Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Case Report
Commentary
Editorial
Guest Editorial
Letter to Editor
Letter to the Editor
Media and News
Medical Innovator
Mini-Review
Original Article
Review Article
Short Communication
Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Case Report
Commentary
Editorial
Guest Editorial
Letter to Editor
Letter to the Editor
Media and News
Medical Innovator
Mini-Review
Original Article
Review Article
Short Communication
Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Case Report
Commentary
Editorial
Guest Editorial
Letter to Editor
Letter to the Editor
Media and News
Medical Innovator
Mini-Review
Original Article
Review Article
Short Communication
View/Download PDF

Translate this page into:

Mini-Review
2020
:15;
2
doi:
10.25259/GJMPBU_11_2020

Mesenchymal Stem Cells Therapy for Coronavirus COVID-19 Induced ARDS: A Promising Concept

Department of Sports Medicine and Orthopedics, Orthopedic Specialty Institute, Fort Lauderdale, Florida, United States
American University of Antigua College of Medicine, University Park Road, Coolidge, Antigua and Barbuda, United States
Department of Pre-Medicine, Florida International University, Miami, Florida, United States
Corresponding author: Jorge Antonio Gonzalez, MD Department of Sports Medicine and Orthopedics, Orthopedic Specialty Institute, Fort Lauderdale 33308, Florida, United States. jgonzalezmd1@gmail.com
Licence
This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-Share Alike 4.0 License, which allows others to remix, tweak, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Gonzalez JA, Gonzalez RR, Ayala LE. Mesenchymal Stem Cells Therapy for Coronavirus COVID-19 Induced ARDS: A Promising Concept. Glob J Med Pharm Biomed Update 2020;15:2.

Abstract

Novel coronavirus severe acute respiratory syndrome (SARS)-CoV-2 pandemic has devastated the world causing an enormous health and financial crisis like none seen before. As many physicians and scientists fight to find a cure and vaccine, the virus continues to spread unchecked with a high mortality rate. Pulmonary involvement and development of severe acute respiratory distress syndrome (ARDS) have been some of the main contributors of mortality and morbidity in SARS-CoV-2 infection. Mesenchymal stem cells (MSC) have gained clinical interest as a treatment option due to their immunomodulatory and antifibrotic properties. Due to the emerging need to find treatment options, multiple MSC clinical trials are undergoing. Cellular treatment may represent a viable option for the treatment of ARDS and coronavirus infection.

Keywords

Coronavirus
COVID-19
SARS-CoV-2
Mesenchymal stem cell
Stem cell therapy
Acute respiratory distress syndrome

INTRODUCTION

Toward the end of the year 2019, a rapidly spreading pulmonary illness was first reported in Wuhan, China. Fudan University first declared that the causative agent of this disease was a new coronavirus strain that has a 5% genetic association with severe acute respiratory syndrome (SARS) and a subset of Sarbecovirus.[1] During this time, the novel virus was named SARS-CoV-2, before the World Health Organization named the virus-associated disease COVID-19.[2]

The disease continues to spread throughout the world, affecting to date (May 2, 2020) over 3 million people with 240,490 deaths in 212 different countries.[3] While these numbers are expected to grow, scientists and clinicians continue to find innovative treatments to fight the infection.

The pathophysiology of SARS-CoV-2 is due to the recognition of angiotensin-converting enzyme two receptor (ACE-2) by its spike protein, which facilitates host cell entry and spread.[4-6] Because ACE-2 receptors are widely expressed in the lungs, infection of the respiratory system leads to a hyperproduction of inflammatory cytokines that have been associated with acute respiratory distress syndrome (ARDS) and cardiopulmonary collapse leading to death.[7,8] This hyperinflammatory response is called cytokine storm and proposed treatment with immunomodulation and immunosuppressant therapies is believed to be beneficial.[9]

ARDS AND MESENCHYMAL STEM CELLS (MSC)

ARDS is a clinical complication of acute lung injury (ALI). It is a devastating condition characterized by a dysfunction of the lung epithelial barrier, the capillary endothelium, causing diffuse alveolar damage, interstitial edema, leading to rapidly progressive acute respiratory hypoxia, failure, and death.[10-12] There have been many preclinical and clinical studies looking into MSC as a treatment option for ARDS [Table 1]. One of the hypothesized mechanisms of action of MSC in the treatment for ARDS is believed to be their anti-inflammatory,[13] anti-fibrotic,[14] and immunomodulatory[15] properties. When analyzing the preclinical and early clinical data, and taking into account the overall safety of these cellular therapies MSC seems like a promising option.[16-19]

Table 1: Summary of preclinical and clinical trials.
Indication Study design Dose Application route Cell source Result References
ARDS - preclinical Escherichia coli induced ARDS in rats 1×107human MSCs/kg i.v. Human MSC’s umbilical cord and bone marrow-derived Reduced inflammatory markers, prolonged animal survival [20]
ARDS - preclinical LPS-induced ARDS in mice 2.5×105MSCs i.v. Syngeneic murine MSCs Reduced inflammation, reduced lung permeability [21]
ARDS - preclinical Bleomycin-induced ARDS in mice 5×105MSCs i.v. Syngeneic murine MSCs Prolonged animal survival, reduced [22]
ARDS - preclinical LPS-induced ARDS in mice 5×105MSCs i.t. Human MSCs Prolonged animal survival, reduced pulmonary edema [23]
ARDS - preclinical LPS-induced ARDS in mice 5×106MSC i.v. Rat MSCs bone marrow-derived Reduced inflammation, reduced lung permeability [24]
ARDS - preclinical LPS-induced ARDS in mice 5×106MSC i.v. Rat MSCs bone marrow-derived Reduced inflammation, reduced lung injury [25]
ARDS - preclinical LPS-induced ARDS in mice 1×106cell i.v. Allogeneic human MSCs, menstrual blood-derived Reduced inflammation, reduced lung permeability, Reduced lung injury [26]
ARDS - Clinical Phase 1, multicenter, open-label, dose-escalation study (n=9) 1×106, 5×106or 10×106MSCs/kg i.v. Allogeneic human MSC’s bone marrow-derived MSC treatment was safe, decrease inflammatory markers [18]
ARDS - Clinical Phase 1, randomized, placebo-controlled pilot study (n=12) 1×106MSCs×kg i.v. Allogeneic human MSCs, adipose tissue-derived MSC treatment was safe [17]
ARDS - Clinical Phase 2a, Double: blinded, randomized, Placebo-controlled (n=60) 1×106MSCs×kg i.v. Allogeneic human MSC’s bone marrow-derived MSC treatment was safe [19]

ARDS: Acute respiratory distress syndrome, MSC: Mesenchymal stem cells, LPS: Lipopolysaccharide

In a study published in the journal of critical care medicine,[20] umbilical cord MSC was injected into rats with Escherichia coli-induced ARDS, and this group noticed improved oxygenation, prolonged survival linked to a reduction of inflammatory markers interleukin-6, tumor necrosis factor-α (TNF-α), and alveolar infiltration of neutrophils. Similar findings were reported by Mei et al. in lipopolysaccharide-induced ARDS. They also found that MSC genetically modified the vascular protective protein angiopoietin-1, which lead to a reversal of lung permeability.[21] The anti-inflammatory properties of MSC were also seen in bleomycin-induced murine ARDS animals.[22] An improvement in ALI and ARDS was seen in mouse models treated with MSCs, and a reduction in macrophage inflammatory protein-2 and TNF-α was seen in bronchoalveolar lavage after infusion.[23] The upregulation of vascular endothelial growth factors and hepatocyte growth factors due to MSC was crucial in preventing vascular permeability and downregulating endothelial cell apoptosis in rat models with ARDS.[24,25] Even in xenotransplantation of human-derived menstrual blood, MSCs into murine models showed a reduction of inflammatory markers and repair of damaged lung tissue.[26]

The safety of MSC in human-clinical trials has thus far demonstrated an excellent safety profile.[17-19] Wilson et al. performed a single-dose allogeneic bone marrow-derived MSC escalation clinical study to assess safety parameters in moderate to severe ARDS patients. Nine patients were randomized to the three dose groups of 1, 5, and 10 × 106 MSC/kg. They reported no adverse events and also showed that the mean lung injury scores and the sequential organ failure assessment score improved in a dose-dependent manner but did not reach statistical significance.[18] The same group went onto perform a double-blinded, multicenter randomized clinical trial of a single-dose MSC infusion for moderate to severe ARDS.[19] They concluded that MSC treatment was safe, but mortality did not differ in treatment groups, which is possibly due to the wide MSC viability range of 36–85% that was reported in their study.

A small randomized placebo-controlled pilot study using allogeneic adipose-derived MSC in 12 patients showed short- term safety profile as well.[17] More randomized controlled trials (RCT) are needed to further assess clinical benefit of MSC treatment in ARDS.

COVID-19 AND MSC

While many pharmaceutical and biotechnology companies throughout the world are working on therapeutics or vaccine options to combat SARS-CoV-2 and COVID-19, doctors and scientists are taking a special interest in cellular treatments. Over a dozen cell-based clinical studies have submitted investigational new drug application applications to the FDA and are in the process of gathering data.[27]

A study published in Beijing, looked at the clinical efficacy of MSC for ARDS due to the epidemic influenza A (H7N9) infection.[28] ARDS, pneumonia, and respiratory failure were the major contributors to mortality and morbidity seen in the Influenza A pandemic in 2013.[28] A single-center open- label clinical trial was performed in China, where 17 patients with H7N9 viral-induced ARDS were treated with allogenic menstrual blood-derived MSC. This study also included a 44 patient control group with similar clinical findings. The study showed lower mortality in the treatment group, which had 17.6% mortality when compared to 54.5 % mortality in the control group.[28] Due to the similarities in viral pathogenesis between Influenza A (H7N9) and SARS-CoV-2 with ARDS, it is believed that MSC treatment for COVID-19 can be beneficial.

Some early cases and small RCT have been reported, with many more expected in the near future. Liang et al. reported a case of a 65-year-old patient with severe ARDS due to COVID-19. After the patient continued to deteriorate despite the experimental treatment of antiviral, antibiotics, IFN-α, methylprednisolone, and immunoglobulin, patient needed intubation. Companionate use of MSC with and without α1 thymosin was given; 5 × 107 cells were injected intravenously.[29] Continued observation of this case showed gradual normalization of inflammatory markers with clinical improvement after the second infusion. Progressive improvement was reported, and the patient was extubated and ultimately discharged from the ICU 2 days after the last MSC treatment.[29]

Another self-reported outcome was published in Israel.[30] After the utilization of a placenta based MSC product, there was a reported 100% survival rate in seven patients with a 66% improvement in respiratory parameters. This company plans on starting a multinational clinical study.

A small randomized control trial published in again and disease treated seven patients with COVID-19 pneumonia with MSC. The study reported improvement in mortality as well as normalization of inflammatory parameters when compared to the three patient control group.[4]

In April, a hospital in New York reported data while using Remestemcel-L in COVID-19 induced ARDS. The allogeneic MSC product had already been accepted for priority review by the FDA for acute graph versus host disease. The results in the companionate use showed an 83% survival rate of the 12 patients treated. Although this was not an RCT, the authors compared their treatment group’s survival rate with the 12% survival rate reported by other New York hospitals in the same time frame.[31] This company has since partnered with Cardiothoracic Surgical Trials Network to participate in a 240 patient RCT .[31]

CONCLUSION

Death and financial struggle due to coronavirus led to a worldwide crisis, as no vaccine or therapeutic options were available. Innovative treatments are being looked into as potential answers for the pandemic. As physicians and researchers battle the novel virus, it is important to understand the treatment options that are being utilized while vaccines are being developed. The disease continues to attack the lungs, and mortality is high for those patients that develop COVID-19 induced ARDS. Cell-based treatment has surfaced as a potential option due to its safety profile and early preclinical evidence. Multiple MSC trials have been registered on clinicaltrials.gov to evaluate clinical safety and efficacy for COVID-19 induced ARDS treatment. It is important to mention that although cellular- based treatment is a promising option to treat COVID-19 infections; these are non-approved treatments in the USA at the moment. Clinic-based treatment and marketing without an investigational new drug application and FDA clinical approval are considered unlawful by the FTC/FDA.[32,33]

The scientific community must continue to work together in an evidence-based approach to find safe and efficacious treatment options for SARS-CoV-2.

Declaration of patient consent

Patient’s consent not required as there are no patients in this study.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

References

  1. , , , , , , et al. A new coronavirus associated with human respiratory disease in China. Nature. 2020;579:265-9.
    [CrossRef] [PubMed] [Google Scholar]
  2. , , . Mesenchymal stem cell therapy for COVID-19: Present or future. Stem Cell Rev Rep. 2020;16:427-33.
    [CrossRef] [PubMed] [Google Scholar]
  3. . Covid-19 Coronavirus Pandemic. Available from: https://www.worldometers.info/coronavirus [Last accessed on 2020 Apr 30]
    [Google Scholar]
  4. , , , , , , et al. Transplantation of ACE2(-) mesenchymal stem cells improves the outcome of patients with COVID-19 pneumonia. Aging Dis. 2020;11:216-28.
    [CrossRef] [PubMed] [Google Scholar]
  5. , , , , , , et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: Implications for virus origins and receptor binding. Lancet. 2020;395:565-74.
    [CrossRef] [Google Scholar]
  6. , , , , , , et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature. 2020;579:270-3.
    [CrossRef] [PubMed] [Google Scholar]
  7. . Mesenchymal stem cells and management of COVID-19 pneumonia. Med Drug Discov. 2020;5:100019.
    [CrossRef] [PubMed] [Google Scholar]
  8. , , , , . Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020;46:846-8.
    [CrossRef] [Google Scholar]
  9. , , , , , , et al. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet. 2020;395:1033-4.
    [CrossRef] [Google Scholar]
  10. , , , , , , et al. Acute respiratory distress syndrome: The Berlin definition. JAMA. 2012;307:2526-33.
    [CrossRef] [Google Scholar]
  11. , , . The acute respiratory distress syndrome. J Clin Invest. 2012;122:2731-40.
    [CrossRef] [PubMed] [Google Scholar]
  12. , , . Acute lung injury: A clinical and molecular review. Arch Pathol Lab Med. 2016;140:345-50.
    [CrossRef] [PubMed] [Google Scholar]
  13. , . Mesenchymal stem/stromal cells (MSCs): Role as guardians of inflammation. Mol Ther. 2012;20:14-20.
    [CrossRef] [PubMed] [Google Scholar]
  14. , . Mesenchymal stromal cells: Sensors and switchers of inflammation. Cell Stem Cell. 2013;13:392-402.
    [CrossRef] [PubMed] [Google Scholar]
  15. , , . Mesenchymal stem cell-based immunomodulation: Properties and clinical application. Stem Cells Int. 2018;2018:3057624.
    [CrossRef] [PubMed] [Google Scholar]
  16. , , , , , , et al. Safety of cell therapy with mesenchymal stromal cells (safecell): A systematic review and meta-analysis of clinical trials. PLoS One. 2012;7:e47559.
    [CrossRef] [PubMed] [Google Scholar]
  17. , , , , , , 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.
    [CrossRef] [PubMed] [Google Scholar]
  18. , , , , , , et al. Mesenchymal stem (stromal) cells for treatment of ARDS: A phase 1 clinical trial. Lancet Respir Med. 2015;3:24-32.
    [CrossRef] [Google Scholar]
  19. , , , , , , 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;7:154-62.
    [CrossRef] [Google Scholar]
  20. , , , , , , et al. Cryopreserved, xeno-free human umbilical cord mesenchymal stromal cells reduce lung injury severity and bacterial burden in rodent Escherichia coli-induced acute respiratory distress syndrome. Crit Care Med. 2017;45:e202-12.
    [CrossRef] [PubMed] [Google Scholar]
  21. , , , , , . Prevention of LPS-induced acute lung injury in mice by mesenchymal stem cells overexpressing angiopoietin 1. PLoS Med. 2007;4:e269.
    [CrossRef] [PubMed] [Google Scholar]
  22. , , , , , , et al. Mesenchymal stem cells stably transduced with a dominant-negative inhibitor of CCL2 greatly attenuate bleomycin-induced lung damage. Am J Pathol. 2011;179:1088-94.
    [CrossRef] [PubMed] [Google Scholar]
  23. , , , , , , et al. Human mesenchymal stem (stromal) cells promote the resolution of acute lung injury in part through lipoxin A4. J Immunol. 2015;195:875-81.
    [CrossRef] [PubMed] [Google Scholar]
  24. , , , , , , et al. The vascular endothelial growth factors-expressing character of mesenchymal stem cells plays a positive role in treatment of acute lung injury in vivo. Mediators Inflamm. 2016;2016:2347938.
    [CrossRef] [PubMed] [Google Scholar]
  25. , , , , , , et al. The hepatocyte growth factor-expressing character is required for mesenchymal stem cells to protect the lung injured by lipopolysaccharide in vivo. Stem Cell Res Ther. 2016;7:66.
    [CrossRef] [PubMed] [Google Scholar]
  26. , , , , , . Transplantation of menstrual blood-derived mesenchymal stem cells promotes the repair of LPS-induced acute lung injury. Int J Mol Sci. 2017;18:E689.
    [CrossRef] [PubMed] [Google Scholar]
  27. . COVID-19, SARS-CoV-2 Clinical Trials. Available from: https://www.clinicaltrials.gov/ct2/results?cond=COVID-19 [Last accessed on 2020 May 02]
    [Google Scholar]
  28. , , , , , , et al. Clinical study of mesenchymal stem cell treating acute respiratory distress syndrome induced by epidemic influenza A (H7N9) infection, a hint for COVID-19 treatment. Engineering (Beijing) 2020
    [CrossRef] [Google Scholar]
  29. , , , . Clinical remission of a critically ill COVID-19 patient treated by human umbilical cord mesenchymal stem cells. ChinaXiv 2020 202002.00084v1
    [Google Scholar]
  30. . Pluristem Reports Preliminary Data from its Covid-19 Compassionate Use Program, Treating Seven Patients with Acute Respiratory Failure. Available from: https://www.pluristem.com/wp-content/uploads/2020/04/PSTI-PR-Follow-up-on-Covid-19-treatments-FINAL-FOR-RELEASE.pdf [Last accessed on 2020 May 02]
    [Google Scholar]
  31. . Mesoblast Partners with the Cardiothoracic Surgical Trials Network Established by the U.S. National Institutes of Health's National Heart, Lung and Blood Institute to Conduct Randomized Controlled Trial of Remestemcel-l for Patients with Acute Respiratory Distress Syndrome Due to Covid-19. Available from: http://www.investorsmedia.mesoblast.com/static-files/e63bf0d5-7dd5-46c0-8381-c2e24bacb130 [Last accessed on 2020 Apr 08]
    [Google Scholar]
  32. . The Office of Compliance and Biologics Quality in the Center for Biologics Evaluation and Research (cber) of the United States Food and Drug Administration (fda), Duncan Ross. Available from: https://www.fda.gov/media/137129/download?utm_campaign=What%27sNew2020-04-20&utm_medium=email&utm_source=Eloqua [Last accessed on 2020 Apr 10]
    [Google Scholar]
  33. . The Office of Compliance and Biologics Quality in the Center for Biologics Evaluation and Research (cber) of the United States Food and Drug Administration (fda), Douglas Spiel. Available from: https://www.fda.gov/media/137396/download?utm_campaign=What%27sNew2020-04-27&utm_medium=email&utm_source=Eloqua [Last accessed 2020 Apr 10]
    [Google Scholar]
Show Sections