Skeletal muscle magnetic resonance biomarkers correlate with function and sentinel events in Duchenne muscular dystrophy

Alison M Barnard, Rebecca J Willcocks, Erika L Finanger, Michael J Daniels, William T Triplett, William D Rooney, Donovan J Lott, Sean C Forbes, Dah-Jyuu Wang, Claudia R Senesac, Ann T Harrington, Richard S Finkel, Barry S Russman, Barry J Byrne, Gihan I Tennekoon, Glenn A Walter, H Lee Sweeney, Krista Vandenborne, Alison M Barnard, Rebecca J Willcocks, Erika L Finanger, Michael J Daniels, William T Triplett, William D Rooney, Donovan J Lott, Sean C Forbes, Dah-Jyuu Wang, Claudia R Senesac, Ann T Harrington, Richard S Finkel, Barry S Russman, Barry J Byrne, Gihan I Tennekoon, Glenn A Walter, H Lee Sweeney, Krista Vandenborne

Abstract

Objective: To provide evidence for quantitative magnetic resonance (qMR) biomarkers in Duchenne muscular dystrophy by investigating the relationship between qMR measures of lower extremity muscle pathology and functional endpoints in a large ambulatory cohort using a multicenter study design.

Methods: MR spectroscopy and quantitative imaging were implemented to measure intramuscular fat fraction and the transverse magnetization relaxation time constant (T2) in lower extremity muscles of 136 participants with Duchenne muscular dystrophy. Measures were collected at 554 visits over 48 months at one of three imaging sites. Fat fraction was measured in the soleus and vastus lateralis using MR spectroscopy, while T2 was assessed using MRI in eight lower extremity muscles. Ambulatory function was measured using the 10m walk/run, climb four stairs, supine to stand, and six minute walk tests.

Results: Significant correlations were found between all qMR and functional measures. Vastus lateralis qMR measures correlated most strongly to functional endpoints (|ρ| = 0.68-0.78), although measures in other rapidly progressing muscles including the biceps femoris (|ρ| = 0.63-0.73) and peroneals (|ρ| = 0.59-0.72) also showed strong correlations. Quantitative MR biomarkers were excellent indicators of loss of functional ability and correlated with qualitative measures of function. A VL FF of 0.40 was an approximate lower threshold of muscle pathology associated with loss of ambulation.

Discussion: Lower extremity qMR biomarkers have a robust relationship to clinically meaningful measures of ambulatory function in Duchenne muscular dystrophy. These results provide strong supporting evidence for qMR biomarkers and set the stage for their potential use as surrogate outcomes in clinical trials.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Representative T 2 maps, 1…
Fig 1. Representative T2 maps, 1H MRS voxel placement, and 1H MR spectra.
A) T2 maps were created from the 40, 60, 80, and 100ms TE spin echo images of the lower leg and thigh. Utilizing the popliteus and biceps femoris short head as standardized anatomical landmarks to guide lower leg and thigh slice selection, the TA, PER, TP, SOL, MG, VL, BFLH, and GRA muscles were traced on three contiguous slices to determine mean T2 for each muscle. B) T1-weighted images demonstrate the location of voxel placement for MR spectroscopy used to quantify FF. Voxels were sized as large as possible while staying completely within the SOL and VL muscles, avoiding the fascia. The voxel position from prior years was referenced to position voxels at an anatomically similar location at follow-up visits. C-F) T1-weighted images of the calf and thigh with corresponding SOL and VL spectra and FFs from a representative participant in each of four functional groups (based on the 10m walk/run test). Participants have increasing levels of SOL and VL fat infiltration (appreciated visually on the T1-weighted images and quantified using spectroscopy) with decreasing ambulatory ability. 10m walk/run times were 3.56s, 7.16s, and 11.32s for the example participants in panels C, D, and E, respectively. (TA = tibialis anterior, PER = peroneus longus and brevis, TP = tibialis posterior SOL = soleus, MG = medial gastrocnemius, VL = vastus lateralis, BFLH = biceps femoris long head, GRA = gracilis, FF = fat fraction).
Fig 2. Scatterplots showing the correlations between…
Fig 2. Scatterplots showing the correlations between VL MRI T2 and FF and functional endpoints.
The qMR biomarkers most strongly correlated to function were VL MRI T2 and VL FF. A) VL MRI T2 versus 10m walk/run, B) VL FF versus 10m walk/run, C) VL MRI T2 versus 6MWD, D) VL FF vs 6MWD. Participants who lost the ability to perform the timed tests were given the maximum time of 45 seconds (open circles) for the first visit in which they were unable to complete the test. Participants who lost the ability to perform the 6 MWT have distances of 0 m (open circles). As indicated in Table 2, all correlations were significant with the 95% bootstrap confidence intervals excluding zero. (VL = vastus lateralis, 6MWD = six minute walk distance, 6MWT = six minute walk test).
Fig 3. FF and quantitative and qualitative…
Fig 3. FF and quantitative and qualitative measures of walking/running ability.
A and B) FF was assessed based on 10m walk/run performance. Data points from participants who could complete the test were divided into bins of 8 sec (n = 127). The LOA group represents data points from subjects that could no longer complete the 10m walk/run at the time of the visit (n = 35). The amount of SOL and VL fatty infiltration was significantly different between functional performance groups. C) Groups were created based on the qualitative rating of participants’ 10m walk/run test at each time point. Mean VL FF was significantly different between each qualitative grade group. * = significantly different; bootstrap confidence intervals excluded zero. (SOL = soleus, VL = vastus lateralis).
Fig 4. Loss of function in participants…
Fig 4. Loss of function in participants with different VL FFs.
Data points were binned by VL FF and further divided by the number of functional skills the participants could complete at the time of the visit. Black = ambulatory, able to climb 4 stairs, able to move supine to stand. Dark gray = ambulatory, able to climb 4 stairs, unable to rise from supine. Light gray = ambulatory, unable to perform stairs, unable to rise from supine. White = nonambulatory. As FF increased, there was a progressive decrease in the percent of participants who could perform the functional tasks. At VL FFs less than 0.19, 99% of participants could perform all functional tests. Apart from one outlier, participants began losing ambulation at VL FFs ≥ 0.40. (VL = vastus lateralis, STS = supine to stand).
Fig 5. qMR biomarkers of muscle pathology…
Fig 5. qMR biomarkers of muscle pathology at the first visit following loss of ambulation.
A) MRI T2 and B) MRS FF are plotted for each muscle at the first time point at which participants were nonambulatory (n = 35). Whiskers represent the 5th-95th percentiles with outliers represented by the filled dots. MRI T2 was highest in the BFLH and VL and lowest in the GRA, TA, and TP. The MRI T2 and FF of the VL were higher than that of the SOL as expected based on the proximal to distal pattern of involvement in DMD.

References

    1. Ruegg U. Pharmacological prospects in the treatment of Duchenne muscular dystrophy. Curr Opin Neurol. 2013;26: 577–584. doi:
    1. Reinig AM, Mirzaei S, Berlau DJ. Advances in the treatment of Duchenne muscular dystrophy: new and emerging pharmacotherapies. Pharmacother. 2017;37: 492–499.
    1. Lynn S, Aartsma-Rus A, Bushby K, Furlong P, Goemans N, De Luca A, et al. Measuring clinical effectiveness of medicinal products for the treatment of Duchenne muscular dystrophy. Neuromuscul Disord. 2015;25: 96–105. doi:
    1. Finanger EL, Russman B, Forbes SC, Rooney WD, Walter GA, Vandenborne K. Use of skeletal muscle MRI in diagnosis and monitoring disease progression in Duchenne muscular dystrophy. Phys Med Rehabil Clin N Am. 2012;23: 1–ix. doi:
    1. Triplett WT, Baligand C, Forbes SC, Willcocks RJ, Lott DJ, DeVos S, et al. Chemical shift-based MRI to measure fat fractions in dystrophic skeletal muscle: MR measurements of fat fraction in dystrophic muscles. Magn Reson Med. 2014;72: 8–19. doi:
    1. Gaeta M, Scribano E, Mileto A, Mazziotti S, Rodolico C, Toscano A, et al. Muscle fat fraction in neuromuscular disorders: dual-echo dual-flip-angle spoiled gradient-recalled MR imaging technique for quantification—a feasibility study. Radiology. 2011;259: 487–494. doi:
    1. Forbes SC, Willcocks RJ, Triplett WT, Rooney WD, Lott DJ, Wang DJ, et al. Magnetic resonance imaging and spectroscopy assessment of lower extremity skeletal muscles in boys with Duchenne muscular dystrophy: a multicenter cross sectional study. PLoS ONE. 2014;9: e106435 doi:
    1. Hollingsworth KG, Garrood P, Eagle M, Bushby K, Straub V. Magnetic resonance imaging in Duchenne muscular dystrophy: longitudinal assessment of natural history over 18 months. Muscle Nerve. 2013;48: 586–588. doi:
    1. Wokke BH, van den Bergen JC, Versluis MJ, Niks EH, Milles J, Webb AG, et al. Quantitative MRI and strength measurements in the assessment of muscle quality in Duchenne muscular dystrophy. Neuromuscul Disord. 2014;24: 409–416. doi:
    1. Dixon WT. Simple proton spectroscopic imaging. Radiology. 1984;153: 189–194. doi:
    1. Kim HK, Laor T, Horn PS, Racadio JM, Wong B, Dardzinski BJ. T2 mapping in Duchenne muscular dystrophy: distribution of disease activity and correlation with clinical assessments 1. Radiology. 2010;255: 899–908. doi:
    1. Arpan I, Forbes SC, Lott DJ, Senesac CR, Daniels MJ, Triplett WT, et al. T2 mapping provides multiple approaches for the characterization of muscle involvement in neuromuscular diseases: a cross-sectional study of lower leg muscles in 5–15-year-old boys with Duchenne muscular dystrophy. NMR Biomed. 2013;26: 320–328. doi:
    1. Maillard SM. Quantitative assessment of MRI T2 relaxation time of thigh muscles in juvenile dermatomyositis. Rheumatology. 2004;43: 603–608. doi:
    1. Forbes SC, Walter GA, Rooney WD, Wang DJ, DeVos S, Pollaro J, et al. Skeletal muscles of ambulant children with Duchenne muscular dystrophy: validation of multicenter study of evaluation with MR imaging and MR spectroscopy. Radiology. 2013;269: 198–207. doi:
    1. Kim HK, Serai S, Lindquist D, Merrow AC, Horn PS, Kim DH, et al. Quantitative skeletal muscle MRI: part 2, MR spectroscopy and T2 relaxation time mapping—comparison between boys with Duchenne muscular dystrophy and healthy boys. Am J Roentgenol. 2015;205: W216–W223.
    1. Johnston JH, Kim HK, Merrow AC, Laor T, Serai S, Horn PS, et al. Quantitative skeletal muscle MRI: part 1, derived T2 fat map in differentiation between boys with Duchenne muscular dystrophy and healthy boys. Am J Roentgenol. 2015;205: W207–W215.
    1. Willcocks RJ, Arpan IA, Forbes SC, et al. Longitudinal measurements of MRI-T2 in boys with Duchenne muscular dystrophy: effects of age and disease progression. Neuromuscul Disord. 2014;24: 393–401. doi:
    1. Willcocks RJ, Rooney WD, Triplett WT, Forbes SC, Lott DJ, Senesac CR, et al. Multicenter prospective longitudinal study of magnetic resonance biomarkers in a large Duchenne muscular dystrophy cohort. Ann Neurol. 2016;79: 535–547. doi:
    1. Godi C, Ambrosi A, Nicastro F, Previtali SC, Santarosa C, Napolitano S, et al. Longitudinal MRI quantification of muscle degeneration in Duchenne muscular dystrophy. Ann Clin Transl Neurol. 2016;3: 607–622. doi:
    1. Arpan I, Willcocks RJ, Forbes SC, Finkel RS, Lott DJ, Rooney WD, et al. Examination of effects of corticosteroids on skeletal muscles of boys with DMD using MRI and MRS. Neurology. 2014;83: 974–980. doi:
    1. Aartsma-Rus A, Ferlini A, Vroom E. Biomarkers and surrogate endpoints in Duchenne: meeting report. Neuromuscul Disord. 2014;24: 743–745. doi:
    1. Fleming TR, Powers JH. Biomarkers and surrogate endpoints in clinical trials. Stat Med. 2012;31: 2973–2984. doi:
    1. Bonati U, Hafner P, Schädelin S, Schmid M, Naduvilekoot Devasia A, Schroeder J, et al. Quantitative muscle MRI: a powerful surrogate outcome measure in Duchenne muscular dystrophy. Neuromuscul. Disord. 2015;25: 679–685. doi:
    1. Fischmann A, Hafner P, Gloor M, Schmid M, Klein A, Pohlman U, et al. Quantitative MRI and loss of free ambulation in Duchenne muscular dystrophy. J Neurol. 2013;260: 969–974. doi:
    1. Gaeta M, Messina S, Mileto A, Vita GL, Ascenti G, Vinci S, et al. Muscle fat-fraction and mapping in Duchenne muscular dystrophy: evaluation of disease distribution and correlation with clinical assessments: preliminary experience. Skeletal Radiol. 2012;41: 955–961. doi:
    1. Torriani M, Townsend E, Thomas BJ, Bredella MA, Ghomi RH, Tseng BS. Lower leg muscle involvement in Duchenne muscular dystrophy: an MR imaging and spectroscopy study. Skeletal Radiol. 2012;41: 437–445. doi:
    1. Wren TAL, Bluml S, Tseng-Ong L, Gilsanz V. Three-point technique of fat quantification of muscle tissue as a marker of disease progression in Duchenne muscular dystrophy: preliminary study. Am J Roentgenol. 2008;190: W8–W12.
    1. Mankodi A, Bishop CA, Auh S, Newbould RD, Fischbeck KH, Janiczek RL. Quantifying disease activity in fatty-infiltrated skeletal muscle by IDEAL-CPMG in Duchenne muscular dystrophy. Neuromuscul Disord. 2016;26: 650–658. doi:
    1. McDonald CM, Henricson EK, Abresch RT, Florence JM, Eagle M, Gappmaier E, et al. The 6-minute walk test and other endpoints in Duchenne muscular dystrophy: longitudinal natural history observations over 48 weeks from a multicenter study. Muscle Nerve. 2013;48: 343–356. doi:
    1. Bushby K, Finkel R, Wong B, Barohn R, Campbell C, Comi G, et al. Ataluren treatment of patients with nonsense mutation dystrophinopathy. Muscle Nerve. 2014;50: 477–487. doi:
    1. Florence JM, Pandya S, King WM, Robison JD, Signore LC, Wentzell M, et al. Clinical trials in Duchenne dystrophy. Standardization and reliability of evaluation procedures. Phys Ther. 1984;64: 41–45.
    1. McDonald CM, Henricson EK, Abresch RT, Florence J, Eagle M, Gappmaier E, et al. The 6-minute walk test and other clinical endpoints in Duchenne muscular dystrophy: reliability, concurrent validity, and minimal clinically important differences from a multicenter study. Muscle Nerve. 2013;48: 357–368. doi:
    1. McDonald CM, Henricson EK, Han JJ, Abresch RT, Nicorici A, Elfring GL, et al. The 6-minute walk test as a new outcome measure in Duchenne muscular dystrophy. Muscle Nerve. 2010;41: 500–510. doi:
    1. Efron B, Tibshirani RJ. An introduction to the bootstrap Chapman & Hall/CRC; 1994.
    1. Pane M, Mazzone ES, Fanelli L, De Sanctis R, Bianco F, Sivo S, et al. Reliability of the performance of upper limb assessment in Duchenne muscular dystrophy. Neuromuscul Disord. 2014;24: 201–206. doi:
    1. Pane M, Mazzone ES, Sivo S, Sormani MP, Messina S, D’Amico A, et al. Long term natural history data in ambulant boys with Duchenne muscular dystrophy: 36-month changes. PLoS ONE. 2014;9: e108205 doi:
    1. Lu Q, Cirak S, Partridge T. What can we learn from clinical trials of exon skipping for DMD? Mol Ther Acids. 2014;3: e152.
    1. Lott DJ, Forbes SC, Mathur S, Germain SA, Senesac CR, Sweeney HL, et al. Assessment of intramuscular lipid and metabolites of the lower leg using magnetic resonance spectroscopy in boys with Duchenne muscular dystrophy. Neuromuscul Disord. 2014;24: 574–582. doi:
    1. D’Angelo MG, Berti M, Piccinini L, Romei M, Guglieri M, Bonato S, et al. Gait pattern in Duchenne muscular dystrophy. Gait Posture. 2009;29: 36–41. doi:
    1. Morrow JM, Sinclair CDJ, Fischmann A, Machado PM, Reilly MM, Yousry T, et al. MRI biomarker assessment of neuromuscular disease progression: a prospective observational cohort study. Lancet Neurol. 2016;15: 65–77. doi:
    1. Willis TA, Hollingsworth KG, Coombs A, Sveen ML, Anderson S, Stojkovic T, et al. Quantitative muscle MRI as an assessment tool for monitoring disease progression in LGMD2I: a multicentre longitudinal study. PLoS ONE. 2013;8: e70993 doi:
    1. Andersen G, Dahlqvist JR, Vissing CR, Heje K, Thomsen C, Vissing J. MRI as outcome measure in facioscapulohumeral muscular dystrophy: 1-year follow-up of 45 patients. J Neurol. 2017;264: 438–447. doi:

Source: PubMed

3
Subscribe