Sundhedsudvalget 2024-25
SUU Alm.del Bilag 187
Offentligt
2990308_0001.png
Sundhedsudvalget
Til
Indenrigs- og sundhedsminister
Sophie Løhde
Kære Jørgen Nielsen, Louise Brown, Peder Hvelplund, Rasmus Lund-Nielsen &
Mathilde Powers.
og sundhedsminister Sophie Løhde
Kære Indenrigs-
24.2.2025
Vedr. ME/CFS vidensafdækningen: Principielle fejl og mangler
ME Foreningen ønsker hermed at udtrykke vores bekymring over den snart afsluttede vidensafdækning
af ME/CFS, som sundhedsministeren igangsatte i 2024. Vi anerkender ministerens intention om at sikre
en grundig og evidensbaseret afdækning, men vi må desværre konstatere, at processen hidtil ikke lever
op til de forventninger, der med rette kan stilles til en så vigtig indsats.
Vores kommentarer beror på de to eksterne leverandørers (Defactum og Implement) præsentationer af
deres foreløbige resultater hhv. d. 22.11.24 og 04.02.25 (vores skriftlige feedback til disse
præsentationer ses i bilag 1 og 2). Rapporterne forventes offentliggjort af Sundhedsstyrelsen i
begyndelsen af marts 2025.
1. Manglende inddragelse af ME-patienter og ME-eksperter
På samrådsmødet den 28. november 2023 understregede ministeren nødvendigheden af at inddrage
ME Foreningen og ME-patienterne. Alligevel blev kun to ME-patienter interviewet af Implement (+ en
repræsentant for ME foreningen). Kun én af de to af ME Foreningen foreslåede ME-eksperter blev
interviewet, og vedkommende har siden sendt en kritisk vurdering af leverandørens foreløbige
resultater, mens Defactum kun indledningsvist inddrog en repræsentant ifm. udarbejdelse af
projektbeskrivelse. Desuden er væsentlig international viden ikke integreret i arbejdet til trods for, at
ME Foreningen løbende har stået til rådighed med litteratur.
2. Ensidigt fokus og mangelfuld litteraturudvælgelse
Vidensafdækningen blev oprindeligt placeret i den afdeling i Sundhedsstyrelsen, der har ansvaret for
funktionelle lidelser
en beslutning, vi fandt påfaldende, da vidensafdækningen ikke skulle omhandle
funktionelle lidelser. Senere blev opgaven dog overdraget til afdelingen for evidensbaseret medicin,
men det reviderede udbudsmateriale udelod det grundlæggende spørgsmål om, hvorvidt ME er en
somatisk eller funktionel lidelse. Denne udeladelse har medført, at op mod 80 % af den biomedicinske
litteratur om ME, som er afgørende for korrekt diagnostik og behandling, er ekskluderet i
vidensafdækningen. De to eksterne leverandører har derfor formentlig ikke anvendt den specifikke,
internationalt anerkendte diagnose ME/CFS, (G93.3), men har i stedet fokuseret på ”fatigue”, dvs.
kronisk træthed eller udmattelse, som herhjemme kategoriseres som en funktionel lidelse under
betegnelsen ”almen træthed” med en anden diagnosekode
(DR688A9).
3. Metodiske mangler og brug af forældede studier
Det ser ikke ud til, at de eksterne leverandørers to rapporter vil belyse ME/CFS biomedicinske natur,
herunder mulige årsager og risikofaktorer, da afklaringen af dette er udeladt i vidensafdækningen. Også
sygdommens epidemiologi (forekomst) afhængig af diagnostiske kriterier og alvorsgrad er udeladt.
Dermed bliver det heller ikke muligt at sammenligne diagnosticeringen og behandlingsindsatsen for
ME/CFS i Danmark med vores nabolande, da sygdommen i disse lande kategoriseres med WHOs
diagnosekode G93.3 og ikke som en funktionel lidelse.
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0002.png
Defactums resultater bygger i høj grad på systematiske reviews, der primært omfatter studier foretaget
før 2011. Disse studier anvender forældede diagnosekriterier, der bl.a. ikke inkluderer PEM
et
nøglesymptom for ME/CFS. Dette svækker resultaternes validitet. Derudover er der inddraget svage,
ikke-blindede studier med subjektive endemål, hvilket yderligere underminerer evidensgrundlaget.
4. Begrænset internationalt perspektiv
Implement formåede ikke at inkludere Storbritannien i deres undersøgelse
det land, hvor
afdækningen af ME-vidensgrundlaget via NICE-rapporten 2021 i dag sætter standarden for ME i andre
lande. Desuden er vigtige internationale konsensusudtalelser og guidelines fra blandt andet USA,
Belgien, Tyskland, Østrig og Schweiz ikke tilstrækkeligt inddraget. Denne manglende inddragelse
forringer muligheden for, at Danmark kan sammenligne sin praksis med internationale standarder.
5. Mangel på gennemsigtighed
ME Foreningen har gentagne gange anmodet om mulighed for at gennemlæse og kommentere de
endelige rapporter fra Defactum og Implement inden offentliggørelse. Disse anmodninger er blevet
afvist, hvilket rejser væsentlige spørgsmål om processens transparens.
6. Konsekvenser for danske patienter
Med den nuværende tilgang risikerer danske ME/CFS-patienter at blive diagnosticeret og behandlet på
et niveau, der ikke er i overensstemmelse med internationale standarder. Det kan betyde, at patienter i
Danmark afskæres fra at få adgang til de nyeste behandlingsmuligheder. Vi kan på baggrund af de to
eksterne leverandørers vidensafdækning konstatere, at den internationale WHO-diagnose (G93.3) for
sygdommen, som resten af verden bruger, i praksis ikke anvendes i Danmark, mens Defactums
fremlæggelse lige så klart viste, at de behandlinger, som anbefales til ME/CFS i Danmark (Cognitive
behavioral therapy og Graded exercise therapy) ikke har nogle positive langtidseffekter, og da heller
ikke anbefales internationalt som helbredende behandling for ME/CFS.
Afsluttende bemærkninger
På baggrund af ovenstående vurderer ME Foreningen, at vidensafdækningen ikke giver et retvisende
billede af den nuværende viden om ME/CFS. Vi finder det særlig problematisk, at vidensafdækningen
ikke udfordrer den danske tilgang til ME som en funktionel lidelse. Dette står i kontrast til internationale
standarder og risikerer at fastholde danske patienter i en uholdbar og effektløs behandlingssituation.
Det er vores håb, at denne tilbagemelding vil blive taget i betragtning i det fremtidige arbejde på
området, der bør omfatte etablering af en arbejdsgruppe med deltagelse af internationale ME-
eksperter således at danske ME/CFS-patienter kan få adgang til diagnose og behandling på linje med
patienter i andre lande. Til slut vil vi gerne henvise til vedhæftede diagnose- og behandlingsvejledning
udarbejdet af ME-ekspert Jesper Mehlsen (se bilag 3, obs. ikke offentliggjort endnu).
ME Foreningen står naturligvis fortsat til rådighed for dialog og vidensdeling, der kan bidrage til en
forbedret praksis fremover.
Med venlig hilsen
ME Foreningen samt medlemmer i ME foreningens Advisory Board (Bernard Jeune, Lektor emeritus,
Epidemiologi, Biostatistik og biodemografi (EBB), Jesper Mehlsen, Klinisk fysiolog
Peter la Cour, prof. Sundhedspsykolog og Vibeke Vind, senior scientist, biokemiker).
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0003.png
Bilag 1
Opfølgning på ’Præsentation af foreløbige resultater fra vidensafdækningen
af ME/CFS’ afholdt i Sundhedsstyrelsen d. 22.11.24
På baggrund af mødet sender vi her en række bekymringspunkter, som også blev fremhævet ved
mødet. Vi håber meget, at I vil tage disse med i det videre arbejde med vidensafdækningerne, så
kvalitet og relevans sikres.
Vi er i særdeleshed bekymrede for de resultater, der blev præsenteret fra Implements afdækning af
erfaringer i relevante lande med organisering af behandlingstilbud samt diagnosticering og behandling
af ME/CFS. Afdækningen besvarer i sin nuværende form på ingen måde det grundspørgsmål, der blev
stillet, ligesom den heller ikke forholder sig kritisk til metode eller inkluderer den viden, der er
tilgængelig. Vi vil understrege, at den oprindelige opgave, der blev stillet af ministeren, var en
afdækning af ME – ikke af Funktionelle lidelser (FL):
”Der er afsat 1,5 millioner kroner i 2024 til en afdækning af evidensgrundlaget for diagnosticering og
behandling af lidelsen ME/CFS (Myalgisk Encephalomyelitis/Chronic Fatigue Syndrome).
Vidensafdækningen bør inddrage international forskning samt erfaringer i relevante lande. I
vidensafdækningen bør inddrages relevante aktører med faglig viden om ME/CFS.”
Dette understreges af ministerens udtalelse på samråd d. 28.11.23 fra Sophie Løhde (Minuttal: 39.40):
Samråd d. 28.11.23
”Funktionelle lidelser er ikke en del af den afdækning, som vi sætter i gang nu. Vi har afsat 1,5 mio. kr.
til vidensafdækning med henblik på at få afdækket evidensgrundlaget for diagnosticering og
behandling af ME og sørger dermed også for at få inddraget international forskning og erfaring fra andre
lande – og at der også inddrages relevante aktører i forbindelse med vidensafdækningen. Dette vil ske
under indkøb af en ekstern leverandør. Vidensafdækningen kommer til at være offentlig tilgængelig.”
Selvom I i Implement søger neutrale beskrivelser, så bør I som vidensorganisation ikke fravælge
hverken en kritisk beskrivelse og tilgang eller at gøre rede for mulige fremtidstendenser og
fremtidsscenarier. Det skal dertil siges, at vi fra ME-foreningen løbende har understøttet med relevant
litteratur og nyeste viden, fx fra de lande, der indgår i afdækningen. Vi fremsender gerne litteraturen
igen. Hvis dette ikke nuanceres, kan vi som forening kun tage afstand fra rapportens metode og
resultater – også offentligt.
Nedenfor følger en række yderligere punkter til hhv. Implements og Defactums præsentationer
samt opsummerende bemærkninger til Sundhedsstyrelsen.
Til Implements afdækning:
Afdækningen beskriver behandlingsforløb for en gruppe patienter, som givetvis ikke har ME, men
FL, da man ikke har forholdt sig til konsensuskriterierne for ME. Hvis der faktisk findes patienter
med ME i FL-gruppen, må de tilhøre den absolut lettest ramte del af ME-spektret, idet de ellers ikke
vil kunne klare de krævende gruppeforløb og transporten til og fra. Der mangler generelt klarhed
over, hvor mange ME-patienter der har en FL-diagnose eller er blevet diagnosticeret på FL-centre.
1
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0004.png
Afdækningen må holde sig til beskrivelsen af forekomsten af WHO-diagnosekoden for ME og ikke
for FL. Når det drejer sig om beskrivelsen af FL, som selvfølgelig må inddrages, da ME pt. ligger
under disse, må det være for at afklare, hvilke af disse patienter der har ME. Hvis det ikke kan
afklares, må det beskrives, hvorfor det ikke kan opregnes. Landspatientregistrets registrering af
WHO-koden for ME må gennemgås (og gerne på hvilke afdelinger denne er registreret). Det bør
undersøges, hvor mange af de 15.000-20.000 ME-patienter i Danmark (ifølge prævalens) der har
ME-diagnosen, hvor de er i systemet, hvilke behandlinger de får, hvor mange der er syge og
sengeliggende, og hvor mange der er børn under 16 år. Ligesom Implement bør beskrive Mehlsen-
klinikkens ME-patientmateriale. Her findes oplysninger på ca. 1.200 patienter, der er
diagnosticeret regelret efter konsensuskriterierne.
Problematikken ift. de anvendte diagnosekoder kan yderligere udfoldes: Ifølge
diagnosevejledningen for FL kan danske læger stille diagnosen DG933A, men på FL-centrene er
ME diagnosen kun stillet fire gange, hvilket ligger meget langt fra prævalensen for ME (0,2-0,4%).
Det er 100% udtrykkeligt angivet af WHO at de danske FL-diagnoser ikke kan bruges til samkodning
med ME-diagnosen under multiple parenting
i
. I opgørelsen kan der derfor ikke pludselig
argumenteres for, at to FL-diagnoser i virkeligheden er ME DG933A. Ingen ved hvad de dækker over.
Kort sagt: Danske patienter fra FL-centre ved ikke, om de har ME, fordi FL-centrene ikke anvender
opdaterede diagnosekriterier med PEM for DG933A – og der kan derfor ikke interviewes mere end
fire patienter (i perioden frem til 10.05.2021) om ME i dette regi. Relevante patienter m.v. bør derfor
findes i andet regi.
Afdækningen bør inkludere og beskrive de sværest syge, herunder de permanent sengeliggende,
bl.a. ved at søge oplysninger om ME-patienter, som har fået førtidspension, evt. også ved at
inddrage avisartikler i BT, Se og Hør, lokale aviser m.fl. Disse er blandt andet også beskrevet her:
https://www.tandfonline.com/doi/full/10.1080/27707571.2024.2359958.
Afdækningen mangler (ud fra det vi fik præsenteret) status på curriculum om ME på medicin og
speciallægeuddannelsen; status på medicinsk behandling af generelle symptomer og af ME
specialiserede symptomer, og status på hvorvidt kurser i FL omfatter viden om ME med PEM.
Afdækningen bør inddrage den danske del af den europæiske interviewundersøgelser af over 500
danske patienter
https://www.europeanmealliance.org/documents/emeaeusurvey/EMEAMEsurveyreport2024.pdf
https://me-foreningen.dk/wp-content/uploads/2024/11/Paneuropaeisk-danske-tal.pdf
ligesom
der også er værdifuld viden i ME-Foreningens medlemsundersøgelser fra 2008, 2014 og 2019 (vi
stiller gerne data til rådighed,
https://me-foreningen.dk/wp-
content/uploads/2016/10/Sp%C3%B8rgeskemaunders%C3%B8gelse-2012-2.0-kopi.pdf,
https://me-foreningen.dk/wp-content/uploads/2020/06/Medlemssurvey-2019-561-patienter.pdf
).
Der findes også tilsvarende i de omkringliggende lande, ikke mindst den selvstændige norske
rapport af over 5000 norske ME-patienter.
Afdækningen af Tysklands tilgang bør inddrage den konsensusvejledning om diagnostik og
behandling af ME, som Tyskland har lavet i samarbejde med Østrig og Schweiz i maj 2024. Dette D-
A-CH Konsensus statement er grundlaget for det videre arbejde med ME i Tyskland.
https://link.springer.com/article/10.1007/s00508-024-02372-y
Som tidligere rejst, så er det meget beklageligt, at Implement ikke har kunnet få fat i relevante
kilder i UK. I den forbindelse blev en rapport fra 2023 om implementeringen af NICE2021 i UK
tilsendt, som der som minimum bør refereres til, da den giver svar på det meste om ME i UK efter
NICE2021.
2
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0005.png
Endelig bør der også henvises til den nyligt publicerede embedslægerapport fra England om
forebyggelse af dødsfald ved ME
https://www.judiciary.uk/prevention-of-future-death-
reports/maeve-boothby-oneill-prevention-of-future-deaths-report/.
Det blev nævnt på mødet, at
der i Danmark er oprettet to højt specialiserede funktioner til de sygeste patienter, men det er
vigtigt at præcisere, at det er til patienter med funktionel lidelse uden specialiseret ME-behandling.
Ligesom I England har Danmark ikke en eneste sengeplads eller anden sundhedsydelse til
patienter med svær ME.
Til Defactums afdækning:
For at være sikker på, at den inkluderede målgruppe er korrekt, bør søgningen begrænses til
undersøgelser, der inddrager PEM.
Det er i orden at fokusere på systematiske reviews og klinisk kontrollerede forsøg, men
afdækningen må kritisk tage stilling til, hvad der ikke kan/bliver afklaret i disse typer studier. Fx de
mange epidemiologiske undersøgelser, som har sammenlignet hyppigheden på samme stikprøver
afhængigt af, hvilke diagnostiske kriterier man anvender. Dette bør inkluderes i en diskussion af
metoden, som er meget væsentlig.
Til Sundhedsstyrelsen:
Som det også blev fremhævet til mødet, vil vi meget gerne deltage i et opfølgende møde, hvor vi får
mulighed for at se Defactums resultater. Hvis dette ikke er muligt, vil vi meget gerne have
rapporten til kommentering. Det samme gælder for den færdige udgave af Implements rapport.
Vi håber (set i lyset af de mange kommentarer til afdækningerne og generel god skik), at I vil sende
begge rapporter i eksternt review blandt internationale ME-eksperter. Hvis det ikke er muligt at
oversætte rapporterne, bør de som minimum sendes til nordiske eksperter. Vi vil meget gerne
bidrage med forslag til specifikke personer.
Desuden vil vi meget gerne høre mere om processen efter rapporterne er afleveret til jer.
Nedsættes der fx en arbejdsgruppe med SST og eksterne ME-eksperter, der kan trække
konklusioner og evt. anbefalinger til Sundhedsministeren fra de to rapporter? Eller hvad vil
processen være? Dette stod ikke helt klart på mødet.
Endelig vil vi gerne høre, hvad processen er for de to leverandører: Vil de udgive rapporterne?
”Multiple parenting” er en paralelldiagnosticering som WHO kun anbefaler til 2 diagnoser som
supplerer hinanden. ME G93.3 og FL-diagnosen ”almen træthed” udelukker gensidigt hinanden og derfor er
”multiple parenting” ikke et begreb der kan anvendes her. Ifølge WHO må der ikke anvendes 2 koder for samme
sygdom/symptomer: Fx udelukker ME G93.3 – ifølge WHO - diagnoser som MG22 træthed og Neurasteni F48.
ii
3
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0006.png
Bilag 2
Opfølgning på mødet: Præsentation af foreløbige resultater fra
vidensafdækningen af ME/CFS gennemført af Defactum d. 04.02.24
Tak for at holde et opfølgende møde, hvor vi fik præsenteret de foreløbige resultater og fik mulighed for
at komme med vores input. Nedenfor følger en skriftlig tilbagemelding med en uddybning af de
punkter, der blev fremhævet på mødet, som vi håber, I vil inkludere i det videre arbejde. Som tidligere
fremført vil vi meget gerne gennemlæse rapporten og komme med skriftlige kommentarer hertil.
Basal viden om sygdommens karakter:
Der er en gennemgående og afgørende mangel på basal viden om sygdommen.
Hvis man
ønsker en hurtig og umiddelbar introduktion, kan man fx spørge OpenAI, som opsummerer det
meget fint. Se bilag 1 til dette dokument.
OpenAI’s rapport giver et solidt biokemisk og patofysiologisk grundlag for forståelsen af ME og
PEM. Det kunne være relevant for Sundhedsstyrelsen at læse denne opsummering
1
.
Som immunologen Derya Unutmaz fra Jackson Laboratory udtrykker det: OpenAI’s dybdegående
forskningsrapporter er “ekstremt imponerende, troværdige og på niveau med eller bedre end
publicerede review-artikler.”
2
Opgavens formål og resultater vedr. diagnose:
Opgaven gik ud på at belyse:”Hvilke
kliniske undersøgelser, parakliniske og diagnostiske test og
kriterier bør indgå ved mistanke om ME”.
Det blev bekræftet, at der ikke findes en simpel biomarkør til diagnosticering, hvilket også gør sig
gældende for mange andre sygdomme, hvor diagnosen stilles på baggrund af kliniske symptomer,
fx migræne.
Manglende inklusion/fokus på diagnosekriterier:
Det fremgik på mødet, at der i rapporten vil
foreligge en række forskellige diagnosekriterier, der bør indgå ved mistanke om ME, men det
fremgik ikke, hvad der vil blive oplyst om dem.
Diagnosekriterierne for ME/CFS har udviklet sig meget de sidste 25 år, idet de er gået fra at være
meget brede kriterier, der medtager træthed af mange typer og af mange årsager og har bevæget
sig frem til langt mere specifikke og dermed snævre kriterier. De internationale konsensuskriterier
(Carruthers et al 2011
3
) er formentlig det afgørende vendepunkt mellem de gamle og de nye
forståelser. Det er her, at PEM bliver et nødvendigt, men ikke tilstrækkeligt hovedsymptom for
https://www.nature.com/articles/d41586-025-00377-9;
https://x.com/DeryaTR_/status/1886487553387430396
3
Carruthers BM, van de Sande MI, De Meirleir KL, et al. Myalgic encephalomyelitis: International Consensus
criteria. J Intern Med. 2011;270(4): 327-38. PMID: 21777306.
1
2
1
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0007.png
ME/CFS. Mayo-klinikkens diagnostiske procedurer for ME/CFS omtaler PEM som patognomonisk
symptom, dvs. både nødvendigt og tilstrækkelig
4
,
5
.
Der er meget store forskelle på de prævalenser, men kommer frem til ved anvendelse af brede og
specifikke kriterier. De anvendte diagnosekriterier er også af afgørende vigtighed for, hvilken gruppe
patienter man undersøger i forhold til behandlinger.
Ved alle andre sygdomme uden biomarkør findes der også en række kliniske kriterier for
sygdommen, fx for demens. Disse kriterier udvikler sig historisk og bliver i reglen mere og mere
specifikke for sygdommen og adskiller den fra andre. Der opstår i tiden konsensus om, hvilke
kriterier, der bør anvendes i forskningen om ME/CFS på nuværende tidspunkt, og kriterierne må
angives specifikt, idet forskning med et sæt kriterier ikke kan sammenlignes direkte med forskning
med andre kriterier.
Som det blev anført på mødet, viser de såkaldte ”cykeltests”, at der kan påvises klare biologiske
forskelle i energiforvaltningen hos patienter defineret nutidsvarende med ME/CFS og immobile
patienter, der lider af andre former for træthed, der kan indfanges af bredere definitioner. De
anvendte kriterier er derfor af yderste vigtighed omkring en nutidig beskrivelse af ME/CFS.
Rapporten bør indeholde en redegørelse for og overblik over udviklingen af (og diskussion om)
diagnosekriterierne for ME/CFS, hvilket er god videnskabelig praksis omkring kliniske
symptomdiagnoser.
Internationale diagnosekriterier:
Der findes forskellige international anerkendte diagnostiske
kriterier, som har udviklet sig over tid, hvor de nyeste alle indeholder symptomet PEM sammen
med en gruppe af andre symptomer. Vores nabolande, der diagnosticerer G93.3 anvender ofte
Canada-kriterierne. Da det er målbart, at patienter med PEM adskiller sig fra kontroller er det en
vigtig parameter. Hvis man anvender kriterier uden PEM fx Oxford eller de danske kriterier for
funktionelle lidelser, som ikke anvendes internationalt, risikerer man at diagnosticere en patient
med ME uden patienten har den sygdom. Det bør fremgå af rapporten
Opgavens formål og resultater vedr. behandling:
Opgaven gik ud på at belyse:
”Hvilken behandling farmakologisk og non-farmakologisk (herunder
pacing), der bør anvendes til voksne med ME”.
Graded exercise therapy (GET):
Dette afsnit hviler meget på Cochranes review, som i 2019 blev
vurderet til at være forældet
6
. Der står således i reviewet: “All studies were conducted with
outpatients diagnosed with 1994 criteria of the Centers for Disease Control and Prevention or the
Oxford criteria, or both. Patients diagnosed using other criteria may experience different effects”.
Reviewet er derfor ikke gældende i sin nuværende form for ME defineret med nyere diagnose-
Consensus recommendations (2021): Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Essentials of
Diagnosis and Management - Mayo Clinic Proceedings
5
Consice review for clinicians (2023):Diagnosis and Management of Myalgic Encephalomyelitis/Chronic Fatigue
Syndrome
6
Publication of Cochrane Review: ‘Exercise therapy for chronic fatigue syndrome’ | Cochrane
4
2
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0008.png
kriterier. Ydermere er reviewet kun gældende for ambulante patienter (outpatients) (mild ME), og
ikke for moderat, severe og very severe ME, der udgør mere end halvdelen af patientgruppen.
Det fremgik af Defactums præsentation at ”alvorlige bivirkninger er sjældne og ligeligt fordelt
mellem grupperne”, men der står i reviewet, at de er dårligt belyst ”the evidence regarding adverse
effects is uncertain”. Da dem, der får det dårligt af træning, er dem, som har PEM, kan man ikke
konkludere, at bivirkninger er sjældne. Dette bør tydeligt fremgå af rapporten.
Cognitive behavioral therapy (CBT):
I dette afsnit er der inkluderet et systematisk review, hvor der
står: “Most studies (k = 10) used the international definition criteria (Centers for Disease Control
and Prevention, [1]) for inclusion of CFS patients, while the other studies either used the Oxford
criteria ([22], k = 3), or cut-off values (k = 2)”. Sætningen fra Cochranes review om GET, må derfor
også gælde her:” Patients diagnosed using other criteria may experience different effects”.
Ydermere er nogle af studierne kun gældende for mild/moderat ME. Dette bør tydeligt fremgå af
rapporten.
Metodediskussion:
Manglende basal viden om sygdommens karakter – og betydning for udvælgelse af studier:
Vi
fik fra præsentationen det indtryk, at det basale overblik over, hvilken sygdom man har med at
gøre, ikke har været retningsgivende i udvælgelsen af hvilke artikler, man udvalgte at gå videre med
i opgavens anden del.
Manglende viden om, hvilke kriterier, der var givet for inklusion i de udvalgte undersøgelser:
Det fremgik af en slide i præsentationen, at der er gennemgået 8 studier, og at de 7 af disse studier
er foretaget før 2011, hvor konsensuskriterierne udkom – og det senere studie fra 2017 angiver, at
anvende helt brede, nu ud-daterede kriterier for CFS (ME er overhovedet ikke nævnt i studiet). Der
er altså anvendt kriterier, hvor PEM ikke er medtaget. Resultaterne fra disse studier kan derfor ikke
overføres til ME/CFS patienter. Det vil være en metodefejl at overføre resultater fra en
patientgruppe til en anden patientgruppe – og der må som minimum gøres ganske udtrykkeligt
opmærksom på dette forhold, hvilket vi også forventer, at det gøres i den endelige rapport. Andet
vil være videnskabeligt forkert.
Inklusion af svage studier:
Alle de enkelt-studier for GET og GBT, der indgår i de systematiske
reviews er meget svage (tæt på ikke-valide), da det er ikke-blindet studier med subjektive endemål.
PACE-studiet indgår i begge reviews, selvom det er fejlbehæftet. Det bør være den korrigerede
genberegning, der anvendes
7
. Dette bør også tydeligt fremgå af rapporten.
Manglende inklusion af europæiske) guidelines/HTA-rapporter:
I inklusionskriterier for begge
review står der, at (europæiske) guidelines/HTA rapporter fra større anerkendte institutioner
ønskes inddraget. Ud fra præsentationen så det ikke ud til, at disse er blevet inddraget. Der findes
Rethinking the treatment of chronic fatigue syndrome—a reanalysis and evaluation of findings from a recent
major trial of graded exercise and CBT | BMC Psychology | Full Text
7
3
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0009.png
for eksempel en europæisk NICE-guideline
8
og en amerikansk IOM-rapport
9
, der beskriver hvordan
ME diagnosticeres og tilgås med hensyn til behandling og pleje. Her fremgår det meget tydeligt, at
pacing ikke er en behandlingsform – det kan derfor ikke undersøges i RCT’er – men en måde at
undgå at udløse PEM og få forværring. Det fremgår også, at simpel symptomlindring er vigtig for at
bedre patienternes meget ringe livskvalitet. Dette tilbydes andre patientgrupper uden der skal
foreligge RCT’er, så det bør også gælde for ME-patientgruppen. Dette bør tydeligt fremgå af
rapporten.
General diskussion af metode:
Defactum bør som vidensorganisation ikke fravælge hverken en
kritisk stillingtagen til formål, metode og resultater – ej heller ikke selvom resultaterne bygger på et
systematisk review. Det er ikke tydeligt for os, hvordan det vil fremgå i rapporten.
Konsensusarbejde fra andre lande:
Opmærksomhed på den internationale konsensus på området:
Andre landes
sundhedsmyndigheder har, uafhængig af hinanden, konkluderet at ME er en kronisk,
multisystemisk sygdom: USA (2014), USA (2015), USA (2016), Nederlandene (2018), Belgien
(2020), Storbritannien (2021), USA (2023) og Tyskland (2023). Tyskland/Østrig/Schweitz (2024)
Dette bør som minimum indgå i baggrund eller diskussion.
Evidence reviews - October 2021 | Myalgic encephalomyelitis (or encephalopathy)/chronic fatigue syndrome:
diagnosis and management | Guidance | NICE
9
Beyond Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Redefining an Illness - PubMed
8
4
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS)
Pathobiology, diagnosis, and treatment
Jesper Mehlsen, MD
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0011.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
About the Author
Jesper Mehlsen is medical doctor graduated from the University of Copenhagen in
1979 and a specialist in Clinical Physiology. As such his primary clinical work and
research has been focused on complex medical conditions – in particular those that
involves dysfunction of the autonomic nervous system.
Jesper Mehlsen has worked clinically with patients suffering from myalgic
encephalomyelitis for more than 10 years and has diagnosed and treated more than
2,000 patients with this disease. Jesper Mehlsen is the current chairman of the
European ME Clinician’s Council.
Jesper Mehlsen has mor than 300 publications - 175 in peer reviewed medical and
technical journals. Jesper Mehlsen is co-chairing the European ME Research Group
and is co-chairing the ME/CFS Clinical Trial Working Group under the National
Institute of Health, USA.
2
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0012.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
Preface
The myalgic encephalomyelitis (ME) stems from Latin: myalgia i.e., pain in a muscle or groups of
muscles and
inflammation of the brain (encephalon) and the spinal cord (myelon). The diagnosis
was first use to describe a multiorgan disease epidemic at the Royal Free Hospital in London in an
editorial in the Lancet in 1956. An outbreak of a similar condition occurred in Nevada 1984 and
was given the more generalized term Chronic Fatigue Syndrome (CFS) by the Centre for Disease
Control in 1984 and hence the short form ME/CFS. The diagnosis of ME has been recognized by
WHO since 1969 and is - in the current disease classification - listed under the group of Postviral
and related fatigue syndromes with the specific code of G93.32.
ME/CFS is a disabling chronic inflammatory condition causing generalized symptoms affecting
multiple organ systems and leading to invalidity ranging from partial incapacitation in daily
activities to a severe condition in which the patient is bedridden, dependent on care 24/7 and
sometimes needing tube feeding. In the severe stage, the patient is hypersensitive to light and
sound, often unable to communicate in a normal fashion and has a very low stress level. Even
though ME/CFS is a life-long condition it is possible with the right care and treatment to achieve a
lower severity level over months to years.
Like in many diseases and illnesses, such as the common cold, migraine headaches, generalized
pain and others, ME/CFS lacks a single and accurate biomarker. Many countries have developed
guidelines for diagnosis, treatment, and care based on scientific evidence and achieved consensus.
In 2011 an international group of clinicians and researchers working with ME/CFS published a
consensus on a medical case definition for the diagnosis of ME/CFS. Quantitative support for the
diagnosis and for grading of severity can be obtained through validated questionnaires as well as
by testing physical and cognitive functions, and through laboratory analyses.
The contents of this expert report are based on clinical experience from more than 2,000 ME/CFS
patients combined with intensive studies of the vast literature on pathobiology and suggested
treatments in this group of patients.
Part 1 of the report is focused on diagnosis, probable causes, and background.
Part 2 of the report describes possible treatment options for mitochondrial dysfunction,
neuroinflammation, and autonomic dysfunction.
Frederiksberg February 2025
3
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
Contents
Section 1: Diagnosis, Probable Causes & Background
1.1.
Diagnosis
1.1.1. Screening diagnosis for ME/CFS – Institute of Medicine Criteria
1.1.1.1. Diagnostic algorithm for ME/CFS
1.1.2. Final diagnosis for ME/CFS - International Consensus Criteria (ICC)
1.1.2.1. Checklist for diagnosing ME/CFS according to ICC.
p. 5
p. 6
p. 7
p.11
1.2.
Probable causes
1.2.1.
1.2.2.
Vira reactivation/infection
Neuroinflammation
p. 12
p. 14
1.3.
Fatigue
1.3.1.
1.3.2.
Mitochondrial dysfunction
Exercise intolerance and post exertional malaise
p. 16
p. 18
p. 20
1.4.
Quantifying disease impact
1.4.1.
Symptom-based questionnaires
p. 22
p. 22
p. 22
p. 23
p. 23
p. 23
p. 26
p. 27
p. 27
p. 29
4
© 2025 JM-Consulting. All rights reserved
1.4.1.1. General health - SF 36
1.4.1.2. General health – EQ-5D-5L/3L
1.4.1.3. Functional capacity – FunCap
1.4.1.4. Fatigue questionnaires
1.4.1.5. Autonomic dysfunction – COMPASS31
1.4.2. Muscle strength and endurance - Hand-grip testing.
1.4.2.1. Autonomic function
1.4.2.2. Tilt-testing and Valsalva manoeuvre
1.4.2.3. Heart rate variability
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
Section 2: Treatment
2.1. Mitochondrial dysfunction
2.1.1 Co-enzyme supplementation
2.1.2 Antioxidants
p. 30
p. 32
2. Neuroinflammation
2.1. Dietary supplements for neuroinflammation.
2.1.1. Palmitoylethanolamide
2.1.2. Pycnogenol
2.2. Treatment of neuroinflammation, medical treatment
2.2.1. Low dose Naltrexone (LDN)
P. 34
p. 34
p. 36
p. 38
p. 38
3. Antiviral treatment
3.1. Valacyclovir
p. 40
p. 40
4. Autonomic dysfunction
4.1. Postural tachycardia syndrome (POTS)
p.42
5
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
Section 1
1. 1. Screening diagnosis – Institute of Medicine Criteria for ME/CFS
The Institute of Medicine (IOM) has proposed simplified criteria for the diagnosis of ME/CFS.
According to these criteria differential diagnoses should be excluded and the patients must have
the following three symptoms and at least one of two other manifestations.
A. A substantial reduction or impairment in the ability to engage in pre-illness levels of
activity (occupational, educational, social, or personal life) that:
a. lasts for more than 6 months,
b. is accompanied by fatigue that is:
i. often profound
ii. of new onset (not life-long)
iii. not the result of ongoing or unusual excessive exertion
iv. not substantially alleviated by rest
B. Post-exertional malaise (PEM) - worsening of symptoms after physical, mental, or
emotional exertion that would not have caused a problem before the illness. PEM often
puts the patient in a relapse that may last days, weeks, or even longer. For some patients,
sensory overload (light and sound) can induce PEM. The symptoms typically get worse 12
to 48 hours after the activity or exposure and can last for days or even weeks.
C. Unrefreshing sleep—patients with ME/CFS may not feel better or less tired even after a full
night of sleep despite the absence of specific objective sleep alterations.
At least one of the following
two additional manifestations
must also be present:
1. Cognitive impairment—patients have problems with thinking, memory, executive function,
and information processing, as well as attention deficit and impaired psychomotor
functions. All can be exacerbated by exertion, effort, prolonged upright posture, stress, or
time pressure, and may have serious consequences on a patient’s ability
to maintain a job
or attend school full time.
2. Orthostatic intolerance—patients develop a worsening of symptoms upon assuming and
maintaining upright posture as measured by objective heart rate and blood pressure
abnormalities during standing, bedside orthostatic vital signs, or head-up tilt testing.
Orthostatic symptoms including lightheadedness, fainting, increased fatigue, cognitive
worsening, headaches, or nausea are worsened with quiet upright posture (either standing
or sitting) during day-to-day life and are improved (though not necessarily fully resolved)
with lying down. Orthostatic intolerance is often the most bothersome manifestation of
ME/CFS among adolescents.
6
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0016.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
1.1.1. Diagnostic algorithm for ME/CFS according to Institute of Medicine, USA
7
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0017.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
1.1.2 Final diagnosis for ME/CFS - International Consensus Criteria (ICC)
Differential diagnoses must have been excluded. Patients must – to receive the diagnosis of myalgic
encephalomyelitis - meet the criteria for post exertional malaise, and have:
A. at least one symptom from three neurological dysfunctions
B. at least one symptom from three immunologic/gastrointestinal/genitourinary change categories
C. at least one symptom from energy metabolism/mobility impairments.
Post-exertional Malaise (PEM): Mandatory
A pathological inability to produce sufficient energy as needed with prominent symptoms in the
neuroimmune areas.
Characteristics are as follows:
Pronounced, rapid physical and/or cognitive fatigue in response to exertion, which may be
minimal such as activities of daily living or simple mental tasks, may be disabling and cause
relapse.
Worsening of symptoms after exertion: e.g., acute flu-like symptoms, pain and worsening of other
symptoms.
Post-exertional fatigue can occur immediately after activity or be delayed by hours or days.
The refund period is extended and usually takes 24 hours or longer. A relapse can last days, weeks
or longer.
Low threshold for physical and mental fatigue (lack of endurance) results in a significant reduction
in activity level before illness.
Notes: To be diagnosed with ME, symptom severity must result in a significant reduction in a patient's
premorbid activity level.
• Mild (approx. 50% reduction in activity level before illness),
• Moderate (mostly at home),
• Severe (mostly bedridden) or
• Very severe (completely bedridden and needs help with basic functions).
There can be marked fluctuations in symptom severity and hierarchy from day to day or hour to hour.
Consider activity, context, and interactive effects.
Regardless of a patient's recovery time after reading for �½ hour, it will take much longer to recover
from grocery shopping for �½ hour and even longer if repeated the next day – if possible.
Those who rest before an activity or have adjusted their activity level to their limited energy may have
shorter recovery periods than those who do not pace their activities sufficiently.
8
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0018.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
Neurological disorders
At least one symptom from three of the following four symptom categories:
Cognitive dysfunction
Difficulty processing information: slowed thinking, reduced concentration, e.g., confusion,
disorientation, cognitive overload, difficulty making decisions, slowed speech, acquired or
effortful dyslexia.
Short-term memory loss: e.g., difficulty remembering what you wanted to say, what you said,
retrieving words, recalling information, poor working memory.
Pain
Headache: e.g., chronic, generalized headache often involves tenderness in the eyes, behind the
eyes, or in the back of the head that may be associated with cervical muscle tension; migraine;
tension headache.
Significant pain can be experienced in muscles, muscle-tendon connections, joints, abdomen, or
chest. It is non-inflammatory in nature and often migrates e.g., generalized hyperalgesia,
widespread pain (may meet fibromyalgia criteria), myofascial or radiating pain.
Disturbed sleep patterns: e.g., insomnia, prolonged sleep including naps, sleeping most of the
day and being awake most of the night, frequent awakenings, waking much earlier than before
onset of illness, vivid dreams/nightmares.
Unrefreshing sleep: e.g., waking up feeling exhausted regardless of the duration of sleep,
daytime sleepiness.
Neurosensory, perceptual, and motor disturbances
Neurosensory and perceptual: e.g., inability to focus vision, sensitivity to light, noise, vibration,
smell, taste, and touch; reduced depth perception.
Engine: e.g., muscle weakness, twitching, poor coordination, unsteadiness on the feet, ataxia.
Notes: Cognitive dysfunction - reported or observed - becomes more pronounced with fatigue.
Overload phenomena can be evident when two tasks are performed simultaneously. Abnormal
pupillary accommodative reactions are common.
Sleep disorders are typically manifested by prolonged sleep, sometimes extreme, in the acute
phase and often develop into significant sleep changes in the chronic phase.
Motor disturbances may not be evident in mild or moderate cases, but abnormal gait and
positive Romberg test may be observed in severe cases.
9
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0019.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
Immunological, gastrointestinal, and genitourinary changes
At least one symptom from three of the following five symptom categories
Flu-like symptoms may be recurrent or chronic and are typically activated or worsened by
exertion e.g., sore throat, sinusitis,
Cervical and/or axillary lymph nodes may be enlarged or tender with accompanying
palpitations.
Susceptibility to viral infections with extended recovery periods
Gastrointestinal tract: e.g., nausea, abdominal pain, bloating, irritable bowel syndrome
Genitourinary: e.g., frequent urge to urinate, incontinence or bladder emptying problem
Hypersensitivity to food, medicine, odors, or chemicals
Notes: Sore throat, tender lymph nodes, and flu-like symptoms are obviously not specific
to ME, but their activation in response to exertion is abnormal. The throat may feel sore,
dry and scratchy. Color changes can be seen in the tonsils, as an indication of immune
activation.
Energy production/mobility disorders
At least one symptom of the following:
Cardiovascular
o
Orthostatic intolerance,
o
neurally mediated hypotension,
o
Postural orthostatic tachycardia syndrome,
o
Palpitations with or without cardiac arrhythmias,
o
Dizziness
Respiratory:
o
air hunger,
o
labored breathing,
o
fatigue of the chest wall muscles.
Loss of thermostatic stability: e.g.
o
subnormal body temperature
o
significant daily fluctuations
o
Sweating episodes
o
Recurrent feeling of fever with or without low fever
o
Cold extremities.
o
Intolerance to extreme temperatures
10
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0020.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
1.1.2.1 Checklist for diagnosing ME/CFS according to ICC
At least 3 positive answers to questions 2-5, at least 3 positive answers to questions 6-10, at least 1
positive answer to questions 11-14
Symptom
1. PEM
2. Cognitive dysfunction
3. Pain
4. Sleep disturbances
5. Sensory/motor disturbances
6. Flu-like symptoms
7. Susceptibility to infections
8. Gastrointestinal problems
9. Bladder problems
10. Sensitivity
11. Cardiovascular disorders
12. Difficulty breathing
13. Temperature sensitivity
14. Difficulties in keeping warm
Yes
No
Uncertain
11
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0021.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
1.2.
Probable causes
1.2.1.
Viral activation/infection
The concept of ME/CFS as being caused by viral infections is strongly supported by a large
number of cases reported in outbreaks of probable viral origin, the most publicized being
outbreaks in Los Angeles
1
, Akureyri in Northern Iceland
2
, the Royal Free Hospital in
London
3
, Incline Village in Nevada
4
, and in Tapanui, New Zealand
5
.
Chronic symptoms following other acute infections are experienced by some patients.
These chronic symptoms resemble ME/CFS, and because of this, ME/CFS is probably a
chronic illness following an infection
6
. A prospective cohort study tracking 253 patients
from the time of acute infection with Epstein-Barr virus, Coxiella burnetii, or Ross River
virus found that 28 of the 253 participants met the diagnostic criteria for chronic fatigue
syndrome at six months
7
. The phenotype was stereotyped and occurred at a similar
incidence after each infection. The syndrome was predicted largely by the severity of the
acute illness rather than demographic, psychological, or microbiological factors
7
.
In a prospectively recruited cohort of 4,501 college students, 238 developed infectious
mononucleosis (IM) and were followed up at six months to determine whether they
recovered or met the criteria for ME/CFS
8
. Those who developed ME/CFS did not have any
significant baseline differences in stress, coping, anxiety, or depression, but before IM, they
had several cytokine markers that were significantly different from those who did not
develop ME/CFS
8
.
ME/CFS following IM is a severely debilitating disease in adolescents, and although the
prognosis seems better than in adults, their condition can fluctuate and significantly impact
their health-related quality of life
9
.
In a cohort of 280 adults with COVID-19, it was observed that fatigue and neurocognitive
dysfunction at a median of four months following initial diagnosis were independently
associated with serological evidence suggesting recent EBV reactivation
10
. The study
concluded that their findings suggest differential effects of chronic viral coinfections on the
likelihood of developing long COVID
10
.
Even though EBV is the primary suspect in ME/CFS, others have pointed to the enterovirus
family as being the main cause of ME/CFS
11
. During the polio epidemic from 1934 to 1935
1
,
60 times as many hospital employees working with polio patients developed a condition
with a cluster of symptoms including pain, fever, headache, nausea, sensory disturbances,
constipation or diarrhoea, vertigo, photophobia, and double vision. Patients also
experienced fatigue when walking short distances, loss of concentration, lapses of memory,
and sleep disturbances, very similar to symptoms reported by patients with ME/CFS; 55%
of the staff were still off duty six months after the peak of the epidemic
1
.
12
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0022.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
References for viral activation.
Gilliam AG. Epidemiological study of an epidemic, diagnosed as poliomyelitis, occurring
among the personnel of the Los Angeles County General Hospital during the summer of
1934.
Public Health Bull.
1938, 130:231–40.
2. Sigurdsson B. Clinical findings six years after outbreak of Akureyri disease.
Lancet.
(1956)
270:766–7. doi: 10.1016/S0140-6736(56)91236-3
3. Staff M. An outbreak of encephalomyelitis in the Royal Free Hospital Group, London, in
1955.
Br Med J.
(1957) 2:895–904. doi: 10.1136/bmj.2.5050.895
4. Levine PH, Jacobson S, Pocinki AG, Cheney P, Peterson D, Connelly RR, et al. Clinical,
epidemiologic, and virologic studies in four clusters of the chronic fatigue syndrome.
Arch
Intern Med.
(1992) 152:1611–6
5. Levine PH, Snow PG, Ranum BA, Paul C, Holmes MJ. Epidemic neuromyasthenia and chronic
fatigue syndrome in west Otago, New Zealand. A 10-year follow-up.
Arch Intern Med.
(1997)
157:750–4.
6. https://www.cdc.gov/me-cfs/causes/index.html#:~:text=injury%2C%20and%20genetics.-
,Infections,an%20illness%20like%20ME%2FCFS.
7. Hickie I, Davenport T, Wakefield D, et al. Dubbo Infection Outcomes Study Group. Post-
infective and chronic fatigue syndromes precipitated by viral and non-viral pathogens:
prospective cohort study. BMJ. 2006, 333: 575.
8. Jason LA, Cotler J, Islam MF, et al. Risks for developing ME/CFS in college students following
Infectious Mononucleosis: A prospective cohort study. Clin Infect Dis 2021; 73(11): e3740–
e3746.
9. Pricoco R, Meidel P, Hofberger T, et al. One-year follow-up of young people with ME/CFS
following infectious mononucleosis by Epstein-Barr virus. Frontiers in Pediatrics 2024, 11,
DOI=10.3389/fped.2023.1266738
10.
Peluso MJ, Deveau T-M, Munter SE, et al. Chronic viral coinfections differentially affect the
likelihood of developing long COVID.
J Clin Invest. 2023,
133:
e163669.
11. Hanson MR. The viral origin of myalgic encephalomyelitis/chronic fatigue syndrome. PLoS
Pathog. 2023 Aug 17;19(8):e1011523.
1.
13
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
1.2.2 Neuroinflammation
Neuroinflammation is a common feature in myalgic encephalomyelitis/chronic fatigue syndrome
(ME/CFS) affecting 85-90% of all patients yet the underlying mechanism(s) responsible for the
initiation and/or promotion of this process is largely unknown. It has been hypothesised to be the
central cause of ME/CFS due to failure of the normal function of neuroglia
1
. An animal study has
suggested that a protein secreted by herpes virus 4 (Epstein-Barr virus) can change the blood brain
barrier and thus initiate the neuroinflammatory reaction
2
.
Following the initial stressor event i.e., a
viral infection, the inflammatory response moves to the brain through neurovascular pathways or
the dysfunctional blood-brain barrier in the central regions of the brain (paraventricular nuclei and
hypothalamus. This elicits a chronic neuroinflammation leading to a sustained illness with chronic
relapse/recovery cycles
3
. This could help explain the widespread pathophysiology of ME/CFS, the
involvement of the hypothalamic/pituitary/adrenal axis
4
, the autonomic nervous system, and the
sensitivity normal life stressors of physical, cognitive, psychological, emotional, and environmental
origin. The hypothalamus connects the nervous system to the endocrine system, regulating both
the HPA axis and homeostatic control of body temperature, fatigue, sleep, and circadian rhythms-
all disturbed in ME/CFS.
The CNS is supported by glia cells (“glue” in Greek) that make up
half the volume of neural tissue
and includes
microglia astrocytes, oligodendrocytes,
and
ependymal cells. Under physiological
conditions, microglia play a role in development and maturation of the CNS and subsequently
serve in immune surveillance as the resting phenotype. The
microglia
can release pro-
inflammatory cytokines, reactive oxygen, and nitrogen species as well as other chemokines in
response to pathogens and/or damage associate debris as they change to
a proinflammatory
phenotype. If overactivated microglia may lead to a neuroinflammatory cascade and may interfere
with nerve repair
5,6
.
Activated microglia will activate astrocytes and cause these cells to secrete proinflammatory
chemokines associated with a loss in neuronal survival and function. Astrocytes also signal the
endothelial cells of the brain vasculature resulting in further disruption of the blood-brain barrier,
increasing perfusion and facilitating immigration of blood immune cells
7
.
Over time, the phenotype of microglia and astrocytes changes to an anti-inflammatory form - M2
and A2 respectively
8
. By virtue of the immune and glial cells’ ability to influence the activity of
other neuroglial cells, effects thereof can be conveyed to distant parts of the brain
8
. On the
functional level, CNS inflammation has been associated with cytokine-mediated sickness
behaviour
9
, excitotoxicity (Dong
et al., 2009)
and dysfunctional connectivity within the brain
(notably due to synaptic loss and demyelination) (Rao
et al., 2012)
that leads to CNS dysfunction
affecting sleep.
Magnetic resonance spectroscopi has shown metabolic abnormality in different brain areas in
MECFS (see reference 11 for full information). Newer diagnostic imaging modalities have been able
to demonstrate changes compatible with neuro-inflammation in ME/CFS. Nakatomi compared PET
scanning of the brain in nine patients with ME/CFS and 10 healthy controls
10
. They used a
11
C-
labelled translocator protein analogue and found
widespread accumulation in brain areas in
14
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
ME/CFS patients, and the level of the inflammatory marker was associated with the severity of
neuropsychologic symptoms
10
. Mueller et al used magnetic resonance spectroscopy to show that
ME/CFS patients had increased temperature in several brain regions, which was not attributable to
increased body temperature or differences in cerebral perfusion
11
. They found correlation between
the brain temperature and metabolic signs of inflammation
11
.
References for Neuroinflammation
1.
Renz-Polster H, Tremblay ME, Bienzle D, Fischer JE. The Pathobiology of Myalgic
Encephalomyelitis/Chronic Fatigue Syndrome: The Case for Neuroglial Failure. Front Cell Neurosci.
2022, 16: 888232.
2.
Williams MV, Cox B, Lafuse WP, Ariza ME. Epstein-Barr Virus dUTPase Induces Neuroinflammatory
Mediators: Implications for Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Clin Ther. 2019, 41:
848-63.
3. Tate W, Walker M, Sweetman E, et al. Molecular Mechanisms of Neuroinflammation in ME/CFS and
Long COVID to Sustain Disease and Promote Relapses. Front Neurol. 2022, 13: 877772.
4.
Stanculescu D, Bergquist J. Perspective: Drawing on Findings From Critical Illness to Explain Myalgic
Encephalomyelitis/Chronic Fatigue Syndrome. Front Med (Lausanne). 2022, 9:818728
5. Wanga J, Heb W, Zhanga J. A richer and more diverse future for microglia phenotypes. 2023 Heliyon 9,
e14713.
6.
Liddelow, S., Guttenplan, K., Clarke, L., et al. Neurotoxic reactive astrocytes are induced by activated
microglia. Nature 2017, 541: 481–87.
7.
Olofsson PS, Rosas-Ballina M, Levine YA, Tracey KJ. Rethinking inflammation: neural circuits in the
regulation of immunity. Immunol Rev. 2012, 248: 188-204.
8.
Norden DM, Trojanowski PJ, Villanueva E, et al. Sequential activation of microglia and astrocyte
cytokine expression precedes increased Iba-1 or GFAP immunoreactivity following systemic immune
challenge. Glia. 2016, 64: 300-16.
9.
Dantzer R. Cytokine, sickness behavior, and depression. Immunol Allergy Clin North Am. 2009, 29:
247-64.
10. Nakatomi Y, Mizuno K, Ishii A, et al. Neuroinflammation in Patients with Chronic Fatigue
Syndrome/Myalgic Encephalomyelitis: An
11
C-(R)-PK11195 PET Study. J Nuclear Med 2014, 55:
945-
950.
11.
Mueller C, Lin JC, Sheriff S, et al. Evidence of widespread metabolite abnormalities in Myalgic
encephalomyelitis/chronic fatigue syndrome: assessment with whole-brain magnetic resonance
spectroscopy. Brain Imaging Behav. 2020, 14: 562-72
15
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
1.3 Fatigue
Intractable or chronic fatigue lasting more than 6 months, unrelieved by sleep, is the most
common complaint in patients seeking medical attention
1-4
. It occurs naturally during aging and is
also an important secondary condition in many clinical diagnoses. The phenomenon of fatigue
(fatigue) has been defined as a multidimensional sensation, and attempts have been made to
determine possible causes of fatigue
5-10
. Most patients understand fatigue as a loss of energy and
the inability to perform even simple tasks without effort. Many medical conditions are associated
with fatigue, including lung, cardiovascular, musculoskeletal, and gastrointestinal diseases as well
as infections and cancer
5-10
.
Fatigue is related to reduction in the efficiency of cellular energy systems found primarily in
mitochondria. Damage to mitochondrial components, mainly by oxidation, can impair their ability
to produce high-energy molecules such as ATP and NADH. This occurs naturally with aging
9-10
and
during chronic diseases, where the production of reactive oxygen species (ROS) can cause oxidative
stress and cellular damage, resulting in oxidation of lipids, proteins, and DNA
7-12
. Upon oxidation,
these molecules change structurally and sometimes functionally. Important targets for ROS
damage are the phospholipid-containing membranes of mitochondria as well as mitochondrial
DNA
7
.
In patients with chronic fatigue syndrome, there is evidence of oxidative damage to DNA and
lipids
10,11
as well as the presence of oxidized blood markers, such as methemoglobin, indicating
excess oxidative stress
12
. In addition, oxidative damage to DNA and membrane lipids has been
found in muscle biopsy samples from patients with chronic fatigue syndrome
13
. These authors also
found increases in antioxidant enzymes, such as glutathione peroxidase, suggesting that this was
an attempt to compensate for excess oxidative stress
13
. Chronic fatigue syndrome patients have
persistently elevated levels of peroxynitrite due to excess nitric oxide, and this has been suggested
to result in lipid peroxidation and loss of mitochondrial function, as well as changes in cytokine
levels that exert a positive feedback on nitric oxide production In addition to mitochondrial
membranes, mitochondrial enzymes are also inactivated by peroxynitrite, and this may contribute
to loss of mitochondrial function
15,16
. Finally, although there are cellular molecules that counteract
the excess oxidative capacity of ROS, such as glutathione and cysteine, these are found at lower
levels in patients with ME/CFS
17
.
16
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
References for Fatigue
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
Kroenke K, Wood DR, Mangelsdorff AD, et al: Chronic fatigue in primary care. Prevalence,
patient characteristics, and outcome. JAMA 260: 929-934, 1988.
Morrison JD: Fatigue as a presenting complaint in family practice. J Family Pract 10: 795-801,
1980.
McDonald E, David AS, Pelosi AJ, Mann AH: Chronic fatigue in primary care attendees. Psychol
Med 23:987-998, 1993.
Nicolson GL: Lipid replacement as an adjunct to therapy for chronic fatigue, anti-aging and
restoration of mitochondrial function. J Am Nutraceut Assoc 6: 22-28, 2003.
Huang H, Manton KG: The role of oxidative damage in aging: a review. Front Biosci 9: 1100-
1117, 2004.
Richter C, Par JW, Ames B: Normal oxidative damage to mitochondrial and nuclear DNA is
extensive. Proc Natl Acad Sci USA 85: 6465-6467, 1998.
Wei YH, Lee HC: Oxidative stress, mitochondrial DNA mutation and impairment of antioxidant
enzymes in aging. Exp Biol Med 227: 671-682, 2002.
Harman D: Aging: A theory based on free radical and radiationchemistry. J Gerontol 2: 298-
300, 1956.
Halliwell B: Role of free radicals in the neurodegenerative diseases: therapeutic implications
for antioxidant treatment. Drugs Aging 18: 685-716, 2001.
Logan AC, Wong C: Chronic fatigue syndrome: oxidative stress and dietary modifications.
Altern Med Rev 6: 450-459, 2001.
Manuel y Keenoy B, Moorkens G, Vertommen J, et al: Antioxidant status and lipoprotein
peroxidation in chronic fatigue syndrome. Life Sci 68: 2037-2049, 2001.
Richards RS, Roberts TK, McGregor NR, et al: Blood parameters indicative of oxidative stress
are associated with symptom expression in chronic fatigue syndrome. Redox Rep 5: 35-41,
2000.
Felle S, Mecocci P, Fano G, et al: Specific oxidative alterations in vastus lateralis muscle of
patients with the diagnosis of chronic fatigue syndrome. Free Radical Biol Med 29: 1252- 1259,
2000.
Pall ML: Elevated, sustained peroxynitrite levels as the cause of chronic fatigue syndrome. Med
Hypotheses 54: 115-125, 2000
Castro L, Rodriguez M, Radi R: Aconitase is readily inactivated by peroxynitrite, but not by its
precursor, nitric oxide. J Biol Chem 269: 29409-29415, 1994.
Radi R, Rodriguez M, Castro L, Telleri R: Inhibition of mitochondrial electronic transport by
peroxynitrite. Arch Biochem Biophys 308: 89-95, 1994.
Manuel y Keenoy B, Moorkens G, Vertommen J, et al: Magnesium status and parameters of the
oxidant-antioxidant balance in patients with chronic fatigue: effects of supple- mentation with
magnesium. J Am Coll Nutr 19: 374-382, 2000.
Nicolson GL, Ellithorpe R: Lipid replacement and antioxidant nutritional therapy for restoring
mitochondrial function and reducing fatigue in chronic fatigue syndrome and other fatiguing
illnesses. J Chronic Fatigue Syndr,
2016 Feb 18;4(1):e54.
Nicolson GL. Lipid Replacement/Antioxidant Therapy as an Adjunct Supplement to Reduce the
Adverse Effects of Cancer Therapy and Restore Mitochondrial Function. Pathology oncology
research 2005 11: 139-44
13.
14.
15.
16.
17.
18.
19.
17
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0027.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
1.3.1 Mitochondrial dysfunction
Metabolic studies indicate differences between ME/CFS patients and healthy controls, with the
most consistent findings being in cellular energy generation and oxidative and nitrosative stress
1
.
Increased mitochondrial damage, reductions in ATP production and impaired oxidative
phosphorylation all point to ME/CFS potentially being a mitochondrial disease. According to
Naviaux et al
2
, many of the pathways and metabolites that have been found abnormal in ME/CFS
are like those of “dauer” (German for
persistence),
a well-studied, hypometabolic state comparable
to hibernation that permits survival and persistence under conditions of environmental stress but
at the cost of severely curtailed function and quality of life.
The main abnormality in mitochondrial function in ME/CFS seems to be a shift in fuel utilization by
reduction in carbohydrate oxidation and a reliance on fat combustion. Like in hibernating animals
this seems primarily mediated by an upregulation of several pyruvate dehydrogenase kinases with
an impairment of the pyruvate dehydrogenase complex
3
. Others have pointed to impairments in
glycolysis in addition to mitochondrial function with lower rates of ATP production across the
board
4
. Based on measurements of ATP production by peripheral blood mononuclear cells
(PBMCs) in the presence of different concentrations of glucose, it has been speculated that
ME/CFS may reduce cellular competency in adapting to changing energetic demands, which would
be consistent with an inability to ramp up ATP production upon exertion. A potential molecular
explanation for this deficiency was provided by Missailidis et al. who identified potential defects in
mitochondrial complex V (ATP synthase) from an analysis of mitochondrial function in
immortalized lymphoblasts generated from 51 ME/CFS patients
5
.
PDK4 (pyruvate dehydrogenase kinase isoform 4) is expressed ubiquitously in the mitochondrion
matrix of tissues with highest expression seen in liver, heart and skeletal muscles. PDK4 is regulated by
glucocorticoids, retinoic acid and insulin, and is found elevated with diabetes, fasting and other
conditions where fatty acids act as energy source. PDK4 inhibits the mitochondrial pyruvate
dehydrogenase complex (PDC) thus contributing to glucose metabolism regulation. Insulin resistance is
associated with PDC dysregulation in skeletal muscles and excessive insulin on the other hand
downregulates PDK4 expression. PDK4 exerts conservation of glucose as well as three-carbon
compounds such as lactate, alanine, pyruvate etc. which serve as substrates for gluconeogenesis and
this conservation helps maintain euglycemia during starvation, however, exacerbates hyperglycemia in
diabetes.
18
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
References - Mitochondrial dysfunction
.
1. Morris G, Maes M. Mitochondrial dysfunctions in Myalgic Encephalomyelitis/chronic fatigue
syndrome explained by activated immuno-inflammatory, oxidative and nitrosative stress pathways.
Metab. Brain Dis., 29 (2014), pp. 19-36.
2. Naviaux RK, et al. Metabolic features of chronic fatigue syndrome. Proc Natl Acad Sci U S
A, 113 (2016), pp. E5472-5480
3. Fluge O, et al. Metabolic profiling indicates impaired pyruvate dehydrogenase function in myalgic
encephalopathy/chronic fatigue syndrome. JCI Insight, 1 (2016), p. e89376
4. C. Tomas JL, et al. The effect of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS)
severity on cellular bioenergetic function. PLoS One, 15 (2020), Article
5. D. Missailidis SJ et al. An isolated complex V inefficiency and dysregulated mitochondrial function in
immortalized lymphocytes from ME/CFS-patients. Int. J. Mol. Sci., 21 (2020).
19
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0029.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
1.3.2 Exercise intolerance and post exertional malaise.
Exercise intolerance
It is often claimed that exertional intolerance in ME/CFS mainly results from deconditioning. In line
with that, some clinicians think that graded exercise can help exercise intolerance in ME/CFS.
However, aerobic capacity is lower in ME/CFS patients than in sedentary control subjects
1
and it
has been demonstrated that ME/CFS patients present clinically significant decrease in peak
maximal oxygen uptake only on day 2 of exercise testing
1
. Lien et al
2
showed that lactate
production per workload was significantly higher in ME/CFS and that the difference was greater on
day 2 of the exercise test
2
. The response to a two-day exercise testing differs significantly between
patients with idiopathic chronic fatigue and those with ME/CFS with the first group improving and
latter group deteriorating on the second day of testing
3
. Deconditioning in otherwise healthy
subjects is characterized by low cardiac output at peak exercise4 but the study by Joseph P et al
5
found increased cardiac output in ME/CFS patients. They also noted low cardiac filling pressures in
ME/CFS rather than the higher pressures seen in detrained individuals. A recent, deep phenotyping
study
4
found previously undescribed dysfunction of brain regions that drive the motor cortex
abnormality apparently impeding the voluntary muscle performance.
Even though deconditioning cannot explain exertional intolerance in ME/CFS, it is important to
prevent secondary deconditioning by encouraging fitness with a careful, individualised exercise
program to help compensate for disease-associated cardiovascular limitations.
Post exertional malaise.
Exertion-induced aggravation of symptoms generally called post-exertional malaise (PEM) is the
hallmark of ME/CFS and often described by the patients as a “crash”. PEM is a delayed worsening
of any or all ME/CFS symptoms irrespective of whether the exertion is physical, social, mental, or a
combination. Focussing on PEM in the clinical situation has significant importance for the outcome
in ME/CFS patients. Cross-sectional surveys covering specialist healthcare services for ME/CFS
patients in Norway found that PEM was infrequently addressed and that failure to address PEM
roughly doubled the risk of health deterioration, following rehabilitation
7
.
A study on lactate levels in ME/CFS and healthy controls
indicates that previous exercise increases
lactate accumulation in ME/CFS as opposed to the reduction seen in healthy controls
8
. This could
be one of the mechanisms responsible for PEM and if repeated could cause inflammation and
myopathy. It has been found that elevated blood lactate in the resting condition correlates with
PEM in ME/CFS
9
.
A study in Long-COVID-patients fulfilling the main ME/CFS criterion of PEM has
shown that local and systemic metabolic disturbances, severe exercise-induced myopathy,
infiltration of amyloid-containing deposits, and immune cells in skeletal muscles were key
characteristics PEM which further underlines the importance of limited exercise programs to avoid
such pathological and perhaps long-term deterioration of physical function in both ME/CFS and
Long-COVID
10
.
Characterizing the nature of post-exertional symptom exacerbation is important for developing
and evaluating rehabilitation strategies, including the potential benefits or harms of exercise for
persons living with ME/CFS. A study by Davenport TE et al
11
indicate clinicians only need to focus
on the presence and duration of just a few symptom categories and prolonged duration to identify
20
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
its existence. According to that study, inquiring about post-exertional cognitive dysfunction,
decline in function, and lack of positive feelings/mood may help identify PEM quickly and
accurately. A newly developed questionnaire addresses the consequences of exertion in ME/CFS
12
,
the premise being that ME/CFS patients can cope with various forms of exertion but at the cost of
deterioration in their health for days to months and perhaps even permanently.
References for Exercise intolerance and post-exertional malaise
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
Keller BA, Pryor JL, Giloteaux L. Inability of myalgic encephalomyelitis/chronic fatigue syndrome
patients to reproduce VO
2
peak indicates functional impairment. J Transl Med. 2014; 12:104.
Lien K, Johansen B, Veierød MB, et al. Abnormal blood lactate accumulation during repeated
exercise testing in myalgic encephalomyelitis/chronic fatigue syndrome. Physiol Rep. 2019
Jun;7(11):e14138.
van Campen CLMC, Visser FC. Comparing Idiopathic Chronic Fatigue and Myalgic
Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) in Males: Response to Two-Day
Cardiopulmonary Exercise Testing Protocol. Healthcare (Basel). 2021 9(6):683.
Saltin B, Blomqvist G, Mitchell JH, Johnson RL Jr, Wildenthal K, Chapman CB. Response to exercise
after bed rest and after training. Circulation. 1968;38(5 Suppl):VII1-VII78.
Joseph P, Arevalo C, Oliveira RKF, Faria-Urbina M, Felsenstein D, Oaklander AL, Systrom DM. Insights
From Invasive Cardiopulmonary Exercise Testing of Patients With Myalgic Encephalomyelitis/Chronic
Fatigue Syndrome. Chest. 2021; 160: 642-651.
Walitt, B., Singh, K., LaMunion, S.R. et al. Deep phenotyping of post-infectious myalgic
encephalomyelitis/chronic fatigue syndrome. Nat Commun 2024, 15: 907.
Wormgoor MEA and Rodenburg SC (2023) Focus on post-exertional malaise when approaching
ME/CFS in specialist healthcare improves satisfaction and reduces deterioration. Front. Neurol.
14:1247698
Lien K, Johansen B, Veierød MB, et al. Abnormal blood lactate accumulation during repeated exercise
testing in myalgic encephalomyelitis/chronic fatigue syndrome. Physiol Rep. 2019 Jun;7(11):e14138.
Ghali A, Lacout C, Ghali M, et al. Elevated blood lactate in resting conditions correlate with post-
exertional malaise severity in patients with myalgic encephalomyelitis/chronic fatigue syndrome. Sci
Rep. 2019 Dec 11;9(1):18817.
Appelman B, Charlton RT, Goulding RP, et al. Muscle abnormalities worsen after post-exertional
malaise in long COVID. Nature Communications 2024 15: 17
Davenport TE, Chu L, Stevens SR, et al. Two symptoms can accurately identify post-exertional malaise
in myalgic encephalomyelitis/chronic fatigue syndrome. Work. 2023;74(4):1199-1213.)
Sommerfelt, K.; Schei, T.; Seton, K.A.; Carding, S.R. Assessing Functional Capacity in ME/CFS: A
Patient Informed Questionnaire. Preprints 2023, 2023092091.
https://doi.org/10.20944/preprints202309.2091.v1
21
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0031.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
1.4
Quantifying disease impact
1.4.1. Symptom-based questionnaires
1.4.1.1. General health - SF 36
Short Form 36-Item Health Survey or SF-36 is a patient-reported health measure that
assesses health-related quality of life in 8 areas: 1) limitations in physical activities
because of health problems; 2) limitations in social activities because of physical or
emotional problems; 3) limitations in usual role activities because of physical health
problems; 4) bodily pain; 5) general mental health; 6) limitations in usual role activities
because of emotional problems; 7) vitality (energy and fatigue); and 8) general health
perceptions. A score of zero represents completely disability, and a score of 100 no
disability. It is available in many languages including Danish with Danish normative
data.
A study including 289 ME/CFS patients diagnose with ME/CFS using the International
Consensus Criteria (ICC) and quantified disease impact by SF36, mean number of steps
per day, and estimated maximal oxygen consumption. They concluded that using these
methods of quantification made the disease impact more comprehensible for the
patient and his/her caretakers, treating physicians and authorities
1
.
1.4.1.2. General Health - EQ-5D-5L/3L
EQ-5D-5L or 3L is a standardised measure of health-related quality of life developed by
the EuroQol Group
2
and assesses health status in terms of five: 1) mobility; 2) self-care;
3) usual activities; 4) pain/discomfort; and 5) anxiety/-depression dimensions. The
questionnaire is available in Danish and was used in study of ME/CFS-patients in
Denmark
3
. Results from the questionnaire was compared to a large Danish study
(177,639 included) of health- related quality of life in the general population i.e. both
healthy subjects and people with various diseases
4
. The study concluded that the
health-related quality of life in patients with ME/CFS is significantly lower than the
population mean and the lowest of all the compared other diseases
3
. The adjusted
analysis confirms that poor quality of life is distinctly different from and not a proxy of
the other included conditions.
A multinational study to assess the impact on the quality of life (QoL) of people with
ME/CFS 2022
5
. The study included 1,418 people with ME/CFS and their 1,418 family
members/partner from 30 countries.
Impact on quality of life was significantly correlated between those with ME/CFS and
their family member. Family members were most impacted emotionally by worry,
frustration and sadness and personally by family activities, holidays, sex life and
finances
5
.
Almost all ME/CFS participants (1,397) had problems performing their usual activities
and more than half were unable to perform usual activities at all. Almost 94%
experienced pain and discomfort and in one-third, pain and discomfort was classified
as extreme. Mobility was affected in 89%, with participants experiencing some
problems (1,063) with walking or were confined to bed (193). In terms of self- care,
22
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0032.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
67% had some problems or were unable to wash or dress themselves. Anxiety and
depression were the least affected dimension, 576 participants were not anxious or
depressed at all, while 678 were either moderately and 164 were extremely anxious or
depressed
5
. The authors concluded that despite the limitations of selection bias in
open participation surveys, this research has revealed the significant worldwide burden
of ME/CFS on the quality of life of people with ME/CFS and on their family members
5
.
1.4.1.3 Functional capacity - FUNCAP
FUNCAP (FC), is a questionnaire primarily developed to accurately assess FC in ME/CFS
patients
6
. The questionnaire consists of eight domains divided by activity types: A)
personal hygiene/basic functions, B) walking/movement, C) being upright, D. activities
in the home, E) communication, F) activities outside the home, G) reactions to light and
sound, and H) concentration. A Danish translation is in progress.
Initial testing in 1263 Norwegian and 1387 English-speaking patients showed FUNCAP
to be a reliable and valid tool for assessing functional capacity in ME/CFS patients.
1.4.1.4 Fatigue questionnaires
Fatigue Severity Scale
There has been limited research comparing the efficacy of different fatigue rating
scales for use with individuals with ME/CFS, but the Fatigue Scale and the Fatigue
Severity Scale (FASS)
7
have commonly been used as a measure of the severity of
specific fatigue-related symptoms, and to assess functional outcomes related to
fatigue, respectively
8
. A comparison between the scales in an ME/CFS-population
found FSS to be more accurate and comprehensive measure of fatigue-related severity,
symptomatology, and functional disability
8
and a study by the same group found that
FSS had the best ability to differentiate ME/CFS from healthy controls
9
. Fatigue severity
scale has been validated in Danish
10
.
Chalder Fatigue Questionnaire (CFQ)
11
was developed to measure severity of fatigue
illnesses and has been used in to study the effect of ME/CFS such as the disputed PACE-
trial
12
. In a recent study in a small group of ME/CFS-patients
9
, CFQ was described as
consisting of one item clearly related to physical symptoms, four items clearly related
to cognitive function, and one item relating to fatigue which could be interpreted as
cognitive and/or physical fatigue. According to participants, the remaining five
questions lacked clarity, were relating to behaviour not symptoms, or relating to
sleepiness not fatigue. The CFQ has not been accepted as a tool for research use by the
US National Institute of Neurologic Diseases and Stroke
9
.
1.4.1.5 Autonomic dysfunction – COMPASS31.
The Composite Autonomic Symptom Score (COMPASS 31) is a validated self-
assessment questionnaire quantifying the severity and distribution of autonomic
symptoms across six domains (orthostatic intolerance, vasomotor, secretomotor,
gastrointestinal, bladder and pupillomotor functions) by scoring 31 clinically selected
questions
7
. It was developed to detect and quantify the degree of autonomic failure, to
23
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
monitor disease progression and to evaluate response to treatment. The total score of
the COMPASS 31 questionnaire ranges 0-100 with higher values mirroring more severe
symptoms of autonomic dysfunction
14
.
The COMPASS 31 questionnaire has been extensively validated in different patient
categories and in healthy subjects. A study comparing COMPASS31 in ME/CFS-patients
to healthy controls showed that a cut off level in total score of 32.5 was a diagnostic
criterion for autonomic dysfunction in CFS patients15. There is some skewing in the
number of questions with few addressing orthostatic intolerance and tree times as
many focussing on gastrointestinal problems. The National Institute of Neurological
Disorders and Stroke rates COMPASS31 as” highly recommended in ME/CFS”
16
but the
shortcomings in orthostatic symptoms seem to require further testing. COMPASS31 has
been translated and validated in Danish
17
.
References on Symptom-based questionnaires
1.
van Campen CLMC, Rowe PC, Visser FC. Validation of the Severity of Myalgic Encephalomyelitis/-
Chronic Fatigue Syndrome by Other Measures than History: Activity Bracelet, Cardiopulmonary
Exercise Testing and a Validated Activity Questionnaire: SF-36. Healthcare (Basel). 2020, 8:273
https://euroqol.org/
Falk Hvidberg M, Brinth LS, Olesen AV, et al. The Health-Related Quality of Life for Patients with
Myalgic Encephalomyelitis / Chronic Fatigue Syndrome (ME/ CFS). PLoS ONE 2015, 10: e0132421
Christensen AI, Ekholm O, Glümer C, et al. The Danish National Health Survey 2010. Study design and
respondent characteristics. Scand J Public Health. 2012; 40: 391
Vyas J, Muirhead N, Singh R, et al. Impact of myalgic encephalomyelitis/ chronic fatigue syndrome
(ME/CFS) on the quality of life of people with ME/CFS and their partners and family members: an
online cross- sectional survey. BMJ Open 2022, 12: e058128. doi:10.1136/ bmjopen-2021-058128
Sommerfelt K, Schei T, Seton KA, Carding SR. Assessing Functional Capacity in Myalgic
Encephalopathy/Chronic Fatigue Syndrome: A Patient-Informed Questionnaire. J Clin Med. 2024 Jun
14;13(12):3486.
Krupp LB, LaRocca NG, Muir-Nash J, Steinberg AD. The fatigue severity scale: Application to patients
with multiple sclerosis and systemic lupus erythematosus. Archives of Neurology. 1989, 46:1121–23
Taylor RR, Jason LA, Torres A. Fatigue rating scales: an empirical comparison. Psychol Med. 2000
Jul;30(4):849-56.
Jason LA, Evans M, Brown M, Porter N, Brown A, Hunnell J, Anderson V, Lerch A. Fatigue Scales and
Chronic Fatigue Syndrome: Issues of Sensitivity and Specificity. Disabil Stud Q. 2011, 31: 1375.
Lorentzen K, Danielsen MA, Kay SD, Voss A. Validation of the Fatigue severity scale in Danish patients
with systemic lupus erythematosus. Dan Med J 2014;61(4):A4808
Chalder T, Berelowitz G, Pawlikowska T, et al. Development of a fatigue scale. Journal of
Psychosomatic Medicine. 199337:147–153.
White, P.D.; Goldsmith, K.A.; Johnson, AL, et al. Comparison of adaptive pacing therapy, cognitive
behaviour therapy, graded exercise therapy, and specialist medical care for chronic fatigue syndrome
(PACE): a randomised trial. The Lancet 2011, 377: 823–836.
Gladwell, P., Harland, M., Adrissi, A., et al. (2024). Exploring the content validity of the Chalder Fatigue
Scale using cognitive interviewing in an ME/CFS population. Fatigue: Biomedicine, Health &
Behavior. 2024, 12: 217–37.
24
© 2025 JM-Consulting. All rights reserved
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
14.
15.
Sletten DM, Suarez GA, Low PA et al. COMPASS 31: a refined and abbreviated Composite Autonomic
Symptom Score. Mayo Clin Proc. 2012, 87:1196-201.
Newton JL, Okonkwo O, Sutcliffe K,et al. Symptoms of autonomic dysfunction in chronic fatigue
syndrome,
QJM: An International Journal of Medicine,
Volume 100, Issue 8, August 2007, Pages 519–
526.
National Institute of Neurological Disorders and Stroke/NIH 2022.
https://www.commondataelements.ninds.nih.gov/report-
viewer/24150/Composite%20Autonomic%20Symptom%20Scale%20(COMPASS-31).
Brinth L, Pors K, Mehlsen J, et al. Translation and linguistic validation of the Composite Autonomic
Symptom Score COMPASS 31 in Danish. Dan Med J 2022, 69: A07210576.
16.
17.
25
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
1.4.2 Muscle strength and endurance – Hand-grip testing
Maximal exercise performances are characterized by the peak oxygen uptake and the maximal
work force, but these procedures will cause post exertional malaise in patients with ME/CFS. A
comparative study in 66 ME/CFS-patients
1
concluded that handgrip strength can predict maximal
exercise performance in these patients.
Hand-grip dynamometry is recommended to perform in sitting position with the arm flexed at the
elbow and cannot be applied to patients following abdominal surgery, musculoskeletal spine or hip
injuries
2
. In a population of young healthy subjects, the reproducibility was nearly perfect
2
.
Normative data have been determined
3
and can be used for comparisons.
Hand grip strength (HGS) has been used as an objective measure of muscle strength and fatigue in
primary symptom of ME/CFS
4
. In a group of 272 British ME/CFS patients, HGS indicators were
associated with having ME/CFS, with magnitudes of association stronger in severely affected than
in mild/moderately affected patients. The association persisted after adjusting for age, sex and
body mass index and there were significant correlations between HGS indicators and clinical
parameters of disease severity, including fatigue analogue scales
4
. A study in 105 German ME/CFS
patients found that - compared to controls - the patients had a significantly lower strength and that
the decline in strength during repeat maximal HGS measurement was significantly higher. Lower
HGS parameters correlated with severity of disease, post-exertional malaise and muscle pain and
with higher blood levels of creatinine kinase and lactate after exertion.
References
1. Jammes Y, Stavris C, Charpin C, et al. Maximal handgrip strength can predict maximal physical
performance in patients with chronic fatigue. Clinical Biomechanics 2020, 73: 162-65.
2.
Huerta Ojeda Á, Fontecilla Díaz B, Yeomans Cabrera MM, Jerez-Mayorga D. Grip power test: A new
valid and reliable method for assessing muscle power in healthy adolescents. PLoS One. 2021, 20
16: e0258720
3.
Massy-Westropp NM, Gill TK, Taylor AW, et al. Hand Grip Strength: age and gender stratified
normative data in a population-based study. BMC Res Notes. 2011, 4: 127.
4.
Nacul LC, Mudie K, Kingdon CC, et al. Hand Grip Strength as a Clinical Biomarker for ME/CFS and
Disease Severity. Front Neurol. 2018, 9:992
5. Jäkel B, Kedor C, Grabowski P, et al. Hand grip strength and fatigability: correlation with clinical
parameters and diagnostic suitability in ME/CFS. J Transl Med. 202119:159.
26
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
1.4.3. Autonomic function
Measurements of autonomic function is primarily based on measurements of
cardiovascular control i.e. heart rate and blood pressure responses to different physical
challenges such as deep breathing, exhaling against an expiratory resistance (the Valsalva
manoeuvre) and changes in position from supine to upright.
Quantitative sweat test is also used in some laboratories to detect abnormalities in the
small nerve fibres of the peripheral autonomic nervous system.
1.4.3.1.
Tilt-testing and Valsalva manoeuvre
In tilt testing the subject is place in the supine position on tilt-table and heart rate and
blood pressure are recorded continuously in a non-invasive manner. After resting for
at least 10 min in the supine position the table is tilted to 60 degrees head-up
position. At this tilt angle the body is submitted to 90% of the gravity force attained in
the full upright position. Dependent on the diagnostic purpose the subject remains in
this position for 10 to 45 min. in the tilted position the force of gravity causse a
displacement of blood from the upper to the lower body compartments causing a
reduction in cardiac filling pressure. As a consequence, there is an immediate
reduction in cardiac stroke volume and hence in blood pressure which is initially
compensated by an increase in heart rate an within the first minute followed by
peripheral vasoconstriction increasing blood pressure and causing heart rate to return
to a level 10-20 beats per minute higher than in the supine. Tilt testing has been
widely covered in consensus satetments
1,2,3
.
In the Valsalva manoeuvre the subject is place in a sitting position with continuous
measurements of heart rate and blood pressure. After a 5 min resting period the
participant is asked to take a deep breath and then exhale through a mouth piece
connected to an aneroid manometer keeping the expiratory and hence the
intrathoracic pressure at 40 mmHg for 15 seconds and then exhale without resistance
and breath normally for 2 min. Then increased intrathoracic pressure reduces the
cardiac filling pressure causing the blood pressure to fall and the heart rate to
increase and in healthy subjects, constriction of the peripheral vasculature then
causes blood pressure to return the level prior to the manoeuvre. At release of the
expiratory pressure blood pressure falls shortly before reaching a higher level
(overshoot) as cardiac filling is normalised and the heart is pumping against an
increased vascular resistance this sequence of events allowing for calculation of the
baroreceptor sensitivity and for extraction of the activity in the parasympathetic and
sympathetic divisions of the autonomic nervous system
4
. The response can also be
classified in different categories relating to the degree of dysfunction in the
autonomic nervous system
5
27
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
References to Tilt test and Valsalva maneuver
1.
Sheldon R, Grubb BP, Olshansky B, et al. 2015 Heart Rhythm Society Expert Consensus Statement
on the Diagnosis and Treatment of Postural Tachycardia Syndrome, Inappropriate Sinus
Tachycardia, and Vasovagal Syncope. Heart Rhythm, 2015, 12: e41.
Raj, Satish R. et al. Canadian Cardiovascular Society Position Statement on Postural Orthostatic
Tachycardia Syndrome (POTS) and Related Disorders of Chronic Orthostatic Intolerance.
Canadian Journal of Cardiology, 2020, 36: 357 – 372.
Thijs RD, Brignole M, Falup-Pecurariu C, et al. Recommendations for tilt table testing and other
provocative cardiovascular autonomic tests in conditions that may cause transient loss of
consciousness: Consensus statement of the European Federation of Autonomic Societies (EFAS)
endorsed by the American Autonomic Society (AAS) and the European Academy of Neurology
(EAN). Clin Auton Res. 2021, 31:369-384.
Randall EB, Billeschou A, Brinth LS, Mehlsen J, Olufsen MS. A model-based analysis of autonomic
nervous function in response to the Valsalva maneuver. J Appl Physiol (1985). 2019 Nov
1;127(5):1386-1402.
Novak P. Assessment of sympathetic index from the Valsalva maneuver. Nurology 2011,
76: 2010-2016.
2.
3.
4.
5.
28
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
1.4.2.3.
Heart rate variability
Heart rate is tightly controlled to adapt to the demands of the body and therefore
show characteristic oscillation over time. There is a diurnal variation with lowest heart
rate in the middle of the sleeping period under normal circumstances. Heart rate also
varies from beat to beat – oscillations than are commonly referred to as heart rate
variability (HRV).
In testing HRV is important to differentiate between variation caused by specific
physiological manoeuvres such a deep breathing or active change in posture from
supine to standing and the spontaneous variation seen over limited time periods in the
resting supine or sitting positions. HRV can also be measured over 24-hour periods or
longer.
The strongest oscillations in heart rate are driven by breathing
1
and the largest
difference in heart rate in response to breathing is attained at a breathing frequency of
6 respirations per minute
1
. The variations in heart rate during deep breathing are
initiated primarily by the parasympathetic nervous system through the vagal nerve and
forms the basis for breathing techniques used to reduce mental stress. The other
strong oscillator causing HRV is the blood pressure control system. The activity of the
two principal oscillators can be quantified through analysis of the frequency
component embedded in HRV – practically separated into high and low frequency
component, where the high frequency component (HF) is commonly equated with
parasympathetic activity and the low frequency component (LF) represents the
sympathetic activity. The ratio between LF can then be equated to the balance
between the two components of the autonomic nervous system
3
. A Danish study on
normative values for HRV has been published
4
.
References for Heart Rate Variability
1. Mehlsen J, Pagh K, Nielsen JS, et al- Heart rate response to breathing: dependency
upon breathing pattern Clinical Physiology 1987 7: 115-124
2. Birdee G, Nelson K, Wallston K, et al. Slow breathing for reducing stress: The effect of
extending exhale. Complement Ther Med. 2023,73:102937.
3. Task Force of The European Society of Cardiology and The North American Society of
Pacing and Electrophysiology. Heart rate variability Standards of measurement,
physiological interpretation, and clinical use European Heart Journal 1996 17: 354-81.
4. Brinth LS, Jørgensen T, Mehlsen J, et al. Normative values of short-term heart rate
variability in a cross-sectional study of a Danish population. The DanFunD study. Scan J
Pub Health 2024, 52:48-57
29
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
Section 2: Treatment
2.1 Mitochondrial Dysfunction
2.1.1.
Co-enzyme supplementation
Coenzyme Q
10
- also called Ubiquinone - is produced in the body and can be obtained
from dietary source mostly localized in the mitochondria. Q
10
is primarily located in the
mitochondria and together with nicotinamide adenine dinucleotide in the oxidized form
(NAD+ or the reduced form NADH), Q
10
is a key component of the electron transport chain
responsible for mitochondrial energy production in the form of ATP. Q
10
also acts as an
antioxidant
1
through removal of reactive oxygen species i.e. unstable molecules that may
oxidize and hence damage other molecules in the cell including the DNA. After acting as
an antioxidant, Q
10
must be reduced via NADH-related enzymes to be reactivated. Q
10
is a
product of the cholesterol synthesis in the liver, and it has been shown that statins
significantly reduce Q
10
levels
2
.
NAD+/NADH is critically involved in the mitochondrial ATP production together with Q
10
and can be synthesized from a multitude of precursors from the normal human diet – the
most prominent collectively known as B3-vitamin (niacin/nicotinamide). Normal dietary
intake of NAD+/NADH precursors is estimated to be 20-40 mg/day and stems from both
animal and plant-based diets
3
.
A recent meta-analysis of Q
10
supplementation in the general population
4
found that it
can reduce inflammatory mediators and that the optimal daily dose seemed to be 300–
400 mg.
A recent review of supplementation with NAD+/NADH
5
in different conditions including
CFS found that their study supported that oral administration of NADH I doses of 5 -
80mg/day can lead to an increase in quality of life and improvements in health
parameters. They also found that NADH supplementation is safe with a low incidence of
side effects.
A prospective randomized, placebo-controlled trial
5
of the combination of Q
10
/NADH in
doses of 200/20mg in 207 ME/CFS patients found reduction in cognitive fatigue
perception and overall using the Fatigue Impact Scale and an improvement in health-
related quality of life from baseline. Significant differences were also shown for sleep
duration at 4 weeks and habitual sleep efficiency at 8 weeks in follow-up visits from
baseline within the experimental group. In a previous study, the same group found
significant reductions in max HR during a cycle ergometer test combined with a decrease
in perception of fatigue in the active group compared to placebo. Both studies found that
the combination of Q
10
/NADH were well tolerated with no serious side-effects.
30
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
References for 2.1.1. Co-enzyme supplementation
1. Quinzii CM, Lopez LC, Gilkerson RW, et al. Reactive oxygen species, oxidative stress, and cell death
correlate with level of CoQ10 deficiency. FASEB J. 2010, 24, 3733–3743.
2. Hofer SJ, Davinelli, Bergmann M, et al. Caloric Restriction Mimetics in Nutrition and Clinical Trials.
Frontiers in Nutrition 2021, 8. DOI: 10.3389/fnut.2021.717343.
3. Hou S, Tian Z, Zhao D, et al. Efficacy and Optimal Dose of Coenzyme Q10 Supplementation on
Inflammation-Related Biomarkers: A GRADE-Assessed Systematic Review and Updated Meta-
Analysis of Randomized Controlled Trials. Mol Nutr Food Res. 2023 Jul;67(13):e2200800. doi:
10.1002/mnfr.202200800.
4. Gindri IM, Ferrari G, Pinto LPS, et al. Evaluation of safety and effectiveness of NAD in different
clinical conditions: a systematic review. Am J Physiol Endocrinol Metab. 2024, 326: E417-E427.
5. Castro-Marrero J, Segundo MJ, Lacasa M, et al. Effect of Dietary Coenzyme Q10 Plus NADH
Supplementation on Fatigue Perception and Health-Related Quality of Life in Individuals with
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A Prospective, Randomized, Double-Blind,
Placebo-Controlled Trial. Nutrients. 2021,13: 2658. doi: 10.3390/nu13082658.
31
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0041.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
2.1.2. Antioxidants
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) play a dual role in being
both deleterious and beneficial. ROS and RNS are normally generated by tightly regulated
enzymes and overproduction results in oxidative stress which causes damage to cell
structures, including lipids, membranes, proteins, and DNA. In contrast, ROS/RNS exerts
beneficial effects at low/moderate concentrations as - for example - defenses against
infectious agents
1
. The reactive oxygen species are counterbalanced by enzymatic and non-
enzymatic antioxidants. Enzymatic antioxidant defenses include - among others -
superoxide dismutase, glutathione peroxidase, and catalase. Non-enzymatic antioxidants
are represented by vitamin C, Vitamin E, glutathione (GSH), carotenoids, and flavonoids. Q
10
also has important antioxidant properties as has selenium and selenium is an important
component of enzymatic antioxidants
2
. Oxidative stress denotes a shift towards increases in
reactive oxygen species relative to antioxidant defense mechanisms and is a hallmark of
mitochondrial dysfunction in ME/CFS
3,4
. Several studies have shown aberrations in
metabolism of lipids, in the redox balance and in energy production in ME/CFS
3,4,5,6
,
probably caused by defects in the pyruvate dehydrogenase complex
4
– an essential key in
the aerobic glucose metabolism.
Vitamin C (ascorbic acid/ascorbate) is a potent antioxidant, important for the formation of
biogenic amines and contributes to immune defense. Infections significantly impact vitamin
C levels due to enhanced inflammation and metabolic requirements. Fatigue, pain,
cognitive disorders, and depression-like symptoms are known symptoms of vitamin C
deficiency
7
. Vitamin C concentrations are tightly controlled with oral ingestion in healthy
subjects with dose-independent plasma levels at intakes of more than 1.000 mg/day. To
achieve higher plasma concentration, intravenous administration (iv) is necessary
8
. A
placebo-controlled study of iv C-vitamin in healthy subjects found significantly lower
measures of oxidative stress and a positive effect on fatigue. There were no differences in
adverse events or side-effects between active and placebo treatment.
Selenium
Selenium (Se) is an essential trace element and has an important role as part of several
selenoproteins with critical roles in thyroid hormone metabolism, DNA synthesis,
reproduction, and protection from oxidative damage and infection. In foods, Se is
predominantly present as selenomethionine, which is an important source of dietary Se in
humans, and as a chemical form that is commonly used for Se supplements in clinical trials.
Daily recommended dose of Selenium in healthy subjects olde than 14 years of age is 55
microg daily. The upper tolerable limit is 400 microg in the same age group
9
Concern for potential deficiency diseases associated with low Se status has led to the
establishment of the recommended daily requirements for Se in many countries. Excess Se
intakes through supplementation and its potential misuse as health therapy could also pose
a risk of adverse health effects if its use is not properly regulated.
32
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
References for antioxidants
1. Valko M, Leibfritz D, Moncola J, et al. Free radicals and antioxidants in normal physiological
functions and human disease. The Internl J Biochem Cell Bio 2007, 39: 44–84.
2. Tingii U. Selenium: its role as antioxidant in human health. Environ Health Prev Med 2008, 13:102–
108.
3. Richards RS, Roberts TK, McGregor NR, et al. Blood parameters indicative of oxidative stress are
associated with symptom expression in chronic fatigue syndrome. Redox Report 2000,
5,
35–41.
4. Germain A, Ruppert D, Levine SM, Hanson MR. Prospective Biomarkers from Plasma Metabolomics
of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome Implicate Redox Imbalance in Disease
Symptomatology. Metabolites 2018, 8: 90.
5. Fluge O, Mella O, Bruland O, et al. Metabolic profiling indicates impaired pyruvate dehydrogenase
function in myalgic encephalopathy/chronic fatigue syndrome. JCI Insight. 2016 1: e89376.
6. Paula BD, Lemled MD, Komaroffe AL, Snyder, SH. Redox imbalance links COVID-19 and myalgic
encephalomyelitis/chronic fatigue syndrome PNAS 2021 Vol. 118 No. 34 e2024358118.
7. Vollbracht C, Kraft K. Feasibility of Vitamin C in the Treatment of Post Viral Fatigue with Focus on
Long COVID, Based on a Systematic Review of IV Vitamin C on Fatigue. Nutrients. 2021,13:1154.
8. Levine M, Padayatty SJ, Espey MG. Vitamin C: a concentration-function approach yields
pharmacology and therapeutic discoveries. Adv Nutr. 2011, 2:78-88.
9. Kumar V, Bhushan D, Supriya S, et al. Efficacy of intravenous vitamin C in management of moderate
and severe COVID-19: A double blind randomized placebo controlled trial. J Family Med Prim Care.
2022, 11:4758-4765.
10. Suh SY, Bae WK, Ahn HY, Choi SE, Jung GC, Yeom CH. Intravenous vitamin C administration reduces
fatigue in office workers: a double-blind randomized controlled trial. Nutr J. 2012, 11:7.
11. Institute of Medicine. Food and Nutrition Board. Dietary Reference Intakes: Vitamin C, Vitamin E,
Selenium, and Carotenoids. Washington, DC: National Academy Press; 2000.
33
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0043.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
2.4.
Treatment of neuroinflammation: Dietary supplements.
2.1.1
Palmitoylethanolamide (PEA)
PEA is a food component first discovered in the late 1950s and shown to have anti-allergic
and anti-inflammatory activity
1
. PEA is an endocannabinoid, and it selectively stimulates
the cannabinoid CB2 receptor avoiding the hallucinogenic effects of stimulating the CB1
receptor. It is generally accepted that PEA has a multitargeted action and – important in the
treatment of ME/CFS and related conditions – an ability to reduce inflammation, pain,
mast-cell activation, and experimental colitis
2
.
Pharmacological properties: Because of the lipidic nature and large particle size, PEA has to
be micronized to give a better adsorption. Experimental inflammation has been shown to
be reduced by oral treatment with micronized and ultramicronized PEA whereas the
nonmicronized PEA had no effect
3
. The plasma elimination half-time of PEA is 12 min in the
rat
4
and most of the PEA is located in the extravascular compartment. PEA is found in the
brain, with a preferential retention in the hypothalamus where CB2 receptors are present
5
.
Effect on inflammation and mast-cell activation: PEA has been reported to decrease the
release of several pro-inflammatory cytokines and that PEA has a curative effect in a model
of acute inflammation
6
. In the brain PEA selectively binds to the cannabinoid CB2 receptor
which is abundantly present on human microglia cells which would account for the possible
beneficial effect of PEA in conditions of neuroinflammation. It has been suggested that
chronic inflammation might develop because of low endocannabinoid tissue concentration
and that a correction of this could be exploited to develop new anti-inflammatory drugs
7
.
Recently, PEA has been reported to down-modulate mast cell activation in vitro by behaving
as an agonist at the peripheral cannabinoid CB2 receptor
8
.
Tolerability of PEA: The current clinical data indicate that there are no ‘very common’ or
‘common’ serious adverse reactions but there is insufficient data to give information in the
‘uncommon’ or ‘rare’ categories
4
. The DTU Food Institute has assessed that no harmful
effects of PEA have been reported and that based on data from experimental studies, a safe
intake of 5 mg/kg body weight per day for PEA can be calculated
9
.
Effect on pain: A meta-analysis of double-blind, controlled, and open-label clinical trials has
shown that PEA elicits a progressive reduction of pain intensity significantly higher than
control with the effects being independent of patient age or gender, and not related to the
type of chronic pain
10
. A large observation study on 600+ patients from a single pain clinic
has shown that PEA significantly reduces pain when added to usual analgesic therapy
11
. A
meta-analysis of six randomized, placebo controlled, clinical trials point to efficacy of PEA
over placebo and when compared to NSAID the effect of PEA lasted longer than that of
NSAID
12
.
The proposed mechanism(s) of action of PEA involve – among others - the effects upon
mast cells
13
and CB2-like cannabinoid receptors
14
.
34
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0044.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
Practical approach: Most of the studies have used a total daily dose micronized PEA of 800
to 1.200 mg. In clinical experience from 500+ patients this is a highly tolerable dosage, and
the onset of effect is to be expected within (several) weeks.
References for PEA
1. Esposito E, Cuzzocrea S. Palmitoylethanolamide is a new possible pharmacological treatment for the
inflammation associated with trauma. Mini Rev Med Chem. 2013 Feb;13(2):237-55. PMID: 22697514.
2. Borelli F, Romano B, Petrosino S, et al. British Journal of Pharmacology (2015)
172
142–158
3. Impellizzeri D, Bruschetta G, Cordaro M, et al. Micronized/ultramicronized palmitoylethanolamide
displays superior oral efficacy compared to nonmicronized palmitoylethanolamide in a rat model of
inflammatory pain. J Neuroinflammation. 2014 Aug 28;11: 136. doi: 10.1186/s12974-014-0136-0.
4. Gabrielsson L, Mattsson S, Fowler CJ. Palmitoylethanolamide for the treatment of pain:
pharmacokinetics, safety and efficacy. Br J Clin Pharmacol (2016) 82 932–942
5. Benito C, Tolón RM, Pazos MR, et al. Cannabinoid CB2 receptors in human brain inflammation. Br J
Pharmacol. 2008 Jan;153(2):277-85. doi: 10.1038/sj.bjp.0707505. Epub 2007 Oct 15. PMID:
17934510; PMCID: PMC2219537.
6. Costa, B., Conti, S., Giagnoni, G. and Colleoni, M. (2002), Therapeutic effect of the endogenous fatty
acid amide, palmitoylethanolamide, in rat acute inflammation: inhibition of nitric oxide and cyclo-
oxygenase systems. British Journal of Pharmacology, 137: 413-420.
7. De Filippis D, D’Amico A, Cipriano M, Petrosino S, Orlando P, Di Marzo V, et al. Levels of
endocannabinoids and palmitoylethanolamide and their pharmacological manipulation in chronic
granulomatous inflammation in rats. Pharmacol Res. Elsevier Ltd; 2010 Apr;61(4):321–8.
8. Mazzari S, Canella R, Petrelli L, Marcolongo G, Leon A. N-(2-hydroxyethyl)hexadecanamide is orally
active in reducing oedema formation and inflammatory hyperalgesia by down-modulating mast cell
activation. Eur J Pharmacol. 1996 Apr 11;300(3):227-36. doi: 10.1016/0014-2999(96)00015-5. PMID:
8739213.
9. DTU National Food Institute, Denmark. Request for risk assessment of palmitoylethanolamide for
food supplements, Danish Food Agency's j.nr.: 2018-29-7100-00439. 2018.
10. Paladini A, Fusco M, Cenacchi T, et al. Palmitoylethanolamide, a Special Food for Medical Purposes,
in the Treatment of Chronic Pain: A Pooled Data Meta-analysis. Pain Physician. 2016 Feb;19(2):11-
24.
11. Gatti A, Lazzari N, Gianfelice V, et al. Palmitoylethanolamide in the treatment of chronic pain caused
by different etiopathogenesis. Pain Medicine 2012; 13: 1121–1130
12. Marini I, Bartolucci ML, Bortolotti F, et al. Palmitoylethanolamide versus a nonsteroidal anti-
inflammatory drug in the treatment of temporomandibular joint inflammatory pain. J Orofac Pain.
2012 Spring;26(2):99-104.
13. Mazzari S, Canella R, Petrelli L, Marcolongo G, Leon A. N-(2-hydroxyethyl) hexadecamide is orally
active in reducing edema formation and inflammatory hyperalgesia by down-modulating mast cell
activation. Eur J Pharmacol 1996; 300: 227–36
14. Rorato R, Ferreira NL, Oliveira FPet al. Prolonged Activation of Brain CB2 Signaling Modulates
Hypothalamic Microgliosis and Astrogliosis in High Fat Diet-Fed Mice. Int J Mol Sci. 2022 May
16;23(10):5527. doi: 10.3390/ijms23105527.
35
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0045.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
2.1.2 Pycnogenol
Pycnogenol is extracted from the bark of a French pine tree (Pinus pinaster) and the main
components are procyanidins (70+/- 5%) and their monomers (catechin and epicatechin) as
well as phenolic acids
1
. Pycnogenol is safe for use in conventional foods, based on the
evaluation of clinical safety and preclinical toxicology data by an independent panel of
toxicology experts. Non-mutagenicity, lethal dose (LD50) > 5.0 g/kg body weight, the no
adverse event level has been determined at > 1,000 mg/kg/day [2], and this translates in a
safe dosage of > 700 mg/day. Typically, oral dosages tested in the literature are in the 30–
200 mg/day range, with some studies exploring higher dosages of 200–450 mg/day. Since it
was introduced into the market in Europe around 1970, there have been no reports of
serious adverse effects in any clinical study or from commercial use of Pycnogenol. Mild
side-effects of gastric discomfort have rarely been reported and linked to stomach-sensitive
patients. No interactions of Pycnogenol with other drugs, alcohol or food intake have been
reported
2
.
Endothelial function: Sandvik MK et al has shown that ME/CFS patients have reduced
macro- and microvascular endothelial function, indicating that vascular homeostasis may
play a role in the clinical presentation of this disease
3
. A study by Newton et al.
4
,
investigating flow-mediated dilation (FMD) for large vessel endothelial function and post-
occlusive reactive hyperemia (PORH) for microvascular function in ME/CFS patients,
concluded that ME/CFS patients have both large and small vessel endothelial dysfunction as
compared to age- and sex-matched controls
4,5
. Clinical studies have showed that
Pycnogenol can improve endothelial function
6,7
. The suggested mechanism of action is
activation of the endothelial nitric oxide synthase amplifying the NO generation from L-
arginine, eventually leading to an increase in vessel lumen and adequate tissue perfusion
7
.
Micro-clots and associated coagulation issues have been found present in ME/CFS and
point to a systemic vascular pathology and potential endothelial inflammation and targeted
therapies to address vascular and endothelial pathology might has been suggested
8
.
Pycnogenol lowers blood platelet aggregation as effectively as aspirin, without increasing
the bleeding time
9,10
. Pycnogenol prevents platelet hyperactivity but does not influence
bleeding time, unlike aspirin
9
.
Antioxidant activity of Pycnogenol has been investigated in several clinical studies
11
and has
been shown to increase the plasma antioxidant capacity and decrease the plasma oxidative
stress.
Practical approach: Treatment with Pycnogenol is recommended at a morning dose of
200mg. Besides hypersensitivity to pine bark there are no known contraindication to
Pycnogenol. Presently, documentation of the optimal duration of treatment and the long-
term effects is lacking
12
.
36
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
References for Pycnogenol
1.
Weichmann F, Rohdewald P. Projected supportive effects of Pycnogenol in patients suffering from
multi-dimensional health impairments after a SARS-CoV2 infection. International Journal of
Antimicrobial Agents. 2020 56 (6): 106191
2.
Oliff H. ABC scientific and clinical Monograph for Pycnogenol 2019 UPDATE. Am Bot Counc
Monogr. 2019:1–46
3.
Sandvik MK, Sørland K, Leirgul E, Rekeland IG, Stavland CS, Mella O, Fluge Ø. Endothelial
dysfunction in ME/CFS patients. PLoS One. 2023 Feb 2;18(2):e0280942.
4.
Newton DJ, Kennedy G, Chan KK, Lang CC, Belch JJ, Khan F. Large and small artery endothelial
dysfunction in chronic fatigue syndrome. Int J Cardiol. 2012; 154(3):335–6.
5.
Thijssen DH, Black MA, Pyke KE, Padilla J, Atkinson G, Harris RA, et al. Assessment of flow-mediated
dilation in humans: a methodological and physiological guideline. American journal of physiology
Heart and circulatory physiology. 2011; 300(1):H2–12. Epub 2010/10/19
6.
Uhlenhut K, Högger P. Facilitated cellular uptake and suppression of inducible nitric oxide synthase
by a metabolite of maritime pine bark extract (Pycnogenol). Free Radic Biol Med. 2012 Jul
15;53(2):305-13.
7.
Nishioka K, Hidaka T, Nakamura S, Umemura T, Jitsuiki D, Soga J, et al. Pycnogenol®, French
maritime pine bark extract, augments endothelium-dependent vasodilation in humans. Hypertens
Res. 2007;30(9):775–780
8.
Nunes JM, Kruger A, Proal A, Kell DB, Pretorius E. The Occurrence of Hyperactivated Platelets and
Fibrinaloid Microclots in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS).
Pharmaceuticals (Basel). 2022 Jul 27;15(8):931
9.
Pütter M, Grotemeyer KHM, Würthwein G, Araghi-Niknam M, Watson RR, Hosseini S, et al.
Inhibition of smoking-induced platelet aggregation by aspirin and pycnogenol. Thromb
Res. 1999;95(4):155–161,
10.
Araghi-Niknam M, Hosseini S, Larson D, Rohdewald P, Watson RR. Pine bark extract reduces platelet
aggregation. Integr Med. 2000;2(00):73–77.
11.
Simpson T, Kure C, Stough C. Assessing the Efficacy and Mechanisms of Pycnogenol
®
on Cognitive
Aging From In Vitro Animal and Human Studies. Front Pharmacol. 2019 Jul 3;10: 694
12.
Malekahmadi M, Moradi Moghaddam O, Firouzi S, Daryabeygi-Khotbehsara R, Shariful Islam SM,
Norouzy A, Soltani S. Effects of pycnogenol on cardiometabolic health: A systematic review and
meta-analysis of randomized controlled trials. Pharmacol Res. 2019 Dec;150: 104472. doi:
10.1016/j.phrs.2019.104472. Epub 2019 Oct 1. PMID: 31585179.
37
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0047.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
2.2
Treatment of neuroinflammation, medical treatment
2.2.1 Medical treatment: Low dose Naltrexone (LDN)
Naltrexone is an opioid receptor antagonist and in low doses (0.4 to 4.5 mg) it has both analgesic
and anti-inflammatory effects
1,2,3
and is referred to as Low-dose naltrexone (LDN). LDN has been
used off-label for treatment of pain and inflammation in multiple sclerosis, Crohn’s disease,
fibromyalgia, and other neuro-pathic/-inflammatory conditions and current evidence supports the
safety and tolerability of LDN in these illnesses
1,2,3
. Studies on quality of life and self-reported pain
demonstrate that LDN has subjective benefits over placebo
1,2,3
Naltrexone is almost completely absorbed orally but the bioavailability ranges between 5 and 40
per cent. The half-life elimination is 4 hours, and it is excreted primarily in the urine, but no dosing
adjustments are needed in mild renal impairment
1
.
Studies have indicated that LDN has a damping effect on the immune cells in the central nervous
system and counteract the proinflammatory activation that occurs in response to pain
2
and leads to
a reversal of neuropathy
3
. A retrospective review on LDN in 215 patients with multiple sclerosis
found that 77% of the participants had no side effects and no participants were hospitalized from
adverse events by LDN. The primary indication was LDN, and this was reduced in approximately 60%.
75% of the participants reported perceiving an increased or stabilized after LDN therapy.
A similar study in 218 patients with ME
5
reported on safety and effectiveness data of LDN in
dosages of 3.0 – 4.5 mg/day. The retrospective analysis of medical records showed positive
response in 74% experiencing improved vigilance/-alertness, improved physical and cognitive
performance. Some patients reported less pain (17%) and fever (15%), while 18% had no
response. Mild adverse effects (insomnia, nausea) were common at the onset of the treatment.
Neither severe adverse effects nor long-term adverse symptoms were reported
5
.
A randomized, double blinded study of LDN in 31 women with fibromyalgia found significant,
reduction of pain compared placebo. LDN was also associated with improved general satisfaction
with life and with improved mood. Thirty-two percent had significant reductions in pain and in
either fatigue or sleep problems contrasted with an 11% response during placebo. LDN was rated
equal to placebo in side-effects, and no serious side effects were reported.
Practical approach: Due to the unpredictable bioavailability in the individual patient it is
recommended to up-titrate the dose slowly starting with 0.5mg with stepwise increases of 0.5 mg
every third week to a maximum of 4.5mg. Should side effects appear the patient could go one step
back and try to proceed with the stepwise increments after three weeks or if there is an effect at a
lower dose the stay at that level.
38
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
References to LDN
1.
Patten DK, Schultz BG, Berlau DJ. The Safety and Efficacy of Low-Dose Naltrexone in the
Management of Chronic Pain and Inflammation in Multiple Sclerosis, Fibromyalgia, Crohn’s Disease,
and Other Chronic Pain Disorders Pharmacotherapy 2018;38(3):382–389).
2.
Toljan K, Vrooman B. Low-Dose Naltrexone (LDN)-Review of Therapeutic Utilization. Med Sci (Basel).
2018 6:82.
3.
Younger J, Parkitny L, McLain D. The use of low-dose naltrexone (LDN) as a novel anti-inflammatory
treatment for chronic pain. Clin Rheumatol. 2014 33:451-9.
4. Hutchinson MR, Zhang Y, Brown K, et al. Non-stereoselective reversal of neuropathic pain by naloxone
and natrexone: involvement of Toll-like receptor 4 (TLR4). Eur J Neruosci 2008 28:20–9.)
5. Turel AP, Oh KH, Zagon IS, McLaughlin PJ. Low dose naltrexone for treatment of multiple sclerosis: a
retrospective chart review of safety and tolerability (Letter). J Clin Psycho-pharmacol 2015 35:609–
11.
6. Polo, O., Pesonen, P., & Tuominen, E. (). Low-dose naltrexone in the treatment of myalgic
encephalomyelitis/chronic fatigue syndrome (ME/CFS). Fatigue: Biomedicine, Health &
Behavior, 2019 7, 207–217
7.
Carruthers BM, Jain AK, De Meirleir KL, et al. Myalgic encephalomyelitis/chronic fatigue syndrome:
clinical working case definition, diagnostic and treatments protocols. J Chronic Fatigue Syndr. 2003
11:7–115.
8.
Younger J, Noor N, McCue R, Mackey S. Low-dose naltrexone for the treatment of fibromyalgia:
findings of a small, randomized, double-blind, placebo-controlled, counterbalanced, crossover trial
assessing daily pain levels. Arthritis Rheum. 2013 65:529-38.
39
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
2990308_0049.png
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
3.
Antiviral treatment
3.1. Valacyclovir
Acyclovir, ganciclovir, and famciclovir are nucleotide analogue inhibitors which inhibit viral
replication during DNA multiplication or RNA multiplication
1
. Valacyclovir is a valine derivative
of acyclovir and is well absorbed from the gastrointestinal tract where it is converted to
acyclovir
2
. Valganciclovir is – like valacyclovir - a valine derivative improving the absorption
from the gastrointestinal tract. Valganciclovir is commonly reserved for preventing and
treating cytomegalovirus-infections in immunosuppressed individuals. Famciclovir is readily
absorbed but does not seem to offer superiority to Valacyclovir
3
and - as is the case with
valganciclovir – far more expensive.
A study in 25 ME/CFS patients has evaluated the safety and efficacy study of valacyclovir given
at 1,000 to 1,500 mg every 6 hours a day for 6 months
4
and this dose achieved serum acyclovir
levels to have a high antiviral activity versus Herpes virus 4 (EBV). The authors concluded that
the 16 ME/CFS patients with persistent infection EBV-infection improved after 6 months of
continuous dosing with valacyclovir. Nine ME/CFS patients with Herpes virus 5 (CMV) co-
infection did not benefit from 6 months of similar treatment.
Safety monitoring data from clinical trials of valacyclovir, involving over 3,000 immuno-
competent and immunocompromised persons receiving long-term therapy for Herpes Simplex
Virus (HSV) suppression, were analysed
5
. Safety profiles of valacyclovir (< 1,000 mg/day),
acyclovir (< 800 mg/day), and placebo were similar. Extensive sensitivity monitoring of HSV
isolates confirmed a very low rate of acyclovir resistance among immunocompetent subjects
(< 0.5%)
References for Valaciclovir
1. De Clercq E, Neyts J. Antiviral agents acting as DNA or RNA chain terminators. Handb Exp
Pharmacol. 2009,189:53-84.
2. Weller S, Blum MR, Doucette M, et al. Pharmacokinetics of the acyclovir pro-drug valacyclovir after
escalating single and multiple-dose administration to normal volunteers. Clin Pharmacol Ther 1993,
54: 595-605
3. Wald A, Selke S, Warren T, et al. Comparative efficacy of famciclovir and valacyclovir for suppression
of recurrent genital herpes and viral shedding. Sex Transm Dis. 2006 Sep;33(9):529-33
4. Lerner AM, Beqaj SH, Deeter RG, et al. A six-month trial of valacyclovir in the Epstein-Barr virus
subset of chronic fatigue syndrome: improvement in left ventricular function. Drugs Today (Barc).
2002,38:549-61.
5. Tyring SK, Baker D, and Snowden W. Valacyclovir for Herpes Simplex Virus Infection: Long-Term
Safetyand Sustained Efficacy after 20 Years’ Experience with Acyclovir. The Journal of Infectious
Diseases 2002;186(Suppl 1): S40–6
40
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
4.
Autonomic dysfunction
A significant minority of patients experience symptoms compatible with dysfunction of the
autonomic nervous system, including orthostatic intolerance (POTS), gastrointestinal
disturbances, bladder dysfunction, and circulation issues.
Using the Composite Autonomic Symptom Scale (COMPASS
1
), a study in ME/CFS found that
symptoms of autonomic dysfunction were strongly and reproducibly associated with the
presence of ME/CFS
2
. They also found a reverse correlation between low frequency variation
in heart rate variability and the COMPASS score Studies in ME/CFS have compared results from
the COMPASS questionnaire and data on heart rate variability HRV and have found A high
COMPASS score correlated to a significant negative correlation between LF-HRV and COMPASS
scores
2
In a case–controlled study of 45 female patients and 25 age- and gender-matched healthy
controls
3
it was found significant relationships between self-reported fatigue symptoms and
indices of heart rate variability in patients with ME/CFS
3
.
4.1. Postural tachycardia syndrome (POTS)
POTS denotes a condition where heart rate increases by more than 30 beats per minute on
assumption of the upright posture in subject older that 18 year of age and more than 40 beats
per minute in the younger age group. Heart rate increases of that magnitude i.e. postural
tachycardia can been seen in dehydrated subject and in astronauts returning after longer
periods in space where they are not submitted to gravity – the most important signal to fluid
retention in humans. POTS is diagnosed if the abnormal hemodynamic measures are
accompanied by returning symptoms of dizziness, nausea, near fainting, cognitive impairment
and other symptoms related to the upright posture
4
.
A study in ME/CFS patients with POTS showed that patients in this subgroup were significantly
younger, had a shorter length of illness, experienced greater task difficulty and were able to
stand for significantly shorter periods compared to those ME/CFS-patients without POTS.
The probable cause for POTS in ME/CFS is most likely and inability to contract the peripheral
vasculature during orthostasis and are thus dependent on increments in heart rate to keep
blood pressure at a level that ensures an adequate perfusion pressure for the cerebral
vasculature. It has also been shown that POTS patients have low frequency oscillations in heart
rate and blood pressure
6
. A study simulating such oscillations in healthy subjects using lower
body negative pressure demonstrated that healthy subject developed brain fog during such
oscillations
7
.
Treatment of POTS
4
is directed at reducing the fluid displacement during the upright posture
by increasing the intake of salt and fluid - 1 g of NaCl pr liter of fluid, reducing the tachycardia
both in the supine and upright position with adrenergic beta-blockers, muscarinic agonists like
Mestinon, and enhancing vasoconstriction with an adrenergic alpha-agonist – Midodrine. Fluid
retention can be amplified by Fludrocortisone and/or a vasopressin analogue.
41
© 2025 JM-Consulting. All rights reserved
SUU, Alm.del - 2024-25 - Bilag 187: Henvendelse af 2/3-25 fra ME Foreningen om igangværende vidensafdækning af ME/CFS
Myalgic Encephalomyelitis (ME/CFS): Pathobiology, diagnosis, and treatment
References for autonomic dysfunction and POTS
1. Sletten DM, Suarez GA, Low PA, et al. COMPASS 31: A refined and abbreviated Composite
Autonomic Symptom Score. Mayo Clinic Proceedings 2012, 87:1196–1201.
2. Newton JL, Okonkwo O, Sutcliffe K, et al. Symptoms of autonomic dysfunction in chronic
fatigue syndrome. QJM. 2007, 100: 519-26.
3. Escorihuela RM, Capdevila L, Castro JR, et al. Reduced heart rate variability predicts fatigue
severity in individuals with chronic fatigue syndrome/myalgic encephalomyelitis. J Transl
Med 2020, 18: 4.
4. Raj, Satish R. et al. Canadian Cardiovascular Society Position Statement on Postural
Orthostatic Tachycardia Syndrome (POTS) and Related Disorders of Chronic Orthostatic
Intolerance. Canadian Journal of Cardiology, 2020, 36: 357 – 372.
5.
Reynolds GK, Lewis DP, Richardson AM, Lidbury BA. Comorbidity of postural orthostatic
tachycardia syndrome and chronic fatigue syndrome in an Australian cohort. J Intern Med.
2014 Apr;275(4):409-17.
6. Geddes JR, Ottesen JT, Mehlsen J, Olufsen MS. Postural orthostatic tachycardia syndrome
explained using a baroreflex response model. - J Royal Society Interface, 2022, 19: 20220220
7. Stewart JM, Balakrishnan K, Visintainer P, et al. Oscillatory lower body negative pressure
impairs task related functional hyperemia in healthy volunteers. Am J Physiol Heart Circ
Physiol 2016, 310: H775–H784.
42
© 2025 JM-Consulting. All rights reserved