Clinical Study of Mesenchymal Stem/Stromal Cell Therapy for the Treatment of Frailty: A Proposed Experimental Design for Therapeutic and Mechanistic Investigation

Duc M Hoang, Kien T Nguyen, Van T Hoang, Lan T M Dao, Hang T Bui, Thanh T K Ho, Thuy T P Nguyen, Anh T L Ngo, Hoa K Nguyen, Liem Nguyen Thanh, Duc M Hoang, Kien T Nguyen, Van T Hoang, Lan T M Dao, Hang T Bui, Thanh T K Ho, Thuy T P Nguyen, Anh T L Ngo, Hoa K Nguyen, Liem Nguyen Thanh

Abstract

Frailty, a specific condition of increased vulnerability and reduced general health associated with aging in older people, is an emerging problem worldwide with major implications for clinical practice and public health. Recent preclinical and clinical studies have supported the safety of mesenchymal stem/stromal cells (MSCs) in the treatment of frailty. Comprehensive study is needed to assess the interrelationship between the condition of frailty and the effects of MSC-based therapy. This randomized controlled phase I/II trial aims to investigate the safety and potential therapeutic efficacy of the allogeneic administration of umbilical cord-derived MSCs (UC-MSCs) in combination with the standard treatment for frailty in Vietnam. Moreover, this study describes the rationales, study designs, methodologies, and analytical strategies currently employed in stem cell research and clinical studies. The primary outcome measures will include the incidences of prespecified administration-associated adverse events and serious adverse events. The potential efficacy will be evaluated based on improvements in frailty conditions (including those determined through a physical examination, patient-reported outcomes, quality of life, immune markers of frailty, metabolism analysis, and cytokine markers from patient plasma). This clinical trial and stem cell analysis associated with patient sampling at different time points aim to identify and characterize the potential effects of UC-MSCs on improving frailty based on the stem cell quality, cytokine/growth factor secretion profiles of UC-MSCs, cellular senescence, and metabolic analysis of patient CD3+ cells providing fundamental knowledge for designing and implementing research strategies in future studies. Clinical Trials Registration Number: NCT04919135.

Keywords: Allogeneic cell therapy; Clinical trial; Stem cell phenotype; Umbilical cord.

© The Author(s) 2021. Published by Oxford University Press on behalf of The Gerontological Society of America.

Figures

Figure 1.
Figure 1.
Interrelationship among hallmarks of aging, clinical symptoms of frailty, and potential regenerative therapy. Aging people are constantly affected by oxidative stress and inflammatory responses throughout their later years. In an extreme condition of this interaction, frailty is developed and defined by 5 major criteria, including unintentional weight loss, exhaustion, slow gait speed, weak grip strength, and low physical activity. Frailty is strongly associated with the aging process, which includes 6 important hallmarks: genomic instability, deregulated proteostasis, nutrient sensing, metabolic malfunctions, cellular senescence, stem cell exhaustion, and compromised cellular communication. Emerging stem cell therapy provides a potential approach to tackle these problems by providing ex vivo cultured stem cells that are able to improve the frailty condition by multiple mechanisms.
Figure 2.
Figure 2.
Proposed functional analysis to reveal the mechanism of action of UC-MSCs in the treatment of frailty. (A) The UC-MSC line was derived from Vinmec Biobank and was intensively characterized and cultured under xeno-free and serum-free conditions. These cells will be expanded to reach the transplantation dose for 22 patients (2 administrations with 1.5 × 106 cells/kg patient body weight) and cryopreserved for long-term storage. Cell viability, MSC marker, karyotype, product sterility (bacterial, fungal, and mycoplasma detection) and endotoxin will be assessed according to the Vietnamese Ministry of Health guideline and the International Society for Cell and Gene Therapy criteria of MSCs. (B) Peripheral blood at baseline and at 1, 3, 6, and 9 months will be collected, and (C) detection of cytokines/chemokines/growth factors using procartaplex technology derived from patient plasma will be performed, whereas CD3+ cells will be isolated for: (D) immunological analysis, (E) cellular senescence, and (F) metabolic analysis using Seahorse XFe96 analyzer. MSCs = mesenchymal stem/stromal cells; UC-MSCs = umbilical cord-derived mesenchymal stem/stromal cells.

References

    1. Hoogendijk EO, Afilalo J, Ensrud KE, Kowal P, Onder G, Fried LP. Frailty: implications for clinical practice and public health. Lancet. 2019;394(10206):1365–1375. doi:10.1016/S0140-6736(19)31786-6
    1. Dung V, Thi Mai Lan N, Thu Trang V, et al. . Quality of life of older adults in nursing homes in Vietnam. Health Psychol Open. 2020;7(2):2055102920954710. doi:10.1177/2055102920954710
    1. Collard RM, Boter H, Schoevers RA, Oude Voshaar RC. Prevalence of frailty in community-dwelling older persons: a systematic review. J Am Geriatr Soc. 2012;60(8):1487–1492. doi:10.1111/j.1532-5415.2012.04054.x
    1. Nguyen TV, Le D, Tran KD, Bui KX, Nguyen TN. Frailty in older patients with acute coronary syndrome in Vietnam. Clin Interv Aging. 2019;14:2213–2222. doi:10.2147/CIA.S234597
    1. Fried LP, Tangen CM, Walston J, et al. ; Cardiovascular Health Study Collaborative Research Group . Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci. 2001;56(3):M146–M156. doi:10.1093/gerona/56.3.m146
    1. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153(6):1194–1217. doi:10.1016/j.cell.2013.05.039
    1. Soysal P, Arik F, Smith L, Jackson SE, Isik AT. Inflammation, frailty and cardiovascular disease. Adv Exp Med Biol. 2020;1216:55–64. doi:10.1007/978-3-030-33330-0_7
    1. Lim JE, Son Y. Endogenous stem cells in homeostasis and aging. Tissue Eng Regen Med. 2017;14(6):679–698. doi:10.1007/s13770-017-0097-3
    1. Sun XL, Hao QK, Tang RJ, Xiao C, Ge ML, Dong BR. Frailty and rejuvenation with stem cells: therapeutic opportunities and clinical challenges. Rejuvenation Res. 2019;22(6):484–497. doi:10.1089/rej.2017.2048
    1. Golpanian S, DiFede DL, Pujol MV, et al. . Rationale and design of the allogeneiC human mesenchymal stem cells (hMSC) in patients with aging fRAilTy via intravenoUS delivery (CRATUS) study: a phase I/II, randomized, blinded and placebo controlled trial to evaluate the safety and potential efficacy of allogeneic human mesenchymal stem cell infusion in patients with aging frailty. Oncotarget. 2016;7(11):11899–11912. doi:10.18632/oncotarget.7727
    1. Tompkins BA, DiFede DL, Khan A, et al. . Allogeneic mesenchymal stem cells ameliorate aging frailty: a phase II randomized, double-blind, placebo-controlled clinical trial. J Gerontol A Biol Sci Med Sci. 2017;72(11):1513–1522. doi:10.1093/gerona/glx137
    1. Beane OS, Fonseca VC, Cooper LL, Koren G, Darling EM. Impact of aging on the regenerative properties of bone marrow-, muscle-, and adipose-derived mesenchymal stem/stromal cells. PLoS One. 2014;9(12):e115963. doi:10.1371/journal.pone.0115963
    1. Zhou S, Greenberger JS, Epperly MW, et al. . Age-related intrinsic changes in human bone-marrow-derived mesenchymal stem cells and their differentiation to osteoblasts. Aging Cell. 2008;7(3):335–343. doi:10.1111/j.1474-9726.2008.00377.x
    1. Nguyen LT, Hoang DM, Nguyen KT, et al. . Type 2 diabetes mellitus duration and obesity alter the efficacy of autologously transplanted bone marrow-derived mesenchymal stem/stromal cells. Stem Cells Transl Med. 2021;10(9):1266–1278. doi:10.1002/sctm.20-0506
    1. Moberg L, Johansson H, Lukinius A, et al. . Production of tissue factor by pancreatic islet cells as a trigger of detrimental thrombotic reactions in clinical islet transplantation. Lancet. 2002;360(9350):2039–2045. doi:10.1016/s0140-6736(02)12020-4
    1. Ben-Porath I, Weinberg RA. When cells get stressed: an integrative view of cellular senescence. J Clin Invest. 2004;113(1):8–13. doi:10.1172/JCI20663
    1. Mather KA, Jorm AF, Parslow RA, Christensen H. Is telomere length a biomarker of aging? A review. J Gerontol A Biol Sci Med Sci. 2011;66(2):202–213. doi:10.1093/gerona/glq180
    1. Haapanen MJ, Perälä MM, Salonen MK, et al. . Telomere length and frailty: the Helsinki Birth Cohort Study. J Am Med Dir Assoc. 2018;19(8):658–662. doi:10.1016/j.jamda.2018.05.011
    1. Liu Y, Sanoff HK, Cho H, et al. . Expression of p16(INK4a) in peripheral blood T-cells is a biomarker of human aging. Aging Cell. 2009;8(4):439–448. doi:10.1111/j.1474-9726.2009.00489.x
    1. Diekman BO, Sessions GA, Collins JA, et al. . Expression of p16INK 4a is a biomarker of chondrocyte aging but does not cause osteoarthritis. Aging Cell. 2018;17(4):e12771. doi:10.1111/acel.12771
    1. Jeck WR, Siebold AP, Sharpless NE. Review: a meta-analysis of GWAS and age-associated diseases. Aging Cell. 2012;11(5):727–731. doi:10.1111/j.1474-9726.2012.00871.x
    1. Lee BY, Han JA, Im JS, et al. . Senescence-associated beta-galactosidase is lysosomal beta-galactosidase. Aging Cell. 2006;5(2):187–195. doi:10.1111/j.1474-9726.2006.00199.x

Source: PubMed

3
Abonnere