1
|
Suhr OB, Grogan M, Silva AMD, Karam C, Garcia-Pavia P, Drachman B, Zago W, Tripuraneni R, Kinney GG. PRX004 in variant amyloid transthyretin (ATTRv) amyloidosis: results of a phase 1, open-label, dose-escalation study. Amyloid 2025; 32:14-21. [PMID: 39472768 DOI: 10.1080/13506129.2024.2420809] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 02/14/2025]
Abstract
BACKGROUND The investigational monoclonal antibody PRX004 is designed to specifically target and deplete TTR amyloid. Here, we report on the safety, tolerability, pharmacokinetics, pharmacodynamics and preliminary clinical activity of PRX004 in patients with ATTRv amyloidosis. METHODS This global, multicentre, phase 1 trial comprised a 3 + 3 dose-escalation phase and a long-term extension (LTE) phase (NCT03336580). In the dose-escalation phase, patients received PRX004 (0.1, 0.3, 1, 3, 10 or 30 mg/kg), administered intravenously every 28 days for 3 months. In the LTE, eligible patients could receive up to 15 additional doses. Patients who received doses of ≥3 mg/kg for ≥9 months were assessed for Global Longitudinal Strain (GLS) and Neuropathy Impairment Score (NIS). The primary objective was to determine the maximum tolerated dose (MTD) of PRX004. RESULTS Overall, 21 patients with ATTRv amyloidosis completed the dose-escalation phase; 17 subsequently enrolled in the LTE. The MTD was not reached. PRX004 was well tolerated at all doses, with dose-proportional exposure. GLS and NIS were improved or maintained over 9 months (n = 7). CONCLUSIONS PRX004 was well tolerated in patients with ATTRv amyloidosis and demonstrated potential clinical activity. A phase 2 randomised controlled trial in ATTR cardiomyopathy is ongoing (NCT05442047).
Collapse
MESH Headings
- Humans
- Male
- Female
- Amyloid Neuropathies, Familial/drug therapy
- Amyloid Neuropathies, Familial/genetics
- Amyloid Neuropathies, Familial/pathology
- Middle Aged
- Aged
- Maximum Tolerated Dose
- Prealbumin/genetics
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/therapeutic use
- Dose-Response Relationship, Drug
- Aged, 80 and over
- Adult
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacokinetics
Collapse
Affiliation(s)
- Ole B Suhr
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | | | - Ana Martins da Silva
- Unidade Corino de Andrade, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Chafic Karam
- University of Pennsylvania, Philadelphia, PA, USA
| | - Pablo Garcia-Pavia
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Hospital Universitario Puerta de Hierro Majadahonda, IDIPHISA, CIBERCV, Madrid, Spain
| | | | | | | | | |
Collapse
|
2
|
Tuvali O, Fassler M, Goland S, Benaim C, Shimoni S, George J. A functional role for spontaneously occurring natural anti-transthyretin antibodies from patients with transthyretin cardiac amyloidosis. Eur J Heart Fail 2024. [PMID: 39563100 DOI: 10.1002/ejhf.3527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 10/24/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Affiliation(s)
- Ortal Tuvali
- Heart Center, Kaplan Medical Center, Rehovot, Affiliated to the Hebrew University, Jerusalem, Israel
| | - Michael Fassler
- Heart Center, Kaplan Medical Center, Rehovot, Affiliated to the Hebrew University, Jerusalem, Israel
| | - Sorel Goland
- Heart Center, Kaplan Medical Center, Rehovot, Affiliated to the Hebrew University, Jerusalem, Israel
| | - Clara Benaim
- Heart Center, Kaplan Medical Center, Rehovot, Affiliated to the Hebrew University, Jerusalem, Israel
| | - Sara Shimoni
- Heart Center, Kaplan Medical Center, Rehovot, Affiliated to the Hebrew University, Jerusalem, Israel
| | - Jacob George
- Heart Center, Kaplan Medical Center, Rehovot, Affiliated to the Hebrew University, Jerusalem, Israel
| |
Collapse
|
3
|
Hancock TJ, Vlasyuk M, Foster JS, Macy S, Wooliver DC, Balachandran M, Williams AD, Martin EB, Kennel SJ, Heidel ER, Wall JS, Jackson JW. Neutrophils enhance the clearance of systemic amyloid deposits in a murine amyloidoma model. Front Immunol 2024; 15:1487250. [PMID: 39600710 PMCID: PMC11588727 DOI: 10.3389/fimmu.2024.1487250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction Amyloid-specific antibodies have been shown to opsonize and enhance amyloid clearance in systemic amyloidosis mouse models. However, the immunological mechanisms by which amyloid is removed have not been clearly defined. Previous reports from preclinical in vivo studies suggest polymorphonuclear cells (i.e., neutrophils) can affect amyloid removal. Therefore, we sought to analyze how neutrophils may contribute to the clearance of human AL amyloid extracts, using a murine amyloidoma model. Methods Immunocompromised nude mice injected subcutaneously with patient-derived AL amyloid extract (generating a localized "amyloidoma") were used to circumvent confounding factors contributed by the adaptive immune system and served as the model system. Two representative AL amyloid extracts were used, ALλ(CLA), which is refractory to clearance, and ALκ(TAL), which is readily cleared in mice. Neutrophil recruitment to the amyloid masses, cellular activation, and propensity to engulf amyloid were assessed. Results Immunophenotyping of amyloidomas from animals implanted with 2 mg of either ALλ or ALκ revealed that more neutrophils were recruited to ALκ amyloid masses as compared to the ALλ material, which was generally devoid of neutrophils. Ex vivo analyses indicated neutrophils do not efficiently phagocytose amyloid directly. However, histological evaluation of the ALκ amyloidoma revealed the abundant presence of neutrophil extracellular traps, which were absent in the ALλ amyloidomas. Using neutrophil depletion experiments in mice, we determined that mice devoid of neutrophils cleared the human amyloid lesions less efficiently. Moreover, mice devoid of neutrophils also had significantly reduced intra-amyloid expression of inflammatory cytokines. Discussion Neutrophils may not directly mediate amyloid clearance through phagocytosis; however, these cells can be stimulated by the amyloid and may function to facilitate phagocytosis and amyloid clearance by professional phagocytes (e.g., macrophages).
Collapse
Affiliation(s)
- Trevor J. Hancock
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Marina Vlasyuk
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - James S. Foster
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Sallie Macy
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Daniel C. Wooliver
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Manasi Balachandran
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Angela D. Williams
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Emily B. Martin
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Stephen J. Kennel
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Eric R. Heidel
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Jonathan S. Wall
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| | - Joseph W. Jackson
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| |
Collapse
|
4
|
Rehman S, Masthan SS, Ibrahim R, Pham HN, Hassan D, Ahmad F, Asif MS, Safdar A, Anwar Z, Raza S, William P. Pharmacological management of transthyretin amyloid cardiomyopathy: a scoping review. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2024; 10:547-556. [PMID: 38830793 DOI: 10.1093/ehjcvp/pvae044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/17/2024] [Accepted: 06/02/2024] [Indexed: 06/05/2024]
Abstract
AIMS Transthyretin amyloid cardiomyopathy (ATTR-CM) is characterized by the accumulation of transthyretin (TTR) protein in the myocardium. The aim of this scoping review is to provide a descriptive summary of the clinical trials and observational studies that evaluated the clinical efficacy and safety of various agents used in ATTR-CM, with a goal of identifying the contemporary gaps in literature and to reveal future research opportunities. METHODS AND RESULTS The search was performed in line with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A literature search using several databases for observational and clinical trials investigating the treatment modalities for ATTR-CM was undertaken. We extracted data including study characteristics, primary endpoints, and adverse events from each study. A total of 19 studies were included in our scoping review. Out of which, 8 were clinical trials and 11 were observational analyses. The drugs evaluated included tafamadis, acoramidis, revusiran, doxycycline and tauroursodeoxycholic acid and doxycycline, diflusinil, inotersan, eplontersen, and patisiran. Tafamidis has shown to be efficacious in the management of ATTR-CM, particularly when initiated at earlier stages. RNA interference and antisense oligonucleotide drugs have shown promising impacts on quality of life. Additionally, this review identified gaps in the literature, particularly among long-term outcomes, comparative effectiveness, and the translation of research into economic contexts. CONCLUSION Multiple pharmacological options are potential disease-modifying therapies for ATTR-CM. However, many gaps exist in the understanding of these various drug therapies, warranting further research. The future directions for management of ATTR-CM are promising in regard to improving prognostic implications.
Collapse
Affiliation(s)
- Shafi Rehman
- D epartment of Histopathology, Khyber Medical University, Road No. 2, University of Rd No. 2, Rahat Abad, Peshawar, Khyber Pakhtunkhwa 25120, Pakistan
| | | | - Ramzi Ibrahim
- Department of Medicine, University of Arizona-Banner University Medical Center, 1625 N Campbell Ave, Tucson, AZ 85719, USA
| | - Hoang Nhat Pham
- Department of Medicine, University of Arizona-Banner University Medical Center, 1625 N Campbell Ave, Tucson, AZ 85719, USA
| | - Danial Hassan
- Harvard TH Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA
| | - Fahad Ahmad
- Sindh Institute of Urology and Transplantation, Chand Bibi Rd, Ranchore Lane, Karachi, Karachi City, Sindh, Pakistan
| | - Mohammad Shahzad Asif
- Department of Medicine, University College of Medicine and Dentistry, Lahore, Punjab, Pakistan
| | - Ahmad Safdar
- Department of Medicine, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| | - Zain Anwar
- University School, Health Education Campus, 9501 Euclid Ave, Cleveland, OH 44106, USA
| | - Shahzad Raza
- Taussig Cancer Center, Cleveland Clinic, 10201 Carnegie Ave, Cleveland, OH 44106, USA
| | - Preethi William
- Department of Cardiovascular Medicine, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
| |
Collapse
|
5
|
Chakrabartty A. Structural Basis for Monoclonal Antibody Therapy for Transthyretin Amyloidosis. Pharmaceuticals (Basel) 2024; 17:1225. [PMID: 39338387 PMCID: PMC11435174 DOI: 10.3390/ph17091225] [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: 07/14/2024] [Revised: 08/21/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
The disease of transthyretin (TTR) amyloidosis (ATTR) has been known since the 1960s, and during the past 60 or so years, there has been a sustained period of steady discoveries that have led to the current model of ATTR pathogenesis. More recent research has achieved major advances in both diagnostics and therapeutics for ATTR, which are having a significant impact on ATTR patients today. Aiding these recent achievements has been the remarkable ability of cryo-electron microscopy (EM) to determine high-resolution structures of amyloid fibrils obtained from individual patients. Here, we will examine the cryo-EM structures of transthyretin amyloid fibrils to explore the structural basis of the two monoclonal antibody therapies for ATTR that are in clinical trials, ALXN-2220 and Coramitug, as well as to point out potential applications of this approach to other systemic amyloid diseases.
Collapse
Affiliation(s)
- Avi Chakrabartty
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9, Canada
- Proteotoxicity Solutions, Toronto, ON L4K 2E1, Canada
| |
Collapse
|
6
|
Köppen J, Kleinschmidt M, Morawski M, Rahfeld JU, Wermann M, Cynis H, Hegenbart U, Daniel C, Roßner S, Schilling S, Schulze A. Identification of isoaspartate-modified transthyretin as potential target for selective immunotherapy of transthyretin amyloidosis. Amyloid 2024; 31:184-194. [PMID: 38801321 DOI: 10.1080/13506129.2024.2358121] [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: 10/25/2023] [Revised: 04/22/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Numerous studies suggest a progressive accumulation of post-translationally modified peptides within amyloid fibrils, including isoaspartate (isoD) modifications. Here, we generated and characterised novel monoclonal antibodies targeting isoD-modified transthyretin (TTR). The antibodies were used to investigate the presence of isoD-modified TTR in deposits from transthyretin amyloidosis patients and to mediate antibody-dependent phagocytosis of TTR fibrils. METHODS Monoclonal antibodies were generated by immunisation of mice using an isoD-modified peptide and subsequent hybridoma generation. The antibodies were characterised in terms of affinity and specificity to isoD-modified TTR using surface plasmon resonance, transmission electron microscopy and immunohistochemical staining of human cardiac tissue. The potential to elicit antibody-dependent phagocytosis of TTR fibrils was assessed using THP-1 cells. RESULTS We developed two mouse monoclonal antibodies, 2F2 and 4D4, with high nanomolar affinity for isoD-modified TTR and strong selectivity over the unmodified epitope. Both antibodies show presence of isoD-modified TTR in human cardiac tissue, but not in freshly purified recombinant TTR, suggesting isoD modification only present in aged fibrillar deposits. Likewise, the antibodies only facilitated phagocytosis of TTR fibrils and not TTR monomers by THP-1 cells. CONCLUSIONS These antibodies label aged, non-native TTR deposits, leaving native TTR unattended and thereby potentially enabling new therapeutic approaches.
Collapse
Affiliation(s)
- Janett Köppen
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| | - Martin Kleinschmidt
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| | - Markus Morawski
- Paul Flechsig Institute - Center of Neuropathology and Brain Research, Leipzig, Germany
| | - Jens-Ulrich Rahfeld
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| | - Michael Wermann
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
| | - Holger Cynis
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
- Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Ute Hegenbart
- Department of Hematology, Oncology and Rheumatology, Amyloidosis Center, University Hospital, Heidelberg, Germany
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Steffen Roßner
- Paul Flechsig Institute - Center of Neuropathology and Brain Research, Leipzig, Germany
| | - Stephan Schilling
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
- Faculty of Applied Biosciences and Bioprocess Technology, Anhalt University of Applied Sciences, Köthen, Germany
| | - Anja Schulze
- Department of Drug Design and Target Validation, Fraunhofer Institute for Cell Therapy and Immunology, Halle (Saale), Germany
- Faculty of Applied Biosciences and Bioprocess Technology, Anhalt University of Applied Sciences, Köthen, Germany
| |
Collapse
|
7
|
Zhou J, Li Y, Geng J, Zhou H, Liu L, Peng X. Recent Progress in the Development and Clinical Application of New Drugs for Transthyretin Cardiac Amyloidosis. J Cardiovasc Pharmacol 2023; 82:427-437. [PMID: 37678276 PMCID: PMC10691666 DOI: 10.1097/fjc.0000000000001478] [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: 06/13/2023] [Accepted: 08/26/2023] [Indexed: 09/09/2023]
Abstract
Transthyretincardiac amyloidosis is a rare disease that has gained significant attention in recent years because of misfolding of transthyretin fibrils produced by the liver, leading to their deposition in the myocardium. The disease has an insidious onset, nonspecific clinical manifestations, and historically lacked effective drugs, making early diagnosis and treatment challenging. The survival time of patients largely depends on the extent of heart involvement at the time of diagnosis, and conventional treatments for cardiovascular disease do not provide significant benefits. Effective management of the disease requires treatment of its underlying cause. Orthotopic liver transplantation and combined hepato-heart transplantation have been clinically effective means of treating transthyretin cardiac amyloidosis mutants for many years. However, transplantation has many limitations in clinical practice. In recent years, the development of new drugs has brought new hope to patients. This review presents the latest advances in drug development and clinical application to provide a reference for clinicians managing transthyretin cardiac amyloidosis.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Cardiology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- Department of Medical, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Yanfang Li
- Department of Gastroenterology, First Hospital Affiliated to Air Force Medical University, Xian, China
| | - Jing Geng
- Department of Medical, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Hong Zhou
- Department of Medical, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Lian Liu
- Department of Pharmacology, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Xiaochun Peng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China; and
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023 Hubei, China
| |
Collapse
|
8
|
Foster JS, Balachandran M, Hancock TJ, Martin EB, Macy S, Wooliver C, Richey T, Stuckey A, Williams AD, Jackson JW, Kennel SJ, Wall JS. Development and characterization of a prototypic pan-amyloid clearing agent - a novel murine peptide-immunoglobulin fusion. Front Immunol 2023; 14:1275372. [PMID: 37854603 PMCID: PMC10580800 DOI: 10.3389/fimmu.2023.1275372] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/13/2023] [Indexed: 10/20/2023] Open
Abstract
Introduction Systemic amyloidosis is a progressive disorder characterized by the extracellular deposition of amyloid fibrils and accessory proteins in visceral organs and tissues. Amyloid accumulation causes organ dysfunction and is not generally cleared by the immune system. Current treatment focuses on reducing amyloid precursor protein synthesis and slowing amyloid deposition. However, curative interventions will likely also require removal of preexisting amyloid deposits to restore organ function. Here we describe a prototypic pan-amyloid binding peptide-antibody fusion molecule (mIgp5) that enhances macrophage uptake of amyloid. Methods The murine IgG1-IgG2a hybrid immunoglobulin with a pan amyloid-reactive peptide, p5, fused genetically to the N-terminal of the immunoglobulin light chain was synthesized in HEK293T/17 cells. The binding of the p5 peptide moiety was assayed using synthetic amyloid-like fibrils, human amyloid extracts and amyloid-laden tissues as substrates. Binding of radioiodinated mIgp5 with amyloid deposits in vivo was evaluated in a murine model of AA amyloidosis using small animal imaging and microautoradiography. The bioactivity of mIgp5 was assessed in complement fixation and in vitro phagocytosis assays in the presence of patient-derived amyloid extracts and synthetic amyloid fibrils as substrates and in the presence or absence of human serum. Results Murine Igp5 exhibited highly potent binding to AL and ATTR amyloid extracts and diverse types of amyloid in formalin-fixed tissue sections. In the murine model of systemic AA amyloidosis, 125I-mIgp5 bound rapidly and specifically to amyloid deposits in all organs, including the heart, with no evidence of non-specific uptake in healthy tissues. The bioactivity of the immunoglobulin Fc domain was uncompromised in the context of mIgp5 and served as an effective opsonin. Macrophage-mediated uptake of amyloid extract and purified amyloid fibrils was enhanced by the addition of mIgp5. This effect was exaggerated in the presence of human serum coincident with deposition of complement C5b9. Conclusion Immunostimulatory, amyloid-clearing therapeutics can be developed by incorporating pan-amyloid-reactive peptides, such as p5, as a targeting moiety. The immunologic functionality of the IgG remains intact in the context of the fusion protein. These data highlight the potential use of peptide-antibody fusions as therapeutics for all types of systemic amyloidosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Jonathan S. Wall
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, United States
| |
Collapse
|
9
|
Durelle C, Delmont E, Michel C, Trabelsi A, Hostin MA, Ogier A, Bendahan D, Attarian S. Quantification of muscle involvement in familial amyloid polyneuropathy using MRI. Eur J Neurol 2023; 30:3286-3295. [PMID: 37422895 DOI: 10.1111/ene.15970] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 06/09/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND AND PURPOSE Transthyretin familial amyloid polyneuropathy (TTR-FAP) is a rare genetic disease with autosomal-dominant inheritance. In this study, we aimed to quantify fatty infiltration (fat fraction [FF]) and magnetization transfer ratio (MTR) in individual muscles of patients with symptomatic and asymptomatic TTR-FAP using magnetic resonance imaging. Secondarily, we aimed to assess correlations with clinical and electrophysiological variables. METHODS A total of 39 patients with a confirmed mutation in the TTR gene (25 symptomatic and 14 asymptomatic) and 14 healthy volunteers were included. A total of 16 muscles were manually delineated in the nondominant lower limb from T1-weighted anatomical images. The corresponding masks were propagated on the MTR and FF maps. Detailed neurological and electrophysiological examinations were conducted in each group. RESULTS The MTR was decreased (42.6 AU; p = 0.001) and FF was elevated (14%; p = 0.003) in the lower limbs of the symptomatic group, with preferential posterior and lateral involvement. In the asymptomatic group, elevated FF was quantified in the gastrocnemius lateralis muscle (11%; p = 0.021). FF was significantly correlated with disease duration (r = 0.49, p = 0.015), neuropathy impairment score for the lower limb (r = 0.42, p = 0.041), Overall Neuropathy Limitations Scale score (r = 0.49, p = 0.013), polyneuropathy disability score (r = 0.57, p = 0.03) and the sum of compound muscle action potential (r = 0.52, p = 0.009). MTR was strongly correlated to FF (r = 0.78, p < 0.0001), and a few muscles with an FF within the normal range had a reduced MTR. CONCLUSION These observations suggest that FF and MTR could be interesting biomarkers in TTR-FAP. In asymptomatic patients, FF in the gastrocnemius lateralis muscle could be a good indicator of the transition from an asymptomatic to a symptomatic form of the disease. MTR could be an early biomarker of muscle alterations.
Collapse
Affiliation(s)
- Clémence Durelle
- Centre de référence des maladies neuromusculaires et de la SLA, hôpitaux universitaires de Marseille, Marseille, France
| | - Emilien Delmont
- Centre de référence des maladies neuromusculaires et de la SLA, hôpitaux universitaires de Marseille, Marseille, France
| | - Constance Michel
- Centre de résonance magnétique biologique et médicale (Crmbm), Marseille, France
| | - Amira Trabelsi
- Aix-Marseille Univ, CNRS, Centrale Marseille, Institute Fresnel, Marseille, France
| | - Marc-Adrien Hostin
- Centre de résonance magnétique biologique et médicale (Crmbm), Marseille, France
| | - Augustin Ogier
- Department of Radiology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - David Bendahan
- Centre de résonance magnétique biologique et médicale (Crmbm), Marseille, France
| | - Shahram Attarian
- Centre de référence des maladies neuromusculaires et de la SLA, hôpitaux universitaires de Marseille, Marseille, France
| |
Collapse
|
10
|
Jackson JW, Foster JS, Martin EB, Macy S, Wooliver C, Balachandran M, Richey T, Heidel RE, Williams AD, Kennel SJ, Wall JS. Collagen inhibits phagocytosis of amyloid in vitro and in vivo and may act as a 'don't eat me' signal. Amyloid 2023; 30:249-260. [PMID: 36541892 DOI: 10.1080/13506129.2022.2155133] [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: 09/21/2022] [Revised: 11/04/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Systemic amyloidosis refers to a group of protein misfolding disorders characterized by the extracellular deposition of amyloid fibrils in organs and tissues. For reasons heretofore unknown, amyloid deposits are not recognized by the immune system, and progressive deposition leads to organ dysfunction. METHODS In vitro and in vivo phagocytosis assays were performed to elucidate the impact of collagen and other amyloid associated proteins (eg serum amyloid p component and apolipoprotein E) had on amyloid phagocytosis. Immunohistochemical and histopathological staining regimens were employed to analyze collagen-amyloid interactions and immune responses. RESULTS Histological analysis of amyloid-laden tissue indicated that collagen is intimately associated with amyloid deposits. We report that collagen inhibits phagocytosis of amyloid fibrils by macrophages. Treatment of 15 patient-derived amyloid extracts with collagenase significantly enhanced amyloid phagocytosis. Preclinical mouse studies indicated that collagenase treatment of amyloid extracts significantly enhanced clearance as compared to controls, coincident with increased immune cell infiltration of the subcutaneous amyloid lesion. CONCLUSIONS These data suggest that amyloid-associated collagen serves as a 'don't eat me' signal, thereby hindering clearance of amyloid. Targeted degradation of amyloid-associated collagen could result in innate immune cell recognition and clearance of pathologic amyloid deposits.
Collapse
Affiliation(s)
- Joseph W Jackson
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - James S Foster
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Emily B Martin
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Sallie Macy
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Craig Wooliver
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Manasi Balachandran
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Tina Richey
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - R Eric Heidel
- Department of Surgery, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Angela D Williams
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Stephen J Kennel
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| | - Jonathan S Wall
- Department of Medicine, University of Tennessee Graduate School of Medicine, Knoxville, TN, USA
| |
Collapse
|
11
|
Onoue K, Ishihara S, Ogura M, Nakano T. Macrophage Phagocytes Amyloid Fibrils in Giant Cell Myocarditis. Intern Med 2023; 62:2291-2292. [PMID: 36476555 PMCID: PMC10465292 DOI: 10.2169/internalmedicine.1020-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/25/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- Kenji Onoue
- Department of Cardiovascular Medicine, Nara Medical University, Japan
| | - Satomi Ishihara
- Department of Cardiovascular Medicine, Nara Medical University, Japan
| | - Moe Ogura
- Department of Cardiovascular Medicine, Nara Medical University, Japan
- Internal Medicine, Yamato-Takada Municipal Hospital, Japan
| | - Tomoya Nakano
- Department of Cardiovascular Medicine, Nara Medical University, Japan
- Internal Medicine, Yamato-Takada Municipal Hospital, Japan
| |
Collapse
|
12
|
Morfino P, Aimo A, Vergaro G, Sanguinetti C, Castiglione V, Franzini M, Perrone MA, Emdin M. Transthyretin Stabilizers and Seeding Inhibitors as Therapies for Amyloid Transthyretin Cardiomyopathy. Pharmaceutics 2023; 15:pharmaceutics15041129. [PMID: 37111614 PMCID: PMC10143494 DOI: 10.3390/pharmaceutics15041129] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/15/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023] Open
Abstract
Transthyretin (TTR) amyloid cardiomyopathy (ATTR-CM) is a progressive and increasingly recognized cause of heart failure which is associated with high mortality and morbidity. ATTR-CM is characterized by the misfolding of TTR monomers and their deposition within the myocardium as amyloid fibrils. The standard of care for ATTR-CM consists of TTR-stabilizing ligands, such as tafamidis, which aim at maintaining the native structure of TTR tetramers, thus preventing amyloid aggregation. However, their efficacy in advanced-staged disease and after long-term treatment is still a source of concern, suggesting the existence of other pathogenetic factors. Indeed, pre-formed fibrils present in the tissue can further accelerate amyloid aggregation in a self-propagating process known as “amyloid seeding”. The inhibition of amyloidogenesis through TTR stabilizers combined with anti-seeding peptides may represent a novel strategy with additional benefits over current therapies. Finally, the role of stabilizing ligands needs to be reassessed in view of the promising results derived from trials which have evaluated alternative strategies, such as TTR silencers and immunological amyloid disruptors.
Collapse
Affiliation(s)
- Paolo Morfino
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Alberto Aimo
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, 56124 Pisa, Italy
| | - Giuseppe Vergaro
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, 56124 Pisa, Italy
| | - Chiara Sanguinetti
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Vincenzo Castiglione
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, 56124 Pisa, Italy
| | - Maria Franzini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Marco Alfonso Perrone
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Michele Emdin
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, 56124 Pisa, Italy
| |
Collapse
|
13
|
Tsoi MR, Lin JH, Patel AR. Emerging Therapies for Transthyretin Amyloidosis. Curr Oncol Rep 2023; 25:549-558. [PMID: 36943555 DOI: 10.1007/s11912-023-01397-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/23/2023]
Abstract
PURPOSE OF REVIEW This review provides an overview of the available therapies for treating neuropathic and/or cardiac manifestations of transthyretin amyloidosis (ATTR), as well as investigational therapeutic agents in ongoing clinical trials. We discuss additional emergent approaches towards thwarting this life-threatening disease that until recently was considered virtually untreatable. RECENT FINDINGS Advances in noninvasive diagnostic methods for detecting ATTR have facilitated easier diagnosis and detection at an earlier stage of disease when therapeutic interventions are likely to be more effective. There are now several ATTR-directed treatments that are clinically available, as well as investigational agents that are being studied in clinical trials. Therapeutic strategies include tetramer stabilization, gene silencing, and fibril disruption. ATTR has been historically underdiagnosed. With advances in diagnostic methods and the advent of disease-modifying treatments, early diagnosis and initiation of treatment is revolutionizing management of this disease.
Collapse
Affiliation(s)
- Melissa R Tsoi
- Department of Medicine, Tufts Medical Center, MA, 02111, Boston, USA
| | - Jeffrey H Lin
- Department of Medicine, Tufts Medical Center, MA, 02111, Boston, USA
| | - Ayan R Patel
- Cardiac Amyloidosis Program, Tufts Medical Center, 800 Washington St., MA, 02111, Boston, USA.
| |
Collapse
|
14
|
Teixeira AC, Saraiva MJ. Selective recognition of human small transthyretin aggregates by a novel monoclonal antibody. Amyloid 2023; 30:74-80. [PMID: 36111397 DOI: 10.1080/13506129.2022.2122034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Biochemical characterisation of transthyretin variant TTR Y78F showed that this variant adopts a tetrameric conformation as normal TTR but exhibits some of the characteristics of an intermediate structure in the fibrillogenesis pathway. It was hypothesised that native Y78F might represent an early event in TTR amyloidogenesis. We immunised TTR knock out mice with recombinant variant TTR Y78F. One stable hybridoma named CE11, of the IgM isotype, was tested for reactivity towards several soluble recombinant TTR variants both amyloidogenic and non-amyloidogenic. CE11 only recognises the highly amyloidogenic TTR variants L55P, S52P, A97S, Y78F or acidified TTR wt preparations. At the same time, this clone was negative for TTR V30M, soluble wild type protein or TTR T119M. The reactivity increased with oligomer formation and decreased as mature fibrils grow. After size exclusion chromatography (SEC) followed by sandwich ELISA and native immunoblotting, the mAb recognised two peaks (i) peak 1 present in acidified and in soluble variant proteins preparations with material above 146 KDa (ii) peak 2 only present in soluble L55P and S52P TTR preparations with material between 66 and 146 KDa. mAb CE11 may be a potential tool to survey therapeutical agents against TTR aggregation.
Collapse
Affiliation(s)
- A C Teixeira
- Molecular Neurobiology Department, Instituto de Investigação e Inovação em Saúde, i3S, Instituto de Biologia Molecular e Celular, IBMC, Porto, Portugal
| | | |
Collapse
|
15
|
Carberry N, Yu S, Fayerman RN, Dugue R, Miller M, Tanji K, Goyal T, Canoll P, Brannagan TH. Leptomeningeal Disease Secondary to Thr60Ala Transthyretin Amyloidosis: Case Report and Review of the Literature. Neurohospitalist 2023; 13:90-95. [PMID: 36531853 PMCID: PMC9755614 DOI: 10.1177/19418744221127849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Abstract
A 31-year-old woman with transthyretin (TTR) amyloidosis secondary to a Thr60Ala mutation developed recurrent stroke-like episodes with fluctuating mental status. Evaluation for stroke and seizures was unrevealing. She was found to have leptomeningeal contrast enhancement on magnetic resonance imaging, which was confirmed to be CNS TTR amyloidosis on histopathology following brain and dura biopsy. While leptomeningeal disease has rarely been known to be associated with TTR amyloidosis, this is the first documented case of leptomeningeal disease secondary to a Thr60Ala mutation in the TTR gene. A literature review of TTR amyloidosis is presented with special focus on the treatment of leptomeningeal TTR amyloidosis.
Collapse
Affiliation(s)
- Nathan Carberry
- Department of Neurology, Neuromuscular Division, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sun Yu
- Department of Pathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Raisy N. Fayerman
- Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
| | - Rachelle Dugue
- Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
| | - Michael Miller
- Department of Pathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kurenai Tanji
- Department of Pathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Tarini Goyal
- Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
| | - Peter Canoll
- Department of Pathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Thomas H. Brannagan
- Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY, USA
| |
Collapse
|
16
|
Bortoletto AS, Graham WV, Trout G, Bonito‐Oliva A, Kazmi MA, Gong J, Weyburne E, Houser BL, Sakmar TP, Parchem RJ. Human Islet Amyloid Polypeptide (hIAPP) Protofibril-Specific Antibodies for Detection and Treatment of Type 2 Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202342. [PMID: 36257905 PMCID: PMC9731688 DOI: 10.1002/advs.202202342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/26/2022] [Indexed: 05/27/2023]
Abstract
Type 2 diabetes mellitus (T2D) is a major public health concern and is characterized by sustained hyperglycemia due to insulin resistance and destruction of insulin-producing β cells. One pathological hallmark of T2D is the toxic accumulation of human islet amyloid polypeptide (hIAPP) aggregates. Monomeric hIAPP is a hormone normally co-secreted with insulin. However, increased levels of hIAPP in prediabetic and diabetic patients can lead to the formation of hIAPP protofibrils, which are toxic to β cells. Current therapies fail to address hIAPP aggregation and current screening modalities do not detect it. Using a stabilizing capping protein, monoclonal antibodies (mAbs) can be developed against a previously nonisolatable form of hIAPP protofibrils, which are protofibril specific and do not engage monomeric hIAPP. Shown here are two candidate mAbs that can detect hIAPP protofibrils in serum and hIAPP deposits in pancreatic islets in a mouse model of rapidly progressing T2D. Treatment of diabetic mice with the mAbs delays disease progression and dramatically increases overall survival. These results demonstrate the potential for using novel hIAPP protofibril-specific mAbs as a diagnostic screening tool for early detection of T2D, as well as therapeutically to preserve β cell function and target one of the underlying pathological mechanisms of T2D.
Collapse
Affiliation(s)
- Angelina S. Bortoletto
- Center for Cell and Gene TherapyStem Cells and Regenerative Medicine CenterDepartment of NeuroscienceDepartment of Molecular and Cellular BiologyTranslational Biology and Molecular Medicine ProgramMedical Scientist Training ProgramBaylor College of MedicineOne Baylor PlazaHoustonTX77030USA
| | - W. Vallen Graham
- Laboratory of Chemical Biology & Signal TransductionThe Rockefeller University1230 York AvenueNew YorkNY10065USA
| | - Gabriella Trout
- Center for Cell and Gene TherapyStem Cells and Regenerative Medicine CenterDepartment of NeuroscienceDepartment of Molecular and Cellular BiologyTranslational Biology and Molecular Medicine ProgramMedical Scientist Training ProgramBaylor College of MedicineOne Baylor PlazaHoustonTX77030USA
| | - Alessandra Bonito‐Oliva
- Laboratory of Chemical Biology & Signal TransductionThe Rockefeller University1230 York AvenueNew YorkNY10065USA
| | - Manija A. Kazmi
- Laboratory of Chemical Biology & Signal TransductionThe Rockefeller University1230 York AvenueNew YorkNY10065USA
| | - Jing Gong
- Celdara Medical16 Cavendish CourtLebanonNH03766USA
| | | | | | - Thomas P. Sakmar
- Laboratory of Chemical Biology & Signal TransductionThe Rockefeller University1230 York AvenueNew YorkNY10065USA
- Department of Neurobiology, Care Sciences, and SocietyCenter for Alzheimer ResearchDivision of NeurogeriatricsKarolinska InstitutetSolna17164Sweden
| | - Ronald J. Parchem
- Center for Cell and Gene TherapyStem Cells and Regenerative Medicine CenterDepartment of NeuroscienceDepartment of Molecular and Cellular BiologyTranslational Biology and Molecular Medicine ProgramMedical Scientist Training ProgramBaylor College of MedicineOne Baylor PlazaHoustonTX77030USA
| |
Collapse
|
17
|
Fassler M, Tshori S, Barac Y, Bowles DE, Benaim C, George J. Dual Targeting of Soluble Oligomeric and Aggregated Transthyretin with a Monoclonal Antibody Ameliorates Experimental Neuropathy. BIOLOGY 2022; 11:biology11101509. [PMID: 36290413 PMCID: PMC9598441 DOI: 10.3390/biology11101509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022]
Abstract
ATTR amyloidosis comprises a spectrum of multiple clinical presentations, including, predominantly, neuropathy and cardiomyopathy. The common triggering pathogenic protein is misfolded transthyretin, a carrier protein that destabilizes misfolds and assembles into mature amyloid fibrils. The current management of ATTR amyloidosis includes the use of agents that stabilize TTR or attenuate its liver inducible production. Herein, we tested the hypothesis that a monoclonal antibody targeting the soluble oligomeric as well as the aggregated TTR would influence experimental neuropathy. We have shown that Ab-A, our previously described humanized IgG monoclonal antibody, dose-dependently ameliorates the toxicity to neurons triggered by misfolded TTR oligomers. Furthermore, the antibody that exhibits wide misTTR epitope recognition that includes the oligomeric and aggregated forms of the protein dose-dependently enhances the uptake of misfolded TTR to microglia, the resident predominant cells of the innate immune system within the CNS. These in vitro mechanistic properties of the antibody were corroborated by experimental in vivo data showing that the antibody rapidly clears human TTR amyloid extracts infiltrated to the sciatic nerves of rats. Thus, the monoclonal antibody targeting soluble and aggregated TTR is effective in experimental neuropathy, likely due its ability to act as a neuroprotective agent, as well its misTTR-mediated clearance via microglia.
Collapse
Affiliation(s)
- Michael Fassler
- Heart Center, Kaplan Medical Center, Rehovot, Hebrew University of Jerusalem, Jerusalem 91905, Israel
- Cognyxx Ltd., Tel Aviv, Israel
| | - Sagi Tshori
- Heart Center, Kaplan Medical Center, Rehovot, Hebrew University of Jerusalem, Jerusalem 91905, Israel
- Research Authority, Kaplan Medical Center, Rehovot, Hebrew University of Jerusalem, Jerusalem 91905, Israel
| | - Yaron Barac
- The Division of Cardiovascular and Thoracic Surgery, Rabin Medical Center, Petach Tikva 4941492, Israel
| | - Dawn E. Bowles
- Surgical Sciences Division, Department of Surgery, Duke University, Durham, NC 27710, USA
| | - Clara Benaim
- Heart Center, Kaplan Medical Center, Rehovot, Hebrew University of Jerusalem, Jerusalem 91905, Israel
- Cognyxx Ltd., Tel Aviv, Israel
| | - Jacob George
- Heart Center, Kaplan Medical Center, Rehovot, Hebrew University of Jerusalem, Jerusalem 91905, Israel
- Cognyxx Ltd., Tel Aviv, Israel
- Correspondence:
| |
Collapse
|
18
|
Abstract
Systemic amyloidoses are characterized by the unrelenting deposition of autologous proteins as highly ordered fibrils in target organs. The ensuing, potentially fatal organ dysfunction is the result of the combined damage caused by the proteotoxic effect of prefibrillar species and by the cytotoxicity and the structural alterations produced by the amyloid fibrils. Current therapy is focused on eliminating the amyloid protein, thus extinguishing the amyloid cascade at its origin. While this approach may end the cell damage caused by prefibrillar aggregates and prevent further amyloid accumulation, the noxious effects of the amyloid fibrils persist and may hamper the recovery of organ function, which is the ultimate goal of therapy as it is necessary to improve the quality of life and extend survival. Preclinical studies indicate that the clearance of amyloid deposits can be accelerated by specific antibodies targeting amyloid fibrils that activate complement-mediated macrophages and giant cell phagocytosis, possibly promoting the recovery of organ function. Measuring the therapeutic effect of anti-amyloid agents is still a matter of research. In recent years, several monoclonal antibodies targeting amyloid deposits have been tested in clinical trials with mixed outcomes. Recent encouraging results from phase I/II trials, new anti-amyloid agents, and new antibody engineering offer hope that effective amyloid removal will be accomplished in the near future, accelerating organ recovery and improving quality of life and survival.
Collapse
|
19
|
Chen J, Cao D, Fortmann SD, Curcio CA, Feist RM, Crosson JN. Transthyretin proteoforms of intraocular origin in human subretinal fluid. Exp Eye Res 2022; 222:109163. [PMID: 35760119 DOI: 10.1016/j.exer.2022.109163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/24/2022] [Accepted: 06/20/2022] [Indexed: 11/26/2022]
Abstract
Understanding the molecular composition of ocular tissues and fluids could inform new approaches to prevalent causes of blindness. Subretinal fluid accumulating between the photoreceptor outer segments and retinal pigment epithelium (RPE) is potentially a rich source of proteins and lipids normally cycling among outer retinal cells and choroid. Herein, intact post-translationally modified proteins (proteoforms) were extracted from subretinal fluids of five patients with rhegmatogenous retinal detachment (RRD), analyzed by tandem mass spectrometry, and compared to published data on these same proteins as synthesized by other organs. Single-nuclei transcriptomic data from non-diseased human retina/RPE were used to identify whether proteins in subretinal fluid were of potential ocular origin. Two human donor eyes with normal maculas were immunoprobed for transthyretin (TTR) with appropriate controls. The three most abundant proteins detected in subretinal fluid were albumin, TTR, and apolipoprotein A-I. Remarkably, TTR relative to the other proteins was more abundant than its serum counterpart, suggestive of TTR being synthesized predominantly locally. Six post-translationally modified protein forms (proteoforms) of TTR were detected, with the relative amount of glutathionylated TTR being much higher in the subretinal fluid (12-43%) than values reported for serum (<5%) and cerebrospinal fluid (0.4-13%). Moreover, a putative glycosylated TTR dimer of 32,428 Da was detected as the fourth most abundant protein. The high abundance of TTR and putative TTR dimer in subretinal fluid was supported by analysis of available single-nuclei transcriptomic data, which showed strong and specific signal for TTR in RPE. Immunohistochemistry further showed strong diffuse TTR immunoreactivity in choroidal stroma that contrasted with vertically aligned signal in the outer segment zone of the subretinal space and negligible signal in RPE cell bodies. These results suggest that TTR in the retina is synthesized intraocularly, and glutathionylation is crucial for its normal function. Further studies on the composition, function, and quantities of TTR and other proteoforms in subretinal fluid could inform mechanisms, diagnostic methods, and treatment strategies for age-related macular degeneration, familial amyloidosis, and other retinal diseases involving dysregulation of physiologic lipid transfer and oxidative stress.
Collapse
Affiliation(s)
- Jianzhong Chen
- Department of Optometry and Vision Science, The University of Alabama at Birmingham, Birmingham, AL, United States.
| | - Dongfeng Cao
- Department of Ophthalmology and Visual Sciences, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Seth D Fortmann
- Department of Ophthalmology and Visual Sciences, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Christine A Curcio
- Department of Ophthalmology and Visual Sciences, The University of Alabama at Birmingham, Birmingham, AL, United States.
| | - Richard M Feist
- Department of Ophthalmology and Visual Sciences, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jason N Crosson
- Department of Ophthalmology and Visual Sciences, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
20
|
Morioka M, Takashio S, Nakashima N, Nishi M, Fujiyama A, Hirakawa K, Hanatani S, Usuku H, Yamamoto E, Kidoh M, Oda S, Matsushita K, Ueda M, Tsujita K. Correlation Between Cardiac Images, Biomarkers, and Amyloid Load in Wild-Type Transthyretin Amyloid Cardiomyopathy. J Am Heart Assoc 2022; 11:e024717. [PMID: 35699194 PMCID: PMC9238652 DOI: 10.1161/jaha.121.024717] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/06/2022] [Indexed: 11/16/2022]
Abstract
Background Several imaging parameters and biomarkers provide diagnostic and prognostic information for wild-type transthyretin amyloid cardiomyopathy. However, the relevance of these parameters and their association with cardiac amyloid load requires further substantiation. We aimed to elucidate the association of imaging parameters obtained using 99mTc-labeled pyrophosphate scintigraphy, cardiovascular magnetic resonance imaging, global longitudinal strain (GLS), and cardiac biomarkers with cardiac amyloid load in patients with wild-type transthyretin amyloid cardiomyopathy. Methods and Results Eighty-eight patients with wild-type transthyretin amyloid cardiomyopathy who underwent 99mTc-labeled pyrophosphate scintigraphy and cardiovascular magnetic resonance were retrospectively evaluated. Quantitative cardiac amyloid load was obtained from 61 patients after myocardial biopsy. Correlations were assessed using Pearson's correlation coefficient applied to medical record data. The mean heart to contralateral ratio, native T1, extracellular volume, and GLS were 1.91±0.36, 1419.4±56.4 ms, 56.5±13.6%, and -9.4±2.5%, respectively. Median high-sensitivity cardiac troponin T (hs-cTnT) and BNP (B-type natriuretic peptide) levels were 0.0478 (0.0334-0.0691) ng/mL and 213.8 (125.8-392.7) pg/mL, respectively. The mean cardiac amyloid load was 22.9±15.0%. The heart to contralateral ratio correlated significantly with native T1 (r=0.397), extracellular volume (r=0.477), GLS (r=0.363), cardiac amyloid load (r=0.379), and Ln (hs-cTnT) (r=0.247). Further, cardiac amyloid load correlated significantly with native T1 (r=0.509), extracellular volume (r=0.310), GLS (r=0.446), and Ln (hs-cTnT) (r=0.354). Compared with BNP, hs-cTnT levels better correlated with several imaging parameters and cardiac amyloid load. Conclusions Increased cardiac amyloid load correlated with increased 99mTc-labeled pyrophosphate positivity, native T1, extracellular volume, and hs-cTnT levels, and an impaired GLS, suggesting that imaging parameters and cardiac biomarkers may reflect histological and functional changes attributable to amyloid deposition in the myocardium.
Collapse
Affiliation(s)
- Mami Morioka
- Departments of Cardiovascular MedicineGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Seiji Takashio
- Departments of Cardiovascular MedicineGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Naoya Nakashima
- Departments of Cardiovascular MedicineGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Masato Nishi
- Departments of Cardiovascular MedicineGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Akira Fujiyama
- Departments of Cardiovascular MedicineGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Kyoko Hirakawa
- Departments of Cardiovascular MedicineGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Shinsuke Hanatani
- Departments of Cardiovascular MedicineGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Hiroki Usuku
- Departments of Cardiovascular MedicineGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Eiichiro Yamamoto
- Departments of Cardiovascular MedicineGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Masafumi Kidoh
- Departments of Diagnostic RadiologyGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Seitaro Oda
- Departments of Diagnostic RadiologyGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Kenichi Matsushita
- Departments of Cardiovascular MedicineGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Mitsuharu Ueda
- Departments of NeurologyGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Kenichi Tsujita
- Departments of Cardiovascular MedicineGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
21
|
Treatment of Transthyretin Amyloid Cardiomyopathy: The Current Options, the Future, and the Challenges. J Clin Med 2022; 11:jcm11082148. [PMID: 35456241 PMCID: PMC9031576 DOI: 10.3390/jcm11082148] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/07/2022] [Accepted: 04/08/2022] [Indexed: 12/13/2022] Open
Abstract
Transthyretin amyloid cardiomyopathy (ATTR-CM) is a progressively debilitating, rare disease associated with high mortality. ATTR-CM occurs when TTR amyloid protein builds up in the myocardium along with different organs, most commonly the peripheral and the autonomic nervous systems. Managing the cardiac complications with standard heart failure medications is difficult due to the challenge to maintain a balance between the high filling pressure associated with restricted ventricular volume and the low cardiac output. To date, tafamidis is the only agent approved for ATTR-CM treatment. Besides, several agents, including green tea, tolcapone, and diflunisal, are used off-label in ATTR-CM patients. Novel therapies using RNA interference also offer clinical promise. Patisiran and inotersen are currently approved for ATTR-polyneuropathy of hereditary origin and are under investigation for ATTR-CM. Monoclonal antibodies in the early development phases carry hope for amyloid deposit clearance. Despite several drug candidates in the clinical development pipeline, the small ATTR-CM patient population raises several challenges. This review describes current and future therapies for ATTR-CM and sheds light on the clinical development hurdles facing them.
Collapse
|
22
|
Ueda M. Transthyretin: Its function and amyloid formation. Neurochem Int 2022; 155:105313. [PMID: 35218869 DOI: 10.1016/j.neuint.2022.105313] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/20/2022] [Accepted: 02/20/2022] [Indexed: 10/19/2022]
Abstract
Transthyretin (TTR), which is one of the major amyloidogenic proteins in systemic amyloidosis, forms extracellular amyloid deposits in the systemic organs such as nerves, ligaments, heart, and arterioles, and causes two kinds of systemic amyloidosis, hereditary ATTR (ATTRv) amyloidosis induced by variant TTR and aging-related wild-type ATTR (ATTRwt) amyloidosis. More than 150 different mutations, most of which are amyloidogenic, have been reported in the TTR gene. Since most disease-associated mutations affect TTR tetramer dissociation rates, destabilization of TTR tetramers is widely believed to be a critical step in TTR amyloid formation. Recently, effective disease-modifying therapies such as TTR tetramer stabilizers and TTR gene silencing therapies have been developed for ATTR amyloidosis. This study reviews the clinical phenotypes of ATTR amyloidosis, TTR features, and recent progress in promising therapies for ATTR amyloidosis.
Collapse
Affiliation(s)
- Mitsuharu Ueda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-0811, Japan.
| |
Collapse
|
23
|
Molecular Mechanisms of Cardiac Amyloidosis. Int J Mol Sci 2021; 23:ijms23010025. [PMID: 35008444 PMCID: PMC8744761 DOI: 10.3390/ijms23010025] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/18/2021] [Accepted: 12/18/2021] [Indexed: 12/22/2022] Open
Abstract
Cardiac involvement has a profound effect on the prognosis of patients with systemic amyloidosis. Therapeutic methods for suppressing the production of causative proteins have been developed for ATTR amyloidosis and AL amyloidosis, which show cardiac involvement, and the prognosis has been improved. However, a method for removing deposited amyloid has not been established. Methods for reducing cytotoxicity caused by amyloid deposition and amyloid precursor protein to protect cardiovascular cells are also needed. In this review, we outline the molecular mechanisms and treatments of cardiac amyloidosis.
Collapse
|
24
|
Chandrashekar P, Desai AK, Trachtenberg BH. Targeted treatments of AL and ATTR amyloidosis. Heart Fail Rev 2021; 27:1587-1603. [PMID: 34783948 DOI: 10.1007/s10741-021-10180-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/12/2021] [Indexed: 12/22/2022]
Abstract
The therapeutic landscape for cardiac amyloidosis is rapidly evolving. In the last decade, our focus has shifted from dealing with the inevitable complications of continued extracellular infiltration of amyloid fibrils to earlier identification of these patients with prompt initiation of targeted therapy to prevent further deposition. Although much of the focus on novel targeted therapies is within the realm of transthyretin amyloidosis, light chain amyloidosis has benefited due to an overlap particularly in the final common pathway of fibrillogenesis and extraction of amyloid fibrils from the heart. Here, we review the targeted therapeutics for transthyretin and light chain amyloidosis. For transthyretin amyloidosis, the list of current and future therapeutics continues to evolve; and therefore, it is crucial to become familiar with the underlying mechanistic pathways of the disease. Although targeted therapeutic choices in AL amyloidosis are largely driven by the hematology team, the cardiac adverse effect profiles of these therapies, particularly in those with advanced amyloidosis, provide an opportunity for early recognition to prevent decompensation and can help inform recommendations regarding therapy changes when required.
Collapse
Affiliation(s)
- Pranav Chandrashekar
- Amyloidosis Center, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Anish K Desai
- Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Barry H Trachtenberg
- Methodist DeBakey Heart and Vascular Center, Houston, TX, USA. .,Cardio-Oncology and Cardiac Amyloidosis Program, Advanced Heart Failure Fellowship Program, Methodist DeBakey Heart and Vascular Centers, J.C. Walter Transplant Center, Houston, USA.
| |
Collapse
|
25
|
The discovery and development of transthyretin amyloidogenesis inhibitors: what are the lessons? Future Med Chem 2021; 13:2083-2105. [PMID: 34633220 DOI: 10.4155/fmc-2021-0248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Transthyretin (TTR) is associated with several human amyloid diseases. Various kinetic stabilizers have been developed to inhibit the dissociation of TTR tetramer and the formation of amyloid fibrils. Most of them are bisaryl derivatives, natural flavonoids, crown ethers and carborans. In this review article, we focus on TTR tetramer stabilizers, genetic therapeutic approaches and fibril remodelers. The binding modes of typical bisaryl derivatives, natural flavonoids, crown ethers and carborans are discussed. Based on knowledge of the binding of thyroxine to TTR tetramer, many stabilizers have been screened to dock into the thyroxine binding sites, leading to TTR tetramer stabilization. Particularly, those stabilizers with unique binding profiles have shown great potential in developing the therapeutic management of TTR amyloidogenesis.
Collapse
|
26
|
Obici L, Mussinelli R. Current and Emerging Therapies for Hereditary Transthyretin Amyloidosis: Strides Towards a Brighter Future. Neurotherapeutics 2021; 18:2286-2302. [PMID: 34850359 PMCID: PMC8804119 DOI: 10.1007/s13311-021-01154-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2021] [Indexed: 12/19/2022] Open
Abstract
The past few years have witnessed an unprecedented acceleration in the clinical development of novel therapeutic options for hereditary transthyretin amyloidosis. Recently approved agents and drugs currently under investigation not only represent a major breakthrough in this field but also provide validation of the therapeutic potential of innovative approaches, like RNA interference and CRISPR-Cas9-mediated gene editing, in rare inherited disorders. In this review, we describe the evolving therapeutic landscape for hereditary transthyretin amyloidosis and discuss how this highly disabling and fatal condition is turning into a treatable disease. We also provide an overview of the molecular mechanisms involved in transthyretin (TTR) amyloid formation and regression, to highlight how a deeper understanding of these processes has contributed to the identification of novel treatment targets. Finally, we focus on major areas of uncertainty and unmet needs that deserve further efforts to improve long-term patients' outcomes and allow for a brighter future.
Collapse
Affiliation(s)
- Laura Obici
- Amyloidosis Research and Treatment Centre, IRCCS Fondazione Policlinico San Matteo, Viale Golgi, 19, 27100, Pavia, Italy.
| | - Roberta Mussinelli
- Amyloidosis Research and Treatment Centre, IRCCS Fondazione Policlinico San Matteo, Viale Golgi, 19, 27100, Pavia, Italy
| |
Collapse
|
27
|
Qin Q, Wei C, Piao Y, Lian F, Wu H, Zhou A, Wang F, Zuo X, Han Y, Lyu J, Guo D, Jia J. Current Review of Leptomeningeal Amyloidosis Associated With Transthyretin Mutations. Neurologist 2021; 26:189-195. [PMID: 34491937 PMCID: PMC8423141 DOI: 10.1097/nrl.0000000000000337] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Leptomeningeal amyloidosis (LA) represents a rare subtype of familial transthyretin (TTR) amyloidosis, characterized by deposition of amyloid in cranial and spinal leptomeninges. Of >120 TTR mutations identified, few have been associated with LA. CASE REPORT A 27-year-old male presented with a 2-year history of progressive symptoms including cognitive decline and right-sided weakness and numbness. Cerebrospinal fluid (CSF) analyses demonstrated high protein level. Gadolinium-enhanced magnetic resonance imaging (MRI) revealed extensive leptomeningeal enhancement over the surface of the brain and spinal cord. Pathologic analyses revealed a TTR mutation c.113A>G (p.D38G). REVIEW SUMMARY Fifteen mutations and genotype-phenotype correlation of 72 LA patients have been summarized to provide an overview of LA associated with transthyretin mutations. The mean age of clinical onset was 44.9 years and the neurological symptoms primarily included cognitive impairment, headache, ataxia seizures and hearing, visual loss. CSF analysis showed elevated high CSF protein level and MRI revealed extensive leptomeningeal enhancement. CONCLUSION Clinicians should be aware of this rare form of familial transthyretin amyloidosis as well as its typical MRI enhancement and high CSF protein. The important role of biopsy, genetic testing and the potential early diagnosis value of contrast MRI were suggested. Early recognition of these characteristics is important to provide misdiagnosis and shorten the time before correct diagnosis. These findings expand the phenotypic spectrum of TTR gene and have implications for the diagnosis, treatment, and systematic study of LA.
Collapse
Affiliation(s)
- Qi Qin
- Department of Neurology, Innovation Center for Neurological Disorders
- Center for Cognitive Disorders, Beijing Geriatric Hospital
| | - Cuibai Wei
- Department of Neurology, Innovation Center for Neurological Disorders
| | | | | | - Hao Wu
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University
| | - Aihong Zhou
- Department of Neurology, Innovation Center for Neurological Disorders
| | - Fen Wang
- Department of Neurology, Innovation Center for Neurological Disorders
| | - Xiumei Zuo
- Department of Neurology, Innovation Center for Neurological Disorders
| | - Yue Han
- Department of Neurology, Innovation Center for Neurological Disorders
| | - Jihui Lyu
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University
| | - Dongmei Guo
- Department of Neurology, Innovation Center for Neurological Disorders
| | - Jianping Jia
- Department of Neurology, Innovation Center for Neurological Disorders
- Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders
- Beijing Key Laboratory of Geriatric Cognitive Disorders
- Neurodegenerative Laboratory of Ministry of Education of the People’s Republic of China, Beijing, China
| |
Collapse
|
28
|
Capustin M, Frishman WH. Transthyretin Cardiac Amyloidosis and Novel Therapies to Treat This Not-so-rare Cause of Cardiomyopathy. Cardiol Rev 2021; 29:263-273. [PMID: 34397539 DOI: 10.1097/crd.0000000000000387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Transthyretin cardiac amyloidosis (ATTR-CA) is typically a late-onset disease caused by the deposit of transthyretin amyloid fibrils throughout the heart. When this occurs, various cardiac sequelae can develop, including hypotension, conduction abnormalities, and valvular lesions. The cardiomyopathy caused by ATTR-CA (ATTR-CM) has proven difficult to treat. Until recently, symptomatic management was the only therapeutic option, and many therapies used to treat congestive heart failure were ineffective or even detrimental to patients with ATTR-CM. In addition, treatment was limited to heart and liver transplantation. As a result, prognosis was poor. Recently, a few drug therapies have come to light as potential treatment modalities for ATTR-CM, most notably tafamidis, sold under the brand names Vyndaqel and Vyndamax. After the phase III Transthyretin Amyloidosis Cardiomyopathy trial displayed the drug's efficacy, it was given breakthrough therapy designation and was approved by the Food and Drug Administration on May 6, 2019, for the treatment of ATTR-CA. This novel therapy, as well as various other therapies in the pipeline, such as inotersen and patisiran, provide hope where, until recently, there was little. Unfortunately, the exorbitant cost of these new therapies may present a barrier to long-term treatment for some patients. However, by further improving diagnostic algorithms and incorporating these new treatments into our existing therapeutic modalities, patients with ATTR-CA should be able to live far longer than previously expected. Finally, further research combining these novel treatment modalities must be done, as they may prove to be additive or even synergistic in their treatment of ATTR amyloidosis.
Collapse
Affiliation(s)
- Matthew Capustin
- From the Department of Medicine, Zucker School of Medicine/Northwell Northshore-Long Island Jewish Medical Center, Manhasset, NY
| | - William H Frishman
- Department of Medicine and Cardiology, New York Medicine and Westchester Medical Center, Valhalla, NY
| |
Collapse
|
29
|
Benbrahim M, Norman K, Sanchorawala V, Siddiqi OK, Hughes D. A Review of Novel Agents and Clinical Considerations in Patients With ATTR Cardiac Amyloidosis. J Cardiovasc Pharmacol 2021; 77:544-548. [PMID: 33657048 DOI: 10.1097/fjc.0000000000001004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/04/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Transthyretin (ATTR) amyloidosis is a multisystem disease caused by organ deposition of amyloid fibrils derived from the misfolded transthyretin (TTR) protein. The purpose of this article is to provide an overview of current treatment regimens and summarize important considerations for each agent. A literature search was performed with the PubMed database for articles published through October 2020. Search criteria included therapies available on the market and investigational therapies used for ATTR amyloidosis treatment. Both prospective clinical trials and retrospective studies have been included in this review. Available therapies discussed in this review article are tafamidis, diflunisal, patisiran, and inotersen. Tafamidis is FDA approved for treatment of wild-type ATTR (ATTRwt) and hereditary ATTR (ATTRv) cardiomyopathy, and patisiran and inotersen are FDA approved for ATTRv polyneuropathy. Diflunisal does not have an FDA-labeled indication for amyloidosis but has been studied in ATTRv polyneuropathy and ATTRwt cardiomyopathy. Investigational therapies include a TTR stabilizer, AG10; 2 antifibril agents, PRX004 and doxycycline/tauroursodeoxycholic acid; and 2 gene silencers, vutrisiran and AKCEA-TTR-LRx; and clinical trials are ongoing. ATTR amyloidosis treatment selection is based on subtype and presence of cardiac or neurological manifestations. Additional considerations such as side effects, monitoring, and administration are outlined in this review.
Collapse
Affiliation(s)
| | - Kelsey Norman
- Department of Pharmacy, Boston Medical Center, Boston, MA
| | - Vaishali Sanchorawala
- Amyloidosis Center, Boston University School of Medicine and Boston Medical Center, Boston, MA; and
| | - Omar K Siddiqi
- Amyloidosis Center, Boston University School of Medicine and Boston Medical Center, Boston, MA; and
- Section of Cardiovascular Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA
| | - David Hughes
- Department of Pharmacy, Boston Medical Center, Boston, MA
| |
Collapse
|
30
|
Diteepeng T, Del Monte F, Luciani M. The long and winding road to target protein misfolding in cardiovascular diseases. Eur J Clin Invest 2021; 51:e13504. [PMID: 33527342 DOI: 10.1111/eci.13504] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/18/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND In the last decades, cardiovascular diseases (CVD) have remained the first leading cause of mortality and morbidity in the world. Although several therapeutic approaches have been introduced in the past, the development of novel treatments remains an important research goal, which is hampered by the lack of understanding of key mechanisms and targets. Emerging evidences in recent years indicate the involvement of misfolded proteins aggregation and the derailment of protein quality control in the pathogenesis of cardiovascular diseases. Several potential interventions targeting protein quality control have been translated from the bench to the bedside to effectively employ the misfolded proteins as promising therapeutic targets for cardiac diseases, but with trivial results. DESIGN In this review, we describe the recent progresses in preclinical and clinical studies of protein misfolding and compromised protein quality control by selecting and reporting studies focusing on cardiovascular diseases including cardiomyopathies, cardiac amyloidosis, atherosclerosis, atrial fibrillation and thrombosis. RESULTS In preclinical models, modulators of several molecular targets (eg heat shock proteins, unfolded protein response, ubiquitin protein system, autophagy and histone deacetylases) have been tested in various conditions with promising results although lacking an adequate transition towards clinical setting. CONCLUSIONS At present, no therapeutic strategies have been reported to attenuate proteotoxicity in patients with CVD due to a lack of specific biomarkers for pinpointing upstream events in protein folding defects at a subclinical stage of the diseases requiring an intensive collaboration between basic scientists and clinicians.
Collapse
Affiliation(s)
- Thamonwan Diteepeng
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Federica Del Monte
- Department of Medicine, Division of Cardiology, Medical University of South Carolina, Charleston, SC, USA.,Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna Alma Mater, Bologna, Italy
| | - Marco Luciani
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland.,Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| |
Collapse
|
31
|
Cruz Rodriguez JB, Tallaj JA. Narrative review of pharmacotherapy for transthyretin cardiac amyloid. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:519. [PMID: 33850916 PMCID: PMC8039703 DOI: 10.21037/atm-20-4636] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Treatment of cardiac amyloidosis is determined by the amyloid type and degree of involvement. Two types of amyloid commonly infiltrate the heart: immunoglobulin light-chain amyloid (AL), and transthyretin amyloid (ATTR), that encompasses other two forms, a hereditary form (hATTR), and a sporadic, age-related wild-type (wtATTR). The prevalence is expected to increase with aging population. The natural history of ATTR cardiomyopathy includes progressive heart failure (HF), complicated by arrhythmias and conduction system disease. New therapies options have been approved or are under investigation. We performed a narrative literature review, manually-searched the reference lists of included articles and relevant reviews. Treatment for cardiac ATTR should be directed towards alleviation of HF symptoms and to slow or stop progressive amyloid deposition. Conventional HF medications are poorly tolerated and may not alter the disease progression or symptoms, except perhaps with the administration of diuretics. There are three approaches of therapy for ATTR cardiomyopathy: tetramer stabilizers, inhibition of ATTR protein synthesis and clearance of deposited fibrils. Tafamidis diminishes the progression of cardiomyopathy, functional parameters, improves overall outcome in patients with early disease stages, irrespective of ATTR status and is well tolerated. Diflunisal has shown promising results in early studies, but at the expense of significant side effects. Two new agents, antisense oligonucleotides, patisiran and inotersen are under investigation in cardiac amyloidosis. Patisiran appears to be the most effective treatment for hATTR, although evidence is limited, with a relatively small cardiac subpopulation. Therapies considering clearance of amyloid fibrils from tissue remain experimental. In conclusion, tafamidis is the only approved agent for the treatment of ATTR cardiomyopathy although multiple other agents have shown promising early results and are undergoing clinical trials. Careful consideration of the type of ATTR, comorbidities and disease stage will be key in deciding the optimal therapy for ATTR patients.
Collapse
Affiliation(s)
- Jose B Cruz Rodriguez
- Division of Cardiovascular Diseases, Texas Tech University Health Science Center El Paso, El Paso, TX, USA
| | - Jose A Tallaj
- Division of Cardiovascular Diseases, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Medicine, Birmingham VA Medical Center, Birmingham, AL, USA
| |
Collapse
|
32
|
Advances in Treatment of ATTRv Amyloidosis: State of the Art and Future Prospects. Brain Sci 2020; 10:brainsci10120952. [PMID: 33316911 PMCID: PMC7763612 DOI: 10.3390/brainsci10120952] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
Hereditary amyloid transthyretin (ATTRv) amyloidosis with polyneuropathy is a progressive disease that is transmitted as an autosomal dominant trait and characterized by multiple organ failure, including axonal sensory-motor neuropathy, cardiac involvement, and autonomic dysfunction. Liver transplantation (LT) and combined heart-liver transplantation, introduced in the 1990s, have been the only therapies for almost two decades. In 2011, tafamidis meglumine became the first specific drug approved by regulatory agencies, since then the attention toward this disease has progressively increased and several drugs with different mechanisms of action are now available. This review describes the drugs already on the market, those that have shown interesting results although not yet approved, and those currently being tested.
Collapse
|
33
|
Spencer-Bonilla G, Alexander KM, Witteles RM. Advances in the Diagnosis and Management of Transthyretin Amyloid Cardiomyopathy. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2020. [DOI: 10.1007/s11936-020-00844-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
34
|
Teng C, Li P, Bae JY, Pan S, Dixon RAF, Liu Q. Diagnosis and treatment of transthyretin-related amyloidosis cardiomyopathy. Clin Cardiol 2020; 43:1223-1231. [PMID: 32725834 PMCID: PMC7661658 DOI: 10.1002/clc.23434] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 12/29/2022] Open
Abstract
Transthyretin-related amyloidosis (ATTR) is a subgroup of amyloidosis that results from extracellular misassembled and toxic amyloid deposits affecting multiple organ systems, and cardiac tissues in particular. Because ATTR often presents as heart failure with preserved ejection fraction (HFpEF), it has been largely underdiagnosed. Once considered incurable with a grave prognosis, ATTR cardiomyopathy has seen the development of promising alternatives for diagnosis and treatment, with early diagnosis and treatment of ATTR cardiomyopathy highly beneficial due to its high mortality rate. For instance, diagnosing ATTR cardiomyopathy previously required a cardiac biopsy, but new modalities, such as cardiac magnetic resonance imaging and radionuclide bone scans, show promise in accurately diagnosing ATTR cardiomyopathy. Ongoing research and clinical trials have focused on identifying new treatments which primarily target amyloid fiber formation by inhibiting TTR gene expression, stabilizing the TTR tetramer, preventing oligomer aggregation, or affecting degradation of amyloid fibers. In this review, we describe the advances made in the diagnosis and treatment of ATTR in order to increase awareness of the disease and encourage a lower threshold for ATTR workup. Our review also highlights the need for improving the screening, diagnosis, and treatment guidelines for ATTR cardiomyopathy.
Collapse
Affiliation(s)
- Catherine Teng
- Department of MedicineYale New Haven Health‐Greenwich HospitalGreenwichConnecticutUSA
| | - Pengyang Li
- Department of MedicineSaint Vincent HospitalWorcesterMassachusettsUSA
| | - Ju Young Bae
- Department of MedicineYale New Haven Health‐Greenwich HospitalGreenwichConnecticutUSA
| | - Su Pan
- Molecular Cardiology ResearchTexas Heart InstituteHoustonTexasUSA
| | | | - Qi Liu
- Molecular Cardiology ResearchTexas Heart InstituteHoustonTexasUSA
| |
Collapse
|
35
|
Spoladore R, Falasconi G, Marcatti M, Di Maio S, Fiore G, Slavich M, Margonato A, Turco A, Fragasso G. Advances in pharmacotherapy for cardiac amyloidosis. Expert Opin Pharmacother 2020; 22:469-481. [PMID: 33043721 DOI: 10.1080/14656566.2020.1836159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Amyloidosis is a group of progressive and devastating disorders resulting from extracellular deposition of misfolded proteins into tissues. When deposition of fibrils occurs in cardiac tissues, this systemic disease can lead to a very poor prognosis. Systemic amyloidosis can be acquired [light chain (AL) amyloidosis; AA amyloidosis], or hereditary [transthyretin (ATTR) amyloidosis]. Cardiac disease in amyloidosis is usually secondary to a systemic disease. The diagnosis of cardiac involvement is often delayed and yields an adverse prognosis. AREAS COVERED in this review, the authors report current literature on advances in pharmacotherapy for cardiac amyloidosis, mainly focused on AL and ATTR amyloidosis treatment. EXPERT OPINION Most pharmacological trials in amyloidosis patients, both AL and TTR, are directed to study the effects of drugs on polyneuropathy. However, since cardiac involvement carries a prominent negative survival impact in amyloidosis patients, future research should be more focused on amyloidosis cardiomyopathy as primary endpoint. Additionally, in AL amyloidosis therapies are mainly derived from experience on multiple myeloma treatment. In this specific setting, possible future research could particularly focus on immunotherapeutic agents able to optimize the standard chemotherapy results and, thus, allowing a larger population of patients to be treated by bone marrow stem cell transplantation.
Collapse
Affiliation(s)
- R Spoladore
- Hypertrophic Cardiomyopathy Unit, IRCCS San Raffaele University Hospital, Milan, Italy.,Clinical Cardiology Unit, IRCCS San Raffaele University Hospital, Milan, Italy
| | - G Falasconi
- Clinical Cardiology Unit, IRCCS San Raffaele University Hospital, Milan, Italy
| | - M Marcatti
- Haematology Unit, IRCCS San Raffaele University Hospital, Milan, Italy
| | - S Di Maio
- Clinical Cardiology Unit, IRCCS San Raffaele University Hospital, Milan, Italy
| | - G Fiore
- Clinical Cardiology Unit, IRCCS San Raffaele University Hospital, Milan, Italy
| | - M Slavich
- Clinical Cardiology Unit, IRCCS San Raffaele University Hospital, Milan, Italy
| | - A Margonato
- Clinical Cardiology Unit, IRCCS San Raffaele University Hospital, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - A Turco
- Division of Cardiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - G Fragasso
- Clinical Cardiology Unit, IRCCS San Raffaele University Hospital, Milan, Italy.,Heart Failure Unit, IRCCS San Raffaele University Hospital, Milan, Italy
| |
Collapse
|
36
|
L'amylose cardiaque à transthyrétine. Rev Med Interne 2020; 41:673-683. [DOI: 10.1016/j.revmed.2020.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/17/2020] [Accepted: 07/01/2020] [Indexed: 12/20/2022]
|
37
|
Review of Transthyretin Silencers, Stabilizers, and Fibril Removal Agents in the Treatment of Transthyretin Cardiac Amyloid. Curr Cardiol Rep 2020; 22:106. [PMID: 32770401 DOI: 10.1007/s11886-020-01374-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW To provide a functional review for practicing clinicians on the current and emerging treatment considerations for transthyretin (TTR) cardiac amyloidosis (ATTR-CA). RECENT FINDINGS Current treatment considerations are characterized as those silencing TTR translation, stabilizing TTR tetramers, and disrupting amyloid fibril deposition. Historically considered a rare disease state, ATTR-CA is increasingly recognized as an important mediator of heart failure morbidity and mortality. The emergence of widely available therapies for ATTR-CA has developed hope for patients where little was previously present. Thus, it is important that all cardiology clinicians have a functional understanding of the disease state and treatment options. This review will discuss agents within each of the above classes with expanded discussion on tafamidis given its favorable efficacy, safety, and availability. ATTR-CA diagnostic considerations are reviewed with regard to the identification of potential tafamidis candidates, and practical economic considerations are also reviewed.
Collapse
|
38
|
Ihne S, Morbach C, Sommer C, Geier A, Knop S, Störk S. Amyloidosis-the Diagnosis and Treatment of an Underdiagnosed Disease. DEUTSCHES ARZTEBLATT INTERNATIONAL 2020; 117:159-166. [PMID: 32295695 DOI: 10.3238/arztebl.2020.0159] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 08/25/2019] [Accepted: 12/12/2019] [Indexed: 01/15/2023]
Abstract
BACKGROUND Systemic amyloidosis is a multi-system disease caused by fibrillary protein deposition with ensuing dysfunction of the affected organ systems. Its diagnosis is often delayed because the manifestations of the disease are variable and non-specific. Its main forms are light chain (AL) amyloidosis and transthyretinrelated ATTR amyloidosis, which, in turn, has both a sporadic subtype (wildtype, ATTRwt) and a hereditary subtype (mutated, ATTRv). METHODS This review is based on pertinent publications that were retrieved by a selective search in PubMed covering the years 2005 to 2019. RESULTS No robust epidemiological figures are available for Germany to date. Both AL amyloidosis and hereditary ATTR amyloidosis are rare diseases, but the prev - alence of ATTRwt amyloidosis is markedly underestimated. The diagnostic algorithm is complex and generally requires histological confirmation of the diagnosis. Only cardiac ATTR amyloidosis can be diagnosed non-invasively with bone scintigraphy once a monoclonal gammopathy has been excluded. AL amyloidosis can be considered a complication of a plasma cell dyscrasia and treated with reference to patterns applied in multiple myeloma. Despite the availability of causally directed treatment, it has not yet been possible to reduce the mortality of advanced cardiac AL amyloidosis. Three drugs (tafamidis, patisiran, and inotersen) are now available to treat grade 1 or 2 polyneuropathy in ATTRv amyloidosis, and further agents are now being tested in clinical trials. It is expected that tafamidis will soon be approved in Germany for the treatment of cardiac ATTR amyloidosis. CONCLUSION The diagnosis of amyloidosis is difficult because of its highly varied presentation. In case of clinical suspicion, a rapid, targeted diagnostic evaluation and subsequent initiation of treatment should be performed in a specialized center. When the new drugs to treat amyloidosis become commercially available, their use and effects should be documented in nationwide registries.
Collapse
Affiliation(s)
- Sandra Ihne
- Interdisciplinary Amyloidosis Center of Northern Bavaria, University Hospital Würzburg, Germany; Medical Clinic and Policlinic II, Dept. of Hemtatology, University Hospital Würzburg, Germany; Comprehensive Heart Failure Center Würzburg, University and University Hospital Würzburg, Germany; Medical Clinic and Policlinic I, Dept. of Cardiology, University Hospital Würzburg, Germany; Department of Neurology, University Hospital Würzburg, Germany; Medical Clinic and Policlinic II, Dept. of Hepatology, University Hospital Würzburg, Germany
| | | | | | | | | | | |
Collapse
|
39
|
Schwartzlow C, Kazamel M. Hereditary Transthyretin Amyloidosis: Clinical Presentation and Management Updates. J Clin Neuromuscul Dis 2020; 21:144-156. [PMID: 32073460 DOI: 10.1097/cnd.0000000000000270] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Hereditary transthyretin amyloidosis, once a rare progressive neuropathy and/or cardiomyopathy, is now recognized with increasing worldwide frequency, various phenotypes, and over 130 gene mutations identified to date. This inherited disorder develops as a result of mutated transthyretin amyloid aggregation and systematic deposition throughout the body. With increasing knowledge about the pathophysiology of this disease, new disease-modifying therapies are being developed. In addition to slowing progression, these new agents were found to improve quality of life and reduce the severity of neuropathic symptoms. Two new gene-modifying therapies recently received Food and Drug Administration approval following the positive results from phase III trials. These include an antisense oligonucleotide, inotersen, and small interfering RNA, patisiran, which were reported to reduce the production of transthyretin and had promising safety profiles. Additional novel therapies are being explored with hopes to prolong survival. Therefore, early diagnosis of this treatable disorder has become increasingly important in clinical practice.
Collapse
Affiliation(s)
- Coreen Schwartzlow
- Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL
| | | |
Collapse
|
40
|
Luigetti M, Romano A, Di Paolantonio A, Bisogni G, Sabatelli M. Diagnosis and Treatment of Hereditary Transthyretin Amyloidosis (hATTR) Polyneuropathy: Current Perspectives on Improving Patient Care. Ther Clin Risk Manag 2020; 16:109-123. [PMID: 32110029 PMCID: PMC7041433 DOI: 10.2147/tcrm.s219979] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/27/2020] [Indexed: 12/16/2022] Open
Abstract
Hereditary transthyretin amyloidosis (hATTR) with polyneuropathy (formerly known as Familial Amyloid Polyneuropathy) is a rare disease due to mutations in the gene encoding transthyretin (TTR) and characterized by multisystem extracellular deposition of amyloid, leading to dysfunction of different organs and tissues. hATTR amyloidosis represents a diagnostic challenge for neurologists considering the great variability in clinical presentation and multiorgan involvement. Generally, patients present with polyneuropathy, but clinicians should consider the frequent cardiac, ocular and renal impairment. Especially a hypertrophic cardiomyopathy, even if usually latent, is identifiable in at least 50% of the patients. Therapeutically, current available options act at different stages of TTR production, including synthesis inhibition (liver transplantation and/or gene-silencing drugs) or tetramer TTR stabilization (TTR stabilizers), increasing survival at different disease stages. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/n8sg_YlGJiA
Collapse
Affiliation(s)
- Marco Luigetti
- Neurology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | | | - Mario Sabatelli
- Università Cattolica del Sacro Cuore, Rome, Italy.,Centro Clinico NEMO Adulti, Rome, Italy
| |
Collapse
|
41
|
Müller ML, Butler J, Heidecker B. Emerging therapies in transthyretin amyloidosis – a new wave of hope after years of stagnancy? Eur J Heart Fail 2020; 22:39-53. [DOI: 10.1002/ejhf.1695] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/07/2019] [Accepted: 10/30/2019] [Indexed: 12/15/2022] Open
Affiliation(s)
- Maximilian L. Müller
- Department of Cardiology, Charité Universitätsmedizin BerlinCampus Benjamin Franklin Berlin Germany
| | - Javed Butler
- Department of MedicineThe Mississippi Medical Center Jackson MS USA
| | - Bettina Heidecker
- Department of Cardiology, Charité Universitätsmedizin BerlinCampus Benjamin Franklin Berlin Germany
| |
Collapse
|
42
|
Gertz MA, Mauermann ML, Grogan M, Coelho T. Advances in the treatment of hereditary transthyretin amyloidosis: A review. Brain Behav 2019; 9:e01371. [PMID: 31368669 PMCID: PMC6749475 DOI: 10.1002/brb3.1371] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/01/2019] [Accepted: 07/03/2019] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION Amyloid transthyretin amyloidosis (ATTR) is a progressive and often fatal disease caused by the buildup of mutated (hereditary ATTR [hATTR]; also known as ATTR variant [ATTRv]) or normal transthyretin (wild-type ATTR) throughout the body. Two new therapies-inotersen, an antisense oligonucleotide therapy, and patisiran, an RNA interference therapy-received marketing authorization and represent a significant advance in the treatment of amyloidosis. Herein, we describe the clinical presentation of ATTR, commonly used procedures in its diagnosis, and current treatment landscape for ATTR, with a focus on hATTR. METHODS A PubMed search from 2008 to September 2018 was conducted to review the literature on ATTR. RESULTS Until recently, there have been few treatment options for polyneuropathy of hATTR. Inotersen and patisiran substantially reduce the amyloidogenic precursor protein transthyretin and have demonstrated efficacy in patients with early- and late-stage disease and in slowing or improving neuropathy progression. In contrast, established therapies, such as liver transplantation, typically reserved for patients with early-stage disease, and tafamidis, indicated for the treatment of early-stage disease in Europe, or diflunisal, a nonsteroidal anti-inflammatory drug that is used off-label, are associated with side effects and/or unclear efficacy in certain patient populations. Thus, inotersen and patisiran are positioned to be the preferred therapeutic modalities. CONCLUSIONS Important differences between inotersen and patisiran, including formulation, dosing, requirements for premedications, and safety monitoring, require an understanding and knowledge of each treatment for informed decision making.
Collapse
Affiliation(s)
| | | | | | - Teresa Coelho
- Centro Hospitalar do Porto, Hospital de Santo António, Porto, Portugal
| |
Collapse
|
43
|
Ablasser K, Verheyen N, Glantschnig T, Agnetti G, Rainer PP. Unfolding Cardiac Amyloidosis –From Pathophysiology to Cure. Curr Med Chem 2019; 26:2865-2878. [DOI: 10.2174/0929867325666180104153338] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 12/13/2022]
Abstract
Deposition of amyloidogenic proteins leading to the formation of amyloid fibrils in the myocardium causes cardiac amyloidosis. Although any form of systemic amyloidosis can affect the heart, light-chain (AL) or transthyretin amyloidosis (ATTR) account for the majority of diagnosed cardiac amyloid deposition. The extent of cardiac disease independently predicts mortality. Thus, the reversal of arrest of adverse cardiac remodeling is the target of current therapies. Here, we provide a condensed overview on the pathophysiology of AL and ATTR cardiac amyloidoses and describe treatments that are currently used or investigated in clinical or preclinical trials. We also briefly discuss acquired amyloid deposition in cardiovascular disease other than AL or ATTR.
Collapse
Affiliation(s)
- Klemens Ablasser
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Nicolas Verheyen
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | | | - Giulio Agnetti
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Peter P. Rainer
- Division of Cardiology, Medical University of Graz, Graz, Austria
| |
Collapse
|
44
|
|
45
|
Gertz MA, Scheinberg M, Waddington-Cruz M, Heitner SB, Karam C, Drachman B, Khella S, Whelan C, Obici L. Inotersen for the treatment of adults with polyneuropathy caused by hereditary transthyretin-mediated amyloidosis. Expert Rev Clin Pharmacol 2019; 12:701-711. [PMID: 31268366 DOI: 10.1080/17512433.2019.1635008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Hereditary transthyretin-mediated amyloidosis (ATTRv; v for variant) is an underdiagnosed, progressive, and fatal multisystemic disease with a heterogenous clinical phenotype that is caused by TTR gene mutations that destabilize the TTR protein, resulting in its misfolding, aggregation, and deposition in tissues throughout the body. Areas covered: Inotersen, an antisense oligonucleotide inhibitor, was recently approved in the United States and Europe for the treatment of the polyneuropathy of ATTRv based on the positive results obtained in the pivotal phase 3 trial, NEURO-TTR. This review will discuss the mechanism of action of inotersen and its pharmacology, clinical efficacy, and safety and tolerability. A PubMed search using the terms 'inotersen,' 'AG10,' 'antisense oligonucleotide,' 'hereditary transthyretin amyloidosis,' 'familial amyloid polyneuropathy,' and 'familial amyloid cardiomyopathy' was performed, and the results were screened for the most relevant English language publications. The bibliographies of all retrieved articles were manually searched to identify additional studies of relevance. Expert opinion: Inotersen targets the disease-forming protein, TTR, and has been shown to improve quality of life and neuropathy progression in patients with stage 1 or 2 ATTRv with polyneuropathy. Inotersen is well tolerated, with a manageable safety profile through regular monitoring for the development of glomerulonephritis or thrombocytopenia.
Collapse
Affiliation(s)
- Morie A Gertz
- a Department of Hematology, Transplant Center, Cancer Center, Mayo Clinic College of Medicine , Rochester , MN , USA
| | - Morton Scheinberg
- b Department of Rheumatology, Hospital Israelita Albert Einstein , Sao Paulo , Brazil
| | - Márcia Waddington-Cruz
- c Neuromuscular Diseases Unit, Federal University of Rio de Janeiro, University Hospital , Rio de Janeiro , Brazil
| | - Stephen B Heitner
- d Hypertrophic Cardiomyopathy Clinic, Knight Cardiovascular Institute , Portland , OR , USA
| | - Chafic Karam
- e Department of Neurology, ALS and Neuromuscular Center, Oregon Health and Science University , Portland , OR , USA
| | - Brian Drachman
- f Department of Cardiovascular Medicine, University of Pennsylvania , Philadelphia , PA , USA
| | - Sami Khella
- g Department of Neurology, University of Pennsylvania , Philadelphia , PA , USA
| | - Carol Whelan
- h Consultant Cardiologist, University College London-National Amyloidosis Centre , London , UK
| | - Laura Obici
- i Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico S. Matteo , Pavia , Italy
| |
Collapse
|
46
|
Ruberg FL, Grogan M, Hanna M, Kelly JW, Maurer MS. Transthyretin Amyloid Cardiomyopathy: JACC State-of-the-Art Review. J Am Coll Cardiol 2019; 73:2872-2891. [PMID: 31171094 PMCID: PMC6724183 DOI: 10.1016/j.jacc.2019.04.003] [Citation(s) in RCA: 666] [Impact Index Per Article: 111.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 12/16/2022]
Abstract
Transthyretin amyloid cardiomyopathy (ATTR-CM) is an under-recognized cause of heart failure (HF) in older adults, resulting from myocardial deposition of misfolded transthyretin (TTR) or pre-albumin. Characteristic patterns of echocardiography and cardiac magnetic resonance can strongly suggest the disease but are not diagnostic. The diagnosis can be made with noninvasive nuclear imaging when there is no evidence of a monoclonal protein. Amyloid fibril formation results from a destabilizing mutation in hereditary ATTR amyloidosis (hATTR) or from an aging-linked process in wild-type ATTR amyloidosis (wtATTR). Recent studies have suggested that up to 10% to 15% of older adults with HF may have unrecognized wtATTR. Associated features, including carpal tunnel syndrome and lumbar spinal stenosis, raise suspicion and may afford a means for early diagnosis. Previously treatable only by organ transplantation, pharmaceutical therapy that slows or halts ATTR-CM progression and favorably affects clinical outcomes is now available. Early recognition remains essential to afford the best treatment efficacy.
Collapse
Affiliation(s)
- Frederick L Ruberg
- Section of Cardiovascular Medicine, Department of Medicine, Amyloidosis Center, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Martha Grogan
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Mazen Hanna
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Jeffery W Kelly
- Departments of Chemistry and Molecular Medicine, Scripps Research Institute, La Jolla, California
| | - Mathew S Maurer
- Division of Cardiology, Department of Medicine, Center for Advanced Cardiac Care, Columbia University Medical Center, New York, New York.
| |
Collapse
|
47
|
Ueda M, Okada M, Mizuguchi M, Kluve-Beckerman B, Kanenawa K, Isoguchi A, Misumi Y, Tasaki M, Ueda A, Kanai A, Sasaki R, Masuda T, Inoue Y, Nomura T, Shinriki S, Shuto T, Kai H, Yamashita T, Matsui H, Benson MD, Ando Y. A cell-based high-throughput screening method to directly examine transthyretin amyloid fibril formation at neutral pH. J Biol Chem 2019; 294:11259-11275. [PMID: 31167790 DOI: 10.1074/jbc.ra119.007851] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/31/2019] [Indexed: 12/12/2022] Open
Abstract
Transthyretin (TTR) is a major amyloidogenic protein associated with hereditary (ATTRm) and nonhereditary (ATTRwt) intractable systemic transthyretin amyloidosis. The pathological mechanisms of ATTR-associated amyloid fibril formation are incompletely understood, and there is a need for identifying compounds that target ATTR. C-terminal TTR fragments are often present in amyloid-laden tissues of most patients with ATTR amyloidosis, and on the basis of in vitro studies, these fragments have been proposed to play important roles in amyloid formation. Here, we found that experimentally-formed aggregates of full-length TTR are cleaved into C-terminal fragments, which were also identified in patients' amyloid-laden tissues and in SH-SY5Y neuronal and U87MG glial cells. We observed that a 5-kDa C-terminal fragment of TTR, TTR81-127, is highly amyloidogenic in vitro, even at neutral pH. This fragment formed amyloid deposits and induced apoptosis and inflammatory gene expression also in cultured cells. Using the highly amyloidogenic TTR81-127 fragment, we developed a cell-based high-throughput screening method to discover compounds that disrupt TTR amyloid fibrils. Screening a library of 1280 off-patent drugs, we identified two candidate repositioning drugs, pyrvinium pamoate and apomorphine hydrochloride. Both drugs disrupted patient-derived TTR amyloid fibrils ex vivo, and pyrvinium pamoate also stabilized the tetrameric structure of TTR ex vivo in patient plasma. We conclude that our TTR81-127-based screening method is very useful for discovering therapeutic drugs that directly disrupt amyloid fibrils. We propose that repositioning pyrvinium pamoate and apomorphine hydrochloride as TTR amyloid-disrupting agents may enable evaluation of their clinical utility for managing ATTR amyloidosis.
Collapse
Affiliation(s)
- Mitsuharu Ueda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Masamitsu Okada
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Mineyuki Mizuguchi
- Laboratory of Structural Biology, Faculty of Pharmacy and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Barbara Kluve-Beckerman
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Kyosuke Kanenawa
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Aito Isoguchi
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yohei Misumi
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Masayoshi Tasaki
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan.,Department of Morphological and Physiological Sciences, Graduate School of Health Sciences, Kumamoto University, Kumamoto 862-0976, Japan
| | - Akihiko Ueda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Akinori Kanai
- Department of Molecular Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
| | - Ryoko Sasaki
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Teruaki Masuda
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yasuteru Inoue
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Toshiya Nomura
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Satoru Shinriki
- Department of Molecular Laboratory Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Tsuyoshi Shuto
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Hirofumi Kai
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Taro Yamashita
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Hirotaka Matsui
- Department of Molecular Laboratory Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Merrill D Benson
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Yukio Ando
- Department of Neurology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
48
|
Michels da Silva D, Langer H, Graf T. Inflammatory and Molecular Pathways in Heart Failure-Ischemia, HFpEF and Transthyretin Cardiac Amyloidosis. Int J Mol Sci 2019; 20:ijms20092322. [PMID: 31083399 PMCID: PMC6540104 DOI: 10.3390/ijms20092322] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/01/2019] [Accepted: 05/07/2019] [Indexed: 02/06/2023] Open
Abstract
Elevated pro-inflammatory biomarkers and cytokines are associated with morbidity and mortality in heart failure (HF). Preclinical and clinical studies have shown multiple inflammatory mechanisms causing cardiac remodeling, dysfunction and chronic failure. Therapeutics in trials targeting the immune response in heart failure and its effects did not result in evident benefits regarding clinical endpoints and mortality. This review elaborates pathways of immune cytokines in pathogenesis and worsening of heart failure in clinical and cellular settings. Besides the well-known mechanisms of immune activation and inflammation in atherosclerosis causing ischemic cardiomyopathy or myocarditis, attention is focused on other mechanisms leading to heart failure such as transthyretin (TTR) amyloidosis or heart failure with preserved ejection fraction. The knowledge of the pathogenesis in heart failure and amyloidosis on a molecular and cellular level might help to highlight new disease defining biomarkers and to lead the way to new therapeutic targets.
Collapse
Affiliation(s)
- Diana Michels da Silva
- Department of Cardiology, Angiology and Intensive Care, Medicine Medical Clinic II, University Heart Center Lübeck, 23562 Lübeck, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany.
| | - Harald Langer
- Department of Cardiology, Angiology and Intensive Care, Medicine Medical Clinic II, University Heart Center Lübeck, 23562 Lübeck, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany.
| | - Tobias Graf
- Department of Cardiology, Angiology and Intensive Care, Medicine Medical Clinic II, University Heart Center Lübeck, 23562 Lübeck, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany.
| |
Collapse
|
49
|
Çakar A, Durmuş-Tekçe H, Parman Y. Familial Amyloid Polyneuropathy. ACTA ACUST UNITED AC 2019; 56:150-156. [PMID: 31223250 DOI: 10.29399/npa.23502] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 12/27/2018] [Indexed: 12/26/2022]
Abstract
Transthyretin-related familial amyloid polyneuropathy (TTR-FAP) is a life-threatening disease caused by the accumulation of amyloidogenic transthyretin (TTR) protein in tissues. Mutations in TTR gene destabilize TTR protein to misfold from its native tetramer form to amyloidogenic monomer form. In endemic countries, TTR-FAP presents with length-dependent small fiber neuropathy, however in non-endemic countries clinical features can be highly variable. Genetic testing for TTR gene is mandatory for the diagnosis. Demonstrating amyloid deposits in tissues may be necessary for distinguishing symptomatic patients from asymptomatic carriers. Routine follow-up should include a wide range of tests to demonstrate systemic involvement. In recent years, treatment of TTR-FAP has significantly improved with new therapeutic approaches. TTR stabilizers and TTR-gene silencing drugs prevent the progression of the disease. Monoclonal antibodies that target amyloid deposits are currently under development. Early initiation of the treatment is important for better functional outcome.
Collapse
Affiliation(s)
- Arman Çakar
- Department of Neurology, İstanbul Faculty of Medicine, İstanbul University, İstanbul, Turkey
| | - Hacer Durmuş-Tekçe
- Department of Neurology, İstanbul Faculty of Medicine, İstanbul University, İstanbul, Turkey
| | - Yeşim Parman
- Department of Neurology, İstanbul Faculty of Medicine, İstanbul University, İstanbul, Turkey
| |
Collapse
|
50
|
Saelices L, Nguyen BA, Chung K, Wang Y, Ortega A, Lee JH, Coelho T, Bijzet J, Benson MD, Eisenberg DS. A pair of peptides inhibits seeding of the hormone transporter transthyretin into amyloid fibrils. J Biol Chem 2019; 294:6130-6141. [PMID: 30733338 DOI: 10.1074/jbc.ra118.005257] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 01/22/2019] [Indexed: 11/06/2022] Open
Abstract
The tetrameric protein transthyretin is a transporter of retinol and thyroxine in blood, cerebrospinal fluid, and the eye, and is secreted by the liver, choroid plexus, and retinal epithelium, respectively. Systemic amyloid deposition of aggregated transthyretin causes hereditary and sporadic amyloidoses. A common treatment of patients with hereditary transthyretin amyloidosis is liver transplantation. However, this procedure, which replaces the patient's variant transthyretin with the WT protein, can fail to stop subsequent cardiac deposition, ultimately requiring heart transplantation. We recently showed that preformed amyloid fibrils present in the heart at the time of surgery can template or seed further amyloid aggregation of native transthyretin. Here we assess possible interventions to halt this seeding, using biochemical and EM assays. We found that chemical or mutational stabilization of the transthyretin tetramer does not hinder amyloid seeding. In contrast, binding of the peptide inhibitor TabFH2 to ex vivo fibrils efficiently inhibits amyloid seeding by impeding self-association of the amyloid-driving strands F and H in a tissue-independent manner. Our findings point to inhibition of amyloid seeding by peptide inhibitors as a potential therapeutic approach.
Collapse
Affiliation(s)
- Lorena Saelices
- From the Departments of Biological Chemistry and Chemistry and Biochemistry, Howard Hughes Medical Institute, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1570
| | - Binh A Nguyen
- From the Departments of Biological Chemistry and Chemistry and Biochemistry, Howard Hughes Medical Institute, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1570
| | - Kevin Chung
- From the Departments of Biological Chemistry and Chemistry and Biochemistry, Howard Hughes Medical Institute, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1570
| | - Yifei Wang
- From the Departments of Biological Chemistry and Chemistry and Biochemistry, Howard Hughes Medical Institute, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1570
| | - Alfredo Ortega
- From the Departments of Biological Chemistry and Chemistry and Biochemistry, Howard Hughes Medical Institute, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1570
| | - Ji H Lee
- From the Departments of Biological Chemistry and Chemistry and Biochemistry, Howard Hughes Medical Institute, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1570
| | - Teresa Coelho
- the Neurophysiology Department and Corino de Andrade Unit, Hospital Santo António, Centro Hospitalar do Porto, Porto 4099-001, Portugal
| | - Johan Bijzet
- the Department of Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, 9713 GZ, The Netherlands
| | - Merrill D Benson
- the Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - David S Eisenberg
- From the Departments of Biological Chemistry and Chemistry and Biochemistry, Howard Hughes Medical Institute, UCLA-DOE Institute, Molecular Biology Institute, UCLA, Los Angeles, California 90095-1570.
| |
Collapse
|