Warning: mkdir(): Permission denied in /home/virtual/lib/view_data.php on line 81

Warning: fopen(upload/ip_log/ip_log_2024-04.txt): failed to open stream: No such file or directory in /home/virtual/lib/view_data.php on line 83

Warning: fwrite() expects parameter 1 to be resource, boolean given in /home/virtual/lib/view_data.php on line 84
What’s new in neuromuscular pathology 2022: myopathy updates and gene therapies
Skip Navigation
Skip to contents

J Pathol Transl Med : Journal of Pathology and Translational Medicine

OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > J Pathol Transl Med > Volume 57(1); 2023 > Article
Newsletter
What’s new in neuromuscular pathology 2022: myopathy updates and gene therapies
Chunyu Caiorcid
Journal of Pathology and Translational Medicine 2022;57(1):79-80.
DOI: https://doi.org/10.4132/jptm.2022.10.14
Published online: December 20, 2022

Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA

Corresponding Author: Chunyu Cai, MD, PhD, Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA, E-mail: 'Chunyu.Cai@utsouthwestern.edu'
This article has been published jointly, with consent, in both Journal of Pathology and Translational Medicine and PathologyOutlines.com.
• Received: September 22, 2022   • Accepted: October 14, 2022

© 2023 The Korean Society of Pathologists/The Korean Society for Cytopathology

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 1,532 Views
  • 155 Download
  • This compilation of new changes in the diagnosis and treatment of muscle and nerve disease is extracted from the latest publications from the European Neuromuscular Centre International workshops, FDA.gov and clinicaltrials.gov.
Classification of idiopathic inflammatory myopathies (IIM)
  • European Neuromuscular Centre (ENMC) clinico-sero-pathological classification divides IIM into 4 subclasses, which are associated with myositis specific antibodies (MSAs), as outlined in Table 1.

  • MSA testing is preferentially performed prior to immune suppression treatment but should also be performed in patients with suspected IIM or interstitial lung disease of unknown etiology without prior MSA testing.

  • Polymyositis no longer exists as an IIM subclass.

Dermatomyositis (DM)
  • A classification of DM cannot be made in the absence of DM rash (Gottron’s sign, Gottron’s papules, heliotrope rash).

  • In the presence of DM-like rash, DM can be diagnosed if a DM-specific autoantibody is present, or if definitive DM muscle biopsy features are present and are combined with clinical signs of proximal muscle weakness or elevated muscle enzymes.

    • - Definitive DM muscle biopsy findings: perifascicular atrophy and/or perifascicular MxA overexpression (Fig. 1) with rare or absent perifascicular necrosis.

    • - Suggestive DM muscle biopsy findings: lymphocytic infiltrates (often perivascular), evidence of perifascicular disease (perifascicular predominant fibers that are pale on COX staining and/or positive on NCAM staining).

    • - DM specific autoantibodies: Mi2, NXP2, TIF1γ, MDA5, SAE. Patients are subclassified according to that autoantibody (e.g., anti-TIF1γ DM, anti-NXP2 DM). Patients who have DM without a DM-specific autoantibody are subclassified as having “autoantibody negative DM.”

Antisynthetase syndrome (ASyS)
  • Defined by the presence of an antibody to aminoacyl tRNA synthetase (ARS), together with a single or combination of the following clinical manifestations:

    • - Myositis

    • - Polyarthritis

    • - Interstitial lung disease

    • - Mechanic hands

    • - Raynaud phenomenon

  • ASyS patients with a DM-like rash are not classified as DM, but as “ASyS with a DM-like rash.”

  • Pathology is characterized by:

    • - Perifascicular pathology with necrotic myofibers and nonnecrotic fibers with sarcolemmal C5b-9 (MAC) expression

    • - Perimysial connective tissue with substantial edema, fragmentation, and mixed mononuclear inflammation

    • - MHC1 sarcoplasmic expression and nuclear actin inclusions in myofibers

Immune mediated necrotizing myopathies (IMNM)
  • Anti-HMGCR and anti-SRP autoantibodies are considered specific for IMNM.

  • Patients with anti-HMGCR antibody and a DM-like rash will be classified as having “anti-HMGCR myopathy with a DM-like rash,” while patients with anti-SRP antibody and a DM-like rash will be classified as having “anti-SRP myopathy with a DM-like rash.”

  • Pathology requires diffusely scattered necrotic fibers at different stages of resolution and macrophage dominant, pauci-lymphocytic inflammation.

Sporadic inclusion body myositis (sIBM)
  • The pathologic criteria for sIBM include:

    • - Endomysial T cell inflammation with invasion of non-necrotic fibers

    • - Rimmed vacuoles

    • - P62 or TDP-43 positive protein aggregates or 15–18 nm filaments (tubulofilamentous inclusions) on electron microscopy

  • Anti-cN1A autoantibody is present in 30%–70% of sIBM patients but has also been found in DM and other systemic autoimmune diseases such as Sjögren’s and lupus.

  • Clinically, elderly patients with asymmetric muscle weakness and atrophy of proximal and distal muscle groups, with predilection for wrist and finger flexors and knee extensors.

  • Usually refractory to immunosuppressive therapies.

Limb girdle muscular dystrophies (LGMD)
  • The definition and nomenclature of LGMD have been re-defined in the 2017 ENMC international workshop as a genetically inherited condition that primarily affects skeletal muscle leading to progressive, predominantly proximal muscle weakness at presentation caused by a loss of muscle fibers.

  • All LGMD subclasses must fulfill all of the following:

    • - Described in at least two unrelated families

    • - Patients have achieved independent walking (to differentiate from congenital muscular dystrophies)

    • - Elevated serum creatine kinase

    • - Degenerative changes on muscle imaging over the course of disease

    • - Dystrophic changes on muscle histology, ultimately leading to end-stage pathology

  • New nomenclature: change from the alphanumeric system to include the name of the affected protein and mode of inheritance (D for dominant, R for recessive, X for X-linked); examples listed in Table 2.

  • Some previous LGMD subclasses no longer fulfill the new LGMD definition (Table 3).

Myofibrillar myopathies (MFM)
  • MFM is a group of disorders associated with myofibrillar degradation that begins in the Z disk.

  • Histologically characterized by sarcoplasmic pleomorphic amorphous, granular or hyaline protein aggregates on Gomori trichrome (Fig. 2), and positive for desmin immunostain.

  • Most contain mutations in Z disk associated protein coding genes: DES, CRYAB, MYOT, ZASP, FLNC, BAG3.

  • Patients with mutations in FHL1, DNAJB6, HSBP8, TTN, ACTA1, PLEC, and LMNA have also been associated with MFM phenotype.

Myotonic dystrophy (DM1/DM2)
  • Autosomal dominant multi-system diseases with the common features of myotonia and progressive muscle weakness. There are two main forms: DM1 and DM2.

  • DM1 is caused by CTG trinucleotide repeats in the 3′ untranslated region of DMPK. DM1 shows striking anticipation, with age at onset decreasing by 20–30 years per generation.

  • DM1 muscle pathology is characterized by markedly increased internalized nuclei, often in chains and ring fibers, in a background of chronic myopathy.

  • DM2 is caused by CCTG repeat expansion in intron 1 of CNBP (ZNF9). Additionally, CLCN1 and SCN4A are disease modifying genes whose mutations may exaggerate DM2 phenotype; they therefore should be included in DM2 genetic screening.

  • DM2 muscle pathology is characterized by type 2 atrophy and frequent internalized nuclei predominantly in type 2 fibers.

Gene replacement therapies for hereditary neuromuscular diseases
  • Adeno-associated virus (AAV) based gene delivery vectors can produce replacement proteins in patients with loss of function mutations, such as spinal muscular atrophy (SMA) or Duchenne’s muscular dystrophy. The vector does not integrate into the patient’s genome and has a low immunogenicity.

  • CRISPR-Cas9-mediated gene editing does incorporate into the patient’s genome and can permanently replace a deleterious mutation in patient with conditions such as hereditary transthyretin-mediated (hATTR) amyloidosis.

  • Both methods entail only a single intravenous injection and thus have a clear advantage over siRNA based and antisense oligonucleotide (ASO) based therapies, which require serial infusions.

FDA approved gene therapy for neuromuscular diseases
  • Zolgensma (Novartis) is the first ever FDA approved (2019), intravenously delivered, AAV9 vector mediated SMN gene therapy for spinal muscular atrophy.

New gene therapies currently in clinical trials
  • NTLA-2001 is a CRISPR-Cas9-mediated gene editing construct that targets hATTR amyloidosis in a phase II–III trial for adults with polyneuropathy or cardiomyopathy (NCT04601051).

  • SRP-9001 (Sarepta), SGT-001 (Solid Biosciences) and PF-06939926 (Pfizer) are AAV based micro-dystrophin constructs in phase III trials for DMD (NCT03375164, NCT03769116, NCT04281485).

  • SPK-3006 (Spark Therapeutics) is an AAV based human GAA gene construct in a phase I/II trial for adult onset Pompe disease (NCT04093349).

Meet the Author
Dr. Chunyu Cai has been part of the PathologyOutlines.com editorial board and the Deputy Editor in Chief for Neuropathology since 2020. He is a pathologist and an Associate Professor at University of Texas Southwestern Medical Center. His research focuses on neuromuscular diseases and brain tumors.
Fig. 1
Sarcoplasmic MxA expression is considered a sensitive and specific marker for dermatomyositis.
jptm-2022-10-14f1.jpg
Fig. 2
Myofibrillar myopathy on Gomori trichrome stain.
jptm-2022-10-14f2.jpg
Table 1
IIM subtypes and their associated autoantibodies
Dermatomyositis (DM) Mi2, NXP2, TIF1γ, MDA5, SAE
Inclusion body myositis (IBM) cN1A*
Immune mediated necrotizing myopathy (IMNM) SRP, HMGCR
Anti-synthetase syndrome (ASys) Jo-1, PL7, PL12, EJ, OJ, KS, Zo, Ha

* MSAs are usually mutually exclusive and specific for IIM subclasses, with the exception of cN1A

Table 2
Old vs new LGMD nomenclature
Previous name Gene New name
LGMD 1D DNAJB6 LGMD D1 DNAJB6-related
LGMD 1I CAPN LGMD D4 Calpain3-related
LGMD 2A CAPN LGMD R1 Calpain3-related
LGMD 2B DYSF LGMD R2 Dysferlin-related
Table 3
Previous LGMD subtypes that are no longer considered LGMD
Previous name Gene New name
LGMD 1A MYOT Myofibrillar myopathy
LGMD 1B LMNA Emery–Dreifuss muscular dystrophy
LGMD 1C CAV3 Rippling muscle disease
LGMD 1E and 2R DES Myofibrillar myopathy
LGMD 1H unknown n/a
LGMD 2V GAA Pompe disease/acid maltase deficiency

n/a, not available

Figure & Data

References

    Citations

    Citations to this article as recorded by  

      • PubReader PubReader
      • ePub LinkePub Link
      • Cite this Article
        Cite this Article
        export Copy Download
        Close
        Download Citation
        Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

        Format:
        • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
        • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
        Include:
        • Citation for the content below
        What’s new in neuromuscular pathology 2022: myopathy updates and gene therapies
        J Pathol Transl Med. 2023;57(1):79-80.   Published online December 20, 2022
        Close
      • XML DownloadXML Download
      Figure
      Related articles

      J Pathol Transl Med : Journal of Pathology and Translational Medicine