Oxaloacetate Treatment For Mental And Physical Fatigue In Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) and Long-COVID fatigue patients: a non-randomized controlled clinical trial

Alan Cash, David Lyons Kaufman, Alan Cash, David Lyons Kaufman

Abstract

Background: There is no approved pharmaceutical intervention for Myalgic Encephalomyelitis/ Chronic Fatigue Syndrome (ME/CFS). Fatigue in these patients can last for decades. Long COVID may continue to ME/CFS, and currently, it is estimated that up to 20 million Americans have significant symptoms after COVID, and the most common symptom is fatigue. Anhydrous Enol-Oxaloacetate, (AEO) a nutritional supplement, has been anecdotally reported to relieve physical and mental fatigue and is dimished in ME/CFS patients. Here, we examine the use of higher dosage AEO as a medical food to relieve pathological fatigue.

Methods: ME/CFS and Long-COVID patients were enrolled in an open label dose escalating "Proof of Concept" non-randomized controlled clinical trial with 500 mg AEO capsules. Control was provided by a historical ME/CFS fatigue trial and supporting meta-analysis study, which showed average improvement with oral placebo using the Chalder Scale of 5.9% improvement from baseline. At baseline, 73.7% of the ME/CFS patients were women, average age was 47 and length of ME/CFS from diagnosis was 8.9 years. The Long-COVID patients were a random group that responded to social media advertising (Face Book) with symptoms for at least 6 months. ME/CFS patients were given separate doses of 500 mg BID (N = 23), 1,000 mg BID (N = 29) and 1000 mg TID (N = 24) AEO for six weeks. Long COVID patients were given 500 mg AEO BID (N = 22) and 1000 mg AEO (N = 21), again over a six-week period. The main outcome measure was to compare baseline scoring with results at 6 weeks with the Chalder Fatigue Score (Likert Scoring) versus historical placebo. The hypothesis being tested was formulated prior to data collection.

Results: 76 ME/CFS patients (73.7% women, median age of 47) showed an average reduction in fatigue at 6 weeks as measured by the "Chalder Fatigue Questionnaire" of 22.5% to 27.9% from baseline (P < 0.005) (Likert scoring). Both physical and mental fatigue were significantly improved over baseline and historical placebo. Fatigue amelioration in ME/CFS patients increased in a dose dependent manner from 21.7% for 500 mg BID to 27.6% for 1000 mg Oxaloacetate BID to 33.3% for 1000 mg TID. Long COVID patients' fatigue was significantly reduced by up to 46.8% in 6-weeks.

Conclusions: Significant reductions in physical and metal fatigue for ME/CFS and Long-COVID patients were seen after 6 weeks of treatment. As there has been little progress in providing fatigue relief for the millions of ME/CFS and Long COVID patients, anhydrous enol oxaloacetate may bridge this important medical need. Further study of oxaloacetate supplementation for the treatment of ME/CFS and Long COVID is warranted. Trial Registration https://ichgcp.net/clinical-trials-registry/NCT04592354 Registered October 19, 2020. 1,000 mg BID Normalized Fatigue Data for Baseline, 2-weeks and 6-weeks evaluated by 3 Validated Fatigue Scoring Questionnaires.

Keywords: Anhydrous enol oxaloacetate; COVID fatigue; Chronic fatigue syndrome; Long COVID; ME/CFS; ME/CFS clinical; ME/CFS treatment; Oxaloacetate; Post viral fatigue.

Conflict of interest statement

Author David Kaufman has no competing interests. Author Alan Cash is an officer in a pharmaceutical company that provided the funding for this clinical trial. Funding was provided by Terra Biological LLC.

© 2022. The Author(s).

Figures

https://www.ncbi.nlm.nih.gov/pmc/articles/instance/9238249/bin/12967_2022_3488_Figa_HTML.jpg
1,000 mg BID Normalized Fatigue Data for Baseline, 2-weeks and 6-weeks evaluated by 3 Validated Fatigue Scoring Questionnaires

References

    1. Evans WJ, Lambert CP. Physiological basis of fatigue. Am J Phys Med Rehabil. 2007;86(1 Suppl):S29–46. doi: 10.1097/PHM.0b013e31802ba53c.
    1. Lock AM, Bonetti DL, Campbell ADK. The psychological and physiological health effects of fatigue. Occup Med (Lond) 2018;68(8):502–511. doi: 10.1093/occmed/kqy109.
    1. Barnett R. Fatigue. Lancet. 2005;366(9479):21. doi: 10.1016/S0140-6736(05)66809-2.
    1. Wan JJ, Qin Z, Wang PY, et al. Muscle fatigue: general understanding and treatment. Exp Mol Med. 2017;49(10):e384. doi: 10.1038/emm.2017.194.
    1. Pennisi M, Malaguarnera G, Di Bartolo G, et al. Decrease in Serum Vitamin D Level of Older Patients with Fatigue. Nutrients. 2019;11(10):2531. doi: 10.3390/nu11102531.
    1. Naviaux RK, Naviaux JC, Li K, et al. Metabolic features of chronic fatigue syndrome. Proc Natl Acad Sci USA. 2016;113(37):E5472–5480. doi: 10.1073/pnas.1607571113.
    1. Paul BD, Lemle MD, Komaroff AL, et al. Redox imbalance links COVID-19 and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Proc Natl Acad Sci USA. 2021;118(34):e2024358118. doi: 10.1073/pnas.2024358118.
    1. Komaroff AL, Bateman L. Will COVID-19 lead to myalgic encephalomyelitis/chronic fatigue syndrome? Front Med (Lausanne) 2020;7:606824. doi: 10.3389/fmed.2020.606824.
    1. Warburg O. On the origin of cancer cells. Science. 1956;123(3191):309–314. doi: 10.1126/science.123.3191.309.
    1. Kornberg MD. The immunologic Warburg effect: Evidence and therapeutic opportunities in autoimmunity. Wiley Interdiscip Rev Syst Biol Med. 2020;12(5):e1486. doi: 10.1002/wsbm.1486.
    1. Icard P, Lincet H, Wu Z, et al. The key role of Warburg effect in SARS-CoV-2 replication and associated inflammatory response. Biochimie. 2020;180:169–177. doi: 10.1016/j.biochi.2020.11.010.
    1. Lawson N, Hsieh CH, March D, et al. Elevated energy production in chronic fatigue syndrome patients. J Nat Sci. 2016;2(10):e221.
    1. Graham T, Sjogaard G, Lollgen H, et al. NAD in muscle of man at rest and during exercise. Pflugers Arch. 1978;376(1):35–39. doi: 10.1007/BF00585245.
    1. Sahlin K, Katz A, Henriksson J. Redox state and lactate accumulation in human skeletal muscle during dynamic exercise. Biochem J. 1987;245(2):551–556. doi: 10.1042/bj2450551.
    1. Sweetman E, Kleffmann T, Edgar C, et al. A SWATH-MS analysis of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome peripheral blood mononuclear cell proteomes reveals mitochondrial dysfunction. J Transl Med. 2020;18(1):365. doi: 10.1186/s12967-020-02533-3.
    1. Afrin LB, Weinstock LB, Molderings GJ. Covid-19 hyperinflammation and post-Covid-19 illness may be rooted in mast cell activation syndrome. Int J Infect Dis. 2020;100:327–332. doi: 10.1016/j.ijid.2020.09.016.
    1. Gupta SC, Kim JH, Kannappan R, et al. Role of nuclear factor kappaB-mediated inflammatory pathways in cancer-related symptoms and their regulation by nutritional agents. Exp Biol Med (Maywood) 2011;236(6):658–671. doi: 10.1258/ebm.2011.011028.
    1. Bower JE, Ganz PA, Irwin MR, et al. Fatigue and gene expression in human leukocytes: increased NF-kappaB and decreased glucocorticoid signaling in breast cancer survivors with persistent fatigue. Brain Behav Immun. 2011;25(1):147–150. doi: 10.1016/j.bbi.2010.09.010.
    1. Morris G, Maes M. Increased nuclear factor-kappaB and loss of p53 are key mechanisms in Myalgic Encephalomyelitis/chronic fatigue syndrome (ME/CFS) Med Hypotheses. 2012;79(5):607–613. doi: 10.1016/j.mehy.2012.07.034.
    1. Filler K, Lyon D, Bennett J, et al. Association of mitochondrial dysfunction and fatigue: a review of the literature. BBA Clin. 2014;1:12–23. doi: 10.1016/j.bbacli.2014.04.001.
    1. Brown AE, Jones DE, Walker M, et al. Abnormalities of AMPK activation and glucose uptake in cultured skeletal muscle cells from individuals with chronic fatigue syndrome. PLoS ONE. 2015;10(4):e0122982. doi: 10.1371/journal.pone.0122982.
    1. Fisicaro F, Di Napoli M, Liberto A, et al. Neurological sequelae in patients with COVID-19: a histopathological perspective. Int J Environ Res Public Health. 2021;18(4):1415. doi: 10.3390/ijerph18041415.
    1. Nogueira L. Acute oxaloacetate exposure enhances resistance to fatigue in in vitro mouse soleus muscle. FASEB J. 2011 doi: 10.1096/fasebj.25.1_supplement.1104.5.
    1. Germain A, Ruppert D, Levine SM, et al. Metabolic profiling of a myalgic encephalomyelitis/chronic fatigue syndrome discovery cohort reveals disturbances in fatty acid and lipid metabolism. Mol Biosyst. 2017;13(2):371–379. doi: 10.1039/C6MB00600K.
    1. Fukuda K, Straus SE, Hickie I, et al. The chronic fatigue syndrome: a comprehensive approach to its definition and study. International Chronic Fatigue Syndrome Study Group. Ann Intern Med. 1994;121(12):953–959. doi: 10.7326/0003-4819-121-12-199412150-00009.
    1. Chalder T, Berelowitz G, Pawlikowska T, et al. Development of a fatigue scale. J Psychosom Res. 1993;37(2):147–153. doi: 10.1016/0022-3999(93)90081-P.
    1. Krupp LB, LaRocca NG, Muir-Nash J, et al. The fatigue severity scale. Application to patients with multiple sclerosis and systemic lupus erythematosus. Arch Neurol. 1989;46(10):1121–1123. doi: 10.1001/archneur.1989.00520460115022.
    1. Yang M, Keller S, Lin JS. Psychometric properties of the PROMIS((R)) Fatigue Short Form 7a among adults with myalgic encephalomyelitis/chronic fatigue syndrome. Qual Life Res. 2019;28(12):3375–3384. doi: 10.1007/s11136-019-02289-4.
    1. Cho HJ, Hotopf M, Wessely S. The placebo response in the treatment of chronic fatigue syndrome: a systematic review and meta-analysis. Psychosom Med. 2005;67(2):301–313. doi: 10.1097/01.psy.0000156969.76986.e0.
    1. Ijare O, Conway D, Cash A, et al. CBMT-49. oxaloacetate alters glucose metabolism in glioblastoma 13C isotopomer study. Neuro-Oncology. 2019;21(Supplement_6):vi43–vi44. doi: 10.1093/neuonc/noz175.171.
    1. Morris G, Maes M. Oxidative and Nitrosative Stress and immune-inflammatory pathways in patients with Myalgic Encephalomyelitis (ME)/Chronic Fatigue Syndrome (CFS) Curr Neuropharmacol. 2014;12(2):168–185. doi: 10.2174/1570159X11666131120224653.
    1. Wilkins HM, Harris JL, Carl SM, et al. Oxaloacetate activates brain mitochondrial biogenesis, enhances the insulin pathway, reduces inflammation and stimulates neurogenesis. Hum Mol Genet. 2014;23(24):6528–6541. doi: 10.1093/hmg/ddu371.
    1. Williams DS, Cash A, Hamadani L, et al. Oxaloacetate supplementation increases lifespan in Caenorhabditis elegans through an AMPK/FOXO-dependent pathway. Aging Cell. 2009;8(6):765–768. doi: 10.1111/j.1474-9726.2009.00527.x.
    1. Haslam JM, Krebs HA. The permeability of mitochondria to oxaloacetate and malate. Biochem J. 1968;107(5):659–667. doi: 10.1042/bj1070659.
    1. Booth NE, Myhill S, McLaren-Howard J. Mitochondrial dysfunction and the pathophysiology of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) Int J Clin Exp Med. 2012;5(3):208–220.
    1. Wilkins HM, Koppel S, Carl SM, et al. Oxaloacetate enhances neuronal cell bioenergetic fluxes and infrastructure. J Neurochem. 2016 doi: 10.1111/jnc.13545.
    1. Brown AE, Dibnah B, Fisher E, 2018. Pharmacological activation of AMPK and glucose uptake in cultured human skeletal muscle cells from patients with ME/CFS. Biosci Rep.
    1. Yoshikawa K. Studies on the anti-diabetic effect of sodium oxaloacetate. Tohoku J Exp Med. 1968;96(2):127–141. doi: 10.1620/tjem.96.127.
    1. Vidoni ED, Choi IY, Lee P, et al. Safety and target engagement profile of two oxaloacetate doses in Alzheimer's patients. Alzheimers Dement. 2020 doi: 10.1002/alz.12156.
    1. Desagher S, Glowinski J, Premont J. Pyruvate protects neurons against hydrogen peroxide-induced toxicity. J Neurosci. 1997;17(23):9060–9067. doi: 10.1523/JNEUROSCI.17-23-09060.1997.
    1. Puntel RL, Nogueira CW, Rocha JB. Krebs cycle intermediates modulate thiobarbituric acid reactive species (TBARS) production in rat brain in vitro. Neurochem Res. 2005;30(2):225–235. doi: 10.1007/s11064-004-2445-7.
    1. Yamamoto HA, Mohanan PV. Effect of alpha-ketoglutarate and oxaloacetate on brain mitochondrial DNA damage and seizures induced by kainic acid in mice. Toxicol Lett. 2003;143(2):115–122. doi: 10.1016/S0378-4274(03)00114-0.

Source: PubMed

3
Prenumerera