Mesenchymal stem cells derived from perinatal tissues for treatment of critically ill COVID-19-induced ARDS patients: a case series

Seyed-Mohammad Reza Hashemian, Rasoul Aliannejad, Morteza Zarrabi, Masoud Soleimani, Massoud Vosough, Seyedeh-Esmat Hosseini, Hamed Hossieni, Saeid Heidari Keshel, Zeinab Naderpour, Ensiyeh Hajizadeh-Saffar, Elham Shajareh, Hamidreza Jamaati, Mina Soufi-Zomorrod, Naghmeh Khavandgar, Hediyeh Alemi, Aliasghar Karimi, Neda Pak, Negin Hossieni Rouzbahani, Masoumeh Nouri, Majid Sorouri, Ladan Kashani, Hoda Madani, Nasser Aghdami, Mohammad Vasei, Hossein Baharvand, Seyed-Mohammad Reza Hashemian, Rasoul Aliannejad, Morteza Zarrabi, Masoud Soleimani, Massoud Vosough, Seyedeh-Esmat Hosseini, Hamed Hossieni, Saeid Heidari Keshel, Zeinab Naderpour, Ensiyeh Hajizadeh-Saffar, Elham Shajareh, Hamidreza Jamaati, Mina Soufi-Zomorrod, Naghmeh Khavandgar, Hediyeh Alemi, Aliasghar Karimi, Neda Pak, Negin Hossieni Rouzbahani, Masoumeh Nouri, Majid Sorouri, Ladan Kashani, Hoda Madani, Nasser Aghdami, Mohammad Vasei, Hossein Baharvand

Abstract

Background: Acute respiratory distress syndrome (ARDS) is a fatal complication of coronavirus disease 2019 (COVID-19). There are a few reports of allogeneic human mesenchymal stem cells (MSCs) as a potential treatment for ARDS. In this phase 1 clinical trial, we present the safety, feasibility, and tolerability of the multiple infusions of high dose MSCs, which originated from the placenta and umbilical cord, in critically ill COVID-19-induced ARDS patients.

Methods: A total of 11 patients diagnosed with COVID-19-induced ARDS who were admitted to the intensive care units (ICUs) of two hospitals enrolled in this study. The patients were critically ill with severe hypoxemia and required mechanical ventilation. The patients received three intravenous infusions (200 × 106 cells) every other day for a total of 600 × 106 human umbilical cord MSCs (UC-MSCs; 6 cases) or placental MSCs (PL-MSCs; 5 cases).

Findings: There were eight men and three women who were 42 to 66 years of age. Of these, six (55%) patients had comorbidities of diabetes, hypertension, chronic lymphocytic leukemia (CLL), and cardiomyopathy (CMP). There were no serious adverse events reported 24-48 h after the cell infusions. We observed reduced dyspnea and increased SpO2 within 48-96 h after the first infusion in seven patients. Of these seven patients, five were discharged from the ICU within 2-7 days (average: 4 days), one patient who had signs of acute renal and hepatic failure was discharged from the ICU on day 18, and the last patient suddenly developed cardiac arrest on day 7 of the cell infusion. Significant reductions in serum levels of tumor necrosis factor-alpha (TNF-α; P < 0.01), IL-8 (P < 0.05), and C-reactive protein (CRP) (P < 0.01) were seen in all six survivors. IL-6 levels decreased in five (P = 0.06) patients and interferon gamma (IFN-γ) levels decreased in four (P = 0.14) patients. Four patients who had signs of multi-organ failure or sepsis died in 5-19 days (average: 10 days) after the first MSC infusion. A low percentage of lymphocytes (< 10%) and leukocytosis were associated with poor outcome (P = 0.02). All six survivors were well with no complaints of dyspnea on day 60 post-infusion. Radiological parameters of the lung computed tomography (CT) scans showed remarkable signs of recovery.

Interpretation: We suggest that multiple infusions of high dose allogeneic prenatal MSCs are safe and can rapidly improve respiratory distress and reduce inflammatory biomarkers in some critically ill COVID-19-induced ARDS cases. Patients that develop sepsis or multi-organ failure may not be good candidates for stem cell therapy. Large randomized multicenter clinical trials are needed to discern the exact therapeutic potentials of MSC in COVID-19-induced ARDS.

Keywords: Acute respiratory distress syndrome; COVID-19; Cell therapy; Mesenchymal stromal cells; Placenta; Pneumonia; SARS-CoV-2; Umbilical cord.

Conflict of interest statement

Massoud Vosough and Hossein Baharvand are regulatory affairs manager and member of the board of directors in CellTech Pharmed Company, respectively. They have no-share in this company.

The other authors have no conflict of interest.

Figures

Fig. 1
Fig. 1
Change in patients’ serum biomarker levels on days 0 (baseline) and 5 after the first infusion. Analysis of biomarkers on before (baseline) and 5 days after the first infusion (24 h after the last infusion) demonstrated a significant reduction in IL-8, tumor necrosis factor-alpha (TNF-α), and C-reactive protein (CRP) in all six survivors. Serum IL-6 levels and interferon gamma (INF-ɣ) reduced in five and four of the recovered patients, respectively. IL-4 and IL-10 levels increased in four cases, but the differences were not statistically significant. *P < 0.05; ns, not significant
Fig. 2
Fig. 2
Chest computed tomography (CT) images in three survivors. a Patient #2: First row is prior to cell infusion. Note the extensive mixed ground glass opacities, crazy paving appearance, vascular dilatation, consolidations with peripheral subpleural dominancy, and bilateral mild pleural effusion. The CT severity score for all five lobes was 24. The second row shows CT images 1 day after the third dose of cell therapy. There is a decrease in extension of consolidations associated with band-like opacities and traction bronchiectasis are evident. CT severity score for all five lobes was 18. The third row shows near complete resolution of opacities and subpleural bands without residual fibrosis 50 days after MSC therapy. The CT severity score for all five lobes was 2. The percentage of lung involvement in each image of the first, second, and third columns was assessed at pretreatment, 1 day after the third post-cell therapy, and after 50 days of treatment, respectively, as follows. First column: about 60%, 25%, and 0%; second column: about 75%, 30%, and 0%; and third column: about 90%, 50%, and 2%. b Patient #3: First row shows chest CT images before cell therapy. Note the patchy areas of ground glass opacity and consolidations in the subpleural regions of the lungs. The CT severity score for all five lobes was 16. The second row shows a significant reduction in the extension of lung involvement 12 days after the third dose of cell therapy. Most consolidations had resolved completely with only band-like opacities and mild tiny residual ground glass opacities present. The CT severity score for all five lobes was 8. The percentage of lung involvement in single images from the first, second, and third columns was assessed at pretreatment and 12 days after the third cell therapy, respectively, as follows. First column: about 15% and 5%; second column: about 35% and 15%; and third column: about 20% and 3%. c Patient #8: Axial CT scan images from the base of the lung before cell therapy. The left image shows peripheral subpleural consolidations and ground glass opacity. At the same level, the right image shows a significant decrease in consolidations 12 days after cell therapy; however, the patient developed bilateral pleural effusion due to acute renal failure during the course of the disease. The CT severity score for all five lobes at the initial lung CT scan was 24, which decreased to 13 at 12 days after cell therapy. The percentage of lung involvement in the pretreatment image was about 60%, which decreased to 20% in the post-treatment image. In all images, different patterns of lung involvement have the following annotations: crazy paving appearance (black boxes), consolidation (black stars), pure ground glass opacity (solid white arrows), vascular dilatation (solid white arrows), traction bronchiectasis (solid black arrows), subpleural band (dashed white arrows), architectural distortion (white boxes), and pleural effusion (dashed black arrows)

References

    1. Wang D, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan. China. JAMA. 2020;323(11):1061–9. doi: 10.1001/jama.2020.1585.
    1. Zhu N, et al. A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med. 2020;382(8):727–733. doi: 10.1056/NEJMoa2001017.
    1. Huang C, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395(10223):497–506. doi: 10.1016/S0140-6736(20)30183-5.
    1. Chen N, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395(10223):507–513. doi: 10.1016/S0140-6736(20)30211-7.
    1. Zhou F, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395(10229):1054–1062. doi: 10.1016/S0140-6736(20)30566-3.
    1. Ramezankhani R, et al. Therapeutic modalities and novel approaches in regenerative medicine for COVID-19. Int J Antimicrob Agents. 2020;56(6):106208. doi: 10.1016/j.ijantimicag.2020.106208.
    1. Hossein-Khannazer N, et al. Novel therapeutic approaches for treatment of COVID-19. J Mol Med (Berl) 2020;98(6):789–803. doi: 10.1007/s00109-020-01927-6.
    1. Qin H, Zhao A. Mesenchymal stem cell therapy for acute respiratory distress syndrome: from basic to clinics. Protein Cell. 2020;11(10):707–722. doi: 10.1007/s13238-020-00738-2.
    1. Keating A. Mesenchymal stromal cells: new directions. Cell Stem Cell. 2012;10(6):709–716. doi: 10.1016/j.stem.2012.05.015.
    1. Zhu H, et al. Therapeutic effects of human umbilical cord-derived mesenchymal stem cells in acute lung injury mice. Sci Rep. 2017;7:39889. doi: 10.1038/srep39889.
    1. Lee DK, Song SU. Immunomodulatory mechanisms of mesenchymal stem cells and their therapeutic applications. Cell Immunol. 2018;326:68–76. doi: 10.1016/j.cellimm.2017.08.009.
    1. Jiang W, Xu J. Immune modulation by mesenchymal stem cells. Cell Prolif. 2020;53(1):e12712.
    1. Fu X, et al. Mesenchymal stem cell migration and tissue repair. Cells. 2019;8(8):784. doi: 10.3390/cells8080784.
    1. Sohni A, Verfaillie CM. Mesenchymal stem cells migration homing and tracking. Stem Cells Int. 2013;2013:130763. doi: 10.1155/2013/130763.
    1. Lee RH, et al. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell Stem Cell. 2009;5(1):54–63. doi: 10.1016/j.stem.2009.05.003.
    1. Wysoczynski M, Khan A, Bolli R. New paradigms in cell therapy: repeated dosing, intravenous delivery, immunomodulatory actions, and new cell types. Circ Res. 2018;123(2):138–158. doi: 10.1161/CIRCRESAHA.118.313251.
    1. Khoury M, et al. Current status of cell-based therapies for respiratory virus infections: applicability to COVID-19. Eur Respir J. 2020;55(6):2000858. doi: 10.1183/13993003.00858-2020.
    1. Sanchez-Guijo F, et al. Adipose-derived mesenchymal stromal cells for the treatment of patients with severe SARS-CoV-2 pneumonia requiring mechanical ventilation. A proof of concept study. EClinicalMed. 2020;25:100454. doi: 10.1016/j.eclinm.2020.100454.
    1. Liang B, et al. Clinical remission of a critically ill COVID-19 patient treated by human umbilical cord mesenchymal stem cells. ChinaXiv. 2020;2:v1.
    1. Leng Z, et al. Transplantation of ACE2(−) mesenchymal stem cells improves the outcome of patients with COVID-19 pneumonia. Aging Dis. 2020;11(2):216–228. doi: 10.14336/AD.2020.0228.
    1. Wu M, et al. Comparison of the biological characteristics of mesenchymal stem cells derived from the human placenta and umbilical cord. Sci Rep. 2018;8(1):5014. doi: 10.1038/s41598-018-23396-1.
    1. W.H.O. Clinical management of severe acute respiratory infection (SARI) when COVID-19 disease is suspected: interim guidance, 13 March 2020. World Health Organization.
    1. Khalifeh Soltani S, et al. Safety and efficacy of allogenic placental mesenchymal stem cells for treating knee osteoarthritis: a pilot study. Cytotherapy. 2019;21(1):54–63. doi: 10.1016/j.jcyt.2018.11.003.
    1. Bauer G, Elsallab M, Abou-El-Enein M. Concise review: a comprehensive analysis of reported adverse events in patients receiving unproven stem cell-based interventions. Stem Cells Transl Med. 2018;7(9):676–685. doi: 10.1002/sctm.17-0282.
    1. Rezoagli, E., et al., The safety and efficiency of addressing ARDS using stem cell therapies in clinical trials, in stem cell-based therapy for lung disease. 2019, Springer. p. 219–238.
    1. Wilson JG, et al. Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. Lancet Respir Med. 2015;3(1):24–32. doi: 10.1016/S2213-2600(14)70291-7.
    1. Matthay MA, et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): a randomised phase 2a safety trial. Lancet Respir Med. 2019;7(2):154–162. doi: 10.1016/S2213-2600(18)30418-1.
    1. Zheng G, et al. Treatment of acute respiratory distress syndrome with allogeneic adipose-derived mesenchymal stem cells: a randomized, placebo-controlled pilot study. Respir Res. 2014;15:39. doi: 10.1186/1465-9921-15-39.
    1. Atluri S, Manchikanti L, Hirsch JA. Expanded umbilical cord mesenchymal stem cells (UC-MSCs) as a therapeutic strategy in managing critically ill COVID-19 patients: the case for compassionate use. Pain Physician. 2020;23(2):E71–E83.
    1. Weiss ARR, Dahlke MH. Immunomodulation by mesenchymal stem cells (MSCs): mechanisms of action of living, apoptotic, and dead MSCs. Front Immunol. 2019;10:1191. doi: 10.3389/fimmu.2019.01191.
    1. Walter J, Ware LB, Matthay MA. Mesenchymal stem cells: mechanisms of potential therapeutic benefit in ARDS and sepsis. Lancet Respir Med. 2014;2(12):1016–1026. doi: 10.1016/S2213-2600(14)70217-6.
    1. Meng SS, et al. mTOR/STAT-3 pathway mediates mesenchymal stem cell-secreted hepatocyte growth factor protective effects against lipopolysaccharide-induced vascular endothelial barrier dysfunction and apoptosis. J Cell Biochem. 2019;120(3):3637–3650. doi: 10.1002/jcb.27642.
    1. Lee JW, et al. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc Natl Acad Sci U S A. 2009;106(38):16357–16362. doi: 10.1073/pnas.0907996106.
    1. Sauler M, Bazan IS, Lee PJ. Cell death in the lung: the apoptosis-necroptosis axis. Annu Rev Physiol. 2019;81:375–402. doi: 10.1146/annurev-physiol-020518-114320.
    1. Naji A, et al. Rationale for determining the functional potency of mesenchymal stem cells in preventing regulated cell death for therapeutic use. Stem Cells Transl Med. 2017;6(3):713–719. doi: 10.5966/sctm.2016-0289.
    1. Tkach M, Thery C. Communication by extracellular vesicles: where we are and where we need to go. Cell. 2016;164(6):1226–1232. doi: 10.1016/j.cell.2016.01.043.
    1. Sengupta V, et al. Exosomes derived from bone marrow mesenchymal stem cells as treatment for severe COVID-19. Stem Cells Dev. 2020;29(12):747–54. doi: 10.1089/scd.2020.0080.
    1. Moll, G., et al., Cryopreserved or fresh mesenchymal stromal cells: only a matter of taste or key to unleash the full clinical potential of MSC therapy?, in Biobanking and cryopreservation of stem cells. 2016, Springer. p. 77–98.
    1. Tang N, et al. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost. 2020;18(4):844–847. doi: 10.1111/jth.14768.

Source: PubMed

3
購読する