1
|
Angel M, Kleinberg Y, Newaz T, Li V, Zaid R, Oved K, Dorot O, Pichinuk E, Avitan-Hersh E, Saada A, Weiss K, Zaremberg V, Tal G, Zalckvar E. Using chanarin-dorfman syndrome patient fibroblasts to explore disease mechanisms and new treatment avenues. Orphanet J Rare Dis 2025; 20:195. [PMID: 40275410 PMCID: PMC12020101 DOI: 10.1186/s13023-025-03711-6] [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: 11/09/2024] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Chanarin-Dorfman syndrome (CDS) is a multisystemic autosomal recessive rare disorder. CDS is caused by variants in the abhydrolase domain containing 5 (ABHD5) encoding gene (CGI-58), which ultimately leads to excessive lipid storage, and therefore a high abundance of cellular lipid droplets (LDs). Although the molecular etiology of the disease was described many years ago, no treatment for CDS is currently available. RESULTS To further characterize the molecular basis of the disease and to uncover new treatment avenues, we used skin fibroblasts originating from a young patient diagnosed with CDS due to a homozygous nonsense mutation. We show that dysfunctional ABHD5 does not only affect LDs, but also influences other metabolic-related organelles; the mitochondria and peroxisomes. Additionally, we found that expressing functional ABHD5 in CDS patient cells reduced LD number. Finally, we developed and applied a high content-based drug repurposing screen based on a collection of ∼2500 FDA approved compounds, yielding several compounds that affected LD total area and size. CONCLUSIONS Our findings enhance the understanding of the dysfunction underlying CDS and propose new avenues for the treatment of CDS patients.
Collapse
Affiliation(s)
- Mor Angel
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Yuval Kleinberg
- Blavatnik Center for Drug Discovery, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Tanmoy Newaz
- Department of Biological Sciences, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Victoria Li
- Department of Biological Sciences, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Rinat Zaid
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
- Clinical Research Institute at Rambam (CRIR), Rambam Health Care Campus, Haifa, Israel
| | - Keren Oved
- Blavatnik Center for Drug Discovery, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Orly Dorot
- Blavatnik Center for Drug Discovery, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Edward Pichinuk
- Blavatnik Center for Drug Discovery, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Emily Avitan-Hersh
- Department of Dermatology, Rambam Health Care Campus, Haifa, Israel
- Metabolic Unit, Ruth Rappaport Children's Hospital, Rambam Health Care Campus, PO Box 9602, Haifa, 3109601, Israel
| | - Ann Saada
- Department of Genetics, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Medical Laboratory Sciences Jerusalem Multidisciplinary College, Jerusalem, Israel
| | - Karin Weiss
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
- Clinical Research Institute at Rambam (CRIR), Rambam Health Care Campus, Haifa, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 3109601, Israel
| | - Vanina Zaremberg
- Department of Biological Sciences, University of Calgary, Calgary, AB, T2N 1N4, Canada
| | - Galit Tal
- Metabolic Unit, Ruth Rappaport Children's Hospital, Rambam Health Care Campus, PO Box 9602, Haifa, 3109601, Israel.
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 3109601, Israel.
| | - Einat Zalckvar
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, 7610001, Israel.
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat- Gan, 52900, Israel.
| |
Collapse
|
2
|
Wang S, Qin L, Liu F, Zhang Z. Unveiling the crossroads of STING signaling pathway and metabolic reprogramming: the multifaceted role of the STING in the TME and new prospects in cancer therapies. Cell Commun Signal 2025; 23:171. [PMID: 40197235 PMCID: PMC11977922 DOI: 10.1186/s12964-025-02169-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/23/2025] [Indexed: 04/10/2025] Open
Abstract
The cGAS-STING signaling pathway serves as a critical link between DNA sensing and innate immunity, and has tremendous potential to improve anti-tumor immunity by generating type I interferons. However, STING agonists have shown decreasing biotherapeutic efficacy in clinical trials. Tumor metabolism, characterized by aberrant nutrient utilization and energy production, is a fundamental hallmark of tumorigenesis. And modulating metabolic pathways in tumor cells has been discovered as a therapeutic strategy for tumors. As research concerning STING progressed, emerging evidence highlights its role in metabolic reprogramming, independent its immune function, indicating metabolic targets as a strategy for STING activation in cancers. In this review, we delve into the interplay between STING and multiple metabolic pathways. We also synthesize current knowledge on the antitumor functions of STING, and the metabolic targets within the tumor microenvironment (TME) that could be exploited for STING activation. This review highlights the necessity for future research to dissect the complex metabolic interactions with STING in various cancer types, emphasizing the potential for personalized therapeutic strategies based on metabolic profiling.
Collapse
Affiliation(s)
- Siwei Wang
- Hepatic Surgery Center, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lu Qin
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Ministry of Education, Huazhong University of Science and Technology), Wuhan, China
| | - Furong Liu
- Hepatic Surgery Center, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Zhanguo Zhang
- Hepatic Surgery Center, Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, 430030, Hubei, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
3
|
Kelpsch DJ, Zhang L, Thierer JH, Koren K, Kumar U, Lin Y, Hensley MR, Sohn M, Liu JO, Lectka T, Mumm JS, Farber SA. A whole-animal phenotypic drug screen identifies suppressors of atherogenic lipoproteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.14.623618. [PMID: 39605440 PMCID: PMC11601432 DOI: 10.1101/2024.11.14.623618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Lipoproteins are essential for lipid transport in all bilaterians. A single Apolipoprotein B (ApoB) molecule is the inseparable structural scaffold of each ApoB-containing lipoprotein (B-lps), which are responsible for transporting lipids to peripheral tissues. The cellular mechanisms that regulate ApoB and B-lp production, secretion, transport, and degradation remain to be fully defined. In humans, elevated levels of vascular B-lps play a causative role in cardiovascular disease. Previously, we have detailed that human B-lp biology is remarkably conserved in the zebrafish using an in vivo chemiluminescent reporter of ApoB (LipoGlo) that does not disrupt ApoB function. Thus, the LipoGlo model is an ideal system for identifying novel mechanisms of ApoB modulation and, due to the ability of zebrafish to generate many progeny, is particularly amenable to large-scale phenotypic drug screening. Here, we report a screen of roughly 3000 compounds that identified 49 unique ApoB-lowering hits. Nineteen hits passed orthogonal screening criteria. A licorice root component, enoxolone, significantly lowered B-lps only in animals that express a functional allele of the nuclear hormone receptor Hepatocyte Nuclear Factor 4α (HNF4α). Consistent with this result, inhibitors of HNF4α also reduce B-lp levels. These data demonstrate that mechanism(s) of action can be rapidly determined from a whole animal zebrafish phenotypic screen. Given the well documented role of HNF4α in human B-lp biology, these data validate the LipoGlo screening platform for identifying small molecule modulators of B-lps that play a critical role in a leading cause of worldwide mortality.
Collapse
Affiliation(s)
- Daniel J. Kelpsch
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Liyun Zhang
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, United States
| | - James H. Thierer
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, United States
| | - Kobe Koren
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Urmi Kumar
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Yuki Lin
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Monica R. Hensley
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Mira Sohn
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Jun O. Liu
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, United States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, United States
| | - Thomas Lectka
- Department of Chemistry, Johns Hopkins University, Baltimore, United States
| | - Jeff S. Mumm
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, United States
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University, Baltimore, United States
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, United States
| | - Steven A. Farber
- Department of Biology, Johns Hopkins University, Baltimore, United States
| |
Collapse
|
4
|
Gu J, Gupta RN, Cheng HK, Xu Y, Raal FJ. Current treatments for the management of homozygous familial hypercholesterolaemia: a systematic review and commentary. Eur J Prev Cardiol 2024; 31:1833-1849. [PMID: 38640433 DOI: 10.1093/eurjpc/zwae144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/15/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
AIMS Homozygous familial hypercholesterolaemia (HoFH) is a rare disorder characterized by markedly elevated circulating low-density lipoprotein cholesterol (LDL-C) from birth. This review aimed to critically evaluate treatments for HoFH with respect to their efficacy, safety, accessibility, overall context and position within the treatment pathway. METHODS AND RESULTS A mixed-methods review was undertaken to systematically identify and characterize primary interventional studies on HoFH, with a focus on LDL-C reduction as the primary outcome. Interventions assessed were ezetimibe, proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), lomitapide, evinacumab, with or without LDL apheresis. Twenty-six seminal studies reporting unique patient data were identified. Four studies were randomized controlled trials (RCTs) with the remainder being single-arm trials or observational registries. Data extracted were heterogeneous and not suitable for meta-analyses. Two RCTs, assessed at being low risk of bias, demonstrated PCSK9i were safe and moderately effective. A randomized controlled trial (RCT) demonstrated evinacumab was safe and effective in all HoFH subgroups. Lomitapide was reported to be efficacious in a single-arm trial, but issues with adverse events, tolerability, and adherence were identified. An RCT on ezetimibe showed it was moderately effective when combined with a statin. LDL apheresis was reported as effective, but its evidence base was at very high risk of bias. All interventions lowered LDL-C, but the magnitude of this, and certainty in the supporting evidence, varied. CONCLUSION In practice, multiple treatments are required to treat HoFH. The sequencing of these should be made on an individualized basis, with consideration made to the benefits of each intervention.
Collapse
Affiliation(s)
- Jing Gu
- Regeneron Pharmaceuticals, Inc, Global Medical Affairs, 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
| | - Rupal N Gupta
- Ultragenyx Pharmaceutical Inc, Global Medical Affairs, 60 Leveroni Court, Novato, CA 94949, USA
| | - Henry K Cheng
- Ultragenyx Pharmaceutical Inc, Global Medical Affairs, 60 Leveroni Court, Novato, CA 94949, USA
| | - Yingxin Xu
- Regeneron Pharmaceuticals, Inc, Global Medical Affairs, 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA
| | - Frederick J Raal
- Department of Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| |
Collapse
|
5
|
Fan N, Zhao F, Meng Y, Chen L, Miao L, Wang P, Tang M, Wu X, Li Y, Li Y, Gao Z. Metal complex lipid-based nanoparticles deliver metabolism-regulating lomitapide to overcome CTC immune evasion via activating STING pathway. Eur J Pharm Biopharm 2024; 203:114467. [PMID: 39173934 DOI: 10.1016/j.ejpb.2024.114467] [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: 05/14/2024] [Revised: 07/23/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Activating the cGAS-STING pathway of circulating tumor cell clusters (CTC clusters) represents a promising strategy to mitigate metastases. To fully exploit the potential of cholesterol-regulating agents in activating CTCs' STING levels, we developed a nanoparticle (NP) composed of metal complex lipid (MCL). This design includes MCL-miriplatin to increase NP stiffness and loads lomitapide (lomi) modulating cholesterol levels, resulting in the creation of PLTs@Pt-lipid@lomi NPs. MCL-miriplatin not only enhances lomi's eliciting efficacy on STING pathway but also increases NPs' stiffness, thus a vital factor affecting the penetration into CTC clusters to further boost lomi's ability. Demonstrated by cy5 tracking experiments, PLTs@Pt-lipid@lomi NPs quickly attach to cancer cell via platelet membrane anchorage, penetrate deep into the spheres, and reach the subcellular endoplasmic reticulum where lomi regulates cholesterol. Additionally, these NPs have been shown to track CTCs in the bloodstream, a capability not demonstrated by the free drug. PLTs@Pt-lipid@lomi NPs more efficiently activate the STING pathway and reduce CTC stemness compared to free lomi. Ultimately, PLTs@Pt-lipid@lomi NPs reduce metastasis in a post-surgery animal model. While cholesterol-regulating agents are limited in efficacy when being repositioned as immunomodulatory agents, this MCL-composing NP strategy demonstrates the potential to effectively deliver these agents to target CTC clusters.
Collapse
Affiliation(s)
- Ni Fan
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Feng Zhao
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuanyuan Meng
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Liqing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lin Miao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ping Wang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Manqing Tang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xuanjun Wu
- Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Qingdao, Shandong University, Shandong 266237, China
| | - Yingpeng Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yunfei Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
6
|
Kalasin S, Surareungchai W. Artificial intelligence-aiding lab-on-a-chip workforce designed oral [3.1.0] bi and [4.2.0] tricyclic catalytic interceptors inhibiting multiple SARS-CoV-2 protomers assisted by double-shell deep learning. RSC Adv 2024; 14:26897-26910. [PMID: 39193274 PMCID: PMC11347926 DOI: 10.1039/d4ra03965c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/20/2024] [Indexed: 08/29/2024] Open
Abstract
While each massive pandemic has claimed the lives of millions of vulnerable populations over the centuries, one limitation exists: that the Edisonian approach (human-directed with trial errors) relies on repurposing pharmaceuticals, designing drugs, and herbal remedies with the violation of Lipinski's rule of five druglikeness. It may lead to adverse health effects with long-term health multimorbidity. Nevertheless, declining birth rates and aging populations will likely cause a shift in society due to a shortage of a scientific workforce to defend against the next pandemic incursion. The challenge of combating the ongoing post-COVID-19 pandemic has been exacerbated by the lack of gold standard drugs to deactivate multiple SARS-CoV-2 protein targets. Meanwhile, there are three FDA-approved antivirals, Remdesivir, Molnupiravir, and Paxlovid, with moderate clinical efficacy and drug resistance. There is a pressing need for additional antivirals and prepared omics technology to combat the current and future devastating coronavirus pandemics. While there is a limitation of existing contemporary inhibitors to deactivate viral RNA replication with minimal rotational bonds, one strategy is to create Lipinski inhibitors with less than 10 rotational bonds and precise halogen bond placement to destabilize multiple viral protomers. This work describes the efforts to design gold-standard oral inhibitors of bi- and tri-cyclic catalytic interceptors with electrophilic heads using double-shell deep learning. Here, KS1 with and KS2 compounds designed by lab-on-a-chip technology attain 5-fold novel filtered-Lipinski, GHOSE, VEBER, EGAN, and MUEGGE druglikeness. The graph neural network (GNN) relies on module-initiation, expansion, relabeling atom index, and termination (METORITE) iterations, while the deep neural network (DNN) engages pinning, extraction, convolution, pooling, and flattening (PROOF) operations. The cyclic compound's specific halogen atom location enhances the nitrile catalytic head, which deactivates several viral protein targets. Initiating this lab-on-a-chip that is not susceptible to the aging process for creating clinical compounds can leverage a new path to many valuable drugs with speedy oral drug discovery, especially to defend the loss of vulnerable population and prevent multimorbidity that is susceptible to hidden viral persistence in the continuing aging times.
Collapse
Affiliation(s)
- Surachate Kalasin
- Faculty of Science and Nanoscience & Nanotechnology Graduate Program, King Mongkut's University of Technology Thonburi 10140 Thailand
| | - Werasak Surareungchai
- Faculty of Science and Nanoscience & Nanotechnology Graduate Program, King Mongkut's University of Technology Thonburi 10140 Thailand
- Pilot Plant Research and Development Laboratory, King Mongkut's University of Technology Thonburi 10150 Bangkok Thailand
- School of Bioresource and Technology, King Mongkut's University of Technology Thonburi 10150 Bangkok Thailand
- Analytical Sciences and National Doping Test Institute, Mahidol University Bangkok 10400 Thailand
| |
Collapse
|
7
|
Tarugi P, Bertolini S, Calandra S, Arca M, Angelico F, Casula M, Cefalù AB, D'Erasmo L, Fortunato G, Perrone-Filardi P, Rubba P, Suppressa P, Averna M, Catapano AL. Consensus document on diagnosis and management of familial hypercholesterolemia from the Italian Society for the Study of Atherosclerosis (SISA). Nutr Metab Cardiovasc Dis 2024; 34:1819-1836. [PMID: 38871496 DOI: 10.1016/j.numecd.2024.05.002] [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: 12/19/2023] [Revised: 04/04/2024] [Accepted: 05/03/2024] [Indexed: 06/15/2024]
Abstract
AIMS Familial Hypercholesterolemia (FH) is a genetic disorder of lipoprotein metabolism that causes an increased risk of premature atherosclerotic cardiovascular disease (ASCVD). Although early diagnosis and treatment of FH can significantly improve the cardiovascular prognosis, this disorder is underdiagnosed and undertreated. For these reasons the Italian Society for the Study of Atherosclerosis (SISA) assembled a Consensus Panel with the task to provide guidelines for FH diagnosis and treatment. DATA SYNTHESIS Our guidelines include: i) an overview of the genetic complexity of FH and the role of candidate genes involved in LDL metabolism; ii) the prevalence of FH in the population; iii) the clinical criteria adopted for the diagnosis of FH; iv) the screening for ASCVD and the role of cardiovascular imaging techniques; v) the role of molecular diagnosis in establishing the genetic bases of the disorder; vi) the current therapeutic options in both heterozygous and homozygous FH. Treatment strategies and targets are currently based on low-density lipoprotein cholesterol (LDL-C) levels, as the prognosis of FH largely depends on the magnitude of LDL-C reduction achieved by lipid-lowering therapies. Statins with or without ezetimibe are the mainstay of treatment. Addition of novel medications like PCSK9 inhibitors, ANGPTL3 inhibitors or lomitapide in homozygous FH results in a further reduction of LDL-C levels. LDL apheresis is indicated in FH patients with inadequate response to cholesterol-lowering therapies. CONCLUSION FH is a common, treatable genetic disorder and, although our understanding of this disease has improved, many challenges still remain with regard to its identification and management.
Collapse
Affiliation(s)
- Patrizia Tarugi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | | | - Sebastiano Calandra
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marcello Arca
- Department of Translational and Precision Medicine (DTPM), Sapienza University of Rome, Policlinico Umberto I, Rome, Italy
| | | | - Manuela Casula
- Department of Pharmacological and Biomolecular Sciences (DisFeB), Epidemiology and Preventive Pharmacology Service (SEFAP), University of Milan, Milan, Italy; IRCCS Multimedica, Sesto San Giovanni (Milan), Italy
| | - Angelo B Cefalù
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Laura D'Erasmo
- Department of Translational and Precision Medicine (DTPM), Sapienza University of Rome, Policlinico Umberto I, Rome, Italy
| | - Giuliana Fortunato
- Department of Medicina Molecolare e Biotecnologie Mediche, University of Naples Federico II and CEINGE Biotecnologie avanzate "Franco Salvatore", Naples, Italy
| | | | - Paolo Rubba
- Department of Internal Medicine and Surgery, Federico II University, Naples, Italy
| | - Patrizia Suppressa
- Department of Internal Medicine and Rare Diseases Centre "C. Frugoni", University of Bari A. Moro, Bari, Italy
| | - Maurizio Averna
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy; Biophysical Institute CNR, Palermo, Italy
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milano, Italy; IRCCS Multimedica, Milano, Italy
| |
Collapse
|
8
|
Gaudet D, Greber-Platzer S, Reeskamp LF, Iannuzzo G, Rosenson RS, Saheb S, Stefanutti C, Stroes E, Wiegman A, Turner T, Ali S, Banerjee P, Drewery T, McGinniss J, Waldron A, George RT, Zhao XQ, Pordy R, Zhao J, Bruckert E, Raal FJ. Evinacumab in homozygous familial hypercholesterolaemia: long-term safety and efficacy. Eur Heart J 2024; 45:2422-2434. [PMID: 38856678 PMCID: PMC11242450 DOI: 10.1093/eurheartj/ehae325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 04/17/2024] [Accepted: 05/12/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND AND AIMS Homozygous familial hypercholesterolaemia (HoFH) is a rare genetic disorder characterized by severely elevated LDL cholesterol (LDL-C) and premature atherosclerotic cardiovascular disease. In the pivotal Phase 3 HoFH trial (NCT03399786), evinacumab significantly decreased LDL-C in patients with HoFH. This study assesses the long-term safety and efficacy of evinacumab in adult and adolescent patients with HoFH. METHODS In this open-label, single-arm, Phase 3 trial (NCT03409744), patients aged ≥12 years with HoFH who were evinacumab-naïve or had previously received evinacumab in other trials (evinacumab-continue) received intravenous evinacumab 15 mg/kg every 4 weeks with stable lipid-lowering therapy. RESULTS A total of 116 patients (adults: n = 102; adolescents: n = 14) were enrolled, of whom 57 (49.1%) were female. Patients were treated for a median (range) duration of 104.3 (28.3-196.3) weeks. Overall, treatment-emergent adverse events (TEAEs) and serious TEAEs were reported in 93 (80.2%) and 27 (23.3%) patients, respectively. Two (1.7%) deaths were reported (neither was considered related to evinacumab). Three (2.6%) patients discontinued due to TEAEs (none were considered related to evinacumab). From baseline to Week 24, evinacumab decreased mean LDL-C by 43.6% [mean (standard deviation, SD), 3.4 (3.2) mmol/L] in the overall population; mean LDL-C reduction in adults and adolescents was 41.7% [mean (SD), 3.2 (3.3) mmol/L] and 55.4% [mean (SD), 4.7 (2.5) mmol/L], respectively. CONCLUSIONS In this large cohort of patients with HoFH, evinacumab was generally well tolerated and markedly decreased LDL-C irrespective of age and sex. Moreover, the efficacy and safety of evinacumab was sustained over the long term.
Collapse
MESH Headings
- Humans
- Female
- Male
- Hyperlipoproteinemia Type II/drug therapy
- Adolescent
- Adult
- Cholesterol, LDL/blood
- Middle Aged
- Anticholesteremic Agents/therapeutic use
- Anticholesteremic Agents/administration & dosage
- Anticholesteremic Agents/adverse effects
- Treatment Outcome
- Young Adult
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/administration & dosage
- Child
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Homozygote
Collapse
Affiliation(s)
- Daniel Gaudet
- Clinical Lipidology and Rare Lipid Disorders Unit, Community Gene Medicine Center, Department of Medicine, Université de Montréal and ECOGENE-21, 930 Jacques-Cartier, Suite 210-B, Chicoutimi, Québec G7H 7K9, Canada
| | - Susanne Greber-Platzer
- Division of Pediatric Pulmonology, Allergology and Endocrinology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Laurens F Reeskamp
- Department of Vascular Medicine, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, University of Naples, Naples, Italy
| | - Robert S Rosenson
- Metabolism and Lipids Unit, Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Samir Saheb
- LDL-Apheresis Unit, Department of Endocrinology, Hôpital de la Pitié-Salpêtrière, Université Paris Diderot, Sorbonne Paris, Paris, France
| | - Claudia Stefanutti
- Department of Molecular Medicine, Extracorporeal Therapeutic Techniques Unit, Lipid Clinic and Atherosclerosis Prevention Centre, Regional Centre for Rare Diseases, Immunohematology and Transfusion Medicine, Umberto I Hospital, ‘Sapienza’ University of Rome, Rome, Italy
| | - Erik Stroes
- Department of Vascular Medicine, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Albert Wiegman
- Department of Paediatrics, Amsterdam University Medical Centers, Location University of Amsterdam, The Netherlands
| | - Traci Turner
- Medpace Reference Laboratories, Cincinnati, OH, USA
| | - Shazia Ali
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | | | | | | | | | - Robert Pordy
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Jian Zhao
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | - Eric Bruckert
- Department of Endocrinology, Hôpital de la Pitié-Salpêtrière, Université Paris Diderot, Sorbonne Paris, Paris, France
| | - Frederick J Raal
- Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
9
|
Wu HT, Wu BX, Fang ZX, Wu Z, Hou YY, Deng Y, Cui YK, Liu J. Lomitapide repurposing for treatment of malignancies: A promising direction. Heliyon 2024; 10:e32998. [PMID: 38988566 PMCID: PMC11234027 DOI: 10.1016/j.heliyon.2024.e32998] [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: 08/04/2023] [Revised: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024] Open
Abstract
The development of novel drugs from basic science to clinical practice requires several years, much effort, and cost. Drug repurposing can promote the utilization of clinical drugs in cancer therapy. Recent studies have shown the potential effects of lomitapide on treating malignancies, which is currently used for the treatment of familial hypercholesterolemia. We systematically review possible functions and mechanisms of lomitapide as an anti-tumor compound, regarding the aspects of apoptosis, autophagy, and metabolism of tumor cells, to support repurposing lomitapide for the clinical treatment of tumors.
Collapse
Affiliation(s)
- Hua-Tao Wu
- Department of General Surgery, the First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Bing-Xuan Wu
- Department of General Surgery, the First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Ze-Xuan Fang
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
- Department of Physiology/Changjiang Scholar's Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Zheng Wu
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
- Department of Physiology/Changjiang Scholar's Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Yan-Yu Hou
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
- Department of Physiology/Changjiang Scholar's Laboratory, Shantou University Medical College, Shantou, 515041, China
| | - Yu Deng
- Department of General Surgery, the First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Yu-Kun Cui
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
| | - Jing Liu
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, 515041, China
- Department of Physiology/Changjiang Scholar's Laboratory, Shantou University Medical College, Shantou, 515041, China
| |
Collapse
|
10
|
Alonso R, Arroyo-Olivares R, Díaz-Díaz JL, Fuentes-Jiménez F, Arrieta F, de Andrés R, Gonzalez-Bustos P, Argueso R, Martin-Ordiales M, Martinez-Faedo C, Illán F, Saenz P, Donate JM, Sanchez Muñoz-Torrero JF, Martinez-Hervas S, Mata P. Improved lipid-lowering treatment and reduction in cardiovascular disease burden in homozygous familial hypercholesterolemia: The SAFEHEART follow-up study. Atherosclerosis 2024; 393:117516. [PMID: 38523000 DOI: 10.1016/j.atherosclerosis.2024.117516] [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: 12/18/2023] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/26/2024]
Abstract
AIM We aimed to describe clinical and genetic characteristics, lipid-lowering treatment and atherosclerotic cardiovascular disease (ASCVD) outcomes over a long-term follow-up in homozygous familial hypercholesterolemia (HoFH). METHODS SAFEHEART (Spanish Familial Hypercholesterolaemia Cohort Study) is a long-term study in molecularly diagnosed FH. Data analyzed in HoFH were prospectively obtained from 2004 until 2022. ASCVD events, lipid profile and lipid-lowering treatment were determined. RESULTS Thirty-nine HoFH patients were analyzed. The mean age was 42 ± 20 years and nineteen (49%) were women. Median follow-up was 11 years (IQR 6,18). Median age at genetic diagnosis was 24 years (IQR 8,42). At enrolment, 33% had ASCVD and 18% had aortic valve disease. Patients with new ASCVD events and aortic valve disease at follow-up were six (15%), and one (3%), respectively. Median untreated LDL-C levels were 555 mg/dL (IQ 413,800), and median LDL-C levels at last follow-up was 122 mg/dL (IQR 91,172). Most patients (92%) were on high intensity statins and ezetimibe, 28% with PCSK9i, 26% with lomitapide, and 23% with lipoprotein-apheresis. Fourteen patients (36%) attained an LDL-C level below 100 mg/dL, and 10% attained an LDL-C below 70 mg/dL in secondary prevention. Patients with null/null variants were youngers, had higher untreated LDL-C and had the first ASCVD event earlier. Free-event survival is longer in patients with defective variant compared with those patients with at least one null variant (p=0.02). CONCLUSIONS HoFH is a severe life threating disease with a high genetic and phenotypic variability. The improvement in lipid-lowering treatment and LDL-C levels have contributed to reduce ASCVD events.
Collapse
Affiliation(s)
- Rodrigo Alonso
- Fundación Hipercolesterolemia Familiar, Madrid, Spain; Center for Advanced Metabolic Medicine and Nutrition, Santiago, Chile.
| | | | | | - Francisco Fuentes-Jiménez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), University of Cordoba, Reina Sofia University Hospital, CIBERObn, Córdoba, Spain
| | | | | | - Pablo Gonzalez-Bustos
- Department of Internal Medicine, Hospital Universitario Virgen de Las Nieves, Granada, Spain
| | - Rosa Argueso
- Department of Endocrinology, Hospital Universitario de Lugo, Lugo, Spain
| | | | | | - Fátima Illán
- Department of Endocrinology, Hospital Morales Meseguer, Murcia, Spain
| | - Pedro Saenz
- Department of Internal Medicine, Hospital de Mérida, Mérida, Spain
| | - José María Donate
- Department of Pediatric Endocrinology, Hospital General Universitario Santa Lucía, Murcia, Spain
| | | | - Sergio Martinez-Hervas
- Department of Endocrinology, Hospital Clínico Universitario de Valencia INCLIVA, CIBER de Diabetes, Spain
| | - Pedro Mata
- Fundación Hipercolesterolemia Familiar, Madrid, Spain.
| |
Collapse
|
11
|
Son SM, Lee HS, Kim J, Kwon RJ. Expression and prognostic significance of microsomal triglyceride transfer protein in brain tumors: a retrospective cohort study. Transl Cancer Res 2024; 13:2282-2294. [PMID: 38881934 PMCID: PMC11170499 DOI: 10.21037/tcr-23-2286] [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: 12/12/2023] [Accepted: 04/10/2024] [Indexed: 06/18/2024]
Abstract
Background Glioblastoma (GBM) is the most common malignant brain tumor and has poor survival. An elevated cholesterol level is involved occurrence and progression of brain tumors. Microsomal triglyceride transfer protein (MTTP) is a target for lowering lipids, and its inhibition helps to improve hyperlipidemia. However, whether the altered expression of MTTP affects the development and prognosis of brain tumors is currently unidentified. The purpose of this study is to determine MTTP as a prognostic marker for brain tumors. Methods Data for patients with brain cancers and control brain tissue were acquired from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). The datasets were analyzed using Mann-Whitney U-test or t-test to compare the expression of MTTP in normal and brain tumor tissues. To examine whether MTTP affected the prognosis of patients with brain tumors, log-rank test and multivariable Cox proportional hazard regression were conducted. Results The expression of MTTP was significantly upregulated in brain tumors and was correlated with age, tumor stage, and isocitrate dehydrogenase (IDH) mutation. Importantly, increased MTTP expression in brain tumors is associated with poor patient survival. Conclusions High MTTP expression is associated with brain tumor development, tumor stage, and prognosis. Therefore, MTTP is an independent prognostic indicator for brain tumors, which can serve as one of the possible targets for adjuvant treatment of GBM.
Collapse
Affiliation(s)
- Soo Min Son
- Family Medicine Clinic and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
- Department of Family Medicine, Pusan National University School of Medicine, Yangsan, Korea
| | - Hye Sun Lee
- Family Medicine Clinic and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Jeongsu Kim
- Division of Cardiology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea
- Division of Cardiology, Department of Internal Medicine, Pusan National University School of Medicine, Yangsan, Korea
| | - Ryuk Jun Kwon
- Family Medicine Clinic and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
- Department of Family Medicine, Pusan National University School of Medicine, Yangsan, Korea
| |
Collapse
|
12
|
Moll TO, Klemek ML, Farber SA. Directly Measuring Atherogenic Lipoprotein Kinetics in Zebrafish with the Photoconvertible LipoTimer Reporter. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596423. [PMID: 38853962 PMCID: PMC11160697 DOI: 10.1101/2024.05.29.596423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Lipoprotein kinetics are a crucial factor in understanding lipoprotein metabolism since a prolonged time in circulation can contribute to the atherogenic character of apolipoprotein-B (ApoB)-containing lipoproteins (B-lps). Here, we report a method to directly measure lipoprotein kinetics in live developing animals. We developed a zebrafish geneticly encoded reporter, LipoTimer, in which endogenous ApoBb.1 is fused to the photoconvertible fluorophore Dendra2 which shift its emission profile from green to red upon UV exposure. By quantifying the red population of ApoB-Dendra2 over time, we found that B-lp turnover in wild-type larvae becomes faster as development proceeds. Mutants with impaired B-lp uptake or lipolysis present with increased B-lp levels and half-life. In contrast, mutants with impaired B-lp triglyceride loading display slightly fewer and smaller-B-lps, which have a significantly shorter B-lp half-life. Further, we showed that chronic high-cholesterol feeding is associated with a longer B-lp half-life in wild-type juveniles but does not lead to changes in B-lp half-life in lipolysis deficient apoC2 mutants. These data support the hypothesis that B-lp lipolysis is suppressed by the flood of intestinal-derived B-lps that follow a high-fat meal.
Collapse
Affiliation(s)
- Tabea O.C. Moll
- Johns Hopkins University, Baltimore, Maryland, United States of America
| | | | - Steven A. Farber
- Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
13
|
Ghose T. Lipoprotein a - Lp(a). Indian Heart J 2024; 76 Suppl 1:S117-S120. [PMID: 38160790 PMCID: PMC11019309 DOI: 10.1016/j.ihj.2023.12.010] [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: 11/17/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024] Open
Abstract
Lp(a) is a genetically determined, heritable, independent and causal risk factor for ASCVD. About 1 in 5 people worldwide have elevated Lp(a) (>50 mg/dL or >125 nmol/L) whereas in Indians it is 25 %. Epidemiological, genome-wide association and mendelian randomization studies have demonstrated an association between elevated Lp(a) levels and increased incidence of myocardial infarction, aortic valve stenosis, ischemic stroke, heart failure, CV and all-cause mortality. The increased Lp(a)-mediated CV risk is mediated by pro-inflammatory, pro-thrombotic and pro-atherogenic processes, leading to progression of atherosclerosis and increased risk of thrombosis. Lp(a) level reaches peak by 5 years of age and remains stable over time. Levels are not much influenced by dietary and environmental factors but it can vary in certain clinical situations like thyroid diseases, chronic kidney disease, inflammation and sepsis. It should be measured at least once in life time. Cascade testing for high Lp(a) is recommended in the settings of FH, family history of (very) high Lp(a), and personal or family history of ASCVD. In the absence of specific Lp(a)-lowering therapies, comprehensive risk factor management is recommended as per guidelines for individuals with elevated Lp(a). PCSK9 inhibitors and Inclisiran reduce Lp(a) by 25%. Pelacarsen is an antisense oligonucleotide and is found to reduce Lp(a) by 80%. In a recent Indian study of 1,021 CAD patients, presence of elevated Lp(a) (>50 mg/dL) correlated with severe angiographic disease. 37% of ACS patients exhibited elevated Lp(a) and it was higher in young CAD patients with FH (43%).
Collapse
Affiliation(s)
- Tapan Ghose
- Director and Head of Cardiology, Fortis Flt Lt Rajan Dhall Hospital New Delhi, 11-0070, India.
| |
Collapse
|
14
|
Al-Ashwal A, Alsagheir A, Al Dubayee M, Al-Khnifsawi M, Al-Sarraf A, Awan Z, Ben-Omran T, Al-Yaarubi S, Almutair A, Habeb A, Maatouk F, Alshareef M, Kholaif N, Blom D. Modern approaches to the management of homozygous familial hypercholesterolemia in the Middle East and North Africa. J Clin Lipidol 2024; 18:e132-e141. [PMID: 38158247 DOI: 10.1016/j.jacl.2023.12.003] [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: 07/06/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024]
Abstract
Homozygous familial hypercholesterolaemia (HoFH) is a severe form of FH in which inheritance of two defective or null mutations in genes associated with metabolism of low-density lipoprotein cholesterol (LDL-C) results in extremely high LDL-C, premature atherosclerotic cardiovascular disease (ASCVD) and mortality. Treatment of HoFH comprises a multi-modal approach of statins, ezetimibe, lipoprotein apheresis; and inhibitors of proprotein convertase subtilisin/kexin type, angiopoietin-like protein 3 (ANGPTL3) and microsomal triglyceride transfer protein. These treatments are generally costly, and patients also often require treatment for ASCVD consequent to HoFH. Therefore, in the interests of both economics and preservation of life, disease prevention via genetic screening and counselling is rapidly becoming a key element in the overall management of HoFH. Guidelines are available to assist diagnosis and treatment of HoFH; however, while advancements have been made in the management of the disease, there has been little systematic attention paid to prevention. Additionally, the Middle East/North Africa (MENA) region has a higher prevalence of HoFH than most other regions - chiefly due to consanguinity. This has led to the establishment of regional lipid clinics and awareness programs that have thrown education and awareness of HoFH into sharp focus. Incorporation of principles of prevention, education, awareness, and data from real-world use of existing therapeutics will significantly enhance the effectiveness of future guidelines for the management of HoFH, particularly in the MENA region.
Collapse
Affiliation(s)
- Abdullah Al-Ashwal
- Medical & Clinical Affairs, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia (Dr Al-Ashwal)
| | - Afaf Alsagheir
- Pediatrics Department, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia (Dr Alsagheir)
| | - Mohammed Al Dubayee
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11426, Saudi Arabia (Dr Al Dubayee)
| | | | - Ahmed Al-Sarraf
- Sabah Al Ahmad Cardiac Center, Department Cardiology, Ministry of Health, Kuwait (Dr Al-Sarraf)
| | - Zuhier Awan
- Division of Clinical Biochemistry, King Abdulaziz University, Abdullah Sulayman, Jeddah, Saudi Arabia (Dr Awan)
| | - Tawfeg Ben-Omran
- Division of Genetics and Genomic Medicine, Sidra Medicine and Hamad Medical Corporation, Doha, Qatar (Dr Ben-Omran)
| | - Saif Al-Yaarubi
- Oman Medical Specialty Board, Muscat, Sultanate of Oman (Dr Al-Yaarubi)
| | - Angham Almutair
- King Abdullah Specialised Children's Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia (Dr Almutair)
| | - Abdelhadi Habeb
- Pediatric Department, Prince Mohamed Bin Abdulaziz Hospital, Madinah, Saudi Arabia (Dr Habeb)
| | - Faouzi Maatouk
- Division of Cardiology, Department of Medicine, Fattouma Bourguiba University Hospital, Tunisia (Dr Maatouk)
| | - Manal Alshareef
- National Guard Hospital, Prince Mutib Ibn Abdullah Rd, National Guard District, Riyadh, Saudi Arabia (Dr Alshareef)
| | - Naji Kholaif
- Heart Centre Cardiology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia (Dr Kholaif); Alfaisal University College of Medicine, Riyadh, Saudi Arabia (Dr Kholaif)
| | - Dirk Blom
- Division of Lipidology, Department of Medicine and Cape Heart Institute, University of Cape Town, Cape Town, South Africa (Dr Blom)
| |
Collapse
|
15
|
Jain P. Traditional and novel non-statin lipid-lowering drugs. Indian Heart J 2024; 76 Suppl 1:S38-S43. [PMID: 37979722 PMCID: PMC11019312 DOI: 10.1016/j.ihj.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023] Open
Abstract
Non-statin drugs find utility in the management of dyslipidaemia in mixed dyslipidaemia, patients with statin intolerance, and when guidelines directed low-density lipoprotein cholesterol (LDL-C) target cannot be achieved despite maximally tolerated statin. The most definite indication of fenofibrate monotherapy is fasting serum triglyceride >500 mg/dl to reduce the risk of acute pancreatitis It offers a modest reduction in cardiovascular events. The statin-ezetimibe combination is commonly used for lipid lowering particularly after ACS. Fish oils reduce serum triglycerides by about 25 %. EPA (and not DHA) seems to have cardioprotective effects. Despite cardiovascular outcome benefits, bile-exchange resins have limited use due to poor tolerance. Bempedoic acid added to maximally tolerated statin therapy is approved to lower LDL-C in adults with primary hypercholesterolemia or mixed dyslipidaemias, HeFH, in patients with ASCVD who require additional lowering of LDL-C, and in patients who are statin-intolerant. Inclisiran is a long-acting double-stranded small interfering RNA (siRNA) that inhibits the transcription of PCSK-9 leading to a decrease in PCSK9 generation in hepatocytes and an increase in LDL receptor expression in the liver cell membrane leading to about 50 % reduction in serum LDL-C levels. Lomitapide lowers plasma levels of all ApoB-containing lipoproteins, including VLDL, LDL, and chylomicrons by inhibiting the enzyme microsomal triglyceride transfer protein (MTP) and approved for the treatment of adult patients with homozygous familial hypercholesterolemia (HoFH). Close monitoring for hepatotoxicity is required. Mipomersen is a single-stranded synthetic antisense oligonucleotide (ASO) that affects the production and secretion of apoB-containing lipoproteins with demonstrated efficacy in both homozygous and heterozygous FH patients. It is approved for restricted use due to risk of hepatotoxicity. Pelacarsen is an antisense oligonucleotide that reduces the production of apo(a) in the liver.
Collapse
Affiliation(s)
- Peeyush Jain
- Department of Preventive Cardiology, Fortis-Escorts Heart Institute, Okhla Road, New Delhi, 110 025, India.
| |
Collapse
|
16
|
Harada-Shiba M, Haruna S, Kogawa N. Real-world safety and efficacy of lomitapide in homozygous familial hypercholesterolemia: interim report of special-use survey in Japan. Future Cardiol 2024; 20:67-80. [PMID: 38420884 DOI: 10.2217/fca-2023-0136] [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: 11/13/2023] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
Aim: To evaluate the safety and efficacy of lomitapide in real-world clinical practice in Japan. Patients & methods: Interim analysis of 39 patients with homozygous familial hypercholesterolemia from an all-case surveillance study. Results: Median lomitapide dose (across 42 months) was 9.8 mg/day. 74 drug-related adverse events (AEs) were reported in 24 (61.5%) patients, including 14 (35.9%) with liver-related AEs, 19 (48.7%) with gastrointestinal disorders and 1 (2.6%) bleeding disorder. Lomitapide dose was reduced for 39.2% of drug-related AEs, withdrawn temporarily for 12.2%, and discontinued for 1 event (1.4%). Mean ± SD blood LDL-C level decreased from 225.9 ± 172.0 mg/dl (5.8 ± 4.5 mmol/l) predose to 159.4 ± 93.0 mg/dl (4.1 ± 2.4 mmol/l) at 12 months (p = 0.0245). Conclusion: This interim analysis suggests lomitapide is safe and effective in real-world clinical practice in Japan.
Collapse
Affiliation(s)
- Mariko Harada-Shiba
- Cardiovascular Center, Osaka Medical & Pharmaceutical University, Osaka, 569-8686, Japan
| | | | - Noriaki Kogawa
- Recordati Rare Diseases Japan K.K., Tokyo, 102-0082, Japan
| |
Collapse
|
17
|
Michaeli DT, Michaeli JC, Albers S, Boch T, Michaeli T. Established and Emerging Lipid-Lowering Drugs for Primary and Secondary Cardiovascular Prevention. Am J Cardiovasc Drugs 2023; 23:477-495. [PMID: 37486464 PMCID: PMC10462544 DOI: 10.1007/s40256-023-00594-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/02/2023] [Indexed: 07/25/2023]
Abstract
Despite treatment with statins, patients with elevated low-density lipoprotein cholesterol (LDL-C) and triglycerides remain at increased risk for adverse cardiovascular events. Consequently, novel pharmaceutical drugs have been developed to control and modify the composition of blood lipids to ultimately prevent fatal cardiovascular events in patients with dyslipidaemia. This article reviews established and emerging lipid-lowering drugs regarding their mechanism of action, development stage, ongoing clinical trials, side effects, effect on blood lipids and reduction in cardiovascular morbidity and mortality. We conducted a keyword search to identify studies on established and emerging lipid modifying drugs. Results were summarized in a narrative overview. Established pharmaceutical treatment options include the Niemann-Pick-C1 like-1 protein (NPC1L1) inhibitor ezetimibe, the protein convertase subtilisin-kexin type 9 (PCSK9) inhibitors alirocumab and evolocumab, fibrates as peroxisome proliferator receptor alpha (PPAR-α) activators, and the omega-3 fatty acid icosapent ethyl. Statins are recommended as the first-line therapy for primary and secondary cardiovascular prevention in patients with hypercholesterinaemia and hypertriglyceridemia. For secondary prevention in hypercholesterinaemia, second-line options such as statin add-on or statin-intolerant treatments are ezetimibe, alirocumab and evolocumab. For secondary prevention in hypertriglyceridemia, second-line options such as statin add-on or statin-intolerant treatments are icosapent ethyl and fenofibrate. Robust data for these add-on therapeutics in primary cardiovascular prevention remains scarce. Recent biotechnological advances have led to the development of innovative small molecules (bempedoic acid, lomitapide, pemafibrate, docosapentaenoic and eicosapentaenoic acid), antibodies (evinacumab), antisense oligonucleotides (mipomersen, volanesorsen, pelcarsen, olezarsen), small interfering RNA (inclisiran, olpasiran), and gene therapies for patients with dyslipidemia. These molecules specifically target new cellular pathways, such as the adenosine triphosphate-citrate lyase (bempedoic acid), PCSK9 (inclisiran), angiopoietin-like 3 (ANGPTL3: evinacumab), microsomal triglyceride transfer protein (MTP: lomitapide), apolipoprotein B-100 (ApoB-100: mipomersen), apolipoprotein C-III (ApoC-III: volanesorsen, olezarsen), and lipoprotein (a) (Lp(a): pelcarsen, olpasiran). The authors are hopeful that the development of new treatment modalities alongside new therapeutic targets will further reduce patients' risk of adverse cardiovascular events. Apart from statins, data on new drugs' use in primary cardiovascular prevention remain scarce. For their swift adoption into clinical routine, these treatments must demonstrate safety and efficacy as well as cost-effectiveness in randomized cardiovascular outcome trials.
Collapse
Affiliation(s)
- Daniel Tobias Michaeli
- Department of Medical Oncology, National Center for Tumour Diseases, Heidelberg University Hospital, Heidelberg, Germany.
| | - Julia Caroline Michaeli
- Department of Obstetrics and Gynaecology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Albers
- Department of Orthopaedics and Sport Orthopaedics, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Tobias Boch
- Department of Medical Oncology, National Center for Tumour Diseases, Heidelberg University Hospital, Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Division of Personalized Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Heidelberg University, Heidelberg, Germany
| | - Thomas Michaeli
- Department of Medical Oncology, National Center for Tumour Diseases, Heidelberg University Hospital, Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
- Division of Personalized Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Personalized Oncology, University Hospital Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
18
|
Abstract
Cholesterol is an essential lipid species of mammalian cells. Cells acquire it through synthesis in the endoplasmic reticulum (ER) and uptake from lipoprotein particles. Newly synthesized cholesterol is efficiently distributed from the ER to other organelles via lipid-binding/transfer proteins concentrated at membrane contact sites (MCSs) to reach the trans-Golgi network, endosomes, and plasma membrane. Lipoprotein-derived cholesterol is exported from the plasma membrane and endosomal compartments via a combination of vesicle/tubule-mediated membrane transport and transfer through MCSs. In this review, we provide an overview of intracellular cholesterol trafficking pathways, including cholesterol flux from the ER to other membranes, cholesterol uptake from lipoprotein donors and transport from the plasma membrane to the ER, cellular cholesterol efflux to lipoprotein acceptors, as well as lipoprotein cholesterol secretion from enterocytes, hepatocytes, and astrocytes. We also briefly discuss human diseases caused by defects in these processes and therapeutic strategies available in such conditions.
Collapse
Affiliation(s)
- Elina Ikonen
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00100 Helsinki, Finland
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
| |
Collapse
|
19
|
Munkhsaikhan U, Kwon YI, Sahyoun AM, Galán M, Gonzalez AA, Ait-Aissa K, Abidi AH, Kassan A, Kassan M. The Beneficial Effect of Lomitapide on the Cardiovascular System in LDLr -/- Mice with Obesity. Antioxidants (Basel) 2023; 12:1287. [PMID: 37372017 PMCID: PMC10295391 DOI: 10.3390/antiox12061287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/26/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
OBJECTIVES Homozygous familial hypercholesteremia (HoFH) is a rare, life-threatening metabolic disease, mainly caused by a mutation in the LDL receptor. If untreated, HoFH causes premature death from acute coronary syndrome. Lomitapide is approved by the FDA as a therapy to lower lipid levels in adult patients with HoFH. Nevertheless, the beneficial effect of lomitapide in HoFH models remains to be defined. In this study, we investigated the effect of lomitapide on cardiovascular function using LDL receptor-knockout mice (LDLr-/-). METHODS Six-week-old LDLr-/- mice were fed a standard diet (SD) or a high-fat diet (HFD) for 12 weeks. Lomitapide (1 mg/Kg/Day) was given by oral gavage for the last 2 weeks in the HFD group. Body weight and composition, lipid profile, blood glucose, and atherosclerotic plaques were measured. Vascular reactivity and markers for endothelial function were determined in conductance arteries (thoracic aorta) and resistance arteries (mesenteric resistance arteries (MRA)). Cytokine levels were measured by using the Mesoscale discovery V-Plex assays. RESULTS Body weight (47.5 ± 1.5 vs. 40.3 ± 1.8 g), % of fat mass (41.6 ± 1.9% vs. 31.8 ± 1.7%), blood glucose (215.5 ± 21.9 vs. 142.3 ± 7.7 mg/dL), and lipid levels (cholesterol: 600.9 ± 23.6 vs. 451.7 ± 33.4 mg/dL; LDL/VLDL: 250.6 ± 28.9 vs. 161.1 ± 12.24 mg/dL; TG: 299.5 ± 24.1 vs. 194.1 ± 28.1 mg/dL) were significantly decreased, and the % of lean mass (56.5 ± 1.8% vs. 65.2 ± 2.1%) was significantly increased in the HFD group after lomitapide treatment. The atherosclerotic plaque area also decreased in the thoracic aorta (7.9 ± 0.5% vs. 5.7 ± 0.1%). After treatment with lomitapide, the endothelium function of the thoracic aorta (47.7 ± 6.3% vs. 80.7 ± 3.1%) and mesenteric resistance artery (66.4 ± 4.3% vs. 79.5 ± 4.6%) was improved in the group of LDLr-/- mice on HFD. This was correlated with diminished vascular endoplasmic (ER) reticulum stress, oxidative stress, and inflammation. CONCLUSIONS Treatment with lomitapide improves cardiovascular function and lipid profile and reduces body weight and inflammatory markers in LDLr-/- mice on HFD.
Collapse
Affiliation(s)
- Undral Munkhsaikhan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Bioscience Research and General Dentistry, College of Dentistry, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Young In Kwon
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Amal M. Sahyoun
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Food Science and Agriculture Chemistry, McGill University, Montreal, QC H9X 3V9, Canada
| | - María Galán
- Faculty of Health Sciences, University Rey Juan Carlos, 28922 Alcorcón, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), ISCIII, 28029 Madrid, Spain
| | - Alexis A. Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso 300, Chile
| | - Karima Ait-Aissa
- College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA
| | - Ammaar H. Abidi
- Department of Bioscience Research and General Dentistry, College of Dentistry, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
- College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA
| | - Adam Kassan
- Department of Pharmaceutical Sciences, School of Pharmacy, West Coast University, Los Angeles, CA 91606, USA
| | - Modar Kassan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- College of Dental Medicine, Lincoln Memorial University, Knoxville, TN 37923, USA
| |
Collapse
|
20
|
Canepari C, Cantore A. Gene transfer and genome editing for familial hypercholesterolemia. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1140997. [PMID: 39086674 PMCID: PMC11285693 DOI: 10.3389/fmmed.2023.1140997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/10/2023] [Indexed: 08/02/2024]
Abstract
Familial hypercholesterolemia (FH) is an autosomal dominant inherited disease characterized by high circulating low-density lipoprotein (LDL) cholesterol. High circulating LDL cholesterol in FH is due to dysfunctional LDL receptors, and is mainly expressed by hepatocytes. Affected patients rapidly develop atherosclerosis, potentially leading to myocardial infarction and death within the third decade of life if left untreated. Here, we introduce the disease pathogenesis and available treatment options. We highlight different possible targets of therapeutic intervention. We then review different gene therapy strategies currently under development, which may become novel therapeutic options in the future, and discuss their advantages and disadvantages. Finally, we briefly outline the potential applications of some of these strategies for the more common acquired hypercholesterolemia disease.
Collapse
Affiliation(s)
- Cesare Canepari
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alessio Cantore
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
21
|
Florance I, Ramasubbu S. Current Understanding on the Role of Lipids in Macrophages and Associated Diseases. Int J Mol Sci 2022; 24:ijms24010589. [PMID: 36614031 PMCID: PMC9820199 DOI: 10.3390/ijms24010589] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 12/31/2022] Open
Abstract
Lipid metabolism is the major intracellular mechanism driving a variety of cellular functions such as energy storage, hormone regulation and cell division. Lipids, being a primary component of the cell membrane, play a pivotal role in the survival of macrophages. Lipids are crucial for a variety of macrophage functions including phagocytosis, energy balance and ageing. However, functions of lipids in macrophages vary based on the site the macrophages are residing at. Lipid-loaded macrophages have recently been emerging as a hallmark for several diseases. This review discusses the significance of lipids in adipose tissue macrophages, tumor-associated macrophages, microglia and peritoneal macrophages. Accumulation of macrophages with impaired lipid metabolism is often characteristically observed in several metabolic disorders. Stress signals differentially regulate lipid metabolism. While conditions such as hypoxia result in accumulation of lipids in macrophages, stress signals such as nutrient deprivation initiate lipolysis and clearance of lipids. Understanding the biology of lipid accumulation in macrophages requires the development of potentially active modulators of lipid metabolism.
Collapse
|
22
|
Zubielienė K, Valterytė G, Jonaitienė N, Žaliaduonytė D, Zabiela V. Familial Hypercholesterolemia and Its Current Diagnostics and Treatment Possibilities: A Literature Analysis. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1665. [PMID: 36422206 PMCID: PMC9692978 DOI: 10.3390/medicina58111665] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/30/2022] [Accepted: 11/13/2022] [Indexed: 09/30/2023]
Abstract
Familial hypercholesterolemia (FH) is a common, inherited disorder of cholesterol metabolism. This pathology is usually an autosomal dominant disorder and is caused by inherited mutations in the APOB, LDLR, and PCSK9 genes. Patients can have a homozygous or a heterozygous genotype, which determines the severity of the disease and the onset age of cardiovascular disease (CVD) manifestations. The incidence of heterozygous FH is 1: 200-250, whereas that of homozygous FH is 1: 100.000-160.000. Unfortunately, FH is often diagnosed too late and after the occurrence of a major coronary event. FH may be suspected in patients with elevated blood low-density lipoprotein cholesterol (LDL-C) levels. Moreover, there are other criteria that help to diagnose FH. For instance, the Dutch Lipid Clinical Criteria are a helpful diagnostic tool that is used to diagnose FH. FH often leads to the development of early cardiovascular disease and increases the risk of sudden cardiac death. Therefore, early diagnosis and treatment of this disease is very important. Statins, ezetimibe, bile acid sequestrants, niacin, PCSK9 inhibitors (evolocumab and alirocumab), small-interfering-RNA-based therapeutics (inclisiran), lomitapide, mipomersen, and LDL apheresis are several of the available treatment possibilities that lower LDL-C levels. It is important to say that the timeous lowering of LDL-C levels can reduce the risk of cardiovascular events and mortality in patients with FH. Therefore, it is essential to increase awareness of FH in order to reduce the burden of acute coronary syndrome (ACS).
Collapse
Affiliation(s)
- Kristina Zubielienė
- Department of Cardiology, Lithuanian University of Health Sciences Kaunas Clinics, LT-50161 Kaunas, Lithuania
- Department of Cardiology, Lithuanian University of Health Sciences Kaunas Hospital, LT-45130 Kaunas, Lithuania
- Kaunas Region Society of Cardiology, LT-44307, Kaunas, Lithuania
| | - Gintarė Valterytė
- Department of Cardiology, Lithuanian University of Health Sciences Kaunas Clinics, LT-50161 Kaunas, Lithuania
| | - Neda Jonaitienė
- Department of Cardiology, Lithuanian University of Health Sciences Kaunas Clinics, LT-50161 Kaunas, Lithuania
| | - Diana Žaliaduonytė
- Department of Cardiology, Lithuanian University of Health Sciences Kaunas Clinics, LT-50161 Kaunas, Lithuania
- Department of Cardiology, Lithuanian University of Health Sciences Kaunas Hospital, LT-45130 Kaunas, Lithuania
- Kaunas Region Society of Cardiology, LT-44307, Kaunas, Lithuania
| | - Vytautas Zabiela
- Department of Cardiology, Lithuanian University of Health Sciences Kaunas Clinics, LT-50161 Kaunas, Lithuania
- Kaunas Region Society of Cardiology, LT-44307, Kaunas, Lithuania
- Institute of Cardiology Kaunas, Cardiology Research Automation Laboratory, Lithuanian University of Health Sciences, LT-44307 Kaunas, Lithuania
| |
Collapse
|
23
|
Duan Y, Gong K, Xu S, Zhang F, Meng X, Han J. Regulation of cholesterol homeostasis in health and diseases: from mechanisms to targeted therapeutics. Signal Transduct Target Ther 2022; 7:265. [PMID: 35918332 PMCID: PMC9344793 DOI: 10.1038/s41392-022-01125-5] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 12/13/2022] Open
Abstract
Disturbed cholesterol homeostasis plays critical roles in the development of multiple diseases, such as cardiovascular diseases (CVD), neurodegenerative diseases and cancers, particularly the CVD in which the accumulation of lipids (mainly the cholesteryl esters) within macrophage/foam cells underneath the endothelial layer drives the formation of atherosclerotic lesions eventually. More and more studies have shown that lowering cholesterol level, especially low-density lipoprotein cholesterol level, protects cardiovascular system and prevents cardiovascular events effectively. Maintaining cholesterol homeostasis is determined by cholesterol biosynthesis, uptake, efflux, transport, storage, utilization, and/or excretion. All the processes should be precisely controlled by the multiple regulatory pathways. Based on the regulation of cholesterol homeostasis, many interventions have been developed to lower cholesterol by inhibiting cholesterol biosynthesis and uptake or enhancing cholesterol utilization and excretion. Herein, we summarize the historical review and research events, the current understandings of the molecular pathways playing key roles in regulating cholesterol homeostasis, and the cholesterol-lowering interventions in clinics or in preclinical studies as well as new cholesterol-lowering targets and their clinical advances. More importantly, we review and discuss the benefits of those interventions for the treatment of multiple diseases including atherosclerotic cardiovascular diseases, obesity, diabetes, nonalcoholic fatty liver disease, cancer, neurodegenerative diseases, osteoporosis and virus infection.
Collapse
Affiliation(s)
- Yajun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Suowen Xu
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Feng Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xianshe Meng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Jihong Han
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, College of Food and Biological Engineering, Hefei University of Technology, Hefei, China. .,College of Life Sciences, Key Laboratory of Bioactive Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China.
| |
Collapse
|
24
|
Lee B, Park SJ, Lee S, Lee J, Lee E, Yoo ES, Chung WS, Sohn JW, Oh BC, Kim S. Lomitapide, a cholesterol-lowering drug, is an anticancer agent that induces autophagic cell death via inhibiting mTOR. Cell Death Dis 2022; 13:603. [PMID: 35831271 PMCID: PMC9279289 DOI: 10.1038/s41419-022-05039-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 06/16/2022] [Accepted: 06/27/2022] [Indexed: 01/21/2023]
Abstract
Autophagy is a biological process that maintains cellular homeostasis and regulates the internal cellular environment. Hyperactivating autophagy to trigger cell death has been a suggested therapeutic strategy for cancer treatment. Mechanistic target of rapamycin (mTOR) is a crucial protein kinase that regulates autophagy; therefore, using a structure-based virtual screen analysis, we identified lomitapide, a cholesterol-lowering drug, as a potential mTOR complex 1 (mTORC1) inhibitor. Our results showed that lomitapide directly inhibits mTORC1 in vitro and induces autophagy-dependent cancer cell death by decreasing mTOR signaling, thereby inhibiting the downstream events associated with increased LC3 conversion in various cancer cells (e.g., HCT116 colorectal cancer cells) and tumor xenografts. Lomitapide also significantly suppresses the growth and viability along with elevated autophagy in patient-derived colorectal cancer organoids. Furthermore, a combination of lomitapide and immune checkpoint blocking antibodies synergistically inhibits tumor growth in murine MC38 or B16-F10 preclinical syngeneic tumor models. These results elucidate the direct, tumor-relevant immune-potentiating benefits of mTORC1 inhibition by lomitapide, which complement the current immune checkpoint blockade. This study highlights the potential repurposing of lomitapide as a new therapeutic option for cancer treatment.
Collapse
Affiliation(s)
- Boah Lee
- grid.37172.300000 0001 2292 0500Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141 Korea ,grid.37172.300000 0001 2292 0500Department of Biological Sciences, KAIST, Daejeon, 34141 Korea ,Present Address: ERSTEQ co., Ltd, Daejeon, 34013 Korea
| | - Seung Ju Park
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, KAIST, Daejeon, 34141 Korea ,Present Address: ERSTEQ co., Ltd, Daejeon, 34013 Korea
| | - Seulgi Lee
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, KAIST, Daejeon, 34141 Korea ,Present Address: ERSTEQ co., Ltd, Daejeon, 34013 Korea
| | - Jinwook Lee
- grid.256155.00000 0004 0647 2973Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, College of Medicine, Incheon, 21999 Korea
| | - Eunbeol Lee
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, KAIST, Daejeon, 34141 Korea
| | - Eun-Seon Yoo
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, KAIST, Daejeon, 34141 Korea
| | - Won-Suk Chung
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, KAIST, Daejeon, 34141 Korea
| | - Jong-Woo Sohn
- grid.37172.300000 0001 2292 0500Department of Biological Sciences, KAIST, Daejeon, 34141 Korea
| | - Byung-Chul Oh
- grid.256155.00000 0004 0647 2973Department of Physiology, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, College of Medicine, Incheon, 21999 Korea
| | - Seyun Kim
- grid.37172.300000 0001 2292 0500Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141 Korea ,grid.37172.300000 0001 2292 0500Department of Biological Sciences, KAIST, Daejeon, 34141 Korea ,grid.37172.300000 0001 2292 0500KAIST Institute for the BioCentury, KAIST, Daejeon, 34141 Korea ,grid.37172.300000 0001 2292 0500KAIST Stem Cell Center, KAIST, Daejeon, 34141 Korea
| |
Collapse
|
25
|
Keshavarz Alikhani H, Pourhamzeh M, Seydi H, Shokoohian B, Hossein-khannazer N, Jamshidi-adegani F, Al-Hashmi S, Hassan M, Vosough M. Regulatory Non-Coding RNAs in Familial Hypercholesterolemia, Theranostic Applications. Front Cell Dev Biol 2022; 10:894800. [PMID: 35813199 PMCID: PMC9260315 DOI: 10.3389/fcell.2022.894800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
Familial hypercholesterolemia (FH) is a common monogenic disease which is associated with high serum levels of low-density lipoprotein cholesterol (LDL-C) and leads to atherosclerosis and cardiovascular disease (CVD). Early diagnosis and effective treatment strategy can significantly improve prognosis. Recently, non-coding RNAs (ncRNAs) have emerged as novel biomarkers for the diagnosis and innovative targets for therapeutics. Non-coding RNAs have essential roles in the regulation of LDL-C homeostasis, suggesting that manipulation and regulating ncRNAs could be a promising theranostic approach to ameliorate clinical complications of FH, particularly cardiovascular disease. In this review, we briefly discussed the mechanisms and pathophysiology of FH and novel therapeutic strategies for the treatment of FH. Moreover, the theranostic effects of different non-coding RNAs for the treatment and diagnosis of FH were highlighted. Finally, the advantages and disadvantages of ncRNA-based therapies vs. conventional therapies were discussed.
Collapse
Affiliation(s)
- Hani Keshavarz Alikhani
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Pourhamzeh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Homeyra Seydi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Bahare Shokoohian
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nikoo Hossein-khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Jamshidi-adegani
- Laboratory for Stem Cell and Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Sulaiman Al-Hashmi
- Laboratory for Stem Cell and Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, Nizwa, Oman
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
- *Correspondence: Massoud Vosough,
| |
Collapse
|
26
|
Ferraro RA, Leucker T, Martin SS, Banach M, Jones SR, Toth PP. Contemporary Management of Dyslipidemia. Drugs 2022; 82:559-576. [PMID: 35303294 PMCID: PMC8931779 DOI: 10.1007/s40265-022-01691-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2022] [Indexed: 12/30/2022]
Abstract
The treatment of dyslipidemia continues to be a dynamic and controversial topic. Even the most appropriate therapeutic range for lipid levels-including that of triglycerides and low-density lipoprotein cholesterol-remain actively debated. Furthermore, with ever-increasing options and available treatment modalities, the management of dyslipidemia has progressed in both depth and complexity. An understanding of appropriate lipid-lowering therapy remains an essential topic of review for practitioners across medical specialties. The goal of this review is to provide an overview of recent research developments and recommendations for patients with dyslipidemia as a means of better informing the clinical practice of lipid management. By utilizing a guideline-directed approach, we provide a reference point on optimal lipid-lowering therapies across the spectrum of dyslipidemia. Special attention is paid to long-term adherence to lipid-lowering therapies, and the benefits derived from instituting appropriate medications in a structured manner alongside monitoring. Novel therapies and their impact on lipid lowering are discussed in detail, as well as potential avenues for research going forward. The prevention of cardiovascular disease remains paramount, and this review provides a roadmap for instituting appropriate therapies in cardiovascular disease prevention.
Collapse
Affiliation(s)
- Richard A Ferraro
- From the Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thorsten Leucker
- From the Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seth S Martin
- From the Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, Lodz, Poland
| | - Steven R Jones
- From the Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter P Toth
- From the Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- CGH Medical Center, 101 East Miller Road, Sterling, IL, 61081, USA.
| |
Collapse
|
27
|
Pirillo A, Catapano AL. Understanding efficacy and safety of lomitapide in homozygous familial hypercholesterolemia. Eur J Prev Cardiol 2022; 29:829-831. [PMID: 35148370 DOI: 10.1093/eurjpc/zwac028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Angela Pirillo
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, Milan, Italy.,IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Alberico Luigi Catapano
- IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy.,Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| |
Collapse
|
28
|
Bicciato G, Arnold M, Gebhardt A, Katan M. Precision medicine in secondary prevention of ischemic stroke: how may blood-based biomarkers help in clinical routine? An expert opinion. Curr Opin Neurol 2022; 35:45-54. [PMID: 34839341 DOI: 10.1097/wco.0000000000001011] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW One in eight patients unfortunately suffers a new stroke within 5 years of their first stroke, even today. Research in precision medicine could lead to a more individualized treatment allocation, possibly achieving lower recurrence rates of ischemic stroke. In this narrative review, we aim to discuss potential clinical implementation of several promising candidate blood biomarkers. RECENT FINDINGS We discuss specifically some promising blood-based biomarkers, which may improve the identification of underlying causes as well as risk stratification of patients according to their specific cerebrovascular risk factor pattern. SUMMARY Multimodal profiling of ischemic stroke patients by means of blood biomarkers, in addition to established clinical and neuroradiological data, may allow in the future a refinement of decision algorithms for treatment allocation in secondary ischemic stroke prevention.
Collapse
Affiliation(s)
- Giulio Bicciato
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | | | | | | |
Collapse
|
29
|
Vieira TF, Magalhães RP, Simões M, Sousa SF. Drug Repurposing Targeting Pseudomonas aeruginosa MvfR Using Docking, Virtual Screening, Molecular Dynamics, and Free-Energy Calculations. Antibiotics (Basel) 2022; 11:185. [PMID: 35203788 PMCID: PMC8868191 DOI: 10.3390/antibiotics11020185] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/10/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic Gram-negative bacterium responsible for acute and chronic infections in planktonic state or in biofilms. The sessile structures are known to confer physical stability, increase virulence, and work as a protective armor against antimicrobial compounds. P. aeruginosa can control the expression of genes, population density, and biofilm formation through a process called quorum sensing (QS), a rather complex and hierarchical system of communication. A recent strategy to try and overcome bacterial resistance is to target QS proteins. In this study, a combined multi-level computational approach was applied to find possible inhibitors against P. aeruginosa QS regulator protein MvfR, also known as PqsR, using a database of approved FDA drugs, as a repurposing strategy. Fifteen compounds were identified as highly promising putative MvfR inhibitors. On those 15 MvfR ligand complexes, molecular dynamic simulations and MM/GBSA free-energy calculations were performed to confirm the docking predictions and elucidate on the mode of interaction. Ultimately, the five compounds that presented better binding free energies of association than the reference molecules (a known antagonist, M64 and a natural inducer, 2-nonyl-4-hydroxyquinoline) were highlighted as very promising MvfR inhibitors.
Collapse
Affiliation(s)
- Tatiana F. Vieira
- UCIBIO/REQUIMTE, BioSIM, Departamento de Medicina, Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (T.F.V.); (R.P.M.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| | - Rita P. Magalhães
- UCIBIO/REQUIMTE, BioSIM, Departamento de Medicina, Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (T.F.V.); (R.P.M.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| | - Manuel Simões
- LEPABE Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal;
| | - Sérgio F. Sousa
- UCIBIO/REQUIMTE, BioSIM, Departamento de Medicina, Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (T.F.V.); (R.P.M.)
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| |
Collapse
|
30
|
Ziółkowska S, Kijek N, Zendran I, Szuster E, Barg E. Familial hypercholesterolemia - treatment update in children, systematic review. Pediatr Endocrinol Diabetes Metab 2022; 28:152-161. [PMID: 35848473 PMCID: PMC10214937 DOI: 10.5114/pedm.2022.116112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 02/26/2022] [Indexed: 06/07/2023]
Abstract
Familial hypercholesterolaemia is one of the most common genetic diseases, and its first symptoms occur in childhood. Proper diagnosis and treatment prevent young patients from severe consequences in their future. The treatment of this dyslipidaemia is still evolving, and new promising agents are being discovered. In this review we summarize the old and new treatment methods of familial hypercholesterolaemia, giving an update estimated on the latest publications.
Collapse
Affiliation(s)
| | | | - Iga Zendran
- Graduate of Wroclaw Medical University, Poland
| | - Ewa Szuster
- Graduate of Wroclaw Medical University, Poland
| | - Ewa Barg
- Department of Basic Medical Sciences, Wroclaw Medical University, Poland
| |
Collapse
|
31
|
Taha HSED, Kandil H, Farag N, Oraby A, Sharkawy ME, Fawzy F, Mahrous H, Bahgat J, Samy M, Aboul M, Abdrabou M, Shaker MM. Egyptian practical guidance in hypertriglyceridemia management 2021. Egypt Heart J 2021; 73:107. [PMID: 34928475 PMCID: PMC8688602 DOI: 10.1186/s43044-021-00235-9] [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: 08/24/2021] [Accepted: 12/14/2021] [Indexed: 11/10/2022] Open
Abstract
Hypertriglyceridemia (HTG) is a very common, yet underappreciated problem in clinical practice. Elevated triglyceride (TG) levels are independently associated with atherosclerotic cardiovascular disease (ASCVD) risk. Furthermore, severe HTG may lead to acute pancreatitis. Although LDL-guided statin therapy has improved ASCVD outcomes, residual risk remains. Recent trials have demonstrated that management of high TG levels, in patients already on statin therapy, reduces the rate of major vascular events. Few guidelines were issued, providing important recommendations for HTG management strategies. The goal of treatment is to reduce the risk of ASCVD and acute pancreatitis. The management stands on lifestyle modification, detection of secondary causes of HTG and pharmacological therapy, when indicated. In this guidance we review the causes and classification of HTG and summarize the current methods for risk estimation, diagnosis and treatment. The present guidance provides a focused update on the management of HTG, outlined in a simple user-friendly format, with an emphasis on the latest available data.
Collapse
Affiliation(s)
- Hesham Salah El Din Taha
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Hossam Kandil
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | | | | | | | - Fouad Fawzy
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Hossam Mahrous
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Juliette Bahgat
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Mina Samy
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Mohamed Aboul
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Mostafa Abdrabou
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| | - Mirna Mamdouh Shaker
- Department of Cardiology, Faculty of Medicine, Cairo University, 27 Nafezet Sheem El Shafae St Kasr Al Ainy, Cairo, 11562 Egypt
| |
Collapse
|
32
|
Apolipoprotein B and Cardiovascular Disease: Biomarker and Potential Therapeutic Target. Metabolites 2021; 11:metabo11100690. [PMID: 34677405 PMCID: PMC8540246 DOI: 10.3390/metabo11100690] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 12/19/2022] Open
Abstract
Apolipoprotein (apo) B, the critical structural protein of the atherogenic lipoproteins, has two major isoforms: apoB48 and apoB100. ApoB48 is found in chylomicrons and chylomicron remnants with one apoB48 molecule per chylomicron particle. Similarly, a single apoB100 molecule is contained per particle of very-low-density lipoprotein (VLDL), intermediate density lipoprotein, LDL and lipoprotein(a). This unique one apoB per particle ratio makes plasma apoB concentration a direct measure of the number of circulating atherogenic lipoproteins. ApoB levels indicate the atherogenic particle concentration independent of the particle cholesterol content, which is variable. While LDL, the major cholesterol-carrying serum lipoprotein, is the primary therapeutic target for management and prevention of atherosclerotic cardiovascular disease, there is strong evidence that apoB is a more accurate indicator of cardiovascular risk than either total cholesterol or LDL cholesterol. This review examines multiple aspects of apoB structure and function, with a focus on the controversy over use of apoB as a therapeutic target in clinical practice. Ongoing coronary artery disease residual risk, despite lipid-lowering treatment, has left patients and clinicians with unsatisfactory options for monitoring cardiovascular health. At the present time, the substitution of apoB for LDL-C in cardiovascular disease prevention guidelines has been deemed unjustified, but discussions continue.
Collapse
|
33
|
D'Erasmo L, Gallo A, Cefalù AB, Di Costanzo A, Saheb S, Giammanco A, Averna M, Buonaiuto A, Iannuzzo G, Fortunato G, Puja A, Montalcini T, Pavanello C, Calabresi L, Vigna GB, Bucci M, Bonomo K, Nota F, Sampietro T, Sbrana F, Suppressa P, Sabbà C, Fimiani F, Cesaro A, Calabrò P, Palmisano S, D'Addato S, Pisciotta L, Bertolini S, Bittar R, Kalmykova O, Béliard S, Carrié A, Arca M, Bruckert E. Long-term efficacy of lipoprotein apheresis and lomitapide in the treatment of homozygous familial hypercholesterolemia (HoFH): a cross-national retrospective survey. Orphanet J Rare Dis 2021; 16:381. [PMID: 34496902 PMCID: PMC8427960 DOI: 10.1186/s13023-021-01999-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 08/24/2021] [Indexed: 01/23/2023] Open
Abstract
Background Homozygous familial hypercholesterolemia (HoFH) is a rare life-threatening condition that represents a therapeutic challenge. The vast majority of HoFH patients fail to achieve LDL-C targets when treated with the standard protocol, which associates maximally tolerated dose of lipid-lowering medications with lipoprotein apheresis (LA). Lomitapide is an emerging therapy in HoFH, but its place in the treatment algorithm is disputed because a comparison of its long-term efficacy versus LA in reducing LDL-C burden is not available. We assessed changes in long-term LDL-C burden and goals achievement in two independent HoFH patients’ cohorts, one treated with lomitapide in Italy (n = 30) and the other with LA in France (n = 29). Results The two cohorts differed significantly for genotype (p = 0.004), baseline lipid profile (p < 0.001), age of treatment initiation (p < 0.001), occurrence of cardiovascular disease (p = 0.003) as well as follow-up duration (p < 0.001). The adjunct of lomitapide to conventional lipid-lowering therapies determined an additional 58.0% reduction of last visit LDL-C levels, compared to 37.1% when LA was added (padj = 0.004).
Yearly on-treatment LDL-C < 70 mg/dl and < 55 mg/dl goals were only achieved in 45.5% and 13.5% of HoFH patients treated with lomitapide. The long-term exposure to LDL-C burden was found to be higher in LA than in Lomitapide cohort (13,236.1 ± 5492.1 vs. 11,656.6 ± 4730.9 mg/dL-year respectively, padj = 0.002). A trend towards fewer total cardiovascular events was observed in the Lomitapide than in the LA cohort. Conclusions In comparison with LA, lomitapide appears to provide a better control of LDL-C in HoFH. Further studies are needed to confirm this data and establish whether this translates into a reduction of cardiovascular risk. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-01999-8.
Collapse
Affiliation(s)
- Laura D'Erasmo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, Italy. .,Department of Endocrinology and Cardiovascular Disease Prevention, Assistance Publique-Hôpitaux de Paris, La Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France.
| | - Antonio Gallo
- Department of Endocrinology and Cardiovascular Disease Prevention, Assistance Publique-Hôpitaux de Paris, La Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France.,Sorbonne Université, UPMC Univ Paris 06, INSERM 1146, - CNRS 7371, Laboratoire d'imagerie Biomédicale, Paris, France
| | - Angelo Baldassare Cefalù
- Dipartimento di Promozione Della Salute, Materno Infantile, Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro" (PROMISE), Università Degli Studi Di Palermo, Palermo, Italy
| | - Alessia Di Costanzo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, Italy
| | - Samir Saheb
- Department of Endocrinology and Cardiovascular Disease Prevention, Assistance Publique-Hôpitaux de Paris, La Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Antonina Giammanco
- Dipartimento di Promozione Della Salute, Materno Infantile, Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro" (PROMISE), Università Degli Studi Di Palermo, Palermo, Italy
| | - Maurizio Averna
- Dipartimento di Promozione Della Salute, Materno Infantile, Medicina Interna e Specialistica di Eccellenza "G. D'Alessandro" (PROMISE), Università Degli Studi Di Palermo, Palermo, Italy
| | - Alessio Buonaiuto
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Giuliana Fortunato
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy.,CEINGE, Advanced Biotechnology, Naples, Italy
| | - Arturo Puja
- Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Tiziana Montalcini
- Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Chiara Pavanello
- Centro Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milan, Italy
| | - Laura Calabresi
- Centro Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università Degli Studi di Milano, Milan, Italy
| | | | - Marco Bucci
- Dipartimento di Medicina e Scienze Dell'Invecchiamento, Università Degli Studi "G. d'annunzio" di Chieti, Pescara, Italy
| | - Katia Bonomo
- Metabolic Disease and Diabetes Unit, AOU San Luigi Gonzaga, Orbassano', Turin, Italy
| | - Fabio Nota
- Metabolic Disease and Diabetes Unit, AOU San Luigi Gonzaga, Orbassano', Turin, Italy
| | - Tiziana Sampietro
- Lipoapheresis Unit-Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana "Gabriele Monasterio", Via Moruzzi 1, Pisa, Italy
| | - Francesco Sbrana
- Lipoapheresis Unit-Reference Center for Diagnosis and Treatment of Inherited Dyslipidemias, Fondazione Toscana "Gabriele Monasterio", Via Moruzzi 1, Pisa, Italy
| | - Patrizia Suppressa
- Department of Internal Medicine and Rare Disease Centre "C.Frugoni", University Hospital of Bari "A. Moro", Piazza G. Cesare 11, Bari, Italy
| | - Carlo Sabbà
- Department of Internal Medicine and Rare Disease Centre "C.Frugoni", University Hospital of Bari "A. Moro", Piazza G. Cesare 11, Bari, Italy
| | - Fabio Fimiani
- Division of Clinical Cardiology, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", A.O.R.N. Sant' Anna e San Sebastiano, 81100, Caserta, Italy
| | - Arturo Cesaro
- Division of Clinical Cardiology, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", A.O.R.N. Sant' Anna e San Sebastiano, 81100, Caserta, Italy
| | - Paolo Calabrò
- Division of Clinical Cardiology, Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli", A.O.R.N. Sant' Anna e San Sebastiano, 81100, Caserta, Italy
| | - Silvia Palmisano
- Hypertension and Atherosclerosis Research Group, Medical and Surgical Sciences Department, Sant'Orsola-Malpighi University Hospital, Via Albertoni 15, 40138, Bologna, Italy
| | - Sergio D'Addato
- Hypertension and Atherosclerosis Research Group, Medical and Surgical Sciences Department, Sant'Orsola-Malpighi University Hospital, Via Albertoni 15, 40138, Bologna, Italy
| | - Livia Pisciotta
- Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS-Polyclinic Hospital San Martino, Genoa, Italy
| | - Stefano Bertolini
- Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS-Polyclinic Hospital San Martino, Genoa, Italy
| | - Randa Bittar
- Inserm, Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, Department of Metabolic Biochemistry, Assistance Publique, Hôpitaux de Paris, Hôpital de La Pitié-Salpêtrière, Sorbonne University, Paris, France
| | - Olga Kalmykova
- Department of Endocrinology and Cardiovascular Disease Prevention, Assistance Publique-Hôpitaux de Paris, La Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Sophie Béliard
- Aix Marseille University, INSERM, INRA, C2VN, Marseille, France.,Department of Nutrition, Metabolic Diseases, Endocrinology, La Conception Hospital, Marseille, France
| | - Alain Carrié
- Inserm, Institute of Cardiometabolism and Nutrition (ICAN), UMR_S1166, APHP, Department of Biochemistry, Obesity and Dyslipidemia Genetics Unit, Hôpital de La Pitié, Sorbonne University, Paris, France
| | - Marcello Arca
- Department of Translational and Precision Medicine, Sapienza University of Rome, Viale del Policlinico 155, Rome, Italy
| | - Eric Bruckert
- Department of Endocrinology and Cardiovascular Disease Prevention, Assistance Publique-Hôpitaux de Paris, La Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| |
Collapse
|
34
|
Han J, Kiss L, Mei H, Remete AM, Ponikvar-Svet M, Sedgwick DM, Roman R, Fustero S, Moriwaki H, Soloshonok VA. Chemical Aspects of Human and Environmental Overload with Fluorine. Chem Rev 2021; 121:4678-4742. [PMID: 33723999 PMCID: PMC8945431 DOI: 10.1021/acs.chemrev.0c01263] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Indexed: 12/24/2022]
Abstract
Over the last 100-120 years, due to the ever-increasing importance of fluorine-containing compounds in modern technology and daily life, the explosive development of the fluorochemical industry led to an enormous increase of emission of fluoride ions into the biosphere. This made it more and more important to understand the biological activities, metabolism, degradation, and possible environmental hazards of such substances. This comprehensive and critical review focuses on the effects of fluoride ions and organofluorine compounds (mainly pharmaceuticals and agrochemicals) on human health and the environment. To give a better overview, various connected topics are also discussed: reasons and trends of the advance of fluorine-containing pharmaceuticals and agrochemicals, metabolism of fluorinated drugs, withdrawn fluorinated drugs, natural sources of organic and inorganic fluorine compounds in the environment (including the biosphere), sources of fluoride intake, and finally biomarkers of fluoride exposure.
Collapse
Affiliation(s)
- Jianlin Han
- Jiangsu
Co-Innovation Center of Efficient Processing and Utilization of Forest
Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Loránd Kiss
- University
of Szeged, Institute of Pharmaceutical Chemistry
and Interdisciplinary Excellence Centre, Eötvös u. 6, 6720 Szeged, Hungary
| | - Haibo Mei
- Jiangsu
Co-Innovation Center of Efficient Processing and Utilization of Forest
Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Attila Márió Remete
- University
of Szeged, Institute of Pharmaceutical Chemistry
and Interdisciplinary Excellence Centre, Eötvös u. 6, 6720 Szeged, Hungary
| | - Maja Ponikvar-Svet
- Department
of Inorganic Chemistry and Technology, Jožef
Stefan Institute, Jamova
cesta 39, 1000 Ljubljana, Slovenia
| | - Daniel Mark Sedgwick
- Departamento
de Química Orgánica, Universidad
de Valencia, 46100 Burjassot, Valencia Spain
| | - Raquel Roman
- Departamento
de Química Orgánica, Universidad
de Valencia, 46100 Burjassot, Valencia Spain
| | - Santos Fustero
- Departamento
de Química Orgánica, Universidad
de Valencia, 46100 Burjassot, Valencia Spain
| | - Hiroki Moriwaki
- Hamari
Chemicals Ltd., 1-19-40, Nankokita, Suminoe-ku, Osaka 559-0034, Japan
| | - Vadim A. Soloshonok
- Department
of Organic Chemistry I, Faculty of Chemistry, University of the Basque Country UPV/EHU, 20018 San Sebastian, Spain
- IKERBASQUE,
Basque Foundation for Science, 48011 Bilbao, Spain
| |
Collapse
|
35
|
Patel NM, Stottlemyer BA, Gray MP, Boyce RD, Kane-Gill SL. A Pharmacovigilance Study of Adverse Drug Reactions Reported for Cardiovascular Disease Medications Approved Between 2012 and 2017 in the United States Food and Drug Administration Adverse Event Reporting System (FAERS) Database. Cardiovasc Drugs Ther 2021; 36:309-322. [PMID: 33599896 DOI: 10.1007/s10557-021-07157-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE Between 2012 and 2017, the FDA approved 29 therapies for a cardiovascular disease (CVD) indication. Due to the limited literature on patient safety outcomes for recently approved CVD medications, this study investigated adverse drug reports (ADRs) reported in the FDA Adverse Event Reporting System (FAERS). METHODS A disproportionality analysis of spontaneously reported ADR was conducted. Reports in FAERS from Quarter 1, 2012, through Quarter 1, 2019, were compiled, allowing a 2-year buffer following drug approval in 2017. Top 10 reported ADRs and reporting odds ratios (ROR; confidence interval (CI)), a measure of disproportionality, were analyzed and compared to drugs available prior to 2012 as appropriate. RESULTS Of 7,952,147 ADR reports, 95,016 (1.19%) consisted of reports for newly approved CVD medications. For oral anticoagulants, apixaban had significantly lower reports for anemia and renal failure compared to dabigatran and rivaroxaban but greater reports for neurological signs/symptoms, and arrhythmias. Evaluating heart failure drugs, sacubitril/valsartan had greater reports for acute kidney injury, coughing, potassium imbalances, and renal impairment but notably, lower for angioedema compared to lisinopril. Assessing familial hypercholesterolemia drugs, alirocumab had greater reports for joint-related-signs/symptoms compared to other agents in this category. A newer pulmonary arterial hypertension treatment, selexipag, had greater reports of reporting for bone/joint-related-signs/symptoms but riociguat had greater reports for hemorrhages and vascular hypotension. CONCLUSION Pharmacovigilance studies allow an essential opportunity to evaluate the safety profile of CVD medications in clinical practice. Additional research is needed to evaluate these reported safety concerns for recently approved CVD medications.
Collapse
Affiliation(s)
- Niti M Patel
- School of Pharmacy, University of Pittsburgh, 3507 Terrace St., Pittsburgh, PA, 15261, USA
| | - Britney A Stottlemyer
- School of Pharmacy, University of Pittsburgh, 3507 Terrace St., Pittsburgh, PA, 15261, USA
| | - Matthew P Gray
- School of Pharmacy, University of Pittsburgh, 3507 Terrace St., Pittsburgh, PA, 15261, USA
| | - Richard D Boyce
- School of Pharmacy, University of Pittsburgh, 3507 Terrace St., Pittsburgh, PA, 15261, USA.,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sandra L Kane-Gill
- School of Pharmacy, University of Pittsburgh, 3507 Terrace St., Pittsburgh, PA, 15261, USA.
| |
Collapse
|
36
|
Sunil B, Foster C, Wilson DP, Ashraf AP. Novel therapeutic targets and agents for pediatric dyslipidemia. Ther Adv Endocrinol Metab 2021; 12:20420188211058323. [PMID: 34868544 PMCID: PMC8637781 DOI: 10.1177/20420188211058323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 10/19/2021] [Indexed: 02/04/2023] Open
Abstract
Landmark studies have convincingly demonstrated that atherosclerosis begins in youth. While generally asymptomatic, an increasing number of youth with disorders of lipid and lipoprotein metabolism, such as familial hypercholesterolemia, are being identified through selective and universal screening. While a heart healthy lifestyle is the foundation of treatment for all youth with dyslipidemia, lipid-lowering therapy may be required by some to prevent morbidity and premature mortality, especially when initiated at a young age. When appropriate, use of statins has become standard of care for reducing low-density lipoprotein cholesterol, while fibrates may be beneficial in helping to lower triglycerides. Many therapeutic options commonly used in adults are not yet approved for use in youth less than 18 years of age. Although currently available lipid-lowering therapy is well tolerated and safe when administered to youth, response to treatment may vary and some conditions lack an efficient therapeutic option. Thus, newer agents are needed to aid in management. Many are in development and clinical trials in youth are currently in progress but will require FDA approval before becoming commercially available. Many utilize novel approaches to favorably alter lipid and lipoprotein metabolism. In the absence of long-term outcome data of youth who were treated beginning at an early age, clinical registries may prove to be useful in monitoring safety and efficacy and help to inform clinical decision-making. In this manuscript, we review currently available and novel therapeutic agents in development for the treatment of elevated cholesterol and triglycerides.
Collapse
Affiliation(s)
- Bhuvana Sunil
- Division of Pediatric Endocrinology &
Diabetes, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Christy Foster
- Division of Pediatric Endocrinology &
Diabetes, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Don P. Wilson
- Cardiovascular Health and Risk Prevention,
Pediatric Endocrinology and Diabetes, Cook Children’s Medical Center, Fort
Worth, TX, USA
| | | |
Collapse
|
37
|
Velvet AJJ, Soran H, Clarke B, Motwani M, Ordoubadi FF, Daniels MJ. Homozygous familial hypercholesterolemia with an update on cholesterol management. Oxf Med Case Reports 2020; 2020:omaa072. [PMID: 32995028 PMCID: PMC7507883 DOI: 10.1093/omcr/omaa072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/02/2020] [Accepted: 07/09/2020] [Indexed: 11/12/2022] Open
Abstract
Familial hypercholesterolemia (FH) is an autosomal dominant condition that increases the risk of premature cardiovascular disease. Despite advances in treatment, it remains under detected and under treated. As an inherited condition, it poses a risk to the patient and family members. Most cases are due to defective low-density lipoprotein receptor (LDLR) activity. Heterozygous mutations are common (1:250-1:300). Homozygous FH is very rare (2-3 in a million), with higher circulating cholesterol levels and a poorer cardiovascular prognosis. We present the management of a case of homozygous hypercholesterolemia due to homozygous LDLR mutation. The patient subsequently developed severe coronary artery and aortic valve disease despite aggressive lipid-lowering therapy. We review advanced lipid management options that include lipoprotein apheresis, Proprotein Convertase Subtilisin/Kexin type 9 inhibition, and the microsomal triglyceride transfer protein inhibitor lomitapide.
Collapse
Affiliation(s)
- Anju J J Velvet
- Manchester Heart Centre, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| | - Handrean Soran
- Department of Endocrinology, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| | - Bernard Clarke
- Manchester Heart Centre, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| | - Manish Motwani
- Manchester Heart Centre, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| | - Farzin F Ordoubadi
- Manchester Heart Centre, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK
| | - Matthew J Daniels
- Manchester Heart Centre, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, UK.,Division of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK.,Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester, Manchester, UK
| |
Collapse
|
38
|
Low cholesterol syndrome and drug development. Curr Opin Cardiol 2020; 35:423-427. [PMID: 32452920 DOI: 10.1097/hco.0000000000000745] [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: 11/25/2022]
Abstract
PURPOSE OF REVIEW Low cholesterol syndromes were considered curiosities. The present article reviews some hypolipidaemic disorders and the drugs developed from the insights they provided. RECENT FINDINGS Abetalipopoproteinaemia and hypobetalipoproteinaemia are associated with low cholesterol concentrations and caused by mutations in apolipoprotein (apo) B or microsomal transfer protein. This led to the development of mipomersen and lomitapide which are used to treat homozygous familial hypercholesterolaemia. Mutations in proprotein convertase subtilisin kexin-9 (PCSK9) can cause either high or low cholesterol. Loss of function PCSK9 mutations prompted the development of antibody therapies to PCSK9 which are now widely used to treat hypercholesterolaemia. Mutations in apolipoprotein C-3 and angiopoietin-like protein 3 (ANGPTL3) cause hypolipoproteinaemia and reduced triglycerides. Antisense therapies to apolipoprotein C-3 and antibodies to ANGPTL3 are in development to treat familial chylomicronaemia syndrome. Activating mutations in apoA-1 result in hyper-functioning high-density lipoprotein (HDL) and suggest that modifying HDL turnover may reduce cardiovascular disease (CVD) risk. SUMMARY Orphan lipid disorders have provided insights into mechanisms involved in lowering cholesterol levels and the potential safety and efficacy of interventional processes. They have been not only enabled development of drugs to treat rare lipid disorders but also those finding wider use in general lowering of CVD risk.
Collapse
|
39
|
Barata‐Vallejo S, Postigo A. New Visible‐Light‐Triggered Photocatalytic Trifluoromethylation Reactions of Carbon–Carbon Multiple Bonds and (Hetero)Aromatic Compounds. Chemistry 2020; 26:11065-11084. [DOI: 10.1002/chem.202000856] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/14/2020] [Indexed: 01/10/2023]
Affiliation(s)
- Sebastian Barata‐Vallejo
- Department of Organic ChemistryUniversidad de Buenos Aires, Facultad de Farmacia y Bioquímica Junin 954 CP 1113 Buenos Aires Argentina
- ISOFConsiglio Nazionale delle Ricerche Via P. Gobetti 101 40129 Bologna Italy
| | - Al Postigo
- Department of Organic ChemistryUniversidad de Buenos Aires, Facultad de Farmacia y Bioquímica Junin 954 CP 1113 Buenos Aires Argentina
| |
Collapse
|
40
|
Esan O, Wierzbicki AS. Volanesorsen in the Treatment of Familial Chylomicronemia Syndrome or Hypertriglyceridaemia: Design, Development and Place in Therapy. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2623-2636. [PMID: 32753844 PMCID: PMC7351689 DOI: 10.2147/dddt.s224771] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/18/2020] [Indexed: 02/04/2023]
Abstract
Severe hypertriglyceridaemia is associated with pancreatitis and chronic pancreatitis-induced diabetes. Familial chylomicronaemia syndrome (FCS) is a rare autosomal recessive disorder of lipid metabolism characterised by high levels of triglycerides (TGs) due to failure of chylomicron clearance. It causes repeated episodes of severe abdominal pain, fatigue and attacks of acute pancreatitis. There are few current options for its long-term management. The only universal long-term therapy is restriction of total dietary fat intake to <10-15% of daily calories (15 to 20g per day). Many patients have been treated with fibrates and statins with a variable response, but many remain susceptible to pancreatitis. Other genetic syndromes associated with hypertriglyceridaemia include familial partial lipodystrophy (FPLD). Targeting apolipoprotein C3 (apoC3) offers the ability to increase clearance of chylomicrons and other triglyceride-rich lipoproteins. Volanesorsen is an antisense oligonucleotide (ASO) inhibitor of apoC3, which reduces TG levels by 70–80% which has been shown also to reduce rates of pancreatitis and improve well-being in FCS and reduce TGs and improve insulin resistance in FPLD. It is now undergoing licensing and payer reviews. Further developments of antisense technology including small interfering RNA therapy to apoC3 as well as other approaches to modulating triglycerides are in development for this rare disorder.
Collapse
Affiliation(s)
- Oluwayemisi Esan
- Department of Metabolic Medicine/Chemical Pathology, Guy's & St Thomas' Hospitals, London SE1 7EH, UK
| | - Anthony S Wierzbicki
- Department of Metabolic Medicine/Chemical Pathology, Guy's & St Thomas' Hospitals, London SE1 7EH, UK
| |
Collapse
|
41
|
Stefanutti C. Lomitapide-a Microsomal Triglyceride Transfer Protein Inhibitor for Homozygous Familial Hypercholesterolemia. Curr Atheroscler Rep 2020; 22:38. [PMID: 32557261 PMCID: PMC7303073 DOI: 10.1007/s11883-020-00858-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Homozygous familial hypercholesterolemia (HoFH) is a rare, genetic condition characterized by high levels of Low density lipoprotein cholesterol (LDL-C); overt, early-onset atherosclerotic cardiovascular disease (ASCVD); and premature cardiovascular events and mortality. Lomitapide is a first-in-class microsomal triglyceride transfer protein inhibitor for the treatment of HoFH. This review provides an update on data emerging from real-world studies of lomitapide following on from its pivotal phase 3 clinical trial in HoFH. RECENT FINDINGS Recent registry data have confirmed that HoFH is characterized by delayed diagnosis, with many patients not receiving effective therapy until they are approaching the age when major adverse cardiovascular events may occur. Data from case series of varying sizes, and from a 163-patient registry of HoFH patients receiving lomitapide, have demonstrated that lomitapide doses are lower and adverse events less severe than in the phase 3 study. Lomitapide enables many patients to reach European Atherosclerosis Society LDL-C targets. Some patients are able to reduce frequency of lipoprotein apheresis or, in some cases, stop the procedure altogether-unless there is significant elevation of lipoprotein (a). Modelling analyses based on historical and clinical trial data indicate that lomitapide has the potential to improve cardiovascular outcomes and survival in HoFH. Real-world clinical experience with lomitapide has shown the drug to be effective with manageable, less marked adverse events than in formal clinical studies. Event modelling data suggest a survival benefit with lomitapide in HoFH.
Collapse
Affiliation(s)
- Claudia Stefanutti
- Extracorporeal Therapeutic Techniques Unit, Lipid Clinic and Atherosclerosis Prevention Centre, Regional Centre (Lazio) for Rare Diseases, Immunohematology and Transfusion Medicine, Department of Molecular Medicine, "Sapienza" University of Rome, "Umberto I" Hospital, Rome, Italy.
| |
Collapse
|
42
|
Gao M, Yu W, Hu H, Liu H, Fan K, Gu C, Wang L, Yu Y. Case Report: Cardiac Surgery and Combined Lipid-Lowering Drug Therapy for Homozygous Familial Hypercholesterolemia. Front Pediatr 2020; 8:535949. [PMID: 33194883 PMCID: PMC7642436 DOI: 10.3389/fped.2020.535949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 09/14/2020] [Indexed: 11/13/2022] Open
Abstract
Homozygous familial hypercholesterolemia (HoFH) is a rare, autosomal dominant, hereditary, metabolic disease. HoFH patients exhibit severe coronary stenosis and valvular disease, which may result in sudden death, even during adolescence. The challenges faced during surgery and the poor curative effect of conventional lipid-lowering therapy create a treatment bottleneck. We report a rare case of HoFH in a 12-years-old boy with acute myocardial infarction, severe mitral insufficiency, and moderate aortic insufficiency. Coronary artery bypass grafting and valvuloplasty resulted in improved heart function. Postoperative combined lipid-lowering drug therapy was able to reduce low-density lipoprotein cholesterol level from 15.37 mm/L to 6.41 mmol/L. Thus, the combination of medical and surgical treatment was considered effective and can be used to inform treatment guidelines for HoFH with severe complications.
Collapse
Affiliation(s)
- Mingxin Gao
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wenyuan Yu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Hui Hu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Hongli Liu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Kangjun Fan
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chengxiong Gu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Lvya Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yang Yu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|