1
|
Singh A, Irfan H, Fatima E, Nazir Z, Verma A, Akilimali A. Revolutionary breakthrough: FDA approves CASGEVY, the first CRISPR/Cas9 gene therapy for sickle cell disease. Ann Med Surg (Lond) 2024; 86:4555-4559. [PMID: 39118728 PMCID: PMC11305803 DOI: 10.1097/ms9.0000000000002146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/24/2024] [Indexed: 08/10/2024] Open
Abstract
Sickle cell disease (SCD) is a hereditary hemoglobinopathy resulting from a β-globin chain mutation that causes abnormal hemoglobin (HbS) polymerization and leads to severe complications. Current treatment options primarily focus on symptom management, with limited curative potential. Recently, Casgevy, the first CRISPR/Cas9-based gene therapy for SCD, has received breakthrough FDA approval. Clinical trials have shown that Casgevy administered to patients aged older than or equal to 12 years enables precise modifications in hematopoietic stem cells, resulting in elevated fetal hemoglobin (HbF) levels and a significant reduction in vaso-occlusive events. Unlike conventional treatments, this therapy offers a curative approach and eliminates the need for recurrent transfusions and transplants, thereby improving the quality of life of patients with SCD. Casgevy has emerged as a beacon of hope for SCD patients and signifies a potential paradigm shift in SCD management due to its safety, curative potential, and transformative impact, positioning it as a groundbreaking intervention. Nevertheless, ethical considerations surrounding CRISPR technology and regulatory frameworks must be addressed to ensure responsible application and equitable access to this one-time gene editing therapy. As the authors celebrate this scientific advancement, sustained interdisciplinary collaboration and ethical scrutiny are essential to navigating the evolving landscape of CRISPR technology in medicine. This review aims to provide a detailed insight into the application of Casgevy, challenges associated with its application, future prospects of this therapy, and its comparison with existing treatment options for SCD.
Collapse
Affiliation(s)
- Ajeet Singh
- Department of Internal Medicine, Dow University of Health Sciences, Karachi
| | - Hamza Irfan
- Department of Medicine, Shaikh Khalifa Bin Zayed Al Nahyan Medical and Dental College Lahore
| | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Zainab Nazir
- Department of Internal Medicine, Dow University of Health Sciences, Karachi
| | - Amogh Verma
- Rama Medical College Hospital and Research Center, Hapur, India
| | - Aymar Akilimali
- Department of research, Medical Research Circle, Goma, Democratic Republic of Congo
| |
Collapse
|
2
|
Kerwash E, Sajic M, Rantell KR, McBlane JW, Johnston JD, Niewiarowska A, Butler AS, Cole S. Regulatory Assessment of Casgevy for the Treatment of Transfusion-Dependent β-Thalassemia and Sickle Cell Disease with Recurrent Vaso-Occlusive Crises. Curr Issues Mol Biol 2024; 46:8209-8225. [PMID: 39194702 DOI: 10.3390/cimb46080485] [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: 06/28/2024] [Revised: 07/23/2024] [Accepted: 07/27/2024] [Indexed: 08/29/2024] Open
Abstract
Sickle cell disease (SCD) and transfusion-dependent β-thalassemia (TDT) are hereditary haemoglobinopathies characterized by a reduction in functional β-globin chains. Both conditions cause tiredness and increase susceptibility to infection, which can lead organ failure, significantly reducing life expectancy and typically requiring those affected to undergo regular erythrocyte transfusion. Recently, a novel therapeutic treatment for SCD and TDT was approved by the UK regulatory body (Medicines and Healthcare products Regulatory Agency; MHRA). Exagamglogene autotemcel (Casgevy) is the first licensed therapy globally to utilize CRIPSR/Cas9 technology and induces an increase in expression of γ-globin chains to compensate for the reduction in functional β-globin. Casgevy represents a first-in-class therapeutic, and numerous considerations were made by the MHRA throughout its assessment of the medicine. These include, but are not limited to, the risk of tumorigenicity and off-target editing, a limited cohort size, the validity of proposed dosing and the conduction of only single-arm studies. The MHRA's analyses of the data to support the proposed indications are presented and discussed throughout this manuscript. Overall, the sponsors claims were considered well supported by their data, and Casgevy was licensed for the treatment of TDT or SCD in patients 12 years of age and older for whom hematopoietic stem cell (HSC) transplantation is appropriate, but a human leukocyte antigen-matched related HSC donor is not available.
Collapse
Affiliation(s)
- Essam Kerwash
- Medicines and Healthcare Products Regulatory Agency (MHRA), 10 South Colonnade, London E14 4PU, UK
| | - Marija Sajic
- Medicines and Healthcare Products Regulatory Agency (MHRA), 10 South Colonnade, London E14 4PU, UK
| | - Khadija Rerhou Rantell
- Medicines and Healthcare Products Regulatory Agency (MHRA), 10 South Colonnade, London E14 4PU, UK
| | - James W McBlane
- Medicines and Healthcare Products Regulatory Agency (MHRA), 10 South Colonnade, London E14 4PU, UK
| | - John D Johnston
- Medicines and Healthcare Products Regulatory Agency (MHRA), 10 South Colonnade, London E14 4PU, UK
| | - Alison Niewiarowska
- Medicines and Healthcare Products Regulatory Agency (MHRA), 10 South Colonnade, London E14 4PU, UK
| | - Andrew S Butler
- Medicines and Healthcare Products Regulatory Agency (MHRA), 10 South Colonnade, London E14 4PU, UK
| | - Susan Cole
- Medicines and Healthcare Products Regulatory Agency (MHRA), 10 South Colonnade, London E14 4PU, UK
| |
Collapse
|
3
|
Li S, Ling S, Wang D, Wang X, Hao F, Yin L, Yuan Z, Liu L, Zhang L, Li Y, Chen Y, Luo L, Dai Y, Zhang L, Chen L, Deng D, Tang W, Zhang S, Wang S, Cai Y. Modified lentiviral globin gene therapy for pediatric β 0/β 0 transfusion-dependent β-thalassemia: A single-center, single-arm pilot trial. Cell Stem Cell 2024; 31:961-973.e8. [PMID: 38759653 DOI: 10.1016/j.stem.2024.04.021] [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: 07/28/2023] [Revised: 01/24/2024] [Accepted: 04/24/2024] [Indexed: 05/19/2024]
Abstract
β0/β0 thalassemia is the most severe type of transfusion-dependent β-thalassemia (TDT) and is still a challenge facing lentiviral gene therapy. Here, we report the interim analysis of a single-center, single-arm pilot trial (NCT05015920) evaluating the safety and efficacy of a β-globin expression-optimized and insulator-engineered lentivirus-modified cell product (BD211) in β0/β0 TDT. Two female children were enrolled, infused with BD211, and followed up for an average of 25.5 months. Engraftment of genetically modified hematopoietic stem and progenitor cells was successful and sustained in both patients. No unexpected safety issues occurred during conditioning or after infusion. Both patients achieved transfusion independence for over 22 months. The treatment extended the lifespan of red blood cells by over 42 days. Single-cell DNA/RNA-sequencing analysis of the dynamic changes of gene-modified cells, transgene expression, and oncogene activation showed no notable adverse effects. Optimized lentiviral gene therapy may safely and effectively treat all β-thalassemia.
Collapse
Affiliation(s)
- Shiqi Li
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, Yunnan 650100, China
| | - Sikai Ling
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China; BDgene Therapeutics, Shanghai 200240, China
| | - Dawei Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | | | - Liufan Yin
- Sequanta Technologies, Shanghai 200131, China
| | - Zhongtao Yuan
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, Yunnan 650100, China
| | - Lin Liu
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, Yunnan 650100, China
| | - Lin Zhang
- BDgene Therapeutics, Shanghai 200240, China
| | - Yu Li
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, Yunnan 650100, China
| | - Yingnian Chen
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, Yunnan 650100, China
| | - Le Luo
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, Yunnan 650100, China
| | - Ying Dai
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, Yunnan 650100, China
| | - Lihua Zhang
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, Yunnan 650100, China
| | - Lvzhe Chen
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, Yunnan 650100, China
| | | | - Wei Tang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sujiang Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sanbin Wang
- 920th Hospital of Joint Logistics Support Force of People's Liberation Army of China, Kunming, Yunnan 650100, China.
| | - Yujia Cai
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
4
|
Mohammadian Gol T, Zahedipour F, Trosien P, Ureña-Bailén G, Kim M, Antony JS, Mezger M. Gene therapy in pediatrics - Clinical studies and approved drugs (as of 2023). Life Sci 2024; 348:122685. [PMID: 38710276 DOI: 10.1016/j.lfs.2024.122685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/17/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Gene therapy in pediatrics represents a cutting-edge therapeutic strategy for treating a range of genetic disorders that manifest in childhood. Gene therapy involves the modification or correction of a mutated gene or the introduction of a functional gene into a patient's cells. In general, it is implemented through two main modalities namely ex vivo gene therapy and in vivo gene therapy. Currently, a noteworthy array of gene therapy products has received valid market authorization, with several others in various stages of the approval process. Additionally, a multitude of clinical trials are actively underway, underscoring the dynamic progress within this field. Pediatric genetic disorders in the fields of hematology, oncology, vision and hearing loss, immunodeficiencies, neurological, and metabolic disorders are areas for gene therapy interventions. This review provides a comprehensive overview of the evolution and current progress of gene therapy-based treatments in the clinic for pediatric patients. It navigates the historical milestones of gene therapies, currently approved gene therapy products by the U.S. Food and Drug Administration (FDA) and/or European Medicines Agency (EMA) for children, and the promising future for genetic disorders. By providing a thorough compilation of approved gene therapy drugs and published results of completed or ongoing clinical trials, this review serves as a guide for pediatric clinicians to get a quick overview of the situation of clinical studies and approved gene therapy products as of 2023.
Collapse
Affiliation(s)
- Tahereh Mohammadian Gol
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Fatemeh Zahedipour
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Paul Trosien
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Guillermo Ureña-Bailén
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Miso Kim
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Justin S Antony
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Markus Mezger
- University Children's Hospital, Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
5
|
Alan S, Kanter J. Advances in pharmacotherapy for sickle cell disease: what is the current state of play? Expert Opin Pharmacother 2024; 25:1325-1334. [PMID: 38973339 DOI: 10.1080/14656566.2024.2377711] [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: 05/21/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
INTRODUCTION Despite over 100 years of neglect and insufficient funding, sickle cell disease has risen to the top of the discussions due to the recent approval of two new genetic therapies. Prior to these approvals, there were only four prior approved medications for sickle cell disease in spite of being the most common inherited blood disorder. The advent and expense of these new genetic therapies have finally brought the trials and tribulations associated with SCD including the suffering and early mortality of affected individuals to the much-needed limelight. Presently, questions about how these therapies will be used and what that means for ongoing pharmaceutical development remain. AREAS COVERED Here, we wish to highlight the current medications and treatments for SCD using already published literature as well as scrutinize the tedious process of implementation for these newly approved commercial genetic therapies. EXPERT OPINION In our expert opinion, despite the progress we have made, significant challenges remain and the most important requirement for any of these treatments is ensuring all affected individuals have access to a sickle cell specialist who can provide comprehensive care.
Collapse
Affiliation(s)
- Sheinei Alan
- Inova Adult Sickle Cell Center, University of Virginia School of Medicine, Inova Fairfax Medical Campus, Fairfax, VA, USA
| | - Julie Kanter
- Lifespan Comprehensive Sickle Cell Center, University of Alabama Birmingham, Birmingham, AL, USA
| |
Collapse
|
6
|
Phuphanitcharoenkun S, Bhukhai K, Phanthong P, Prasongtanakij S, Linn AK, Sutjarit N, Anurathapan U, Leboulch P, Payen E, Hongeng S, Borwornpinyo S. Droplet digital polymerase chain reaction-based quantitation of therapeutic lentiviral vector copies in transduced hematopoietic stem cells. Cytotherapy 2024; 26:586-591. [PMID: 38551525 DOI: 10.1016/j.jcyt.2024.02.018] [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: 11/08/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND AIMS Gene therapy using lentiviral vectors (LVs) that harbor a functional β-globin gene provides a curative treatment for hemoglobinopathies including beta-thalassemia and sickle cell disease. Accurate quantification of the vector copy number (VCN) and/or the proportion of transduced cells is critical to evaluate the efficacy of transduction and stability of the transgene during treatment. Moreover, commonly used techniques for LV quantification, including real-time quantitative polymerase chain reaction (PCR) or fluorescence-activated cell sorting, require either a standard curve or expression of a reporter protein for the detection of transduced cells. In the present study, we describe a digital droplet PCR (ddPCR) technique to measure the lentiviral VCN in transduced hematopoietic stem and progenitor cells (HSPCs). METHODS After HSPCs were transduced with an LV encoding the therapeutic β-globin (βA-T87Q) gene, the integrated lentiviral sequence in the host genome was amplified with primers that targeted a sequence within the vector and the human RPP30 gene. The dynamic range of ddPCR was between 5 × 10-3 ng and 5 × 10-6 ng of target copy per reaction. RESULTS We found that the ddPCR-based approach was able to estimate VCN with high sensitivity and a low standard deviation. Furthermore, ddPCR-mediated quantitation of lentiviral copy numbers in differentiated erythroblasts correlated with the level of βA-T87Q protein detected by reverse-phase high-performance liquid chromatography. CONCLUSIONS Taken together, the ddPCR technique has the potential to precisely detect LV copy numbers in the host genome, which can be used for VCN estimation, calculation of infectious titer and multiplicity of infection for HSPC transduction in a clinical setting.
Collapse
Affiliation(s)
| | - Kanit Bhukhai
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Phetcharat Phanthong
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Somsak Prasongtanakij
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Aung Khine Linn
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Nareerat Sutjarit
- Graduate Program in Nutrition, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Usanarat Anurathapan
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Philippe Leboulch
- Harvard Medical School and Genetics Division, Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts, USA
| | - Emmanuel Payen
- Paris-Saclay University, CEA, INSERM, Center for Immunology of Viral, Auto - immune, Hematological and Bacterial Diseases, Fontenay aux Roses, France
| | - Suradej Hongeng
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand; Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| | - Suparerk Borwornpinyo
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok, Thailand; Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
7
|
Locatelli F, Cavazzana M, Frangoul H, Fuente JDL, Algeri M, Meisel R. Autologous gene therapy for hemoglobinopathies: From bench to patient's bedside. Mol Ther 2024; 32:1202-1218. [PMID: 38454604 PMCID: PMC11081872 DOI: 10.1016/j.ymthe.2024.03.005] [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: 05/05/2023] [Revised: 01/31/2024] [Accepted: 03/05/2024] [Indexed: 03/09/2024] Open
Abstract
In recent years, a growing number of clinical trials have been initiated to evaluate gene therapy approaches for the treatment of patients with transfusion-dependent β-thalassemia and sickle cell disease (SCD). Therapeutic modalities being assessed in these trials utilize different molecular techniques, including lentiviral vectors to add functional copies of the gene encoding the hemoglobin β subunit in defective cells and CRISPR-Cas9, transcription activator-like effector protein nuclease, and zinc finger nuclease gene editing strategies to either directly address the underlying genetic cause of disease or induce fetal hemoglobin production by gene disruption. Here, we review the mechanisms of action of these various gene addition and gene editing approaches and describe the status of clinical trials designed to evaluate the potentially for these approaches to provide one-time functional cures to patients with transfusion-dependent β-thalassemia and SCD.
Collapse
Affiliation(s)
- Franco Locatelli
- Department of Pediatric Haematology/Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, 00165 Rome, Italy; Catholic University of the Sacred Heart, 00168 Rome, Italy.
| | - Marina Cavazzana
- Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), University of Paris, 75006 Paris, France
| | - Haydar Frangoul
- Sarah Cannon Center for Blood Cancer at The Children's Hospital at TriStar Centennial, Nashville, TN 37203, USA
| | - Josu de la Fuente
- Imperial College Healthcare NHS Trust, St Mary's Hospital, London W21NY, UK
| | - Mattia Algeri
- Department of Pediatric Haematology/Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, 00165 Rome, Italy; Department of Health Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| |
Collapse
|
8
|
Jones-Wonni B, Kelkar AH, Achebe MO. A Review of Gene Therapies for Hemoglobinopathies. Hemoglobin 2024; 48:141-152. [PMID: 39145521 DOI: 10.1080/03630269.2024.2369534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 08/16/2024]
Abstract
Due to the significant morbidity and mortality of hemoglobinopathies, curative options have long been pursued. The overall goal of gene therapy is to modify a patient's own hematopoietic stem cells to overcome the deleterious effects of the underlying genetic defect by gene addition, gene editing, or gene silencing. Gene addition incorporates genes with superior function than the abnormal gene; gene editing takes advantage of molecular tools such as zinc finger proteins, Transcription Activator-Like Effector Nucleases and Clustered Regularly Interspaced Short Palindromic Repeats coupled with Cas9 proteins (CRISPR-Cas9) which allow for sequence-specific breaks in DNA that disrupt gene function; and gene silencing suppresses gene expression by interference with mRNA transcription/protein translation or epigenetic modification. The majority of gene therapy strategies for hemoglobinopathies have targeted erythroid-specific BCL11A, a major regulator of fetal hemoglobin repression at the gamma-globin locus, in the normal fetal-to-adult hemoglobin switch that occurs shortly after birth. Other goals have involved the incorporation of anti-sickling globins, such as βT87Q or βAS3. Landmark clinical trials of gene therapy in transfusion-dependent thalassemia and sickle cell disease have shown remarkable efficacy and acceptable safety and culminated in recent regulatory approvals of gene therapy for both diseases in Europe and the United States.
Collapse
Affiliation(s)
| | - Amar H Kelkar
- Dana-Farber Cancer Institute - Department of Medical Oncology, Boston, MA, USA
| | - Maureen O Achebe
- Dana-Farber Cancer Institute - Department of Medical Oncology, Boston, MA, USA
- Brigham and Women's Hospital - Division of Hematology, Boston, MA, USA
| |
Collapse
|
9
|
Wang X, McKillop WM, Dlugi TA, Faber ML, Alvarez-Argote J, Chambers CB, Wilber A, Medin JA. A mass spectrometry assay for detection of endogenous and lentiviral engineered hemoglobin in cultured cells and sickle cell disease mice. J Gene Med 2024; 26:e3567. [PMID: 37455676 DOI: 10.1002/jgm.3567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/18/2023] Open
Abstract
Sickle cell disease (SCD) results from a sequence defect in the β-globin chain of adult hemoglobin (HbA) leading to expression of sickle hemoglobin (HbS). It is traditionally diagnosed by cellulose-acetate hemoglobin electrophoresis or high-performance liquid chromatography. While clinically useful, these methods have both sensitivity and specificity limitations. We developed a novel mass spectrometry (MS) method for the rapid, sensitive and highly quantitative detection of endogenous human β-globin and sickle hβ-globin, as well as lentiviral-encoded therapeutic hβAS3-globin in cultured cells and small quantities of mouse peripheral blood. The MS methods were used to phenotype homozygous HbA (AA), heterozygous HbA-HbS (AS) and homozygous HbS (SS) Townes SCD mice and detect lentiviral vector-encoded hβAS3-globin in transduced mouse erythroid cell cultures and transduced human CD34+ cells after erythroid differentiation. hβAS3-globin was also detected in peripheral blood 6 weeks post-transplant of transduced Townes SS bone marrow cells into syngeneic Townes SS mice and persisted for over 20 weeks post-transplant. As several genome-editing and gene therapy approaches for severe hemoglobin disorders are currently in clinical trials, this MS method will be useful for patient assessment before treatment and during follow-up.
Collapse
Affiliation(s)
- Xuejun Wang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - William M McKillop
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Theresa A Dlugi
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Mary L Faber
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Juliana Alvarez-Argote
- Department of Medicine, Division of Hematology-Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Christopher B Chambers
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Andrew Wilber
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Jeffrey A Medin
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
10
|
Pollock G, Negre O, Ribeil JA. Gene-addition/editing therapy in sickle cell disease. Presse Med 2023; 52:104214. [PMID: 38000628 DOI: 10.1016/j.lpm.2023.104214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2023] Open
Abstract
Gene therapy is an innovative strategy that offers potential cure for patients with sickle cell disease, and no appropriate donor for transplant consideration. While we await long term data from these clinical trials, we remain optimistic that gene therapy will become a standard of care for curative treatment in sickle cell disease. As gene therapy becomes a standard of treatment in sickle cell disease, we must also acknowledge the potential for financial burden to patients. We also must acknowledge the prevalence of sickle cell disease in low-resource settings. Hopefully, as we learn more about gene therapy, we can assess ways to overcome the financial toxicity that comes with this therapy.
Collapse
Affiliation(s)
- Galia Pollock
- Section of Hematology and Medical Oncology, Boston University Aram V. Chobanian & Edward Avedisian School of Medicine, Boston Medical Center, Center of Excellence in Sickle Cell Disease, Boston, MA, USA
| | | | - Jean-Antoine Ribeil
- Section of Hematology and Medical Oncology, Boston University Aram V. Chobanian & Edward Avedisian School of Medicine, Boston Medical Center, Center of Excellence in Sickle Cell Disease, Boston, MA, USA.
| |
Collapse
|
11
|
Hardouin G, Magrin E, Corsia A, Cavazzana M, Miccio A, Semeraro M. Sickle Cell Disease: From Genetics to Curative Approaches. Annu Rev Genomics Hum Genet 2023; 24:255-275. [PMID: 37624668 DOI: 10.1146/annurev-genom-120122-081037] [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: 08/27/2023]
Abstract
Sickle cell disease (SCD) is a monogenic blood disease caused by a point mutation in the gene coding for β-globin. The abnormal hemoglobin [sickle hemoglobin (HbS)] polymerizes under low-oxygen conditions and causes red blood cells to sickle. The clinical presentation varies from very severe (with acute pain, chronic pain, and early mortality) to normal (few complications and a normal life span). The variability of SCD might be due (in part) to various genetic modulators. First, we review the main genetic factors, polymorphisms, and modifier genes that influence the expression of globin or otherwise modulate the severity of SCD. Considering SCD as a complex, multifactorial disorder is important for the development of appropriate pharmacological and genetic treatments. Second, we review the characteristics, advantages, and disadvantages of the latest advances in gene therapy for SCD, from lentiviral-vector-based approaches to gene-editing strategies.
Collapse
Affiliation(s)
- Giulia Hardouin
- Laboratory of Chromatin and Gene Regulation During Development, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France; ,
- Centre d'Investigation Clinique Spécialisé en Biothérapie, Département de Biothérapie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; ,
- Human Lymphohematopoiesis Laboratory, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France;
| | - Elisa Magrin
- Centre d'Investigation Clinique Spécialisé en Biothérapie, Département de Biothérapie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; ,
| | - Alice Corsia
- Human Lymphohematopoiesis Laboratory, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France;
| | - Marina Cavazzana
- Centre d'Investigation Clinique Spécialisé en Biothérapie, Département de Biothérapie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France; ,
- Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France
- Université Paris Cité, Paris, France
| | - Annarita Miccio
- Laboratory of Chromatin and Gene Regulation During Development, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France; ,
| | - Michaela Semeraro
- Université Paris Cité, Paris, France
- Centre d'Investigation Clinique and Unité de Recherche Clinique, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France;
| |
Collapse
|
12
|
Piel FB, Rees DC, DeBaun MR, Nnodu O, Ranque B, Thompson AA, Ware RE, Abboud MR, Abraham A, Ambrose EE, Andemariam B, Colah R, Colombatti R, Conran N, Costa FF, Cronin RM, de Montalembert M, Elion J, Esrick E, Greenway AL, Idris IM, Issom DZ, Jain D, Jordan LC, Kaplan ZS, King AA, Lloyd-Puryear M, Oppong SA, Sharma A, Sung L, Tshilolo L, Wilkie DJ, Ohene-Frempong K. Defining global strategies to improve outcomes in sickle cell disease: a Lancet Haematology Commission. Lancet Haematol 2023; 10:e633-e686. [PMID: 37451304 PMCID: PMC11459696 DOI: 10.1016/s2352-3026(23)00096-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/31/2023] [Accepted: 04/12/2023] [Indexed: 07/18/2023]
Abstract
All over the world, people with sickle cell disease (an inherited condition) have premature deaths and preventable severe chronic complications, which considerably affect their quality of life, career progression, and financial status. In addition, these people are often affected by stigmatisation or structural racism, which can contribute to stress and poor mental health. Inequalities affecting people with sickle cell disease are also reflected in the distribution of the disease—mainly in sub-Saharan Africa, India, and the Caribbean—whereas interventions, clinical trials, and funding are mostly available in North America, Europe, and the Middle East. Although some of these characteristics also affect people with other genetic diseases, the fate of people with sickle cell disease seems to be particularly unfair. Simple, effective interventions to reduce the mortality and morbidity associated with sickle cell disease are available. The main obstacle preventing better outcomes in this condition, which is a neglected disease, is associated with inequalities impacting the patient populations. The aim of this Commission is to highlight the problems associated with sickle cell disease and to identify achievable goals to improve outcomes both in the short and long term. The ambition for the management of people with sickle cell disease is that curative treatments become available to every person with the condition. Although this would have seemed unrealistic a decade ago, developments in gene therapy make this potentially achievable, albeit in the distant future. Until these curative technologies are fully developed and become widely available, health-care professionals (with the support of policy makers, funders, etc) should make sure that a minimum standard of care (including screening, prophylaxis against infection, acute medical care, safe blood transfusion, and hydroxyurea) is available to all patients. In considering what needs to be achieved to reduce the global burden of sickle cell disease and improve the quality of life of patients, this Commission focuses on five key areas: the epidemiology of sickle cell disease (Section 1 ); screening and prevention (Section 2 ); established and emerging treatments for the management of the disease (Section 3 ); cellular therapies with curative potential (Section 4 ); and training and education needs (Section 5 ). As clinicians, researchers, and patients, our objective to reduce the global burden of sickle cell disease aligns with wider public health aims to reduce inequalities, improve health for all, and develop personalised treatment options. We have observed in the past few years some long-awaited momentum following the development of innovative point-of-care testing devices, new approved drugs, and emerging curative options. Reducing the burden of sickle cell disease will require substantial financial and political commitment, but it will impact the lives of millions of patients and families worldwide and the lessons learned in achieving this goal would unarguably benefit society as a whole.
Collapse
Affiliation(s)
- Frédéric B Piel
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
| | - David C Rees
- Department of Paediatric Haematology, King's College London, King's College Hospital, London, UK
| | - Michael R DeBaun
- Department of Pediatrics, Vanderbilt-Meharry Center of Excellence for Sickle Cell Disease, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Obiageli Nnodu
- Department of Haematology and Blood Transfusion, College of Health Sciences and Centre of Excellence for Sickle Cell Disease Research and Training, University of Abuja, Abuja, Nigeria
| | - Brigitte Ranque
- Department of Internal Medicine, Georges Pompidou European Hospital, Assistance Publique-Hopitaux de Paris Centre, University of Paris Cité, Paris, France
| | - Alexis A Thompson
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Russell E Ware
- Division of Hematology and Global Health Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Miguel R Abboud
- Department of Pediatrics and Adolescent Medicine, and Sickle Cell Program, American University of Beirut, Beirut, Lebanon
| | - Allistair Abraham
- Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, USA
| | - Emmanuela E Ambrose
- Department of Paediatrics and Child Health, Bugando Medical Centre, Mwanza, Tanzania
| | - Biree Andemariam
- New England Sickle Cell Institute, University of Connecticut Health, Connecticut, USA
| | - Roshan Colah
- Department of Haematogenetics, Indian Council of Medical Research National Institute of Immunohaematology, Mumbai, India
| | - Raffaella Colombatti
- Pediatric Oncology Hematology Unit, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Nicola Conran
- Department of Clinical Medicine, School of Medical Sciences, Center of Hematology and Hemotherapy (Hemocentro), University of Campinas-UNICAMP, Campinas, Brazil
| | - Fernando F Costa
- Department of Clinical Medicine, School of Medical Sciences, Center of Hematology and Hemotherapy (Hemocentro), University of Campinas-UNICAMP, Campinas, Brazil
| | - Robert M Cronin
- Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Mariane de Montalembert
- Department of Pediatrics, Necker-Enfants Malades Hospital, Assistance Publique-Hopitaux de Paris Centre, Paris, France
| | - Jacques Elion
- Paris Cité University and University of the Antilles, Inserm, BIGR, Paris, France
| | - Erica Esrick
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA, USA
| | - Anthea L Greenway
- Department Clinical Haematology, Royal Children's Hospital, Parkville and Department Haematology, Monash Health, Clayton, VIC, Australia
| | - Ibrahim M Idris
- Department of Hematology, Aminu Kano Teaching Hospital/Bayero University Kano, Kano, Nigeria
| | - David-Zacharie Issom
- Department of Business Information Systems, School of Management, HES-SO University of Applied Sciences and Arts of Western Switzerland, Geneva, Switzerland
| | - Dipty Jain
- Department of Paediatrics, Government Medical College, Nagpur, India
| | - Lori C Jordan
- Department of Pediatrics, Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zane S Kaplan
- Department of Clinical Haematology, Monash Health and Monash University, Melbourne, VIC, Australia
| | - Allison A King
- Departments of Pediatrics and Internal Medicine, Divisions of Pediatric Hematology and Oncology and Hematology, Washington University School of Medicine, St Louis, MO, USA
| | - Michele Lloyd-Puryear
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Samuel A Oppong
- Department of Obstetrics and Gynecology, University of Ghana Medical School, Accra, Ghana
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Lillian Sung
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Leon Tshilolo
- Institute of Biomedical Research/CEFA Monkole Hospital Centre and Official University of Mbuji-Mayi, Mbuji-Mayi, Democratic Republic of the Congo
| | - Diana J Wilkie
- Department of Biobehavioral Nursing Science, College of Nursing, University of Florida, Gainesville, FL, USA
| | - Kwaku Ohene-Frempong
- Division of Hematology, Children's Hospital of Philadelphia, Pennsylvania, USA; Sickle Cell Foundation of Ghana, Kumasi, Ghana
| |
Collapse
|
13
|
Papaioannou I, Owen JS, Yáñez‐Muñoz RJ. Clinical applications of gene therapy for rare diseases: A review. Int J Exp Pathol 2023; 104:154-176. [PMID: 37177842 PMCID: PMC10349259 DOI: 10.1111/iep.12478] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 03/08/2023] [Accepted: 04/16/2023] [Indexed: 05/15/2023] Open
Abstract
Rare diseases collectively exact a high toll on society due to their sheer number and overall prevalence. Their heterogeneity, diversity, and nature pose daunting clinical challenges for both management and treatment. In this review, we discuss recent advances in clinical applications of gene therapy for rare diseases, focusing on a variety of viral and non-viral strategies. The use of adeno-associated virus (AAV) vectors is discussed in the context of Luxturna, licenced for the treatment of RPE65 deficiency in the retinal epithelium. Imlygic, a herpes virus vector licenced for the treatment of refractory metastatic melanoma, will be an example of oncolytic vectors developed against rare cancers. Yescarta and Kymriah will showcase the use of retrovirus and lentivirus vectors in the autologous ex vivo production of chimeric antigen receptor T cells (CAR-T), licenced for the treatment of refractory leukaemias and lymphomas. Similar retroviral and lentiviral technology can be applied to autologous haematopoietic stem cells, exemplified by Strimvelis and Zynteglo, licenced treatments for adenosine deaminase-severe combined immunodeficiency (ADA-SCID) and β-thalassaemia respectively. Antisense oligonucleotide technologies will be highlighted through Onpattro and Tegsedi, RNA interference drugs licenced for familial transthyretin (TTR) amyloidosis, and Spinraza, a splice-switching treatment for spinal muscular atrophy (SMA). An initial comparison of the effectiveness of AAV and oligonucleotide therapies in SMA is possible with Zolgensma, an AAV serotype 9 vector, and Spinraza. Through these examples of marketed gene therapies and gene cell therapies, we will discuss the expanding applications of such novel technologies to previously intractable rare diseases.
Collapse
Affiliation(s)
| | - James S. Owen
- Division of MedicineUniversity College LondonLondonUK
| | - Rafael J. Yáñez‐Muñoz
- AGCTlab.orgCentre of Gene and Cell TherapyCentre for Biomedical SciencesDepartment of Biological SciencesSchool of Life Sciences and the EnvironmentRoyal Holloway University of LondonEghamUK
| |
Collapse
|
14
|
Jiang T, Gonzalez KM, Cordova LE, Lu J. Nanotechnology-enabled gene delivery for cancer and other genetic diseases. Expert Opin Drug Deliv 2023; 20:523-540. [PMID: 37017558 PMCID: PMC10164135 DOI: 10.1080/17425247.2023.2200246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 04/04/2023] [Indexed: 04/06/2023]
Abstract
INTRODUCTION Despite gene therapy is ideal for genetic abnormality-related diseases, the easy degradation, poor targeting, and inefficiency in entering targeted cells are plaguing the effective delivery of gene therapy. Viral and non-viral vectors have been used for delivering gene therapeutics in vivo by safeguarding nucleic acid agents to target cells and to reach the specific intracellular location. A variety of nanotechnology-enabled safe and efficient systems have been successfully developed to improve the targeting ability for effective therapeutic delivery of genetic drugs. AREAS COVERED In this review, we outline the multiple biological barriers associated with gene delivery process, and highlight recent advances to gene therapy strategy in vivo, including gene correction, gene silencing, gene activation and genome editing. We point out current developments and challenges exist of non-viral and viral vector systems in association with chemical and physical gene delivery technologies and their potential for the future. EXPERT OPINION This review focuses on the opportunities and challenges to various gene therapy strategy, with specific emphasis on overcoming the challenges through the development of biocompatibility and smart gene vectors for potential clinical application.
Collapse
Affiliation(s)
- Tong Jiang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Karina Marie Gonzalez
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Leyla Estrella Cordova
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, Arizona, 85721, United States
- NCI-designated University of Arizona Comprehensive Cancer Center, Tucson, Arizona, 85721, United States
- BIO5 Institute, The University of Arizona, Tucson, Arizona, 85721, United States
- Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, 85721, United States
| |
Collapse
|
15
|
Christakopoulos GE, Telange R, Yen J, Weiss MJ. Gene Therapy and Gene Editing for β-Thalassemia. Hematol Oncol Clin North Am 2023; 37:433-447. [PMID: 36907613 PMCID: PMC10355137 DOI: 10.1016/j.hoc.2022.12.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
After many years of intensive research, emerging data from clinical trials indicate that gene therapy for transfusion-dependent β-thalassemia is now possible. Strategies for therapeutic manipulation of patient hematopoietic stem cells include lentiviral transduction of a functional erythroid-expressed β-globin gene and genome editing to activate fetal hemoglobin production in patient red blood cells. Gene therapy for β-thalassemia and other blood disorders will invariably improve as experience accumulates over time. The best overall approaches are not known and perhaps not yet established. Gene therapy comes at a high cost, and collaboration between multiple stakeholders is required to ensure that these new medicines are administered equitably.
Collapse
Affiliation(s)
- Georgios E Christakopoulos
- Department of Oncology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS #355, Memphis, TN 38105, USA
| | - Raul Telange
- Department of Hematology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS #355, Memphis, TN 38105, USA
| | - Jonathan Yen
- Department of Hematology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS #355, Memphis, TN 38105, USA
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS #355, Memphis, TN 38105, USA.
| |
Collapse
|
16
|
Hu S, Xu S, Lu W, Si Y, Wang Y, Du Z, Wang Y, Feng Z, Tang X. The research on the treatment of primary immunodeficiency diseases by hematopoietic stem cell transplantation: A bibliometric analysis from 2013 to 2022. Medicine (Baltimore) 2023; 102:e33295. [PMID: 37000105 PMCID: PMC10063298 DOI: 10.1097/md.0000000000033295] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 04/01/2023] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is curative in patients with primary immunodeficiency syndrome. The safety and efficacy of HSCT as a therapeutic option for primary immunodeficiency diseases (PID) have been studied by many research groups. The purpose of our study was to perform a bibliometric analysis of research on HSCT for the treatment of PID, to assess research trends in this field, and note future research priorities. The Web of Science Core Collection (WOSCC) was used to identify relevant publications. VOSviewer and CiteSpace software were used to analyze bibliometric parameters, such as yearly records, authors, grouped authors, countries, institutions, categories and keywords. There are 602 relevant records for the last decade (2013-2022). The top 5 productive authors and high-quality paper journals are listed. Reference co-citations analysis demonstrated recent research trends were "inborn errors of immunity," "gene editing," and "enteropathy." Research on HSCT for the treatment of PID has increased rapidly in the last decade, and bibliometrics are valuable for researchers to obtain an overview of hot categories, academic collaborations and trends in this study field.
Collapse
Affiliation(s)
- Siqi Hu
- Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
- Institute of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
- Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
| | - Shixia Xu
- Department of Pediatrics, Eden Hospital, Beijing, China
| | - Wei Lu
- Institute of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
- Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
- Department of Hematology and Transplantation, Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
- Department of Children’s Internal Medicine, Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
| | - Yingjian Si
- Institute of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
- Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
- Department of Hematology and Transplantation, Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
- Department of Children’s Internal Medicine, Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
| | - Ya Wang
- Institute of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
- Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
- Department of Hematology and Transplantation, Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
- Department of Children’s Internal Medicine, Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
| | - Zhenlan Du
- Institute of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
- Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
- Department of Hematology and Transplantation, Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
- Department of Children’s Internal Medicine, Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
| | - Yi Wang
- Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
- Institute of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
- Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
| | - Zhichun Feng
- Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
- Institute of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
- Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
| | - Xiangfeng Tang
- Institute of Pediatrics, the Seventh Medical Center of PLA General Hospital, Beijing, China
- National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China
- Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
- Department of Hematology and Transplantation, Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
- Department of Children’s Internal Medicine, Faculty of Pediatrics, the Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
17
|
Segura EER, Ayoub PG, Hart KL, Kohn DB. Gene Therapy for β-Hemoglobinopathies: From Discovery to Clinical Trials. Viruses 2023; 15:713. [PMID: 36992422 PMCID: PMC10054523 DOI: 10.3390/v15030713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/03/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Investigations to understand the function and control of the globin genes have led to some of the most exciting molecular discoveries and biomedical breakthroughs of the 20th and 21st centuries. Extensive characterization of the globin gene locus, accompanied by pioneering work on the utilization of viruses as human gene delivery tools in human hematopoietic stem and progenitor cells (HPSCs), has led to transformative and successful therapies via autologous hematopoietic stem-cell transplant with gene therapy (HSCT-GT). Due to the advanced understanding of the β-globin gene cluster, the first diseases considered for autologous HSCT-GT were two prevalent β-hemoglobinopathies: sickle cell disease and β-thalassemia, both affecting functional β-globin chains and leading to substantial morbidity. Both conditions are suitable for allogeneic HSCT; however, this therapy comes with serious risks and is most effective using an HLA-matched family donor (which is not available for most patients) to obtain optimal therapeutic and safe benefits. Transplants from unrelated or haplo-identical donors carry higher risks, although they are progressively improving. Conversely, HSCT-GT utilizes the patient's own HSPCs, broadening access to more patients. Several gene therapy clinical trials have been reported to have achieved significant disease improvement, and more are underway. Based on the safety and the therapeutic success of autologous HSCT-GT, the U.S. Food and Drug Administration (FDA) in 2022 approved an HSCT-GT for β-thalassemia (Zynteglo™). This review illuminates the β-globin gene research journey, adversities faced, and achievements reached; it highlights important molecular and genetic findings of the β-globin locus, describes the predominant globin vectors, and concludes by describing promising results from clinical trials for both sickle cell disease and β-thalassemia.
Collapse
Affiliation(s)
- Eva Eugenie Rose Segura
- Molecular Biology Interdepartmental Doctoral Program, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA;
| | - Paul George Ayoub
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Kevyn Lopez Hart
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Donald Barry Kohn
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Pediatrics (Hematology/Oncology), David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center for Stem Cell Research and Regenerative Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
18
|
Applying the CRISPR/Cas9 for treating human and animal diseases: a comprehensive review. ANNALS OF ANIMAL SCIENCE 2023. [DOI: 10.2478/aoas-2023-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Abstract
Recently, genome editing tools have been extensively used in many biomedical sciences. The gene editing system is applied to modify the DNA sequences in the cellular system to comprehend their physiological response. A developing genome editing technology like clustered regularly short palindromic repeats (CRISPR) is widely expended in medical sciences. CRISPR and CRISPR-associated protein 9 (CRISPR/Cas9) system is being exploited to edit any DNA mutations related to inherited ailments to investigate in animals (in vivo) and cell lines (in vitro). Remarkably, CRISPR/Cas9 could be employed to examine treatments of many human genetic diseases such as Cystic fibrosis, Tyrosinemia, Phenylketonuria, Muscular dystrophy, Parkinson’s disease, Retinoschisis, Hemophilia, β-Thalassemia and Atherosclerosis. Moreover, CRISPR/Cas9 was used for disease resistance such as Tuberculosis, Johne’s diseases, chronic enteritis, and Brucellosis in animals. Finally, this review discusses existing progress in treating hereditary diseases using CRISPR/Cas9 technology and the high points accompanying obstacles.
Collapse
|
19
|
Mollashahi B, Latifi-Navid H, Owliaee I, Shamdani S, Uzan G, Jamehdor S, Naserian S. Research and Therapeutic Approaches in Stem Cell Genome Editing by CRISPR Toolkit. Molecules 2023; 28:1982. [PMID: 36838970 PMCID: PMC9961668 DOI: 10.3390/molecules28041982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
The most widely used genome editing toolkit is CRISPR (clustered regularly interspaced short palindromic repeats). It provides the possibility of replacing and modifying DNA and RNA nucleotides. Furthermore, with advancements in biological technology, inhibition and activation of the transcription of specific gene(s) has become possible. Bioinformatics tools that target the evolution of CRISPR-associated protein 9 (Cas9) turn this protein into a vehicle that is specific for a DNA or RNA region with single guide RNA (sgRNA). This toolkit could be used by researchers to investigate the function of stem cell gene(s). Here, in this review article, we cover recent developments and applications of this technique in stem cells for research and clinical purposes and discuss different CRISPR/Cas technologies for knock-out, knock-in, activation, or inhibition of gene expression. Additionally, a comparison of several deliveries and off-target detecting strategies is discussed.
Collapse
Affiliation(s)
- Behrouz Mollashahi
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy
| | - Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran 14965/161, Iran
| | - Iman Owliaee
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamedan 6517838636, Iran
| | - Sara Shamdani
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Paris-Saclay University, 94807 Villejuif, France
- CellMedEx, 94100 Saint Maur Des Fossés, France
| | - Georges Uzan
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Paris-Saclay University, 94807 Villejuif, France
| | - Saleh Jamehdor
- Department of Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamedan 6517838636, Iran
| | - Sina Naserian
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Paris-Saclay University, 94807 Villejuif, France
- CellMedEx, 94100 Saint Maur Des Fossés, France
| |
Collapse
|
20
|
CRISPR Gene Therapy: A Promising One-Time Therapeutic Approach for Transfusion-Dependent β-Thalassemia—CRISPR-Cas9 Gene Editing for β-Thalassemia. THALASSEMIA REPORTS 2023. [DOI: 10.3390/thalassrep13010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
β-Thalassemia is an inherited hematological disorder that results from genetic changes in the β-globin gene, leading to the reduced or absent synthesis of β-globin. For several decades, the only curative treatment option for β-thalassemia has been allogeneic hematopoietic cell transplantation (allo-HCT). Nonetheless, rapid progress in genome modification technologies holds great potential for treating this disease and will soon change the current standard of care for β-thalassemia. For instance, the emergence of the CRISPR/Cas9 genome editing platform has opened the door for precision gene editing and can serve as an effective molecular treatment for a multitude of genetic diseases. Investigational studies were carried out to treat β-thalassemia patients utilizing CRISPR-based CTX001 therapy targeting the fetal hemoglobin silencer BCL11A to restore γ-globin expression in place of deficient β-globin. The results of recently carried out clinical trials provide hope of CTX001 being a promising one-time therapeutic option to treat β-hemoglobinopathies. This review provides an insight into the key scientific steps that led to the development and application of novel CRISPR/Cas9–based gene therapies as a promising therapeutic platform for transfusion-dependent β-thalassemia (TDT). Despite the resulting ethical, moral, and social challenges, CRISPR provides an excellent treatment option against hemoglobin-associated genetic diseases.
Collapse
|
21
|
Suwito BE, Adji AS, Widjaja JS, Angel SCS, Al Hajiri AZZ, Salamy NFW, Choirotussanijjah C. A Review of CRISPR Cas9 for SCA: Treatment Strategies and Could Target β-globin Gene and BCL11A Gene using CRISPR Cas9 Prevent the Patient from Sickle Cell Anemia? Open Access Maced J Med Sci 2023. [DOI: 10.3889/oamjms.2023.11435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND: Sickle cell anemia is a hereditary globin chain condition that leads to hemolysis and persistent organ damage. Chronic hemolytic anemia, severe acute and chronic pain, and end-organ destruction occur throughout the lifespan of sickle cell anemia. SCD is associated with a higher risk of mortality. Genome editing with CRISPR-associated regularly interspersed short palindromic repeats (CRISPR/Cas9) have therapeutic potential for sickle cell anemia thala.
AIM: This research aimed to see if using CRISPR/Cas9 to target β-globin gene is an effective therapeutic and if it has a long-term effect on Sickle Cell Anemia.
METHODS: The method used in this study summarizes the article by looking for keywords that have been determined in the title and abstract. The authors used official guidelines from Science Direct, PubMed, Google Scholar, and Journal Molecular Biology to select full-text articles published within the last decade, prioritizing searches within the past 10 years.
RESULTS: CRISPR/Cas9-mediated genome editing in clinical trials contributes to α-globin gene deletion correcting β-thalassemia through balanced α- and β-globin ratios and inhibiting disease progression.
CONCLUSION: HBB and BCL11A targeting by CRISPR/Cas9 deletion effectively inactivate BCL11A, a repressor of fetal hemoglobin production. However, further research is needed to determine its side effects and safety.
Collapse
|
22
|
Kanter J, Thompson AA, Pierciey FJ, Hsieh M, Uchida N, Leboulch P, Schmidt M, Bonner M, Guo R, Miller A, Ribeil JA, Davidson D, Asmal M, Walters MC, Tisdale JF. Lovo-cel gene therapy for sickle cell disease: Treatment process evolution and outcomes in the initial groups of the HGB-206 study. Am J Hematol 2023; 98:11-22. [PMID: 36161320 PMCID: PMC10092845 DOI: 10.1002/ajh.26741] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/14/2022] [Accepted: 09/21/2022] [Indexed: 02/04/2023]
Abstract
lovo-cel (bb1111; LentiGlobin for sickle cell disease [SCD]) gene therapy (GT) comprises autologous transplantation of hematopoietic stem and progenitor cells transduced with the BB305 lentiviral vector encoding a modified β-globin gene (βA-T87Q ) to produce anti-sickling hemoglobin (HbAT87Q ). The efficacy and safety of lovo-cel for SCD are being evaluated in the ongoing phase 1/2 HGB-206 study (ClinicalTrials.gov: NCT02140554). The treatment process evolved over time, using learnings from outcomes in the initial patients to optimize lovo-cel's benefit-risk profile. Following modest expression of HbAT87Q in the initial patients (Group A, n = 7), alterations were made to the treatment process for patients subsequently enrolled in Group B (n = 2, patients B1 and B2), including improvements to cell collection and lovo-cel manufacturing. After 6 months, median Group A peripheral blood vector copy number (≥0.08 c/dg) and HbAT87Q levels (≥0.46 g/dL) were inadequate for substantial clinical effect but stable and sustained over 5.5 years; both markedly improved in Group B (patient B1: ≥0.53 c/dg and ≥2.69 g/dL; patient B2: ≥2.14 c/dg and ≥6.40 g/dL, respectively) and generated improved biologic and clinical efficacy in Group B, including higher total hemoglobin and decreased hemolysis. The safety of the lovo-cel for SCD treatment regimen largely reflected the known side effects of HSPC collection, busulfan conditioning regimen, and underlying SCD; acute myeloid leukemia was observed in two patients in Group A and deemed unlikely related to insertional oncogenesis. Changes made during development of the lovo-cel treatment process were associated with improved outcomes and provide lessons for future SCD GT studies.
Collapse
Affiliation(s)
- Julie Kanter
- Department of Hematology-Oncology, University of Alabama Birmingham, Birmingham, Alabama, USA
| | - Alexis A Thompson
- Division of Hematology, Oncology, and Stem Cell Transplantation, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.,Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | | | - Matthew Hsieh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute/National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Naoya Uchida
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute/National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Philippe Leboulch
- Commissariat à l'énergie atomique et aux énergies alternatives, Institute of Emerging Disease and Innovative Therapies, Fontenay-aux-Roses, France.,Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Ruiting Guo
- bluebird bio, Inc., Somerville, Massachusetts, USA
| | - Alex Miller
- bluebird bio, Inc., Somerville, Massachusetts, USA
| | | | | | | | - Mark C Walters
- Division of Hematology, University of California San Francisco Benioff Children's Hospital, Oakland, California, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute/National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
23
|
Cavazzana M. [Advances in gene therapy in genetic diseases of the hematopoietic system]. Med Sci (Paris) 2022; 38:768-771. [PMID: 36219075 DOI: 10.1051/medsci/2022120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Marina Cavazzana
- Département de biothérapie, hôpital universitaire Necker-Enfants malades, groupe hospitalier Paris Centre, AP-HP, Paris, France - Centre d'investigation clinique biothérapie, groupe hospitalier universitaire Paris centre, AP-HP, Inserm CIC 1416 Paris, France - Institut Imagine, université Paris Cité, Paris, France
| |
Collapse
|
24
|
Lin Q, Xie Y, Zhong X, Sun X, Wang D. RS12574989 and haplotype associated with α/β-chain imbalance and population HbA2 reduction. BMC Med Genomics 2022; 15:179. [PMID: 35971149 PMCID: PMC9377088 DOI: 10.1186/s12920-022-01333-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/08/2022] [Indexed: 11/10/2022] Open
Abstract
Determining the associated relationship of genotype and phenomenon would benefit the understanding of disease and renew disease intervention means. 14,518 patients who underwent haemoglobin electrophoresis from June 2020 to December 2020 were enrolled in our study, and additional data including sex, age and routine blood examination results were collected. We focused on individuals with normal red blood cell indices and no common thalassemia pathogenic mutation and selected three groups for the following study: the control group (2.5% ≤ HbA2 ≤ 3.5%), the HbA2 under 2.5 group (HbA2 < 2.5%) and the HbA2 under 2.4 group (HbA2 < 2.4%). Four regions of β-globin regulation were sequenced. Statistical analysis was conducted to compare the collected information of the three groups and the genotype distributions in the control group and sequenced group. The HbA2 under 2.5 group was characterized by a majority of females and lower red blood cell counts and haemoglobin compared with the control group. There were genotypes associated with the grouping as the T of rs12574989 and TTTAGC of the haplotype were significantly increased in the HbA2 under 2.4 group and CTTAGC was significantly decreased in the HbA2 under 2.4 group. This study demonstrated that the genotypes of the population associated with HbA2 were reduced in southern China.
Collapse
Affiliation(s)
- Qiyin Lin
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Guangzhou, 510150, Guangdong, China.,Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Yingjun Xie
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Guangzhou, 510150, Guangdong, China.,Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Xuan Zhong
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, 510000, Guangdong, China
| | - Xiaofang Sun
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Guangzhou, 510150, Guangdong, China.,Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China
| | - Ding Wang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Guangzhou, 510150, Guangdong, China. .,Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China.
| |
Collapse
|
25
|
Chauhan W, Shoaib S, Fatma R, Zaka‐ur‐Rab Z, Afzal M. β‐thalassemia, and the advent of new Interventions beyond Transfusion and Iron chelation. Br J Clin Pharmacol 2022; 88:3610-3626. [DOI: 10.1111/bcp.15343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/10/2022] [Accepted: 03/29/2022] [Indexed: 01/19/2023] Open
Affiliation(s)
- Waseem Chauhan
- Human Genetics and Toxicology Laboratory, Department of Zoology Aligarh Muslim University Aligarh India
| | - Shoaib Shoaib
- Department of Biochemistry, JNMC Aligarh Muslim University Aligarh India
| | - Rafat Fatma
- Human Genetics and Toxicology Laboratory, Department of Zoology Aligarh Muslim University Aligarh India
| | - Zeeba Zaka‐ur‐Rab
- Department of Pediatrics, JNMC Aligarh Muslim University Aligarh India
| | - Mohammad Afzal
- Human Genetics and Toxicology Laboratory, Department of Zoology Aligarh Muslim University Aligarh India
| |
Collapse
|
26
|
Abbasalipour M, Khosravi MA, Zeinali S, Khanahmad H, Azadmanesh K, Karimipoor M. Lentiviral vector containing beta-globin gene for beta thalassemia gene therapy. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
27
|
Kanter J, Walters MC, Krishnamurti L, Mapara MY, Kwiatkowski JL, Rifkin-Zenenberg S, Aygun B, Kasow KA, Pierciey FJ, Bonner M, Miller A, Zhang X, Lynch J, Kim D, Ribeil JA, Asmal M, Goyal S, Thompson AA, Tisdale JF. Biologic and Clinical Efficacy of LentiGlobin for Sickle Cell Disease. N Engl J Med 2022; 386:617-628. [PMID: 34898139 DOI: 10.1056/nejmoa2117175] [Citation(s) in RCA: 173] [Impact Index Per Article: 86.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Sickle cell disease is characterized by the painful recurrence of vaso-occlusive events. Gene therapy with the use of LentiGlobin for sickle cell disease (bb1111; lovotibeglogene autotemcel) consists of autologous transplantation of hematopoietic stem and progenitor cells transduced with the BB305 lentiviral vector encoding a modified β-globin gene, which produces an antisickling hemoglobin, HbAT87Q. METHODS In this ongoing phase 1-2 study, we optimized the treatment process in the initial 7 patients in Group A and 2 patients in Group B with sickle cell disease. Group C was established for the pivotal evaluation of LentiGlobin for sickle cell disease, and we adopted a more stringent inclusion criterion that required a minimum of four severe vaso-occlusive events in the 24 months before enrollment. In this unprespecified interim analysis, we evaluated the safety and efficacy of LentiGlobin in 35 patients enrolled in Group C. Included in this analysis was the number of severe vaso-occlusive events after LentiGlobin infusion among patients with at least four vaso-occlusive events in the 24 months before enrollment and with at least 6 months of follow-up. RESULTS As of February 2021, cell collection had been initiated in 43 patients in Group C; 35 received a LentiGlobin infusion, with a median follow-up of 17.3 months (range, 3.7 to 37.6). Engraftment occurred in all 35 patients. The median total hemoglobin level increased from 8.5 g per deciliter at baseline to 11 g or more per deciliter from 6 months through 36 months after infusion. HbAT87Q contributed at least 40% of total hemoglobin and was distributed across a mean (±SD) of 85±8% of red cells. Hemolysis markers were reduced. Among the 25 patients who could be evaluated, all had resolution of severe vaso-occlusive events, as compared with a median of 3.5 events per year (range, 2.0 to 13.5) in the 24 months before enrollment. Three patients had a nonserious adverse event related or possibly related to LentiGlobin that resolved within 1 week after onset. No cases of hematologic cancer were observed during up to 37.6 months of follow-up. CONCLUSIONS One-time treatment with LentiGlobin resulted in sustained production of HbAT87Q in most red cells, leading to reduced hemolysis and complete resolution of severe vaso-occlusive events. (Funded by Bluebird Bio; HGB-206 ClinicalTrials.gov number, NCT02140554.).
Collapse
Affiliation(s)
- Julie Kanter
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Mark C Walters
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Lakshmanan Krishnamurti
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Markus Y Mapara
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Janet L Kwiatkowski
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Stacey Rifkin-Zenenberg
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Banu Aygun
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Kimberly A Kasow
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Francis J Pierciey
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Melissa Bonner
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Alex Miller
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Xinyan Zhang
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Jessie Lynch
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Dennis Kim
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Jean-Antoine Ribeil
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Mohammed Asmal
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Sunita Goyal
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - Alexis A Thompson
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| | - John F Tisdale
- From the University of Alabama Birmingham, Birmingham (J.K.); UCSF Benioff Children's Hospital, Oakland, CA (M.C.W.); Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta (L.K.); the Division of Hematology-Oncology, Columbia Center for Translational Immunology, Columbia University Medical Center, New York (M.Y.M.), the Division of Pediatric Hematology, Oncology and Cellular Therapy, Cohen Children's Medical Center, New Hyde Park (B.A.), and Zucker School of Medicine at Hofstra-Northwell, Hempstead (B.A.) - all in New York; the Division of Hematology, Children's Hospital of Philadelphia, and the Department of Pediatrics, University of Pennsylvania Perelman School of Medicine - both in Philadelphia (J.L.K.); Hackensack University Medical Center, Hackensack, NJ (S.R.-Z.); the University of North Carolina at Chapel Hill, Chapel Hill (K.A.K.); Bluebird Bio, Cambridge, MA (F.J.P., M.B., A.M., X.Z., J.L., D.K., J.-A.R., M.A., S.G.); Northwestern University Feinberg School of Medicine and Ann and Robert H. Lurie Children's Hospital - both in Chicago (A.A.T.); and the Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute and National Institute of Diabetes and Digestive and Kidney Diseases (NHLBI-NIDDK), National Institutes of Health, Bethesda, MD (J.F.T.)
| |
Collapse
|
28
|
Locatelli F, Thompson AA, Kwiatkowski JL, Porter JB, Thrasher AJ, Hongeng S, Sauer MG, Thuret I, Lal A, Algeri M, Schneiderman J, Olson TS, Carpenter B, Amrolia PJ, Anurathapan U, Schambach A, Chabannon C, Schmidt M, Labik I, Elliot H, Guo R, Asmal M, Colvin RA, Walters MC. Betibeglogene Autotemcel Gene Therapy for Non-β 0/β 0 Genotype β-Thalassemia. N Engl J Med 2022; 386:415-427. [PMID: 34891223 DOI: 10.1056/nejmoa2113206] [Citation(s) in RCA: 105] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Betibeglogene autotemcel (beti-cel) gene therapy for transfusion-dependent β-thalassemia contains autologous CD34+ hematopoietic stem cells and progenitor cells transduced with the BB305 lentiviral vector encoding the β-globin (βA-T87Q) gene. METHODS In this open-label, phase 3 study, we evaluated the efficacy and safety of beti-cel in adult and pediatric patients with transfusion-dependent β-thalassemia and a non-β0/β0 genotype. Patients underwent myeloablation with busulfan (with doses adjusted on the basis of pharmacokinetic analysis) and received beti-cel intravenously. The primary end point was transfusion independence (i.e., a weighted average hemoglobin level of ≥9 g per deciliter without red-cell transfusions for ≥12 months). RESULTS A total of 23 patients were enrolled and received treatment, with a median follow-up of 29.5 months (range, 13.0 to 48.2). Transfusion independence occurred in 20 of 22 patients who could be evaluated (91%), including 6 of 7 patients (86%) who were younger than 12 years of age. The average hemoglobin level during transfusion independence was 11.7 g per deciliter (range, 9.5 to 12.8). Twelve months after beti-cel infusion, the median level of gene therapy-derived adult hemoglobin (HbA) with a T87Q amino acid substitution (HbAT87Q) was 8.7 g per deciliter (range, 5.2 to 10.6) in patients who had transfusion independence. The safety profile of beti-cel was consistent with that of busulfan-based myeloablation. Four patients had at least one adverse event that was considered by the investigators to be related or possibly related to beti-cel; all events were nonserious except for thrombocytopenia (in 1 patient). No cases of cancer were observed. CONCLUSIONS Treatment with beti-cel resulted in a sustained HbAT87Q level and a total hemoglobin level that was high enough to enable transfusion independence in most patients with a non-β0/β0 genotype, including those younger than 12 years of age. (Funded by Bluebird Bio; HGB-207 ClinicalTrials.gov number, NCT02906202.).
Collapse
Affiliation(s)
- Franco Locatelli
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Alexis A Thompson
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Janet L Kwiatkowski
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - John B Porter
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Adrian J Thrasher
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Suradej Hongeng
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Martin G Sauer
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Isabelle Thuret
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Ashutosh Lal
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Mattia Algeri
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Jennifer Schneiderman
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Timothy S Olson
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Ben Carpenter
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Persis J Amrolia
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Usanarat Anurathapan
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Axel Schambach
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Christian Chabannon
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Manfred Schmidt
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Ivan Labik
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Heidi Elliot
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Ruiting Guo
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Mohammed Asmal
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Richard A Colvin
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Mark C Walters
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| |
Collapse
|
29
|
Abdelnour SA, Xie L, Hassanin AA, Zuo E, Lu Y. The Potential of CRISPR/Cas9 Gene Editing as a Treatment Strategy for Inherited Diseases. Front Cell Dev Biol 2022; 9:699597. [PMID: 34977000 PMCID: PMC8715006 DOI: 10.3389/fcell.2021.699597] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR) is a promising innovative technology for genomic editing that offers scientists the chance to edit DNA structures and change gene function. It has several possible uses consisting of editing inherited deficiencies, treating, and reducing the spread of disorders. Recently, reports have demonstrated the creation of synthetic RNA molecules and supplying them alongside Cas9 into genome of eukaryotes, since distinct specific regions of the genome can be manipulated and targeted. The therapeutic potential of CRISPR/Cas9 technology is great, especially in gene therapy, in which a patient-specific mutation is genetically edited, or in the treating of human disorders that are untreatable with traditional treatments. This review focused on numerous, in vivo, in vitro, and ex vivo uses of the CRISPR/Cas9 technology in human inherited diseases, discovering the capability of this versatile in medicine and examining some of the main limitations for its upcoming use in patients. In addition to introducing a brief impression of the biology of the CRISPR/Cas9 scheme and its mechanisms, we presented the utmost recent progress in the uses of CRISPR/Cas9 technology in editing and treating of human genetic diseases.
Collapse
Affiliation(s)
- Sameh A Abdelnour
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China.,Animal Production Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Long Xie
- Center for Animal Genomics, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Abdallah A Hassanin
- Genetics Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Erwei Zuo
- Center for Animal Genomics, Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yangqing Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning, China
| |
Collapse
|
30
|
Development and clinical translation of ex vivo gene therapy. Comput Struct Biotechnol J 2022; 20:2986-3003. [PMID: 35782737 PMCID: PMC9218169 DOI: 10.1016/j.csbj.2022.06.015] [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: 02/08/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 11/27/2022] Open
Abstract
Retroviral gene therapy has emerged as a promising therapeutic modality for multiple inherited and acquired human diseases. The capability of delivering curative treatment or mediating therapeutic benefits for a long-term period following a single application fundamentally distinguishes this medical intervention from traditional medicine and various lentiviral/γ-retroviral vector-mediated gene therapy products have been approved for clinical use. Continued advances in retroviral vector engineering, genomic editing, synthetic biology and immunology will broaden the medical applications of gene therapy and improve the efficacy and safety of the treatments based on genetic correction and alteration. This review will summarize the advent and clinical translation of ex vivo gene therapy, with the focus on the milestones during the exploitation of genetically engineered hematopoietic stem cells (HSCs) tackling a variety of pathological conditions which led to marketing approval. Finally, current statue and future prospects of gene editing as an alternative therapeutic approach are also discussed.
Collapse
|
31
|
Wang X, Ma C, Rodríguez Labrada R, Qin Z, Xu T, He Z, Wei Y. Recent advances in lentiviral vectors for gene therapy. SCIENCE CHINA-LIFE SCIENCES 2021; 64:1842-1857. [PMID: 34708326 DOI: 10.1007/s11427-021-1952-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/19/2021] [Indexed: 02/05/2023]
Abstract
Lentiviral vectors (LVs), derived from human immunodeficiency virus, are powerful tools for modifying the genes of eukaryotic cells such as hematopoietic stem cells and neural cells. With the extensive and in-depth studies on this gene therapy vehicle over the past two decades, LVs have been widely used in both research and clinical trials. For instance, third-generation and self-inactive LVs have been used to introduce a gene with therapeutic potential into the host genome and achieve targeted delivery into specific tissue. When LVs are employed in leukemia, the transduced T cells recognize and kill the tumor B cells; in β-thalassemia, the transduced CD34+ cells express normal β-globin; in adenosine deaminase-deficient severe combined immunodeficiency, the autologous CD34+ cells express adenosine deaminase and realize immune reconstitution. Overall, LVs can perform significant roles in the treatment of primary immunodeficiency diseases, hemoglobinopathies, B cell leukemia, and neurodegenerative diseases. In this review, we discuss the recent developments and therapeutic applications of LVs. The safe and efficient LVs show great promise as a tool for human gene therapy.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cuicui Ma
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Roberto Rodríguez Labrada
- Department Clinical Neurophysiology, Centre for the Research and Rehabilitation of Hereditary Ataxias, Holguín, 80100, Cuba
| | - Zhou Qin
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Xu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Zhiyao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Yuquan Wei
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
32
|
Rout-Pitt N, Donnelley M, Parsons D. In vitro optimization of miniature bronchoscope lentiviral vector delivery for the small animal lung. Exp Lung Res 2021; 47:417-425. [PMID: 34632894 DOI: 10.1080/01902148.2021.1989523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Current gene therapy delivery protocols for small animal lungs typically utilize indirect dose delivery via the nasal airways, or bolus delivery directly into the trachea. Both methods can result in variable transduction throughout the lung, as well as between animals, and cannot be applied in a targeted manner. To minimize variability and improve lung coverage we previously developed and validated a method to visualize and dose gene vectors into pre-selected lobes of rat lungs using a mini-bronchoscope. Lentiviral (LV) vectors are known to be fragile and can be inactivated easily by temperature or the application of shear stresses. There are several ways that the bronchoscope could be configured to deliver the LV vector, and these could result in different amounts of functional LV vector being delivered to the lung. This study evaluated several methods of LV vector delivery through the bronchoscope, and how flow rates and LV vector stabilizing diluents impact LV vector delivery. NIH-3T3 cells were exposed to LV vector containing the green fluorescent protein (GFP) reporter gene using various bronchoscopic delivery techniques and the number of GFP-positive cells produced by each was quantified by flow cytometry. The results showed that directly drawing the LV vector into the bronchoscope tip resulted in 80-90% recovery of viable vector, and was also the simplest method of delivery. The fluid delivery rate and the use of stabilizing serum in the vector diluent had no effect on the viability of the LV vector delivered. These findings can be used to optimize LV vector dose delivery into individual lung lobes of small animal models.
Collapse
Affiliation(s)
- Nathan Rout-Pitt
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia.,Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia
| | - Martin Donnelley
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia.,Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia
| | - David Parsons
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia.,Adelaide Medical School, University of Adelaide, Adelaide, South Australia.,Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia
| |
Collapse
|
33
|
Wild-type HIV infection after treatment with lentiviral gene therapy for β-thalassemia. Blood Adv 2021; 5:2701-2706. [PMID: 34196676 DOI: 10.1182/bloodadvances.2020003680] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Betibeglogene autotemcel (beti-cel) gene therapy (GT) for patients with transfusion-dependent β-thalassemia uses autologous CD34+ cells transduced with BB305 lentiviral vector (LVV), which encodes a modified β-globin gene. BB305 LVV also contains select HIV sequences for viral packaging, reverse transcription, and integration. This case report describes a patient successfully treated with beti-cel in a phase 1/2 study (HGB-204; #NCT01745120) and subsequently diagnosed with wild-type (WT) HIV infection. From 3.5 to 21 months postinfusion, the patient stopped chronic red blood cell transfusions; total hemoglobin (Hb) and GT-derived HbAT87Q levels were 6.6 to 9.5 and 2.8 to 3.8 g/dL, respectively. At 21 months postinfusion, the patient resumed transfusions for anemia that coincided with an HIV-1 infection diagnosis. Quantitative polymerase chain reaction assays detected no replication-competent lentivirus. Next-generation sequencing confirmed WT HIV sequences. Six months after starting antiretroviral therapy, total Hb and HbAT87Q levels recovered to 8.6 and 3.6 g/dL, respectively, and 3.5 years postinfusion, 13.4 months had elapsed since the patient's last transfusion. To our knowledge, this is the first report of WT HIV infection in an LVV-based GT recipient and demonstrates persistent long-term hematopoiesis after treatment with beti-cel and the ability to differentiate between WT HIV and BB305-derived sequences.
Collapse
|
34
|
Gene Therapies for Transfusion-Dependent β-Thalassemia. Indian Pediatr 2021. [DOI: 10.1007/s13312-021-2263-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
35
|
Zheng B, Wille L, Peppel K, Hagen D, Matteson A, Ahlers J, Schaff J, Hua F, Yuraszeck T, Cobbina E, Apgar JF, Burke JM, Roberts J, Das R. A systems pharmacology model for gene therapy in sickle cell disease. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:696-708. [PMID: 34139105 PMCID: PMC8302248 DOI: 10.1002/psp4.12638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/11/2021] [Accepted: 03/27/2021] [Indexed: 11/17/2022]
Abstract
We developed a mathematical model for autologous stem cell therapy to cure sickle cell disease (SCD). Experimental therapies using this approach seek to engraft stem cells containing a curative gene. These stem cells are expected to produce a lifelong supply of red blood cells (RBCs) containing an anti‐sickling hemoglobin. This complex, multistep treatment is expensive, and there is limited patient data available from early clinical trials. Our objective was to quantify the impact of treatment parameters, such as initial stem cell dose, efficiency of lentiviral transduction, and degree of bone marrow preconditioning on engraftment efficiency, peripheral RBC numbers, and anti‐sickling hemoglobin levels over time. We used ordinary differential equations to model RBC production from progenitor cells in the bone marrow, and hemoglobin assembly from its constituent globin monomers. The model recapitulates observed RBC and hemoglobin levels in healthy and SCD phenotypes. Treatment simulations predict dynamics of stem cell engraftment and RBC containing the therapeutic gene product. Post‐treatment dynamics show an early phase of reconstitution due to short lived stem cells, followed by a sustained RBC production from stable engraftment of long‐term stem cells. This biphasic behavior was previously reported in the literature. Sensitivity analysis of the model quantified relationships between treatment parameters and efficacy. The initial dose of transduced stem cells, and the intensity of myeloablative bone marrow preconditioning are predicted to most positively impact long‐term outcomes. The quantitative systems pharmacology approach used here demonstrates the value of model‐assisted therapeutic design for gene therapies in SCD.
Collapse
Affiliation(s)
- Bo Zheng
- CSL Behring, King of Prussia, Pennsylvania, USA
| | - Lucia Wille
- Applied BioMath LLC, Concord, Massachusetts, USA
| | | | - David Hagen
- Applied BioMath LLC, Concord, Massachusetts, USA
| | | | | | - James Schaff
- Applied BioMath LLC, Concord, Massachusetts, USA
| | - Fei Hua
- Applied BioMath LLC, Concord, Massachusetts, USA
| | - Theresa Yuraszeck
- CSL Behring, King of Prussia, Pennsylvania, USA.,Applied BioMath LLC, Concord, Massachusetts, USA
| | | | | | - John M Burke
- Applied BioMath LLC, Concord, Massachusetts, USA
| | | | - Raibatak Das
- Applied BioMath LLC, Concord, Massachusetts, USA
| |
Collapse
|
36
|
Garg H, Tatiossian KJ, Peppel K, Kato GJ, Herzog E. Gene therapy as the new frontier for Sickle Cell Disease. Curr Med Chem 2021; 29:453-466. [PMID: 34047257 DOI: 10.2174/0929867328666210527092456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/28/2021] [Accepted: 04/11/2021] [Indexed: 11/22/2022]
Abstract
Sickle Cell Disease (SCD) is one of the most common monogenic disorders caused by a point mutation in the β-globin gene. This mutation results in polymerization of hemoglobin (Hb) under reduced oxygenation conditions, causing rigid sickle-shaped RBCs and hemolytic anemia. This clearly defined fundamental molecular mechanism makes SCD a prototypical target for precision therapy. Both the mutant β-globin protein and its downstream pathophysiology are pharmacological targets of intensive research. SCD also is a disease well-suited for biological interventions like gene therapy. Recent advances in hematopoietic stem cell (HSC) transplantation and gene therapy platforms, like Lentiviral vectors and gene editing strategies, expand the potentially curative options for patients with SCD. This review discusses the recent advances in precision therapy for SCD and the preclinical and clinical advances in autologous HSC gene therapy for SCD.
Collapse
Affiliation(s)
- Himanshu Garg
- CSL Behring, 1020 1St Ave, King of Prussia, PA 19406, United States
| | | | - Karsten Peppel
- CSL Behring, 1020 1St Ave, King of Prussia, PA 19406, United States
| | - Gregory J Kato
- CSL Behring, 1020 1St Ave, King of Prussia, PA 19406, United States
| | - Eva Herzog
- CSL Behring, 1020 1St Ave, King of Prussia, PA 19406, United States
| |
Collapse
|
37
|
Grech L, Borg K, Borg J. Novel therapies in β-thalassaemia. Br J Clin Pharmacol 2021; 88:2509-2524. [PMID: 34004015 DOI: 10.1111/bcp.14918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 04/30/2021] [Accepted: 05/08/2021] [Indexed: 01/19/2023] Open
Abstract
Beta-thalassaemia is one of the most significant haemoglobinopathies worldwide resulting in the synthesis of little or no β-globin chains. Without treatment, β-thalassaemia major is lethal within the first decade of life due to the complex pathophysiology, which leads to wide clinical manifestations. Current clinical management for these patients depends on repeated transfusions followed by iron-chelating therapy. Several novel approaches to correct the resulting α/β-globin chain imbalance, treat ineffective erythropoiesis and improve iron overload are currently being developed. Up to now, the only curative treatment for β-thalassemia is haematopoietic stem-cell transplantation, but this is a risky and costly procedure. Gene therapy, gene editing and base editing are emerging as a powerful approach to treat this disease. In β-thalassaemia, gene therapy involves the insertion of a vector containing the normal β-globin or γ-globin gene into haematopoietic stem cells to permanently produce normal red blood cells. Gene editing and base editing involves the use of zinc finger nucleases, transcription activator-like nucleases and clustered regularly interspaced short palindromic repeats/Cas9 to either correct the causative mutation or else insert a single nucleotide variant that will increase foetal haemoglobin. In this review, we will examine the current management strategies used to treat β-thalassaemia and focus on the novel therapies targeting ineffective erythropoiesis, improving iron overload and correction of the globin chain imbalance.
Collapse
Affiliation(s)
- Laura Grech
- Centre for Molecular Medicine and Biobanking, University of Malta, Malta
| | - Karen Borg
- Department of Public Health Medicine, Ministry for Health, Malta
| | - Joseph Borg
- Centre for Molecular Medicine and Biobanking, University of Malta, Malta.,Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Malta
| |
Collapse
|
38
|
Myelodysplastic syndrome unrelated to lentiviral vector in a patient treated with gene therapy for sickle cell disease. Blood Adv 2021; 4:2058-2063. [PMID: 32396618 DOI: 10.1182/bloodadvances.2019001330] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/27/2020] [Indexed: 12/12/2022] Open
Abstract
Key Points
Ability to accurately attribute adverse events post–gene therapy is required to describe the benefit-risk of these novel treatments. A SCD patient developed myelodysplastic syndrome post-LentiGlobin treatment; we show how insertional oncogenesis was excluded as the cause.
Collapse
|
39
|
Nualkaew T, Sii-Felice K, Giorgi M, McColl B, Gouzil J, Glaser A, Voon HPJ, Tee HY, Grigoriadis G, Svasti S, Fucharoen S, Hongeng S, Leboulch P, Payen E, Vadolas J. Coordinated β-globin expression and α2-globin reduction in a multiplex lentiviral gene therapy vector for β-thalassemia. Mol Ther 2021; 29:2841-2853. [PMID: 33940155 PMCID: PMC8417505 DOI: 10.1016/j.ymthe.2021.04.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 03/08/2021] [Accepted: 04/27/2021] [Indexed: 01/30/2023] Open
Abstract
A primary challenge in lentiviral gene therapy of β-hemoglobinopathies is to maintain low vector copy numbers to avoid genotoxicity while being reliably therapeutic for all genotypes. We designed a high-titer lentiviral vector, LVβ-shα2, that allows coordinated expression of the therapeutic βA-T87Q-globin gene and of an intron-embedded miR-30-based short hairpin RNA (shRNA) selectively targeting the α2-globin mRNA. Our approach was guided by the knowledge that moderate reduction of α-globin chain synthesis ameliorates disease severity in β-thalassemia. We demonstrate that LVβ-shα2 reduces α2-globin mRNA expression in erythroid cells while keeping α1-globin mRNA levels unchanged and βA-T87Q-globin gene expression identical to the parent vector. Compared with the first βA-T87Q-globin lentiviral vector that has received conditional marketing authorization, BB305, LVβ-shα2 shows 1.7-fold greater potency to improve α/β ratios. It may thus result in greater therapeutic efficacy and reliability for the most severe types of β-thalassemia and provide an improved benefit/risk ratio regardless of the β-thalassemia genotype.
Collapse
Affiliation(s)
- Tiwaporn Nualkaew
- Hudson Institute of Medical Research, Clayton, Melbourne, VIC 3168, Australia; Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand; Murdoch Children's Research Institute, Parkville, Melbourne, VIC 3052, Australia
| | - Karine Sii-Felice
- Division of Innovative Therapies, CEA François Jacob Biology Institute, 18 route du Panorama, 92260, Fontenay-aux-Roses, France; Paris-Saclay University, CEA, INSERM, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 18 route du Panorama, 92260 Fontenay-aux-Roses & Le Kremlin Bicêtre, France
| | - Marie Giorgi
- Division of Innovative Therapies, CEA François Jacob Biology Institute, 18 route du Panorama, 92260, Fontenay-aux-Roses, France
| | - Bradley McColl
- Murdoch Children's Research Institute, Parkville, Melbourne, VIC 3052, Australia
| | - Julie Gouzil
- Division of Innovative Therapies, CEA François Jacob Biology Institute, 18 route du Panorama, 92260, Fontenay-aux-Roses, France
| | - Astrid Glaser
- Murdoch Children's Research Institute, Parkville, Melbourne, VIC 3052, Australia
| | - Hsiao P J Voon
- Department of Biochemistry and Molecular Biology, Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Hsin Y Tee
- Hudson Institute of Medical Research, Clayton, Melbourne, VIC 3168, Australia
| | - George Grigoriadis
- Hudson Institute of Medical Research, Clayton, Melbourne, VIC 3168, Australia
| | - Saovaros Svasti
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand; Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Philippe Leboulch
- Division of Innovative Therapies, CEA François Jacob Biology Institute, 18 route du Panorama, 92260, Fontenay-aux-Roses, France; Genetics Division, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Emmanuel Payen
- Division of Innovative Therapies, CEA François Jacob Biology Institute, 18 route du Panorama, 92260, Fontenay-aux-Roses, France; Paris-Saclay University, CEA, INSERM, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial Diseases (IMVA-HB/IDMIT), 18 route du Panorama, 92260 Fontenay-aux-Roses & Le Kremlin Bicêtre, France.
| | - Jim Vadolas
- Hudson Institute of Medical Research, Clayton, Melbourne, VIC 3168, Australia; Murdoch Children's Research Institute, Parkville, Melbourne, VIC 3052, Australia.
| |
Collapse
|
40
|
Cromer MK, Camarena J, Martin RM, Lesch BJ, Vakulskas CA, Bode NM, Kurgan G, Collingwood MA, Rettig GR, Behlke MA, Lemgart VT, Zhang Y, Goyal A, Zhao F, Ponce E, Srifa W, Bak RO, Uchida N, Majeti R, Sheehan VA, Tisdale JF, Dever DP, Porteus MH. Gene replacement of α-globin with β-globin restores hemoglobin balance in β-thalassemia-derived hematopoietic stem and progenitor cells. Nat Med 2021; 27:677-687. [PMID: 33737751 PMCID: PMC8265212 DOI: 10.1038/s41591-021-01284-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/09/2021] [Indexed: 12/12/2022]
Abstract
β-Thalassemia pathology is due not only to loss of β-globin (HBB), but also to erythrotoxic accumulation and aggregation of the β-globin-binding partner, α-globin (HBA1/2). Here we describe a Cas9/AAV6-mediated genome editing strategy that can replace the entire HBA1 gene with a full-length HBB transgene in β-thalassemia-derived hematopoietic stem and progenitor cells (HSPCs), which is sufficient to normalize β-globin:α-globin messenger RNA and protein ratios and restore functional adult hemoglobin tetramers in patient-derived red blood cells. Edited HSPCs were capable of long-term and bilineage hematopoietic reconstitution in mice, establishing proof of concept for replacement of HBA1 with HBB as a novel therapeutic strategy for curing β-thalassemia.
Collapse
Affiliation(s)
- M Kyle Cromer
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Joab Camarena
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Renata M Martin
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Benjamin J Lesch
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | | | - Nicole M Bode
- Integrated DNA Technologies, Inc., Coralville, IA, USA
| | - Gavin Kurgan
- Integrated DNA Technologies, Inc., Coralville, IA, USA
| | | | | | - Mark A Behlke
- Integrated DNA Technologies, Inc., Coralville, IA, USA
| | - Viktor T Lemgart
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Yankai Zhang
- Division of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Ankush Goyal
- Division of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Feifei Zhao
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Ezequiel Ponce
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Waracharee Srifa
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Rasmus O Bak
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| | - Naoya Uchida
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Vivien A Sheehan
- Division of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel P Dever
- Department of Pediatrics, Stanford University, Stanford, CA, USA.
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA, USA.
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
41
|
Moscatelli I, Almarza E, Schambach A, Ricks D, Schulz A, Herzog CD, Henriksen K, Askmyr M, Schwartz JD, Richter J. Gene therapy for infantile malignant osteopetrosis: review of pre-clinical research and proof-of-concept for phenotypic reversal. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 20:389-397. [PMID: 33575431 PMCID: PMC7848732 DOI: 10.1016/j.omtm.2020.12.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Infantile malignant osteopetrosis is a devastating disorder of early childhood that is frequently fatal and for which there are only limited therapeutic options. Gene therapy utilizing autologous hematopoietic stem and progenitor cells represents a potentially advantageous therapeutic alternative for this multisystemic disease. Gene therapy can be performed relatively rapidly following diagnosis, will not result in graft versus host disease, and may also have potential for reduced incidences of other transplant-related complications. In this review, we have summarized the past sixteen years of research aimed at developing a gene therapy for infantile malignant osteopetrosis; these efforts have culminated in the first clinical trial employing lentiviral-mediated delivery of TCIRG1 in autologous hematopoietic stem and progenitor cells.
Collapse
Affiliation(s)
- Ilana Moscatelli
- Department of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund University, Lund, Sweden
| | | | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - David Ricks
- Rocket Pharmaceuticals, Inc., New York, NY, USA
| | - Ansgar Schulz
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Germany
| | | | | | - Maria Askmyr
- Department of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund University, Lund, Sweden
| | | | - Johan Richter
- Department of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund University, Lund, Sweden
| |
Collapse
|
42
|
Ouyang W, Dong G, Zhao W, Li J, Zhou Z, Yang G, Liu R, Li Y, Zhang Q, Du X, Sun H, Gu Y, Lai Y, Liu S, Liu C. Restoration of β-Globin Expression with Optimally Designed Lentiviral Vector for β-Thalassemia Treatment in Chinese Patients. Hum Gene Ther 2021; 32:481-494. [PMID: 33256481 DOI: 10.1089/hum.2020.204] [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: 01/02/2023] Open
Abstract
β-Thalassemia is one of the most prevalent genetic diseases worldwide. The current treatment for β-thalassemia is allogeneic hematopoietic stem cell transplantation, which is limited due to lack of matched donors. Gene therapy has been developed as an alternative therapeutic option for transfusion-dependent β-thalassemia (TDT). However, successful gene therapy for β-thalassemia patients in China has not been reported. Here, we present the results of preclinical studies of an optimally designed lentiviral vector (LV) named LentiHBBT87Q in hematopoietic stem and progenitor cells (HSPCs) derived from Chinese TDT patients. LentiHBBT87Q was selected from a series of LVs with optimized backbone and de novo cloning strategy. It contains an exogenous T87Q β-globin gene (HBBT87Q) driven by a specific reconstituted locus control region, and efficiently expresses HBB mRNA and HBB protein in erythroblasts derived from cord blood HSPCs. To facilitate clinical transformation, we manufactured clinical-grade LentiHBBT87Q (cLentiHBBT87Q) and optimized its transduction procedure. Importantly, transduction of cLentiHBBT87Q restored expression of HBB monomer and adult hemoglobin tetramer to relatively normal level in erythroblasts from bone marrow HSPCs of Chinese TDT patients that carry the most common mutation types and cover various genotypes, including β0/β0. Furthermore, viral integration sites (VISs) of cLentiHBBT87Q were similar to other LVs safely used in previous clinical trials, and gene-ontology (term) analysis of VIS targeted genes suggests that no tumor-associated pathways were enriched in treated samples. Taken together, we have engineered the cLentiHBBT87Q that can restore β-globin expression in the HSPCs-derived erythroblasts of Chinese TDT patients with minimal risk of tumorigenesis, providing a favorable starting point for future clinical application.
Collapse
Affiliation(s)
- Wenjie Ouyang
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China.,Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen, China
| | - Guoyi Dong
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China.,Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen, China.,BGI Education Center, University of Chinese Academy Sciences, Shenzhen, China
| | - Weihua Zhao
- Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Jing Li
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China.,Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen, China.,BGI Education Center, University of Chinese Academy Sciences, Shenzhen, China
| | - Ziheng Zhou
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Gaohui Yang
- Department of Hematology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Rongrong Liu
- Department of Hematology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yue Li
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Qiaoxia Zhang
- Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Xin Du
- Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Haixi Sun
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Ying Gu
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China.,Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen, China
| | - Yongrong Lai
- Department of Hematology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Sixi Liu
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, China
| | - Chao Liu
- BGI-Shenzhen, Shenzhen, China.,China National GeneBank, BGI-Shenzhen, Shenzhen, China.,Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen, China
| |
Collapse
|
43
|
Zittersteijn HA, Harteveld CL, Klaver-Flores S, Lankester AC, Hoeben RC, Staal FJT, Gonçalves MAFV. A Small Key for a Heavy Door: Genetic Therapies for the Treatment of Hemoglobinopathies. Front Genome Ed 2021; 2:617780. [PMID: 34713239 PMCID: PMC8525365 DOI: 10.3389/fgeed.2020.617780] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/14/2020] [Indexed: 12/26/2022] Open
Abstract
Throughout the past decades, the search for a treatment for severe hemoglobinopathies has gained increased interest within the scientific community. The discovery that ɤ-globin expression from intact HBG alleles complements defective HBB alleles underlying β-thalassemia and sickle cell disease, has provided a promising opening for research directed at relieving ɤ-globin repression mechanisms and, thereby, improve clinical outcomes for patients. Various gene editing strategies aim to reverse the fetal-to-adult hemoglobin switch to up-regulate ɤ-globin expression through disabling either HBG repressor genes or repressor binding sites in the HBG promoter regions. In addition to these HBB mutation-independent strategies involving fetal hemoglobin (HbF) synthesis de-repression, the expanding genome editing toolkit is providing increased accuracy to HBB mutation-specific strategies encompassing adult hemoglobin (HbA) restoration for a personalized treatment of hemoglobinopathies. Moreover, besides genome editing, more conventional gene addition strategies continue under investigation to restore HbA expression. Together, this research makes hemoglobinopathies a fertile ground for testing various innovative genetic therapies with high translational potential. Indeed, the progressive understanding of the molecular clockwork underlying the hemoglobin switch together with the ongoing optimization of genome editing tools heightens the prospect for the development of effective and safe treatments for hemoglobinopathies. In this context, clinical genetics plays an equally crucial role by shedding light on the complexity of the disease and the role of ameliorating genetic modifiers. Here, we cover the most recent insights on the molecular mechanisms underlying hemoglobin biology and hemoglobinopathies while providing an overview of state-of-the-art gene editing platforms. Additionally, current genetic therapies under development, are equally discussed.
Collapse
Affiliation(s)
- Hidde A. Zittersteijn
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Cornelis L. Harteveld
- Department of Human and Clinical Genetics, The Hemoglobinopathies Laboratory, Leiden University Medical Center, Leiden, Netherlands
| | | | - Arjan C. Lankester
- Department of Pediatrics, Stem Cell Transplantation Program, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands
| | - Rob C. Hoeben
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Frank J. T. Staal
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | |
Collapse
|
44
|
Breda L, Ghiaccio V, Tanaka N, Jarocha D, Ikawa Y, Abdulmalik O, Dong A, Casu C, Raabe TD, Shan X, Danet-Desnoyers GA, Doto AM, Everett J, Bushman FD, Radaelli E, Assenmacher CA, Tarrant JC, Hoepp N, Kurita R, Nakamura Y, Guzikowski V, Smith-Whitley K, Kwiatkowski JL, Rivella S. Lentiviral vector ALS20 yields high hemoglobin levels with low genomic integrations for treatment of beta-globinopathies. Mol Ther 2021; 29:1625-1638. [PMID: 33515514 DOI: 10.1016/j.ymthe.2020.12.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/11/2020] [Accepted: 12/30/2020] [Indexed: 10/22/2022] Open
Abstract
Ongoing clinical trials for treatment of beta-globinopathies by gene therapy involve the transfer of the beta-globin gene, which requires integration of three to four copies per genome in most target cells. This high proviral load may increase genome toxicity, potentially limiting the safety of this therapy and relegating its use to total body myeloablation. We hypothesized that introducing an additional hypersensitive site from the locus control region, the complete sequence of the second intron of the beta-globin gene, and the ankyrin insulator may enhance beta-globin expression. We identified a construct, ALS20, that synthesized significantly higher adult hemoglobin levels than those of other constructs currently used in clinical trials. These findings were confirmed in erythroblastic cell lines and in primary cells isolated from sickle cell disease patients. Bone marrow transplantation studies in beta-thalassemia mice revealed that ALS20 was curative at less than one copy per genome. Injection of human CD34+ cells transduced with ALS20 led to safe, long-term, and high polyclonal engraftment in xenograft experiments. Successful treatment of beta-globinopathies with ALS20 could potentially be achieved at less than two copies per genome, minimizing the risk of cytotoxic events and lowering the intensity of myeloablation.
Collapse
Affiliation(s)
- Laura Breda
- Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Valentina Ghiaccio
- Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Naoto Tanaka
- Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Danuta Jarocha
- Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Yasuhiro Ikawa
- Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Osheiza Abdulmalik
- Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Alisa Dong
- Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Carla Casu
- Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Tobias D Raabe
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiaochuan Shan
- Stem and Xenograft Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gwenn A Danet-Desnoyers
- Stem and Xenograft Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aoife M Doto
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Everett
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles A Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James C Tarrant
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Natalie Hoepp
- Clinical Pathology Laboratory, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryo Kurita
- RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | | | - Virginia Guzikowski
- Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Kim Smith-Whitley
- Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Janet L Kwiatkowski
- Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stefano Rivella
- Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Cell and Molecular Biology Affinity Group (CAMB), University of Pennsylvania, Philadelphia, PA, USA; Raymond G. Perelman Center for Cellular and Molecular Therapeutics, CHOP, Philadelphia, PA, USA; Penn Center for Musculoskeletal Disorders, CHOP, Philadelphia, PA, USA.
| |
Collapse
|
45
|
Kim HS, Hwang GH, Lee HK, Bae T, Park SH, Kim YJ, Lee S, Park JH, Bae S, Hur JK. CReVIS-Seq: A highly accurate and multiplexable method for genome-wide mapping of lentiviral integration sites. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 20:792-800. [PMID: 33768124 PMCID: PMC7961857 DOI: 10.1016/j.omtm.2020.10.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/17/2020] [Indexed: 12/16/2022]
Abstract
Lentiviruses have been widely used as a means of transferring exogenous DNAs into human cells to treat various genetic diseases. Lentiviral vectors are fundamentally integrated into the host genome, but their integration sites are generally unpredictable, which may increase the uncertainty for their use in therapeutics. To determine the viral integration sites in the host genome, several PCR-based methods have been developed. However, the sensitivities of the PCR-based methods are highly dependent on the primer sequences, and optimized primer design is required for individual target sites. In order to address this issue, we developed an alternative method for genome-wide mapping of viral insertion sites, named CReVIS-seq (CRISPR-enhanced Viral Integration Site Sequencing). The method is based on the sequential steps: fragmentation of genomic DNAs, in vitro circularization, cleavage of target sequence in a CRISPR guide RNA-specific manner, high-throughput sequencing of the linearized DNA fragments in an unbiased manner, and identification of viral insertion sites via sequence analysis. By design, CReVIS-seq is not affected by biases that could be introduced during the target enrichment step via PCR amplification using site specific primers. Furthermore, we found that multiplexed CReVIS-seq, using collections of different single-guide RNAs (sgRNAs), enables simultaneous identification of multiple target sites and structural variations (i.e., circularized viral genome), in both single cell clones and heterogeneous cell populations.
Collapse
Affiliation(s)
- Heon Seok Kim
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Gue-Ho Hwang
- Department of Chemistry, Hanyang University, Seoul 04763, South Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 04763, South Korea
| | - Hyomin K Lee
- Department of Medicine, Graduate School, Hanyang University, Seoul 04763, South Korea
| | - Taegeun Bae
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, South Korea
| | - Seong-Ho Park
- Department of Medicine, Graduate School, Hanyang University, Seoul 04763, South Korea
| | - Yong Jun Kim
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul 02447, South Korea.,Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, South Korea
| | - Sun Lee
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul 02447, South Korea.,Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, South Korea
| | - Jae-Hoon Park
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul 02447, South Korea.,Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, South Korea
| | - Sangsu Bae
- Department of Chemistry, Hanyang University, Seoul 04763, South Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul 04763, South Korea
| | - Junho K Hur
- Department of Genetics, College of Medicine, Hanyang University, Seoul 04763, South Korea.,Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, South Korea
| |
Collapse
|
46
|
Papizan JB, Porter SN, Sharma A, Pruett-Miller SM. Therapeutic gene editing strategies using CRISPR-Cas9 for the β-hemoglobinopathies. J Biomed Res 2021; 35:115-134. [PMID: 33349624 PMCID: PMC8038529 DOI: 10.7555/jbr.34.20200096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
With advancements in gene editing technologies, our ability to make precise and efficient modifications to the genome is increasing at a remarkable rate, paving the way for scientists and clinicians to uniquely treat a multitude of previously irremediable diseases. CRISPR-Cas9, short for clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9, is a gene editing platform with the ability to alter the nucleotide sequence of the genome in living cells. This technology is increasing the number and pace at which new gene editing treatments for genetic disorders are moving toward the clinic. The β-hemoglobinopathies are a group of monogenic diseases, which despite their high prevalence and chronic debilitating nature, continue to have few therapeutic options available. In this review, we will discuss our existing comprehension of the genetics and current state of treatment for β-hemoglobinopathies, consider potential genome editing therapeutic strategies, and provide an overview of the current state of clinical trials using CRISPR-Cas9 gene editing.
Collapse
Affiliation(s)
- James B Papizan
- Department of Cellular and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.,Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shaina N Porter
- Department of Cellular and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.,Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cellular and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.,Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
47
|
Gill KP, Denham M. Optimized Transgene Delivery Using Third-Generation Lentiviruses. CURRENT PROTOCOLS IN MOLECULAR BIOLOGY 2020; 133:e125. [PMID: 32986282 PMCID: PMC7583475 DOI: 10.1002/cpmb.125] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The lentivirus system enables efficient genetic modification of both dividing and non-dividing cells and therefore is a useful tool for elucidating developmental processes and disease pathogenesis. The development of third-generation lentiviruses has resulted in improved biosafety, low immunogenicity, and substantial packaging capabilities. However, because third-generation lentiviruses require successful co-transfection with four plasmids, this typically means that lower titers are attained. This is problematic, as it is often desirable to produce purified lentiviruses with high titers (>1 × 108 TU/ml), especially for in vivo applications. The manufacturing process for lentiviruses involves several critical experimental factors that can influence titer, purity, and transduction efficiency. Here, we describe a straightforward, stepwise protocol for the reproducible manufacture of high-titer third-generation lentiviruses (1 × 108 to 1 × 109 TU/ml). This optimized protocol enhances transgene expression by use of Lipofectamine transfection and optimized serum replacement medium, a single ultracentrifugation step, use of a sucrose cushion, and addition of a histone deacetylation inhibitor. Furthermore, we provide alternate methods for titration analyses, including functional and genomic integration analyses, using common laboratory techniques such as FACS as well as genomic DNA extraction and qPCR. These optimized methods will be beneficial for investigating developmental processes and disease pathogenesis in vitro and in vivo. © 2020 The Authors. Basic Protocol 1: Lentivirus production Support Protocol: Lentivirus concentration Basic Protocol 2: Lentivirus titration Alternate Protocol 1: Determination of viral titration by FACS analysis Alternate Protocol 2: Determination of viral titration by genome integration analysis.
Collapse
Affiliation(s)
- Katherine P. Gill
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular MedicineAarhus UniversityAarhusDenmark
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | - Mark Denham
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular MedicineAarhus UniversityAarhusDenmark
- Department of BiomedicineAarhus UniversityAarhusDenmark
| |
Collapse
|
48
|
Xie M, Viviani M, Fussenegger M. Engineering precision therapies: lessons and motivations from the clinic. Synth Biol (Oxf) 2020; 6:ysaa024. [PMID: 33817342 PMCID: PMC7998714 DOI: 10.1093/synbio/ysaa024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/18/2022] Open
Abstract
In the past decade, gene- and cell-based therapies have been at the forefront of the biomedical revolution. Synthetic biology, the engineering discipline of building sophisticated 'genetic software' to enable precise regulation of gene activities in living cells, has been a decisive success factor of these new therapies. Here, we discuss the core technologies and treatment strategies that have already gained approval for therapeutic applications in humans. We also review promising preclinical work that could either enhance the efficacy of existing treatment strategies or pave the way for new precision medicines to treat currently intractable human conditions.
Collapse
Affiliation(s)
- Mingqi Xie
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zheijang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zheijang, China
| | - Mirta Viviani
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zheijang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zheijang, China
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Faculty of Science, University of Basel, Basel, Switzerland
| |
Collapse
|
49
|
Iqubal A, Iqubal MK, Khan A, Ali J, Baboota S, Haque SE. Gene Therapy, A Novel Therapeutic Tool for Neurological Disorders: Current Progress, Challenges and Future Prospective. Curr Gene Ther 2020; 20:184-194. [DOI: 10.2174/1566523220999200716111502] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/02/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023]
Abstract
:
Neurological disorders are one of the major threat for health care system as they put enormous
socioeconomic burden. All aged populations are susceptible to one or other neurological problems
with symptoms of neuroinflammation, neurodegeneration and cognitive dysfunction. At present,
available pharmacotherapeutics are insufficient to treat these diseased conditions and in most cases,
they provide only palliative effect. It was also found that the molecular etiology of neurological disorders
is directly linked with the alteration in genetic makeup, which can be inherited or triggered by the
injury, environmental toxins and by some existing disease. Therefore, to take care of this situation,
gene therapy has emerged as an advanced modality that claims to permanently cure the disease by deletion,
silencing or edition of faulty genes and by insertion of healthier genes. In this modality, vectors
(viral and non-viral) are used to deliver targeted gene into a specific region of the brain via various
routes. At present, gene therapy has shown positive outcomes in complex neurological disorders, such
as Parkinson's disease, Alzheimer's disease, Huntington disease, Multiple sclerosis, Amyotrophic lateral
sclerosis and in lysosomal storage disease. However, there are some limitations such as immunogenic
reactions non-specificity of viral vectors and a lack of effective biomarkers to understand the efficacy
of therapy. Considerable progress has been made to improve vector design, gene selection and
targeted delivery. This review article deals with the current status of gene therapy in neurological disorders
along with its clinical relevance, challenges and future prospective.
Collapse
Affiliation(s)
- Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Mohammad Kashif Iqubal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Aamir Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| | - Syed Ehtaishamul Haque
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi- 110062, India
| |
Collapse
|
50
|
Osunkwo I, Manwani D, Kanter J. Current and novel therapies for the prevention of vaso-occlusive crisis in sickle cell disease. Ther Adv Hematol 2020; 11:2040620720955000. [PMID: 33062233 PMCID: PMC7534097 DOI: 10.1177/2040620720955000] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 08/10/2020] [Indexed: 12/28/2022] Open
Abstract
Individuals with sickle cell disease (SCD) are living further into adulthood in high-resource countries. However, despite increased quantity of life, recurrent, acute painful episodes cause significant morbidity for affected individuals. These SCD-related painful episodes, also referred to as vaso-occlusive crises (VOCs), have multifactorial causes, and they often occur as a result of multicellular aggregation and vascular adherence of red blood cells, neutrophils, and platelets, leading to recurrent and unpredictable occlusion of the microcirculation. In addition to severe pain, long-term complications of vaso-occlusion may include damage to muscle and/or bone, in addition to vital organs such as the liver, spleen, kidneys, and brain. Severe pain associated with VOCs also has a substantial detrimental impact on quality of life for individuals with SCD, and is associated with increased health care utilization, financial hardship, and impairments in education and vocation attainment. Previous treatments have targeted primarily SCD symptom management, or were broad nontargeted therapies, and include oral or parenteral hydration, analgesics (including opioids), nonsteroidal anti-inflammatory agents, and various other types of nonpharmacologic pain management strategies to treat the pain associated with VOC. With increased understanding of the pathophysiology of VOCs, there are several new potential therapies that specifically target the pathologic process of vaso-occlusion. These new therapies may reduce cell adhesion and inflammation, leading to decreased incidence of VOCs and prevention of end-organ damage. In this review, we consider the benefits and limitations of current treatments to reduce the occurrence of VOCs in individuals with SCD and the potential impact of emerging treatments on future disease management.
Collapse
Affiliation(s)
- Ifeyinwa Osunkwo
- Non-Malignant Hematology Section, The Levine Cancer Institute and Atrium Health, Charlotte, NC, USA
| | - Deepa Manwani
- Division of Pediatric Hematology and Oncology, The Children’s Hospital at Montefiore, Albert Einstein College of Medicine, New York, NY, USA
| | - Julie Kanter
- Division of Hematology and Oncology, University of Alabama Birmingham, 1720 2nd Avenue S, NP 2510, Birmingham, AL 35294, USA
| |
Collapse
|