First-in-human high-cumulative-dose stem cell therapy in idiopathic pulmonary fibrosis with rapid lung function decline

Alexander Averyanov, Irina Koroleva, Mikhail Konoplyannikov, Veronika Revkova, Victor Lesnyak, Vladimir Kalsin, Olesya Danilevskaya, Alexey Nikitin, Anna Sotnikova, Svetlana Kotova, Vladimir Baklaushev, Alexander Averyanov, Irina Koroleva, Mikhail Konoplyannikov, Veronika Revkova, Victor Lesnyak, Vladimir Kalsin, Olesya Danilevskaya, Alexey Nikitin, Anna Sotnikova, Svetlana Kotova, Vladimir Baklaushev

Abstract

Previous phase I studies demonstrated safety and some beneficial effects of mesenchymal stem cells (MSCs) in patients with mild to moderate idiopathic pulmonary fibrosis (IPF). The aim of our study was to evaluate the safety, tolerability, and efficacy of a high cumulative dose of bone marrow MSCs in patients with rapid progressive course of severe to moderate IPF. Twenty patients with forced ventilation capacity (FVC) ≥40% and diffusing capacity of the lung for carbon monoxide (DLCO) ≥20% with a decline of both >10% over the previous 12 months were randomized into two groups: one group received two intravenous doses of allogeneic MSCs (2 × 108 cells) every 3 months, and the second group received a placebo. A total amount of 1.6 × 109 MSCs had been administered to each patient after the study completion. There were no significant adverse effects after administration of MSCs in any patients. In the group of MSC therapy, we observed significantly better improvement for the 6-minute walk distance in 13 weeks, for DLCO in 26 weeks, and for FVC in 39 weeks compared with placebo. FVC for 12 months in the MSCs therapy group increased by 7.8% from baseline, whereas it declined by 5.9% in the placebo group. We did not find differences between the groups in mortality (two patients died in each group) or any changes in the high-resolution computed tomography fibrosis score. In patients with IPF and a rapid pulmonary function decline, therapy with high doses of allogeneic MSCs is a safe and promising method to reduce disease progression.

Keywords: adult human bone marrow; adult stem cells; bone marrow stromal cells; cellular therapy; clinical trials; lung; stem cell transplantation; transplantation tolerance.

Conflict of interest statement

V.K. and V.B. declared researched funding from Federal Medical and Biological Agency, Order 20.001.13.800. The other authors indicated no potential conflict of interest.

© The Authors. Stem Cells Translational Medicine published by Wiley Periodicals, Inc. on behalf of AlphaMed Press.

Figures

Figure 1
Figure 1
Characterization of cultured human bone marrow‐derived mesenchymal stem cell (MSCs; passage 4) before the transplantation. A, CD‐immunophenotyping of MSCs using flow cytometry. Red histograms represent isotype specific Ig control; blue histograms: fluorescein isothiocyanate/PE‐conjugated antihuman CD antibodies. B‐D, Differentiation analysis. MSCs were characterized by their differentiation potential by staining with Oil Red O—adipogenic lineage, B, Alcian blue—specific to sulfated GAGs, C, and alizarin red—osteogenic lineage, D. Magnification ×200, B‐D. E, Karyotype analysis of human MSCs, mFISH visualization method
Figure 2
Figure 2
CONSORT flow diagram
Figure 3
Figure 3
Secondary endpoints dynamics. A‐C, Differences (delta) of real values (y‐axis) of forced ventilation capacity (FVC; A), diffusing capacity of the lung for carbon monoxide, B, and 6MWD, C, from the baseline level, during the treatment period (median, minimum and maximum, 25% and 75% quartiles are shown). D, Change from the baseline in FVC median (% of predicted) during 12 months before the treatment and during the treatment (by week 52) in the main group and the control. In the legend, 3, 6, 9, and 12 mean the values after 3, 6, 9, and 12 months, respectively

References

    1. Raghu G, Collard HR, Egan JJ et al. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence‐based guidelines for diagnosis and management. Am J Respir Crit Care Med. 2011;183:788‐824.
    1. Navaratnam V, Fleming KM, West J et al. The rising incidence of idiopathic pulmonary fibrosis in the UK. Thorax. 2011;66:462‐467.
    1. Reichmann WM, Yu YF, Macaulay D et al. Change in forced vital capacity and associated subsequent outcomes in patients with newly diagnosed idiopathic pulmonary fibrosis. BMC Pulm Med. 2015;15:167.
    1. Kim HJ, Perlman D, Tomic R. Natural history of idiopathic pulmonary fibrosis. Respir Med. 2015;109:661‐670.
    1. Ley B, Collard HR, King TE. Clinical course and prediction of survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2011;183:431‐440.
    1. Kim DS, Collard HR, King TE Jr. Classification and natural history of the idiopathic interstitial pneumonias. Proc Am Thorac Soc. 2006;3:285‐292.
    1. Naira G, Matelaa A, Kurbanov D et al. Newer developments in idiopathic pulmonary fibrosis in the era of anti‐fibrotic medications. Exp Rev Respir Med. 2016;10:699‐711.
    1. Song JW, Hong SB, Lim CM et al. Acute exacerbation of idiopathic pulmonary fibrosis: incidence, risk factors and outcome. Eur Respir J. 2011;37:356‐363.
    1. Paterniti MO, Bi Y, Rekić D et al. Acute exacerbation and decline in forced vital capacity are associated with increased mortality in idiopathic pulmonary fibrosis. Ann Am Thorac Soc. 2017;14:1395‐1402.
    1. Richeldi L, du Bois RM, Raghu G et al. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med. 2014;370:2071‐2082.
    1. King T, Bradford W, Castro‐Bernardini S et al. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med. 2014;370:2083‐2092.
    1. Liu M, Ren D, Wu D et al. Stem cell and idiopathic pulmonary fibrosis: mechanisms and treatment. Curr Stem Cell Res Ther. 2015;10:466‐476.
    1. Toonkel RL, Hare JM, Matthay MA et al. Mesenchymal stem cells and idiopathic pulmonary fibrosis: potential for clinical testing. Am J Respir Crit Care Med. 2013;188:133‐140.
    1. Chambers DC, Enever D, Ilic N et al. A phase 1b study of placenta‐derived mesenchymal stromal cells in patients with idiopathic pulmonary fibrosis. Respirology. 2014;19:1013‐1018.
    1. Tzouvelekis A, Paspaliaris V, Koliakos G et al. A prospective, non‐randomized, no placebo‐controlled, phase Ib clinical trial to study the safety of the adipose derived stromal cells‐stromal vascular fraction in idiopathic pulmonary fibrosis. J Transl Med. 2013;11:171.
    1. Glassberg MK, Minkiewicz J, Toonkel RL et al. Allogeneic human mesenchymal stem cells in patients with idiopathic pulmonary fibrosis via intravenous delivery (AETHER): a phase I, safety, clinical trial. Chest. 2017;151:971‐981.
    1. Miller MR, Hankinson J, Brusasco V et al. Standardisation of spirometry. Eur Respir J. 2005;26:319‐338.
    1. Macintyre N, Crapo RO, Viegi G et al. Standardisation of the single‐breath determination of carbon monoxide uptake in the lung. Eur Respir J. 2005;26:720‐735.
    1. ATS Committee on Proficiency Standards for Clinical Pulmonary Function Laboratories . ATS statement: guidelines for the six‐minute walk test. Am J Respir Crit Care Med. 2002;166:111‐117.
    1. Oda K, Ishimoto H, Yatera K et al. High‐resolution CT scoring system‐based grading scale predicts the clinical outcomes in patients with idiopathic pulmonary fibrosis. Respir Res. 2014;15:10.
    1. Lennon DP, Caplan AI. Mesenchymal stem cells for tissue engineering In: Freshney RI, Vunjak‐Novakovic G, eds. Culture of Cells for Tissue Engineering. Hoboken, NJ: Wiley; 2006.
    1. Liehr T, Weise A, Hamid AB et al. Multicolor FISH methods in current clinical diagnostics. Expert Rev Mol Diagn. 2013;13:251‐255.
    1. Kundel HL, Polansky M. Measurement of observer agreement. Radiology. 2003;228:303‐308.
    1. Wilson JG, Liu KD, Zhuo H et al. Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. Lancet Respir Med. 2015;3:24‐32.
    1. Pätilä T, Lehtinen M, Vento A et al. Autologous bone marrow mononuclear cell transplantation in ischemic heart failure: a prospective, controlled, randomized, double‐blind study of cell transplantation combined with coronary bypass. J Heart Lung Transplant. 2014;33:567‐574.
    1. Zhao R, Su Z, Wu J et al. Serious adverse events of cell therapy for respiratory diseases: a systematic review and meta‐analysis. Oncotarget. 2017;8:30511‐30523.
    1. Meng M, Liu Y, Wang W et al. Umbilical cord mesenchymal stem cell transplantation in the treatment of multiple sclerosis. Am J Transl Res. 2018;10:212‐223.
    1. Kong D, Zhuang X, Wang D et al. Umbilical cord mesenchymal stem cell transfusion ameliorated hyperglycemia in patients with type 2 diabetes mellitus. Clin Lab. 2014;60:1969‐1976.
    1. Afzal MR, Samanta A, Shah ZI et al. Adult bone marrow cell therapy for ischemic heart disease: evidence and insights from randomized controlled trials. Circ Res. 2015;117:558‐575.
    1. Lee SH, Jang AS, Kim YE et al. Modulation of cytokine and nitric oxide by mesenchymal stem cell transfer in lung injury/fibrosis. Respir Res. 2010;11:16.
    1. Ortiz LA, DuTreil M, Fattman C et al. Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proc Natl Acad Sci USA. 2007;104:11002‐11007.
    1. Cahill EF, Kennelly H, Carty F et al. Hepatocyte growth factor is required for mesenchymal stromal cell protection against bleomycin‐induced pulmonary fibrosis. Stem Cells Translational Medicine. 2016;5:1307‐1318.
    1. Wang X, Gao JL, Zhao MM et al. Therapeutic effects of conditioned medium from bone marrow‐derived mesenchymal stem cells on epithelial‐mesenchymal transition in A549 cells. Int J Mol Med. 2018;41:659‐668.
    1. Andersson‐Sjöland A, de Alba CG, Nihlberg K et al. Fibrocytes are a potential source of lung fibroblasts in idiopathic pulmonary fibrosis. Int J Biochem Cell Biol. 2008;40:2129‐2140.
    1. Badri L, Murray S, Liu LX et al. Mesenchymal stromal cells in bronchoalveolar lavage as predictors of bronchiolitis obliterans syndrome. Am J Respir Crit Care Med. 2011;183:1062‐1070.
    1. Huntsman HD, Zachwieja N, Zou K et al. Mesenchymal stem cells contribute to vascular growth in skeletal muscle in response to eccentric exercise. Am J Physiol Heart Circ Physiol. 2013;304:H72‐H81.
    1. Hedhli J, Konopka CJ, Schuh S et al. Multimodal assessment of mesenchymal stem cell therapy for diabetic vascular complications. Theranostics. 2017;7:3876‐3888.
    1. Li LF, Chang YL, Chen NH et al. Inhibition of Src and forkhead box O1 signaling by induced pluripotent stem‐cell therapy attenuates hyperoxia‐augmented ventilator‐induced diaphragm dysfunction. Transl Res. 2016;173:131‐147.
    1. de Mendonça L, Felix NS, Blanco NG et al. Mesenchymal stromal cell therapy reduces lung inflammation and vascular remodeling and improves hemodynamics in experimental pulmonary arterial hypertension. Stem Cell Res Ther. 2017;8:220.
    1. Maffioli E, Nonnis S, Angioni R et al. Proteomic analysis of the secretome of human bone marrow‐derived mesenchymal stem cells primed by pro‐inflammatory cytokines. J Proteomics. 2017;166:115‐126.
    1. Samuelsson H, Ringden O, Lonnies H et al. Optimizing in vitro conditions for immunomodulation and expansion of mesenchymal stromal cells. Cytotherapy. 2009;11:129‐136.
    1. Kuçi Z, Bönig H, Kreyenberg H et al. Mesenchymal stromal cells from pooled mononuclear cells of multiple bone marrow donors as rescue therapy in pediatric severe steroid‐refractory graft‐versus‐host disease: a multicenter survey. Haematologica. 2016;101:985‐994.

Source: PubMed

3
Se inscrever