1
|
Zhang Z, Wang L, Liu Z, Yang C, Chen M, Han B. Long-Term Experience with Luspatercept in Relapsed/Refractory Myelodysplastic Neoplasms: A Chinese Real-World Study. Adv Ther 2025; 42:1907-1918. [PMID: 40025395 DOI: 10.1007/s12325-025-03141-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 02/10/2025] [Indexed: 03/04/2025]
Abstract
INTRODUCTION Luspatercept has been shown to be efficacious for patients with relapsed or refractory lower-risk myelodysplastic neoplasms (LR-MDS) in both clinical trials and real-world studies. Nevertheless, long-term follow-up data in real-world settings remain scarce, particularly in Asia. METHODS Data from patients diagnosed with relapsed or refractory LR-MDS who had been treated with luspatercept at our center between June 2022 and May 2024 were retrospectively collected. RESULTS In total, 60 patients were included in this study (63.4% males). The median duration of luspatercept exposure was 9 (range 3-25) months, and the median follow-up time was 15 (range 3-26) months. The hematologic improvement-erythroid (HI-E) rate was 46.7%, 51.0%, 48.6%, and 43.3% at the 3rd, 6th, and 12th months, and at the end of follow-up, respectively. Among patients who were transfusion-dependent prior to luspatercept, 48.3%, 38.7%, and 25.8% achieved transfusion independence for 8, 12, and 16 weeks or longer at the 6th month. Over time, patients treated with luspatercept had a significant increase in hemoglobin level compared with that of the baseline from the 1st month to the end of follow-up (all P < 0.05). At the end of follow-up, 5 of 32 (15.6%) patients who had response had experienced a relapse, 1 patient (1.7%) had progressed to higher-risk myelodysplastic neoplasms (MDS), and 2 patients (3.3%) had progressed to acute myeloid leukemia. Three patients (5.0%) died of pulmonary infection. Serum erythropoietin (EPO) ≤ 500 IU/l at baseline was the only independent predictive factor for HI-E at the 3rd month (P = 0.007). CONCLUSION Luspatercept is proved efficacious and well tolerated in relapsed/refractory LR-MDS and appears to be beneficial in reducing disease progression and prolonging survival.
Collapse
Affiliation(s)
- Zhuxin Zhang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100730, China
| | - Leyu Wang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100730, China
| | - Ziwei Liu
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100730, China
| | - Chen Yang
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100730, China
| | - Miao Chen
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100730, China
| | - Bing Han
- Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science, No.1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
2
|
Frangoul H, Stults A, Bruce K, Domm J, Carroll C, Aide S, Duckworth M, Evans M, McManus M. Best Practices in Gene Therapy for Sickle Cell Disease and Transfusion-dependent β-Thalassemia. Transplant Cell Ther 2025:S2666-6367(25)01061-9. [PMID: 40058646 DOI: 10.1016/j.jtct.2025.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/26/2025]
Abstract
Sickle cell disease (SCD) and transfusion-dependent β-thalassemia (TDT) are inherited blood disorders caused by pathogenic variants of the β-globin gene. Historically, allogeneic hematopoietic stem cell transplantation (HSCT) from human leukocyte antigen (HLA)-matched donors has been the only curative option. However, as most patients with SCD or TDT lack HLA-matched donors, autologous or patient-derived HSCT can provide an alternative, transformative option. Gene therapy-based autologous HSCT for the treatment of SCD and TDT entails a complex patient journey and requires the careful implementation of numerous policies and procedures. As gene therapies for these diseases are now commercially available, there is great value in institutions with developed and implemented approaches sharing their best practices. Here, we describe standardized approaches and best practices for the optimized implementation of gene therapies based on our experience in administering this novel class of medicines.
Collapse
Affiliation(s)
- Haydar Frangoul
- Sarah Cannon Research Institute at TriStar Centennial Children's Hospital, Nashville, Tennessee.
| | - Amanda Stults
- Sarah Cannon Research Institute at TriStar Centennial Children's Hospital, Nashville, Tennessee
| | - Katie Bruce
- Sarah Cannon Research Institute at TriStar Centennial Children's Hospital, Nashville, Tennessee
| | - Jennifer Domm
- Sarah Cannon Research Institute at TriStar Centennial Children's Hospital, Nashville, Tennessee
| | - Clinton Carroll
- Sarah Cannon Research Institute at TriStar Centennial Children's Hospital, Nashville, Tennessee
| | - Shelby Aide
- Sarah Cannon Research Institute at TriStar Centennial Children's Hospital, Nashville, Tennessee
| | - Morgan Duckworth
- Sarah Cannon Research Institute at TriStar Centennial Children's Hospital, Nashville, Tennessee
| | - Misty Evans
- Sarah Cannon Research Institute at TriStar Centennial Children's Hospital, Nashville, Tennessee
| | - Meghann McManus
- Sarah Cannon Research Institute at TriStar Centennial Children's Hospital, Nashville, Tennessee
| |
Collapse
|
3
|
Cappellini MD, Viprakasit V, Georgiev P, Coates TD, Origa R, Khelif A, Liew HK, Tantiworawit A, Chew LP, Khalil A, Ho PJ, Kuo KHM, Holot N, Perin M, Giuseppi AC, Kuo WL, Lai Y, Medlin LF, Bueno LM, Kattamis A, Taher AT. Long-term efficacy and safety of luspatercept for the treatment of anaemia in patients with transfusion-dependent β-thalassaemia (BELIEVE): final results from a phase 3 randomised trial. Lancet Haematol 2025; 12:e180-e189. [PMID: 39947215 DOI: 10.1016/s2352-3026(24)00376-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 03/08/2025]
Abstract
BACKGROUND Treatments to reduce red blood cell (RBC) transfusion burden among patients with transfusion-dependent β-thalassaemia remain limited. Here, we report long-term follow-up data from the phase 3 BELIEVE trial of luspatercept for transfusion-dependent β-thalassaemia. METHODS BELIEVE was a phase 3, randomised, double-blind, placebo-controlled study performed at 65 sites in 15 countries. The trial included adults with transfusion-dependent β-thalassaemia or haemoglobin E/β-thalassaemia and Eastern Cooperative Oncology Group score of 0-1. Patients were randomly assigned (2:1) using integrated response technology stratified by region to luspatercept (1·0-1·25 mg/kg) or placebo administered subcutaneously once every 21 days. After study unblinding, patients could receive luspatercept in the open-label extension phase (crossover allowed). The primary endpoint results (proportion of patients with reduction in transfusion burden of ≥33% and ≥2 RBC units during weeks 13-24) are described elsewhere; herein we present an update to the primary endpoint analysis consequent to late-reported transfusion events. We also report long-term efficacy (intention-to-treat population) and safety data (safety population) for patients followed up for approximately 3 years. This trial is registered on ClinicalTrials.gov (NCT02604433) and is completed. FINDINGS Between May 2, 2016, and May 16, 2017, 336 patients were randomly assigned to luspatercept (n=224) or placebo (n=112). The median age of patients was 30 years (IQR 23-40); 195 (58%) were female and 141 (42%) male. As of Jan 5, 2021, the median duration of treatment in the luspatercept group was 153·6 weeks (IQR 81·0-171·0) and median study follow-up was 163·1 weeks (140·5-176·2). Due to the difference in treatment duration between the luspatercept and placebo groups, no comparative analyses between the two groups were performed after week 96. Patients in the luspatercept group showed a sustained reduction in RBC transfusion burden from baseline through week 192, with mean decreases of 6·2 RBC units (SD 5·7) during weeks 97-144 and 6·4 RBC units (4·3) during weeks 145-192. In the luspatercept group, a 33% or greater reduction in transfusion burden from baseline was observed in 173 (77%) patients over any 12-week interval and in 116 (52%) patients over any 24-week interval. The median total duration of 33% or greater transfusion burden reduction response during any period of at least 12 weeks was 586·0 days (IQR 264·0-1010·0). The most common grade 3 or worse treatment-emergent adverse events (TEAEs) among all patients who received luspatercept (n=315, including 92 patients who crossed over after study unblinding) were anaemia (nine [3%]), increased liver iron concentration (seven [2%]), and bone pain (seven [2%]); serious TEAEs occurred in 71 (23%) patients. No treatment-related deaths occurred in any group during the study. INTERPRETATION These long-term results affirm luspatercept's efficacy in addressing key unmet needs of patients with transfusion-dependent β-thalassaemia with a manageable safety profile. FUNDING Celgene and Acceleron Pharma.
Collapse
Affiliation(s)
| | | | - Pencho Georgiev
- St. George University Hospital for Active Treatment and Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Thomas D Coates
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles and USC Keck School of Medicine, Los Angeles, CA, USA
| | - Raffaella Origa
- University of Cagliari, Ospedale Pediatrico Microcitemico 'A. Cao', Cagliari, Italy
| | - Abderrahim Khelif
- Department of Clinical Hematology, Farhat Hached Hospital and Ibn El Jazzar Faculty of Medicine, Sousse University, Sousse, Tunisia
| | - Hong-Keng Liew
- Department of Medicine, Hospital Sultanah Bahiyah, Alor Setar, Kedah, Malaysia
| | - Adisak Tantiworawit
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Lee-Ping Chew
- Department of Medicine, Haematology Unit, Sarawak General Hospital, Kuching, Sarawak, Malaysia
| | - Abdalla Khalil
- Department of Pediatric Hematology Oncology, Rambam Medical Center, Haifa, Israel
| | - P Joy Ho
- Royal Prince Alfred Hospital and the University of Sydney, Sydney, NSW, Australia
| | - Kevin H M Kuo
- Division of Medical Oncology and Hematology, Department of Medicine, University Health Network and Division of Hematology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Martina Perin
- Celgene International Sàrl, a Bristol-Myers Squibb Company, Boudry, Switzerland
| | | | | | - Yinzhi Lai
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Loyse Felber Medlin
- Celgene International Sàrl, a Bristol-Myers Squibb Company, Boudry, Switzerland
| | - Luciana Moro Bueno
- Celgene International Sàrl, a Bristol-Myers Squibb Company, Boudry, Switzerland
| | - Antonis Kattamis
- Thalassaemia Unit, First Department of Paediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
4
|
Fu C, Yang X. Cardiac injury caused by iron overload in thalassemia. Front Pediatr 2025; 13:1514722. [PMID: 39931654 PMCID: PMC11808023 DOI: 10.3389/fped.2025.1514722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025] Open
Abstract
Cardiac iron overload affects approximately 25% of patients with β-thalassemia major, which is associated with increased morbidity and mortality. Two mechanisms are responsible for iron overload in β-thalassemia: increased iron absorption due to ineffective erythropoiesis and blood transfusions. This review examines the mechanisms of myocardial injury caused by cardiac iron overload and role of various clinical examination techniques in assessing cardiac iron burden and functional impairment. Early identification and intervention for cardiac injury and iron overload in β-thalassemia have the potential to prevent and reverse or delay its progression in the early stages, playing a crucial role in its prognosis.
Collapse
Affiliation(s)
- Chunxi Fu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Xue Yang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Musallam KM, Taher AT. Luspatercept: a treatment for ineffective erythropoiesis in thalassemia. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:419-425. [PMID: 39644029 PMCID: PMC11665503 DOI: 10.1182/hematology.2024000567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
Patients with β-thalassemia continue to have several unmet needs. In non-transfusion-dependent patients, untreated ineffective erythropoiesis and anemia have been associated with a variety of clinical sequelae, with no treatment currently available beyond supportive transfusions. In transfusion-dependent forms, lifelong transfusion and iron chelation therapy are associated with considerable clinical, psychological, and economic burden on the patient and health care system. Luspatercept is a novel disease-modifying agent targeting ineffective erythropoiesis that became recently available for patients with β-thalassemia. Data from randomized clinical trials confirmed its efficacy and safety in reducing transfusion burden in transfusion-dependent patients and increasing total hemoglobin level in non-transfusion-dependent patients. Secondary clinical benefits in patient-reported outcomes and iron overload were also observed on long-term therapy, and further data from real-world evidence studies are awaited.
Collapse
Affiliation(s)
- Khaled M Musallam
- Center for Research on Rare Blood Disorders, Burjeel Medical City, Abu Dhabi, United Arab Emirates
- Division of Hematology/Oncology, Department of Pediatrics, Weill Cornell Medicine, New York, NY
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
6
|
Gattermann N. Iron overload in acquired sideroblastic anemias and MDS: pathophysiology and role of chelation and luspatercept. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:443-449. [PMID: 39644054 DOI: 10.1182/hematology.2024000569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
Besides transfusion therapy, ineffective erythropoiesis contributes to systemic iron overload in myelodysplastic syndromes with ring sideroblasts (MDS-RS) via erythroferrone-induced suppression of hepcidin synthesis in the liver, leading to increased intestinal iron absorption. The underlying pathophysiology of MDS-RS, characterized by disturbed heme synthesis and mitochondrial iron accumulation, is less well understood. Several lines of evidence indicate that the mitochondrial transporter ABCB7 is critically involved. ABCB7 is misspliced and underexpressed in MDS-RS, due to somatic mutations in the splicing factor SF3B1. The pathogenetic significance of ABCB7 seems related to its role in stabilizing ferrochelatase, the enzyme incorporating iron into protoporphyrin IX to make heme. Although iron-related oxidative stress is toxic, many patients with MDS do not live long enough to develop clinical complications of iron overload. Furthermore, it is difficult to determine the extent to which iron overload contributes to morbidity and mortality in older patients with MDS, because iron-related complications overlap with age-related medical problems. Nevertheless, high-quality registry studies showed that transfusion dependency is associated with the presence of toxic iron species and inferior survival and confirmed a significant survival benefit of iron chelation therapy. The most widely used iron chelator in patients with MDS is deferasirox, owing to its effectiveness and convenient oral administration. Luspatercept, which can reduce SMAD2/SMAD3-dependent signaling implicated in suppression of erythropoiesis, may obviate the need for red blood cell transfusion in MDS-RS for more than a year, thereby diminishing further iron loading. However, luspatercept cannot be expected to substantially reduce the existing iron overload.
Collapse
|
7
|
Rabie MAF, El Benhawy SA, Masoud IM, Arab ARR, Saleh SAM. Impact of met-haemoglobin and oxidative stress on endothelial function in patients with transfusion dependent β-thalassemia. Sci Rep 2024; 14:25328. [PMID: 39455629 PMCID: PMC11512062 DOI: 10.1038/s41598-024-74930-3] [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: 08/05/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Transfusion dependent β-thalassemia is a genetic blood disorder characterized by chronic anaemia. Blood transfusion is lifesaving but comes at a cost. Iron overload emerges as a prime culprit as a free radicals damage endothelial cells. Chronic anaemia further disrupts oxygen delivery, exacerbating the oxidative stress. Increased levels of met-haemoglobin and malondialdehyde compromise endothelial function. This research sheds light on the impact of met-haemoglobin and oxidative stress on endothelial function in 50 patients with transfusion dependent β-thalassemia major compared to 50 healthy individuals as control. Blood samples were collected & subjected to CBC, biochemical analysis including creatinine, ferritin, CRP, LDH, and HCV antibodies. Oxidative stress was assessed using met-haemoglobin & malondialdehyde. Endothelial dysfunction was evaluated by endothelial activation and stress index (EASIX). EASIX, met-haemoglobin and malondialdehyde were significantly increased in patients (1.44 ± 0.75, 2.07 ± 0.2, 4.8 ± 0.63; respectively) compared to the control (0.52 ± 0.24,0.88 ± 0.34,0.8 ± 0.34; respectively). Significant strong positive correlation was found between EASIX and met-haemoglobin, malondialdehyde, serum ferritin and CRP (P = 0.00, r = 0.904, P = 0.00, r = 0.948, P = 0.00, r = 0.772, P = 0.00, r = 0.971; respectively. Met-haemoglobin as well as EASIX should be routinely estimated to assess endothelial function especially before the decision of splenectomy. Antioxidant drugs should be supplemented.
Collapse
Affiliation(s)
- Maha Abubakr Feissal Rabie
- Department of Basic Science, Pharos University in Alexandria, Canal El Mahmoudia Street, Beside Green Plaza Complex, Alexandria, Egypt.
| | - Sanaa A El Benhawy
- Radiation Sciences Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Inas M Masoud
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Amal R R Arab
- Department of Applied Medical Chemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Sally A M Saleh
- Department of Clinical Haematology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
8
|
Guerra A, Hamilton N, Rivera A, Demsko P, Guo S, Rivella S. Combination of a TGF-β ligand trap (RAP-GRL) and TMPRSS6-ASO is superior for correcting β-thalassemia. Am J Hematol 2024; 99:1300-1312. [PMID: 38659383 PMCID: PMC11166515 DOI: 10.1002/ajh.27332] [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: 12/11/2023] [Revised: 03/11/2024] [Accepted: 03/22/2024] [Indexed: 04/26/2024]
Abstract
A recently approved drug that induces erythroid cell maturation (luspatercept) has been shown to improve anemia and reduce the need for blood transfusion in non-transfusion-dependent as well as transfusion-dependent β-thalassemia (BT) patients. Although these results were predominantly positive, not all the patients showed the expected increase in hemoglobin (Hb) levels or transfusion burden reduction. Additional studies indicated that administration of luspatercept in transfusion-dependent BT was associated with increased erythropoietic markers, decreased hepcidin levels, and increased liver iron content. Altogether, these studies suggest that luspatercept may necessitate additional drugs for improved erythroid and iron management. As luspatercept does not appear to directly affect iron metabolism, we hypothesized that TMPRSS6-ASO could improve iron parameters and iron overload when co-administered with luspatercept. We used an agent analogous to murine luspatercept (RAP-GRL) and another novel therapeutic, IONIS TMPRSS6-LRx (TMPRSS6-ASO), a hepcidin inducer, to treat non-transfusion-dependent BT-intermedia mice. Our study shows that RAP-GRL alone improved red blood cell (RBC) production, with no or limited effect on splenomegaly and iron parameters. In contrast, TMPRSS6-ASO improved RBC measurements, ameliorated splenomegaly, and improved iron overload most effectively. Our results provide pre-clinical support for combining TMPRSS6-ASO and luspatercept in treating BT, as these drugs together show potential for simultaneously improving both erythroid and iron parameters in BT patients.
Collapse
Affiliation(s)
- Amaliris Guerra
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Nolan Hamilton
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Ariel Rivera
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Perry Demsko
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Cell and Molecular Biology affinity group (CAMB), University of Pennsylvania, Philadelphia, PA, USA
| | - Shuling Guo
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, USA
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
- University of Pennsylvania, Perelman School of Medicine, 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
- Penn Center for Musculoskeletal Disorders, CHOP, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
9
|
de Haan LR, van Golen RF, Heger M. Molecular Pathways Governing the Termination of Liver Regeneration. Pharmacol Rev 2024; 76:500-558. [PMID: 38697856 DOI: 10.1124/pharmrev.123.000955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/24/2024] [Accepted: 02/08/2024] [Indexed: 05/05/2024] Open
Abstract
The liver has the unique capacity to regenerate, and up to 70% of the liver can be removed without detrimental consequences to the organism. Liver regeneration is a complex process involving multiple signaling networks and organs. Liver regeneration proceeds through three phases: the initiation phase, the growth phase, and the termination phase. Termination of liver regeneration occurs when the liver reaches a liver-to-body weight that is required for homeostasis, the so-called "hepatostat." The initiation and growth phases have been the subject of many studies. The molecular pathways that govern the termination phase, however, remain to be fully elucidated. This review summarizes the pathways and molecules that signal the cessation of liver regrowth after partial hepatectomy and answers the question, "What factors drive the hepatostat?" SIGNIFICANCE STATEMENT: Unraveling the pathways underlying the cessation of liver regeneration enables the identification of druggable targets that will allow us to gain pharmacological control over liver regeneration. For these purposes, it would be useful to understand why the regenerative capacity of the liver is hampered under certain pathological circumstances so as to artificially modulate the regenerative processes (e.g., by blocking the cessation pathways) to improve clinical outcomes and safeguard the patient's life.
Collapse
Affiliation(s)
- Lianne R de Haan
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, China (L.R.d.H., M.H.); Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands (L.R.d.H.); Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands (R.F.v.G.); Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands (M.H.); and Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands (M.H.)
| | - Rowan F van Golen
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, China (L.R.d.H., M.H.); Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands (L.R.d.H.); Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands (R.F.v.G.); Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands (M.H.); and Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands (M.H.)
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, China (L.R.d.H., M.H.); Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands (L.R.d.H.); Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands (R.F.v.G.); Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands (M.H.); and Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, The Netherlands (M.H.)
| |
Collapse
|
10
|
Kontoghiorghes GJ. The Importance and Essentiality of Natural and Synthetic Chelators in Medicine: Increased Prospects for the Effective Treatment of Iron Overload and Iron Deficiency. Int J Mol Sci 2024; 25:4654. [PMID: 38731873 PMCID: PMC11083551 DOI: 10.3390/ijms25094654] [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: 04/02/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
The supply and control of iron is essential for all cells and vital for many physiological processes. All functions and activities of iron are expressed in conjunction with iron-binding molecules. For example, natural chelators such as transferrin and chelator-iron complexes such as haem play major roles in iron metabolism and human physiology. Similarly, the mainstay treatments of the most common diseases of iron metabolism, namely iron deficiency anaemia and iron overload, involve many iron-chelator complexes and the iron-chelating drugs deferiprone (L1), deferoxamine (DF) and deferasirox. Endogenous chelators such as citric acid and glutathione and exogenous chelators such as ascorbic acid also play important roles in iron metabolism and iron homeostasis. Recent advances in the treatment of iron deficiency anaemia with effective iron complexes such as the ferric iron tri-maltol complex (feraccru or accrufer) and the effective treatment of transfusional iron overload using L1 and L1/DF combinations have decreased associated mortality and morbidity and also improved the quality of life of millions of patients. Many other chelating drugs such as ciclopirox, dexrazoxane and EDTA are used daily by millions of patients in other diseases. Similarly, many other drugs or their metabolites with iron-chelation capacity such as hydroxyurea, tetracyclines, anthracyclines and aspirin, as well as dietary molecules such as gallic acid, caffeic acid, quercetin, ellagic acid, maltol and many other phytochelators, are known to interact with iron and affect iron metabolism and related diseases. Different interactions are also observed in the presence of essential, xenobiotic, diagnostic and theranostic metal ions competing with iron. Clinical trials using L1 in Parkinson's, Alzheimer's and other neurodegenerative diseases, as well as HIV and other infections, cancer, diabetic nephropathy and anaemia of inflammation, highlight the importance of chelation therapy in many other clinical conditions. The proposed use of iron chelators for modulating ferroptosis signifies a new era in the design of new therapeutic chelation strategies in many other diseases. The introduction of artificial intelligence guidance for optimal chelation therapeutic outcomes in personalised medicine is expected to increase further the impact of chelation in medicine, as well as the survival and quality of life of millions of patients with iron metabolic disorders and also other diseases.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol 3021, Cyprus
| |
Collapse
|
11
|
Bhat AA, Moglad E, Bansal P, Kaur H, Deorari M, Thapa R, Almalki WH, Kazmi I, Alzarea SI, Kukreti N, Ali H. Pollutants to pathogens: The role of heavy metals in modulating TGF-β signaling and lung cancer risk. Pathol Res Pract 2024; 256:155260. [PMID: 38493726 DOI: 10.1016/j.prp.2024.155260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024]
Abstract
Lung cancer is a malignant tumor that develops in the lungs due to the uncontrolled growth of aberrant cells. Heavy metals, such as arsenic, cadmium, mercury, and lead, are metallic elements characterized by their high atomic weights and densities. Anthropogenic activities, such as industrial operations and pollution, have the potential to discharge heavy metals into the environment, hence presenting hazards to ecosystems and human well-being. The TGF-β signalling pathways have a crucial function in controlling several cellular processes, with the ability to both prevent and promote tumor growth. TGF-β regulates cellular responses by interacting in both canonical and non-canonical signalling pathways. Research employing both in vitro and in vivo models has shown that heavy metals may trigger TGF-β signalling via complex molecular pathways. Experiments conducted in a controlled laboratory environment show that heavy metals like cadmium and arsenic may directly bind to TGF-β receptors, leading to alterations in their structure that enable the receptor to be phosphorylated. Activation of this route sets in motion subsequent signalling cascades, most notably the canonical Smad pathway. The development of lung cancer has been linked to heavy metals, which are ubiquitous environmental pollutants. To grasp the underlying processes, it is necessary to comprehend their molecular effect on TGF-β pathways. With a particular emphasis on its consequences for lung cancer, this abstract delves into the complex connection between exposure to heavy metals and the stimulation of TGF-β signalling.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur 302017, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh 247341, India; Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand 831001, India
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur 302017, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Aljouf 72341, Saudi Arabia
| | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan.
| |
Collapse
|
12
|
Girelli D, Marchi G. Deironing the spleen with luspatercept. Blood 2023; 142:1856-1858. [PMID: 38032674 DOI: 10.1182/blood.2023022548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023] Open
Affiliation(s)
- Domenico Girelli
- University of Verona
- Azienda Ospedaliera Universitaria Integrata Verona
| | - Giacomo Marchi
- University of Verona
- Azienda Ospedaliera Universitaria Integrata Verona
| |
Collapse
|