175692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/16/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
This article provides an overview of conventional, new, and future treatment options for sickle cell disease (SCD), a genetic disorder affecting the production of hemoglobin. Current treatments include hydroxyurea, a conventional SCD treatment that increases the levels of fetal hemoglobin, and new treatments such as voxelotor, a recently approved SCD treatment that selectively binds hemoglobin, preventing formation of sickled red blood cells. In addition to discussing the mechanisms of action of current SCD treatments, potential side effects are also discussed, highlighting the need for new treatments that can address the limitations of current treatments and improve the quality of life for people with SCD. Future treatments, such as gene therapy, are also explored as promising treatment options for SCD patients.
Collapse
Affiliation(s)
- Fahd A Kuriri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra, Riyadh 15572, Saudi Arabia
| |
Collapse
|
7
|
Houghton OM, Carter J, Dhamija R. Retinal Vasculopathy With Cerebral Leukoencephalopathy and Systemic Manifestations: Critical Role of Retina Specialists. JOURNAL OF VITREORETINAL DISEASES 2023; 7:171-177. [PMID: 37006667 PMCID: PMC10037756 DOI: 10.1177/24741264221129095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Purpose To describe a case of retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations (RVCL-S) to enhance early recognition of this often-missed diagnosis. Methods A case report is presented. Results A 50-year-old woman with a history of Raynaud phenomenon, memory difficulties, and a family history of strokes was referred for evaluation of a bilateral, small-vessel, occlusive disease refractory to immunosuppressive therapy. An extensive workup for treatable causes was unrevealing. Fifteen months after presentation, brain imaging showed white-matter lesions and dystrophic calcification, which led to the discovery of a pathogenic variant in TREX1 and the diagnosis of RVCL-S. Conclusions Retina specialists play a critical role in the timely diagnosis of RVCL-S. Although the findings in this condition can mimic those in other common retinal vascular disorders, there are key characteristics that increase the suspicion for RVCL-S. Early recognition might decrease unnecessary therapies and procedures.
Collapse
Affiliation(s)
| | | | - Radhika Dhamija
- Department of Clinical Genomics, Mayo Clinic,
Scottsdale, AZ, USA
| |
Collapse
|
8
|
Muzazu SGY, Chirwa M, Khatanga-Chihana S, Munyinda M, Simuyandi M. Sickle Cell Disease in Early Infancy: A Case Report. Pediatric Health Med Ther 2022; 13:377-383. [PMID: 36536766 PMCID: PMC9759007 DOI: 10.2147/phmt.s388147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/02/2022] [Indexed: 08/29/2023] Open
Abstract
Sickle cell disease (SCD) refers to a group of hereditary disorders that result in faulty hemoglobin carriage by the red blood cells. This paper discusses an atypical presentation of SCD in early infancy. Despite current literature suggesting protection by fetal hemoglobin in the first few months of life, we report a diagnosis of SCD at 2 months of age with severe symptoms requiring hospitalization. It is therefore important for clinicians to raise their clinical index of suspicion of SCD in children presenting with severe anemia even though they are less than 6 months old and do not present with classic dactylitis or pain syndromes. Expansion and sustained newborn screening programs for SCD in developing countries could help clinicians and parents plan for early treatment, appropriate prophylaxis, and improved management of SCD complications.
Collapse
Affiliation(s)
- Seke G Y Muzazu
- Enteric Disease and Vaccines Research Unit, Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka, Zambia
| | - Masuzyo Chirwa
- Enteric Disease and Vaccines Research Unit, Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka, Zambia
| | - Shalom Khatanga-Chihana
- Enteric Disease and Vaccines Research Unit, Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka, Zambia
| | - Masiliso Munyinda
- Enteric Disease and Vaccines Research Unit, Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka, Zambia
| | - Michelo Simuyandi
- Enteric Disease and Vaccines Research Unit, Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka, Zambia
| |
Collapse
|
9
|
Alshurafa A, Yassin MA. Short- and long-term follow-up and additional benefits in a sickle cell disease patient experienced severe crizanlizumab infusion-related vaso-occlusive crisis: A case report. Front Med (Lausanne) 2022; 9:1048571. [PMID: 36523780 PMCID: PMC9745667 DOI: 10.3389/fmed.2022.1048571] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/02/2022] [Indexed: 09/20/2023] Open
Abstract
Sickle cell disease is an autosomal recessive disorder characterized by the presence of sickle hemoglobin that leads to chronic hemolysis and vaso-occlusive crisis. After decades of limited therapy options, crizanlizumab is a humanized monoclonal antibody approved by the Food and Drug Administration (FDA) in 2019 for sickle cell-related pain crises for patients 16 years of age and above. Although rare, infusion-related reactions, including painful crises, occurred in 3% as per the package insert. However, the data on how to deal with such reactions and about further treatment outcomes are limited as most patients stopped crizanlizumab after the reaction. Herein, we report the good outcome of 13 doses of crizanlizumab in a 19-year-old female patient with sickle cell disease on hydroxyurea, despite experiencing a severe infusion-related painful crisis during the second infusion. Additional benefits of crizanlizumab, in this case, were preventing new episodes of acute chest syndrome, quitting chronic narcotics use, and a remarkable improvement in quality of life and overall performance.
Collapse
|
10
|
Ibrahim A, Muhammad SA. Antioxidant-Rich Nutraceutical as a Therapeutic Strategy for Sickle Cell Disease. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2022:1-10. [PMID: 36069788 DOI: 10.1080/27697061.2022.2108930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 07/23/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Sickle cell disease (SCD) is a genetically inherited disease in which the "SS" individual possesses two copies of the abnormal beta-globin gene. This disease is one of the most dominant genetic diseases in the world. SCD is marked by the propensity of red cell hemoglobin to polymerize and distort the red cell from a biconcave disk shape into a sickle shape, resulting in a typical vaso-occlusive episode and accelerated hemolysis. Plants are rich sources of bioactive compounds that are promising anti-sickling agents to scavenge free radicals, thereby ensuring oxidative balance. The current review highlights the potential therapeutic benefits of antioxidant-rich nutraceutical in the treatment and management of sickle cell disease. The anti-sickling potential of nutraceutical is attributed to the presence of antioxidant bioactive chemicals such as alkaloids, polyphenols, vitamins, and minerals, which acts as scavengers of free radicals that prevent oxidative damage of the hemoglobin and prevent hemolysis, facilitating longer erythrocyte lifespan. The challenges of current therapies for SCD and future directions are also discussed.KEY TEACHING POINTSSickle cell disease is a genetically inherited disease in which SS individuals possess two copies of the abnormal beta-globin gene.Oxidative stress contributes to the pathophysiology of secondary dysfunction in sickle cell patients.Antioxidants can play a vital role in maintaining a balance between oxidant and antioxidant defense systems.Nutraceutical rich in antioxidants such as alkaloids, polyphenols, vitamins, and minerals is potential therapeutic agents for sickle cell disease.An antioxidant-rich nutraceutical may act to reduce vaso-occlusive crises.
Collapse
Affiliation(s)
- Abdulwasiu Ibrahim
- Department of Biochemistry and Molecular Biology, Usmanu Danfodiyo University, Sokoto, Nigeria
| | | |
Collapse
|
11
|
Dehghani T, Thai PN, Sodhi H, Ren L, Sirish P, Nader CE, Timofeyev V, Overton JL, Li X, Lam KS, Chiamvimonvat N, Panitch A. Selectin-targeting glycosaminoglycan-peptide conjugate limits neutrophil-mediated cardiac reperfusion injury. Cardiovasc Res 2022; 118:267-281. [PMID: 33125066 PMCID: PMC8932156 DOI: 10.1093/cvr/cvaa312] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 09/10/2020] [Accepted: 10/16/2020] [Indexed: 01/13/2023] Open
Abstract
AIMS One of the hallmarks of myocardial infarction (MI) is excessive inflammation. During an inflammatory insult, damaged endothelial cells shed their glycocalyx, a carbohydrate-rich layer on the cell surface which provides a regulatory interface to immune cell adhesion. Selectin-mediated neutrophilia occurs as a result of endothelial injury and inflammation. We recently designed a novel selectin-targeting glycocalyx mimetic (termed DS-IkL) capable of binding inflamed endothelial cells. This study examines the capacity of DS-IkL to limit neutrophil binding and platelet activation on inflamed endothelial cells, as well as the cardioprotective effects of DS-IkL after acute myocardial infarction. METHODS AND RESULTS In vitro, DS-IkL diminished neutrophil interactions with both recombinant selectin and inflamed endothelial cells, and limited platelet activation on inflamed endothelial cells. Our data demonstrated that DS-IkL localized to regions of vascular inflammation in vivo after 45 min of left anterior descending coronary artery ligation-induced MI. Further, findings from this study show DS-IkL treatment had short- and long-term cardioprotective effects after ischaemia/reperfusion of the left anterior descending coronary artery. Mice treated with DS-IkL immediately after ischaemia/reperfusion and 24 h later exhibited reduced neutrophil extravasation, macrophage accumulation, fibroblast and endothelial cell proliferation, and fibrosis compared to saline controls. CONCLUSIONS Our findings suggest that DS-IkL has great therapeutic potential after MI by limiting reperfusion injury induced by the immune response.
Collapse
Affiliation(s)
- Tima Dehghani
- Department of Biomedical Engineering, University of California, 451 Health Sciences Drive, GBSF 2303, Davis, CA 95616, USA
| | - Phung N Thai
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA, USA
| | - Harkanwalpreet Sodhi
- Department of Biomedical Engineering, University of California, 451 Health Sciences Drive, GBSF 2303, Davis, CA 95616, USA
| | - Lu Ren
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA, USA
| | - Padmini Sirish
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA, USA
| | - Carol E Nader
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA, USA
| | - Valeriy Timofeyev
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA, USA
| | - James L Overton
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA, USA
| | - Xiaocen Li
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, University of California, Davis, CA, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, CA, USA
- Department of Veterans Affairs, Northern California Health Care System, Mather, CA, USA
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, 451 Health Sciences Drive, GBSF 2303, Davis, CA 95616, USA
- Department of Surgery, University of California, Davis, CA, USA
| |
Collapse
|
12
|
Karki NR, Kutlar A. P-Selectin Blockade in the Treatment of Painful Vaso-Occlusive Crises in Sickle Cell Disease: A Spotlight on Crizanlizumab. J Pain Res 2021; 14:849-856. [PMID: 33833562 PMCID: PMC8019662 DOI: 10.2147/jpr.s278285] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/13/2021] [Indexed: 01/01/2023] Open
Abstract
Microvascular vaso-occlusion driven pain crisis is the hallmark of sickle cell disease with profound morbidity and increased mortality. Selectins, most notably P-selectins have an integral role in this phenomenon. P-selection was first identified in 1989. In 2019, after 3 decades of basic, translational, and clinical work with this pathway, the US Food and Drug Administration approved a P-selectin antibody, crizanlizumab to reduce frequency of pain crisis in patients more than 16 years with sickle cell disease. We review the fundamentals of P-selectin pathobiology, P-selectin blocking agents, clinical data with the use of crizanlizumab and prospects of this novel class of drugs in the context of other treatments for painful vaso-occlusive episodes.
Collapse
Affiliation(s)
- Nabin Raj Karki
- Division of Hematology/Oncology, Augusta University, Augusta, GA, USA
| | - Abdullah Kutlar
- Division of Hematology/Oncology, Augusta University, Augusta, GA, USA
| |
Collapse
|
13
|
L-Selectin P213S and Integrin Alpha 2 C807T Genetic Polymorphisms in Pediatric Sickle Cell Disease Patients. J Pediatr Hematol Oncol 2020; 42:e707-e711. [PMID: 32433445 DOI: 10.1097/mph.0000000000001839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Sickle cell disease (SCD) is an autosomal recessive hemoglobinopathy characterized by increased cellular adhesiveness. Vaso-occlusion (VOC) is the most prevalent disease complication of SCD that could be altered by genetic factors. L-Selectin and integrin alpha 2 (ITGA2) are 2 adhesion molecules linked to vasculopathy and inflammation. The current study aimed at detecting the prevalence of genetic variants of L-selectin and ITGA2 as possible molecular modulators and novel therapeutic targets in a cohort of pediatric SCD patients. Genotyping was performed by polymerase chain reaction restriction fragment length polymorphism technique for 100 SCD patients and 100 age and gender-matched unrelated healthy controls. The homomutant genotype of ITGA2 C807T was significantly higher in SCD patients compared with controls (P=0.001) and confirmed almost a 3-fold increased risk of moderate and severe attacks of VOC. There are significant adverse effects caused by the polymorphisms of ITGA2, and hence Egyptian SCD patients could benefit from the targeted therapies specifically against ITGA2 to ameliorate the severe course of the disease and improve the quality of life. However, further studies of genotypes and expression levels of these adhesion molecules during the attacks of VOC are recommended.
Collapse
|
14
|
Dehghani T, Panitch A. Endothelial cells, neutrophils and platelets: getting to the bottom of an inflammatory triangle. Open Biol 2020; 10:200161. [PMID: 33050789 PMCID: PMC7653352 DOI: 10.1098/rsob.200161] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023] Open
Abstract
Severe fibrotic and thrombotic events permeate the healthcare system, causing suffering for millions of patients with inflammatory disorders. As late-state consequences of chronic inflammation, fibrosis and thrombosis are the culmination of pathological interactions of activated endothelium, neutrophils and platelets after vessel injury. Coupling of these three cell types ensures a pro-coagulant, cytokine-rich environment that promotes the capture, activation and proliferation of circulating immune cells and recruitment of key pro-fibrotic cell types such as myofibroblasts. As the first responders to sterile inflammatory injury, it is important to understand how endothelial cells, neutrophils and platelets help create this environment. There has been a growing interest in this intersection over the past decade that has helped shape the development of therapeutics to target these processes. Here, we review recent insights into how neutrophils, platelets and endothelial cells guide the development of pathological vessel repair that can also result in underlying tissue fibrosis. We further discuss recent efforts that have been made to translate this knowledge into therapeutics and provide perspective as to how a compound or combination therapeutics may be most efficacious when tackling fibrosis and thrombosis that is brought upon by chronic inflammation.
Collapse
Affiliation(s)
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California, Davis, 451 Health Sciences Drive, GBSF 2303, Davis, CA, USA
| |
Collapse
|
15
|
Cannon M, Phillips H, Smith S, Williams K, Brinton L, Gregory C, Landes K, Desai P, Byrd J, Lapalombella R. Large-Scale Drug Screen Identifies FDA-Approved Drugs for Repurposing in Sickle-Cell Disease. J Clin Med 2020; 9:E2276. [PMID: 32708954 PMCID: PMC7408993 DOI: 10.3390/jcm9072276] [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: 06/20/2020] [Revised: 07/07/2020] [Accepted: 07/15/2020] [Indexed: 12/26/2022] Open
Abstract
Sickle-cell disease (SCD) is a debilitating hematological disorder with very few approved treatment options. Therapeutic reactivation of fetal hemoglobin (HbF) is one of the most pursued methods for ameliorating the systemic manifestations of SCD. Despite this, very few pharmacological agents have advanced to clinical trials or marketing for use. In this study, we report the development of an HbF in situ intracellular immunoblot assay coupled to a high-throughput drug screen to identify Food and Drug Administration (FDA) approved drugs that can be repurposed clinically for treatment of SCD. Using this assay we evaluated the National Institute of Health (NIH) Clinical Collection (NCC), a publicly available library of 725 small molecules, and found nine candidates that can significantly re-express HbF in erythroid cell lines as well as primary erythroblasts derived from SCD patients. Furthermore, we show the strong effects on HbF expression of these candidates to occur with minimal cytotoxicity in 7 of the 9 drugs. Given these data and their proven history of use for other indications, we hypothesize that several of these candidate drugs warrant further investigation for use in SCD.
Collapse
Affiliation(s)
- Matthew Cannon
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Hannah Phillips
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Sidney Smith
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Katie Williams
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Lindsey Brinton
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Charles Gregory
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Kristina Landes
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Payal Desai
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - John Byrd
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
- College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Rosa Lapalombella
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| |
Collapse
|