Minireviews Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Sep 26, 2021; 13(9): 1318-1337
Published online Sep 26, 2021. doi: 10.4252/wjsc.v13.i9.1318
Mesenchymal stem cells and COVID-19: What they do and what they can do
Ejlal Abu-El-Rub, Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg R2H2A6, Canada
Ejlal Abu-El-Rub, Department of Physiology and Pathophysiology, Basic Medical Sciences, Yarmouk University, IRBID 21163, Jordan
Ramada R Khasawneh, Department of Anatomy and Histology, Basic Medical Sciences, Yarmouk University, IRBID 21163, Jordan
Fatimah Almahasneh, Department of Physiology and Pharmacology, Basic Medical Sciences, Yarmouk University, IRBID 21163, Jordan
Zaid Altaany, Department of Biochemistry and Genetics, Basic Medical Sciences, Yarmouk University, IRBID 21163, Jordan
Nesreen Bataineh, Department of Pathology, Basic Medical Sciences, Yarmouk University, IRBID 21163, Jordan
Hana Zegallai, Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg R2H2A6, Canada
Saravanan Sekaran, Department of Pharmacology, Saveetha Dental College and Hospitals to be University, Chennai 600077, India
ORCID number: Ejlal Abu-El-Rub (0000-0001-9217-2560); Ramada R Khasawneh (0000-0003-3873-925X); Fatimah Almahasneh (0000-0002-8799-2721); Zaid Altaany (0000-0002-3260-9078); Nesreen Bataineh (0000-0003-1666-6854); Hana Zegallai (0000-0001-9959-7318); Saravanan Sekaran (0000-0002-2212-0180).
Author contributions: Abu-El-Rub E and Altaany Z conceptualized the review subtopics; Abu-El-Rub E, Khasawneh RR, Almahasneh F, Altaany Z, Bataineh N, Zegallai H and Sekaran S collected the literature used to write the review and drafted the manuscript; Abu-El-Rub E, Almahasneh F and Sekaran S revised and formatted the content of the manuscript and verified spelling, punctuation and grammatical errors; all authors have read and approved the final manuscript.
Conflict-of-interest statement: Authors of this manuscript have no conflicts of interest to disclose.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ejlal Abu-El-Rub, PharmD, PhD, Assistant Professor, Department of Physiology and Pathophysiology, Faculty of Medicine, Yarmouk University, Shafiq Irshidat st, IRBID 21163, Jordan. ejlal.abuelrub@yu.edu.jo
Received: March 14, 2021
Peer-review started: March 15, 2021
First decision: June 5, 2021
Revised: June 15, 2021
Accepted: August 27, 2021
Article in press: August 27, 2021
Published online: September 26, 2021

Abstract

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or coronavirus disease 2019 (COVID-19) pandemic has exhausted the health systems in many countries with thousands cases diagnosed daily. The currently used treatment guideline is to manage the common symptoms like fever and cough, but doesn’t target the virus itself or halts serious complications arising from this viral infection. Currently, SARS-CoV-2 exhibits many genetic modulations which have been associated with the appearance of highly contagious strains. The number of critical cases of COVID-19 increases markedly, and many of the infected people die as a result of respiratory failure and multiple organ dysfunction. The regenerative potential of mesenchymal stem cells (MSCs) has been extensively studied and confirmed. The impressive immunomodulation and anti-inflammatory activity of MSCs have been recognized as a golden opportunity for the treatment of COVID-19 and its associated complications. Moreover, MSCs regenerative and repairing abilities have been corroborated by many studies with positive outcomes and high recovery rates. Based on that, MSCs infusion could be an effective mechanism in managing and stemming the serious complications and multiple organ failure associated with COVID-19. In the present review, we discuss the commonly reported complications of COVID-19 viral infection and the established and anticipated role of MSCs in managing these complications.

Key Words: SARS-CoV-2, COVID-19, Mesenchymal stem cells, Inflammation, Acute respiratory distress syndrome, Pulmonary fibrosis, Pneumonia, Renal injury, Cardiovascular diseases, Regeneration, Anti-inflammatory

Core Tip: Coronavirus disease 2019 (COVID-19) pandemic continues its rampant spread as more vengeant strains emerged in many countries. Severe cases of COVID-19 have been complicated by respiratory failure and multiple organ dysfunction with high mortality rate. Mesenchymal stem cells regenerative and anti-inflammatory abilities can be an innovative approach in repairing the damaged organs and improve the survival rate for critically ill COVID-19 patients.



INTRODUCTION

The 2020 severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), coronavirus disease 2019 (COVID-19) pandemic catastrophe continues to strike many countries severely with million cases and thousands of death reported daily by the World Health Organization. This disease urged many clinicians and researchers to sprint to find effective treatments so as to control and manage the rampant spread of COVID-19 virus worldwide. Despite the fact that COVID-19 is primarily a pulmonary disease, it can hit other organs leading to hematological, hepatic, neurological, cardiac and renal complications[1]. There are no approved remedy to treat COVID-19 and its associated complications. There are many treatment recommendations that are considered as a “leap-of-faith” approach to save the life of many COVID-19 patients[2]. The most commonly used medications to treat symptomatic COVID-19 patients are oxygen, corticosteroids, remdesivir, chloroquine, hydroxychloroquine, lopinavir/ritonavir, nitazoxanide, vitamin super B-complex, zinc and vitamin D[2]. All these suggested medications have many limitations, including: (1) The lack of information regarding the initiation and duration of treatment regimen; (2) Many studies that have been conducted to evaluate the efficacy of these treatments recruited young patients who had relatively asymptomatic disease; (3) The long-run side effects have not been addressed yet; and (4) There is a dearth of data regarding the effect of these treatments in preventing post-infection complications which can be serious and life-threatening[2,3]. Recently, mesenchymal stem cells (MSCs) have been used as one of the promising therapeutic strategies for COVID-19 symptoms and complications[4]. MSCs are one of the most trustworthy stem cells in regenerating and repairing the damages of many organs. MSCs can be isolated and transplanted even in unrelated donor-recipient due to their hypoimmunogenic characteristics[5]. MSCs can repair and restore the function of many injured tissues and improve the quality of life for patients with chronic disorders[5]. MSCs exert their therapeutic effects via different mechanisms[5]. MSCs have the ability to subdue severe inflammation by releasing anti-inflammatory factors such as interleukin-10 (IL-10), IL-4, Indoleamine 2, 3-dioxygenase (IDO), prostaglandin E2 (PGE2), and Heme oxygenase-1 (HO-1)[6]. Furthermore, MSCs release many anti-fibrotic factors, including Transforming growth factor beta-1 (TGF-β1), matrix metalloprotein (MMP) 1, 2 and 9, tissue inhibitor of metalloproteinases-1 (TIMP1) and Collagen Type III Alpha 1 Chain (COL3A1)[7]. MSCs can also promote the angiogenesis and proliferation of cells mediated by releasing many growth factors such as vascular endothelial growth factor (VEGF), Insulin-like growth factor 1 (IGF-1), hepatocyte growth factor (HGF), and platelet-derived growth factor (PDGF)[8]. As MSCs are multipotent stem cells, they can differentiate into many cell types, including cardiomyocytes-like cells, adipocytes, chondrocytes, Clara cells or alveolar cells, endothelial and epithelial cells, which can restore the normal function of impaired organs[9]. The encouraging therapeutic outcomes of MSCs transplantation are expected to be effective in treating COVID-19 complications and decreasing the death rate even in seriously ill patients. Numerous preclinical and clinical trials have been carried out to study the role of MSCs in alleviating COVID-19 symptoms and complications, including acute respiratory distress syndrome (ARDS), pneumonia, pulmonary fibrosis, systemic inflammation, hypercoagulability, and cardiovascular complications. The search for effective COVID-19 management strategies continues to evolve, therefore, in the current review article, we discussed the research outcomes and therapeutic potential of MSCs for COVID-19 patients to provide an updated reference for many clinicians and scientific researchers.

COVID-19 INFLAMMATORY CASCADE AND THE ANTI-INFLAMMATORY ROLE OF MSCS

Current understanding of SARS-CoV-2 mechanisms suggests a central role for exaggerated activation of the innate immune system as an important contributor to COVID-19 adverse outcomes. COVID-19 progression can be divided into three distinct phases, including: (1) Early infection phase, wherein the virus infiltrates host cells in the lung parenchyma; (2) Pulmonary phase, in which viral propagation causes lung tissue injury as the host immune response is activated; and (3) The inflammatory cascade which is triggered by pathogen-associated molecular patterns (i.e., viral RNA) and damage-associated molecular patterns (DAMPs, i.e., cellular debris is released during pyroptosis) are exposed during active viral replication and release[10]. The inflammatory response plays a crucial role in the clinical manifestations and subsequent complications of COVID-19. Post SARS-CoV-2 entry, host factors trigger an immune response against the virus, which, if it is left uncontrolled, may result in pulmonary tissue damage, pulmonary functional impairment, and reduced lung capacity[11]. In addition to respiratory failure, other complicated feature among patients with severe COVID-19 infection is a sudden decline of the patient’s health status approximately two weeks after onset. Continuous infiltration of monocytes and macrophages accompanied by extremely high levels of inflammatory response leads to atrophy of the spleen and lymph nodes, along with reduced lymphocytes in lymphoid organs, hypercoagulability, thrombosis, and multiple organ damage[12]. Cytokine storm syndrome (CSS) has been proposed as underlying the etiology of respiratory failure and multiple organ impairment in patients with COVID-19[13]. CSS is delineated as a significant pro-inflammatory cytokines gush that leads to inflammatory cells recruitment and widespread tissue damage. High levels of the cytokines have been reported, IL-2, IL-7, IL-10, granulocyte colony-stimulating factor, interferon γ-induced protein 10, monocyte chemoattractant protein 1 (MCP1)[13]. The disproportionately high levels of these cytokines were found to be associated with more severe forms of COVID-19 and increased need for intensive care units (ICU) admission.

MSCs remarkable immunomodulatory capacity is one of the most important therapeutic mechanism elicited by MSCs[14]. MSCs immunomodulation abilities can be effective through controlling both innate and adaptive immune systems[14]. Therefore, MSCs have the aptitude to “turn on and turn off” the immune system based on the body needs. Numerous studies on the immune regulation potential of MSCs reported a potent ability of MSCs to control and regulate the functions of different types of immune cells, including B lymphocytes, natural killer (NK) cells, dendritic cells, macrophages, and T lymphocytes by secreting many immunomodulatory factors, primarily TGF-β, HGF, IL-10, IDO, and PGE2[15-17]. Many studies verified the usefulness of MSCs to treat autoimmune diseases and inflammation. There is a lot of preclinical and clinical evidence for the anti-inflammatory effect of MSCs in mitigating virally engendered lung injury and mortality in mice. Several studies have illustrated that MSCs are capable of significantly reducing acute lung injury (ALI) supervening the infection of H9N2 and H5N1 viruses by decreasing the levels of pro-inflammatory cytokines and chemokines, as well as lessening the recruitment of inflammatory cells into the lungs[18]. Applying MSCs to interfere in endotoxin liposaccharide (LPS)-induced ALI mouse model proved that MSCs can remarkably lead to reduction of inflammatory cell infiltration in lung tissue, alleviate inflammation, and regenerate the damaged lung tissues[18]. To date, there is limited published literature regarding the potential efficacy of MSCs in COVID-19 disease. In February 2020, a clinical study from China has been published in which they evaluated a single dose infusion of bone marrow (BM) MSCs (obtained commercially) in seven COVID-19 patients aged 45 to 65[17]. The recruited patients, who were either in critical condition or had mild disease, did not improve with the treatment protocol. At 14 d post-infusion, all critically ill patients were weaned off mechanical ventilation. These positive outcomes regarding the clinical efficacy of infused MSCs were also accompanied by marked decrease in the levels of C-reactive protein, NK cells, T-cells, and tumor necrosis factor-α (TNF-α). Over-activated cytokine-secreting immune cells, CXCR3+CD4+ T cells, CXCR3+CD8+ T cells, and CXCR3+ NK cells were not existent after 3-6 Dy post-infusion[17]. In May 2020, the results of a clinical trial conducted in Liaocheng People’s Hospital in China, using human umbilical cord (UC) Wharton’s jelly-derived MSCs was published[19]. In the trial, a single female patient was involved who tested positive for COVID-19. With standard treatment, the patient vital physical signs improved substantially, but after that the patient took a turn for the worse. Considering the severe organ injury caused by an inflammatory response, human UC Wharton’s jelly-derived MSCs from a healthy donor was intravenously transfused to the patient. Two days after the infusion, the pulmonary function and symptoms of the patient with COVID-19 pneumonia significantly improved, and chest computerized tomography (CT) imaging showed a great amelioration. The patient recovered rapidly and was discharged 7 days after treatment[19]. The results showed that the frequency of CD3+ T, CD4+ T and CD8+ T cells increased, and serum C-reactive protein, IL-6 and TNF-α levels were reduced. Six days after MSC treatment, the patient became negative for SARS-CoV-2. Therefore, therapy based on the use of Wharton’s jelly-derived MSCs may be effective in the treatment of patients with COVID-19[19].

Liang et al[20] and colleagues reported that transplantation of human UC-derived MSCs could modulate the immune response and promote the functional recovery in a 65-year-old female patient with critically ill COVID-19 and severe complications such as respiratory failure and multiple organ failure. The patient received three doses of allogeneic UC-MSCs intravenously at day 9 of infection which were followed by two infusions with an interval of three days. Following the second dose, the vital signs were improved, and she did not require the ventilator. Two days after the third dose, she was transferred out of the ICU[20]. After the administration of UC-MSCs, many clinical indexes and symptoms of the patient were improved. The counts of CD3+ T cell, CD4+ T cell, and CD8+ T cell remarkably increased to the normal level indicating the reversal of lymphopenia, which is a common feature of the COVID-19 patients and is associated with disease severity and mortality. The patient showed a significant decreased level of C-reaction protein, white blood, neutrophil, and alanine aminotransferase/aspartate aminotransferase along with increased level of total lymphocyte count. The study also suggested that the infusion of thymosin α1 will greatly enhance the immunomodulation potential of MSCs.

The safety of human UC-derived MSCs infusions for the treatment of patients with moderate and severe COVID-19 pulmonary disease was evaluated. A total of 18 patients were enrolled in clinical trial, nine of whom (five with moderate disease and four with severe disease) received three cycles of UC-derived MSCs treatment. The patients in the MSCs group displayed a reduction of serum IL-6. Moreover, this trial demonstrated that transplantation of MSCs in patients with COVID-19 was safe, and no serious adverse events were reported[21]. The data show that intravenous UC-derived MSCs infusion in patients with moderate and severe COVID-19 is safe and well tolerated.

The published clinical study and case report have demonstrated that intravenous transplantation of MSCs was safe and effective in treating COVID-19, and remarkably, MSCs may be resistant to virus infection, whereas more clinical trials with larger samples are warranted for more convincing evidence.

COVID-19 RESPIRATORY COMPLICATIONS AND MSCS
COVID-19 pneumonia and MSCs

The most common manifestations of COVID-19 primarily target the respiratory system. According to a large cohort study from China, 81% of COVID-19 patients showed mild to moderate symptoms, including dry cough and moderate pneumonia, 14% had severe presentation, such as dyspnea, hypoxia or more than 50% lung involvement on imaging, and 5% were in critical condition manifested by having respiratory failure, shock, or multi-organ system dysfunction[22]. Besides being the first target for SARS-CoV-2 virus, the respiratory system is also involved in the transmission of SARS-CoV-2 virus, which occurs through respiratory droplets emitted via sneezing and coughing. SARS-CoV-2 virus was found to be stable in aerosols and different surface materials, and was detected in stool, tears and conjunctival secretions[23]. These can add to certain extent to its transmission. To enter the host organism, the SARS-CoV-2 virus passes through the mucous membranes of the nose and larynx, gaining access to the respiratory tract and eventually resides in the lungs. The lung alveolar epithelial cells are considered the site where the spike proteins of the virus bind to the cell surface angiotensin converting enzyme 2 (ACE2) receptors[23]. This leads to the activation of various proteases, which cleave the spike proteins at a site close to the S1/S2 subunit boundary, inducing viral fusion and subsequent internalization by endocytosis[24]. Some patients with COVID-19 pneumonia rapidly progress to critical illness and ARDS[25]. ARDS is the most severe form of ALI which is a form of diffuse alveolar injury. ARDS is defined as “an acute condition characterized by bilateral pulmonary infiltrates and severe hypoxemia (PaO2/FiO2 ratio < 200) in the absence of evidence for cardiogenic pulmonary edema”[26].

COVID-19 ARDS shares mechanistic and pathophysiologic properties with influenza A virus-induced pneumonia. The presence of viral RNA in the cytoplasm of host epithelial cells stimulates antiviral pathways which then activate a number of molecular and cellular mechanisms of immunity[27]. This includes elevated serum cytokines (especially IL-1β, IL-6 and TNF-α), impaired interferon I and III responses, cytokines storm and peripheral lymphopenia[28,29]. Although these immune processes eliminate the virus, they also cause prolonged inflammation and destruction of the host alveolar epithelial cells, which result in severe lung injury and ARDS.

Compared to ARDS caused by other pathologies, the ARDS induced by SARS-CoV-2 seems to be caused by an injury to the alveolar epithelial cells with almost no effect is exerted on endothelial cells. This produces less exudation and, hence, dry cough. Clinical symptoms are often unpredictable and inconsistent with the severity of laboratory and imaging findings. The time of the onset of ARDS in COVID-19 cases is 8-12 d, which is longer compared to other respiratory conditions (up to 7 d)[25]. Mortality associated with COVID-19 is attributable to the development of pneumonia-induced ARDS, consequently, it would be of paramount importance to find effective prevention and treatment strategies for pneumonia-induced ARDS complication in COVID-19 patients[27]. MSCs are known to be an excellent candidate for cell therapy that targets lung diseases. MSCs had been successfully tested in the setting of different lung disease where they regulated the immune response by suppressing infiltrated cells and decreasing edema[30]. Plenty of studies have investigated the effects of MSCs in animal models of infection-induced ALI/ARDS. A recently published meta-analysis found that MSCs can reduce lung injury score of ALI, improve animal’s survival, decrease lung edema and improve lung compliance. In addition, MSCs reduce levels of pro-inflammatory factors such as IL-1β, IL-6 and TNF-α, while increasing the level of anti-inflammatory IL-10[31]. Comparable results were observed in human clinical trials. In an open-label study in 2013, administration of allogeneic menstrual-blood-derived MSCs lowered the death rate in patients with influenza H7N9-induced ARDS, with no significant undesirable effects were reported in the treated patients over a five-year follow-up period[32]. Other clinical trials[33-35] failed to prove the therapeutic effectiveness of MSCs in ARDS due to the limited number of recruited patients or inappropriate dose of MSCs. However, they confirmed the safety of such therapy; as no MSC-related hemodynamic, respiratory adverse events, or infusion toxicities were reported. Also, there were no significant differences in the overall number of adverse events between the treatment and control groups.

Due to the lack of effective therapies against COVID-19 virus complications, and based on the promising results of many preclinical and clinical studies of MSCs in the setting of ARDS, a number of patients with severe COVID-19 were empirically treated with MSCs and their cases were reported and published[19,20,36-41]. The majority of these cases involved male patients older than 50 and were reported in China. The source of MSCs injected included Wharton’s jelly, UC blood, and BM. The route of MSCs administration was intravenous infusions, and in one of the cases having brain manifestations, MSCs were delivered intrathecally[36]. Indeed, IV infusion is the preferred route of administration of UC-MSCs since most of the infused cells will be trapped in the lungs, the organ most affected by COVID-19[4]. All patients received other recommended lines of therapy, such as antivirals, antibiotics and/or corticosteroids. There was one patient who was treated with both convalescent plasma and MSCs[39]. In all reported cases, MSCs therapy caused no adverse effects, and patients showed complete resolving of COVID-19 symptoms.

The database clinicaltrials.gov lists 71 clinical trials of MSCs in COVID-19, which are summarized in Table 1. Eight of these trials were completed and their results disclosed. The outcome of one clinical trial has been reported on the same website[42], while the results of a few others have been published. Meng and colleagues, for example, reported the findings of a non-randomized phase 1 clinical trial which enrolled 18 patients with moderate to severe COVID-19. The patients were divided into a control and an UC-MSCs treatment group. No serious adverse events were reported, although two patients receiving UC-MSCs developed transient facial flushing and fever, and one developed transient hypoxia. Treatment with UC-MSCs decreased the need for mechanical ventilation, repaired lung pathological changes and improved pulmonary functions[21]. Lanzoni et al[43] and colleagues conducted a double-blind, phase 1/2a, controlled trial that recruited 24 patients with COVID-19-induced ARDS. The study was randomized to either the UC-MSCs treatment or the control group. UC-MSCs infusions were found to be safe and remarkably improved the patients’ survival and shortened the recovery time[43]. An open-label, individually randomized, controlled trial was carried out by Shu et al[44] which included 12 patients in the treatment group (UC-MSCs) and 29 in the control group. The treatment group had significantly shorter recovery time, rapid symptomatic improvement, faster alleviation of lung inflammation, and without any safety issues. In Spain, 13 patients with severe COVID-19 on mechanical ventilation were treated with adipose tissue-derived MSCs (AD-MSCs). The therapy was associated with clinical, radiological and ventilation improvements[45]. Leng et al[17] studied the therapeutic effects of injecting ACE2- MSCs in 7 patients with severe COVID-19 pneumonia. The patients exhibited significant clinical improvements without observed adverse effects.

Table 1 Summary of clinical trials listed in clinicaltrials.org involving mesenchymal stem cells in the treatment of coronavirus disease 2019.
NCT number
Title
Phases
Study designs
Cells/products
No. of cells
No. of infusions
Delivery route
No. of patients
Status
Location
1NCT04366271Clinical trial of allogeneic mesenchymal cells from umbilical cord tissue (UC) in patients with COVID-19Phase 2Randomized/parallel assignment/open labelUC-MSCsNot reported1Not reported106RecruitingSpain
2NCT04444271Mesenchymal stem cell infusion for COVID-19 infectionPhase 2Randomized/parallel assignment/open labelBM-MSCs2 × 106/kg1 or 2IV20RecruitingPakistan
3NCT04416139Mesenchymal stem cell for acute respiratory distress syndrome (ARDS) due for COVID-19Phase 2Non-randomized/parallel assignment/open labelUC-MSCs1 × 106/kg1IV10RecruitingMexico
4NCT04713878Mesenchymal stem cells therapy in patients with COVID-19 pneumoniaNot applicableRandomized/parallel assignment/open labelMSCs1 × 106/kg3IV21CompletedTurkey
5NCT04352803Adipose mesenchymal cells for abatement of SARS-COV-2 respiratory compromise in COVID-19 diseasePhase 1Non-randomized/sequential assignment/open labelAutologous AD-MSCs5 × 105/kg1IV20Not yet recruitingUnited States
6NCT04565665Cord blood-derived mesenchymal stem cells (MSCs) for the treatment of COVID-19 related ARDSPhase 1Randomized/parallel assignment/open labelUC-MSCsNot reported1 or 2IV70RecruitingUnited States
7NCT04429763Safety and efficacy of MSCs in the management of severe COVID-19 pneumoniaPhase 2Randomized/parallel assignment/masking: TripleUC-MSCs1 × 106/kg1IV30Not yet recruitingColombia
8NCT04456361Use of MSCs in ARDS caused by COVID-19Early Phase 1Single group assignment/open labelWJ-MSCs1 × 1081IV9Active, not recruitingMexico
9NCT04315987NestaCell® MSC to treat patients with severe COVID-19 pneumoniaPhase 2Randomized/parallel assignment/quadruple maskingNestaCell®2 × 107/kg4IV90Not yet recruitingBrazil
10NCT04366323Clinical trial to assess the safety and efficacy of intravenous administration of allogeneic adult MSCs of expanded adipose tissue in patients with severe pneumonia due to COVID-19Phase 1/phase 2Randomized/parallel assignment/open labelAD-MSCs8 × 1072IV26Active, not recruitingSpain
11NCT04611256MSCs in patients diagnosed with COVID-19Phase 1Randomized/parallel assignment/open labelAD-MSCs1 × 106/kg2IV20RecruitingMexico
12NCT04625738Efficacy of infusions of MSC from Wharton jelly in the SARS-Cov-2 (COVID-19) related ARDSPhase 2Randomized/parallel assignment/masking: QuadrupleWJ-MSCs2 × 10/kg3IV30Not yet recruitingFrance
13NCT04252118MSC treatment for pneumonia patients infected with COVID-19Phase 1Non-randomized/parallel assignment/open labelMSCs3 × 107/kg3IV20RecruitingChina
14NCT04346368Bone marrow-derived MSC treatment for severe patients with COVID-19Phase 1/phase 2Randomized/parallel assignment/masking: single (participant)BM-MSCs1 × 106/kg1IV20Not yet recruitingChina
15NCT04313322Treatment of COVID-19 patients using Wharton's jelly-MSCsPhase 1Single group assignment/open labelWJ-MSCs1 × 106/kg3IV5RecruitingJordan
16NCT04288102Treatment with human UC-derived MSCs for severe COVID-19Phase 2Randomized/parallel assignment/double-blind/placebo controlledUC-MSCs4 × 107/kg3IV100CompletedChina
17NCT04629105Regenerative medicine for COVID-19 and flu-elicited ARDS using longeveron MSCs (LMSCs) (recover)Phase 1Randomized/parallel assignment/double-blindLMSCs1 × 1083IV70RecruitingUnited States
18NCT04382547Treatment of COVID-19 associated pneumonia with allogenic pooled olfactory mucosa-derived MSCsPhase 1/phase 2Non-randomized/parallel assignment/open labelAllogenic pooled olfactory mucosa-derived MSCsNot reportedNot reportedIV40Enrolling by invitationBelarus
19NCT04336254Safety and efficacy study of allogeneic human dental pulp MSCs to treat severe COVID-19 patientsPhase 1/phase 2Randomized/parallel assignment/masking: TripleAllogeneic human dental pulp stem cells3 × 107/kg3IV20RecruitingChina
20NCT04527224Study to evaluate the efficacy and safety of AstroStem-V in treatment of COVID-19 pneumoniaPhase 1/phase 2Single group assignment/open labelAstroStem-V (AD-MSCs)Not reportedNot reportedNot reported10Not yet recruitingNA
21NCT04273646Study of human UC MSCs in the treatment of severe COVID-19Not applicableRandomized/parallel assignment/open labelUC-MSCs0.5 × 106/kg4IV48Not yet recruitingChina
22NCT04302519Novel coronavirus induced severe pneumonia treated by dental pulp MSCsEarly phase 1Single group assignment/open labelDental pulp MSCs1 × 106/kg3IV24Not yet recruitingChina
23NCT04728698Study of intravenous administration of allogeneic adipose-derived MSCs for COVID-19-induced acute respiratory distressPhase 2Randomized/parallel assignment/masking: DoubleCOVI-MSC (AD-MSCs)1-1.5 × 106/kg1IV100Not yet recruitingUnited States
24NCT04457609Administration of allogenic UC-MSCs as adjuvant therapy for critically-ill COVID-19 PatientsPhase 1Randomized/parallel assignment/masking: TripleUC-MSCs1 × 106/kg1IV40RecruitingIndonesia
25NCT04348435A randomized, double-blind, placebo-controlled clinical trial to determine the safety and efficacy of Hope Biosciences allogeneic adipose-derived mesenchymal stem cell therapy (HB-adMSCs) to provide protection against COVID-19Phase 2Randomized/parallel assignment/masking: QuadrupleAD-MSCsG1: 2 × 108; G2: 1 × 108; G3: 5 × 1075IV100Enrolling by invitationUnited States
26NCT04349631A clinical trial to determine the safety and efficacy of HB-adMSCs to provide protection against COVID-19Phase 2Single group assignment/open label/primary purpose: PreventionAD-MSCsNot reported5IV56Active, not recruitingUnited States
27NCT04366063MSC therapy for SARS-CoV-2-related ARDSPhase 2/phase 3Randomized/parallel assignment/open labelMSCs and EVs from MSCsG1: 1 × 108 MSC; G2: 1 × 108 MSCs + EVs2IV60RecruitingIran
28NCT04339660Clinical research of human MSCs in the treatment of COVID-19 pneumoniaPhase 1/phase 2Randomized/parallel assignment/masking: TripleUC-MSCs1 × 106/kg1IV30RecruitingChina
29NCT04428801Autologous adipose-derived stem cells (AdMSCs) for COVID-19Phase 2Randomized/parallel assignment/masking: DoubleAutologous AD-MSCs2 × 1083IV200Not yet recruitingUnited States
30NCT04573270MSCs for the treatment of COVID-19Phase 1Randomized/single group assignment/masking: triplePrimePro (UC-MSCs)Not reported1IV40CompletedUnited States
31NCT04490486UC derived MSCs versus placebo to treat acute pulmonary inflammation due to COVID-19Phase 1Randomized/parallel assignment/masking: DoubleUC-MSCs1 × 1082IV21Not yet recruitingUnited States
32NCT04355728Use of UC-MSCs for COVID-19 patientsPhase 1/Phase 2Randomized/parallel assignment/masking: tripleUC-MSCs + heparin1 × 1082IV24CompletedUnited States
33NCT04371601Safety and effectiveness of MSCs in the treatment of pneumonia of COVID-19Early Phase 1Randomized/parallel assignment/open labelUC-MSCs1 × 106/kg4IV60Active, not recruitingChina
34NCT04522986An exploratory study of adr-001 in patients with severe pneumonia caused by SARS-CoV-2 InfectionPhase 1Single group assignment/open labelAD-MSCs (ADR-001)1 × 1084IV6Not yet recruitingJapan
35NCT04390152Safety and efficacy of intravenous Wharton's jelly derived MSCs in ARDS due to COVID 19Phase 1/Phase 2Randomized/parallel assignment/masking: QuadrupleWJ-MSCs5 × 1072IV40RecruitingColombia
36NCT04461925Treatment of coronavirus COVID-19 pneumonia (pathogen SARS-CoV-2) with cryopreserved allogeneic P_MMSCs and UC-MMSCsPhase 1/phase 2Non-randomized/parallel assignment/open labelPlacenta-derived MSCs1 × 106/kg3IV30RecruitingUkraine
37NCT04348461BAttLe against COVID-19 using mesenchymal stromal cellsPhase 2Randomized/parallel assignment/masking: QuadrupleAllogeneic and expanded AD-MSCs1.5 × 106/kg2IV100Not yet recruitingSpain
38NCT04535856Therapeutic study to evaluate the safety and efficacy of DW-MSC in COVID-19 patientsPhase 1Randomized/parallel assignment/masking: QuadrupleDW-MSCsG1: 5 × 107; G2: 10 × 1071IV9CompletedIndonesia
39NCT04362189Efficacy and safety study of allogeneic HB-adMSCs for the treatment of COVID-19Phase 2Randomized/parallel assignment/masking: QuadrupleAD-MSCs1 × 1084IV100Active, not recruitingUnited States
40NCT04494386UC lining stem cells (ULSC) in patients with COVID-19 ARDSPhase 1/Phase 2Randomized/parallel assignment/masking: TripleUC-MSCs1 × 1081 or 2IV60RecruitingUnited States
41NCT04397796Study of the safety of therapeutic tx with immunomodulatory MSC in adults with COVID-19 infection requiring mechanical ventilationPhase 1Randomized/parallel assignment/masking: QuadrupleBM-MSCsNot reportedNot reportedNot reported45RecruitingUnited States
42NCT04345601Mesenchymal stromal cells for the treatment of SARS-CoV-2 induced acute respiratory failure (COVID-19 Disease)Phase 1/Phase 2Randomized/parallel assignment/open labelMSCs1 × 108/kgUp to 2IV30RecruitingUnited States
43NCT04452097Use of hUC-MSC product (BX-U001) for the treatment of COVID-19 with ARDSPhase 1/phase 2Non-randomized/sequential assignment/open labelUC-MSCsG1: 0.5 × 106; G2: 1 × 106; G3: 1.5 × 1061IV39Not yet recruitingUnited States
44NCT04492501Investigational treatments for COVID-19 in tertiary care hospital of PakistanNot applicableNon-randomized/factorial assignment/open labelBM-MSCs (+ TPE and convalescent plasma)2 × 106/kg1IV600CompletedPakistan
45NCT04377334MSCs in inflammation-resolution programs of COVID-19 induced ARDSPhase 2Randomized/parallel assignment/open labelBM-MSCsNot reportedNot reportedIV40Not yet recruitingGermany
46NCT04390139Efficacy and safety evaluation of MSCs for the treatment of patients with respiratory distress due to COVID-19Phase 1/phase 2Randomized/parallel assignment/masking: QuadrupleWJ-MSCs1 × 106/kg2IV30RecruitingSpain
47NCT04467047Safety and feasibility of allogenic MSC in the treatment of COVID-19Phase 1Single group assignment/open labelMSCs1 x 106/kg1IV10Not yet recruitingBrazil
48NCT04392778Clinical use of stem cells for the treatment of COVID-19Phase 1/phase 2Randomized/parallel assignment/masking: QuadrupleUC-MSCs3 × 106/kg3IV30RecruitingTurkey
49NCT04537351The MEseNchymal COVID-19 trial: a pilot study to investigate early efficacy of MSCs in adults with COVID-19Phase 1/phase 2Randomized/parallel assignment/open labelCYP-001 (Cymerus® MSC)up to 2 × 106/kg2IV24RecruitingAustralia
50NCT04361942Treatment of severe COVID-19 pneumonia with allogeneic mesenchymal stromal cells (COVID_MSV)Phase 2Randomized/parallel assignment/masking: TripleMSCs1 × 106/kg1IV24RecruitingSpain
51NCT04398303ACT-20 in patients with severe COVID-19 pneumoniaPhase 1/phase 2Randomized/Parallel Assignment/Double-blindUC-MSCs and conditioned UC-MSCsG1: 1 × 106 G2: 100 mL conditioned media (MD)1IV70Not yet recruitingUnited States
52NCT03042143Repair of ARDS by stromal cell administration (realist) (COVID-19)Phase 1/phase 2Randomized/parallel assignment/masking: QuadruplehUC-derived CD362 enriched MSCs400 × 1061IV75RecruitingUnited Kingdom
53NCT04269525UC-derived MSCs treatment for the 2019-novel coronavirus (nCOV) pneumoniaPhase 2Single group assignment/open label/primary purpose: PreventionUC-MSCs9.9 × 1074IV16RecruitingChina
54NCT04602442Safety and efficiency of method of exosome inhalation in COVID-19 associated pneumoniaPhase 2Randomized/parallel assignment/masking: DoubleEXO-1 and EXO-2 (exosomes from MSC)0.5-2 × 1010 nanoparticles20 (2/d for 20 d)Aerosol inhalation90Enrolling by invitationRussia
55NCT04437823Efficacy of intravenous infusions of stem cells in the treatment of COVID-19 patientsPhase 2Randomized/parallel assignment/open labelUC-MSCs5 × 105/kg3IV20RecruitingPakistan
56NCT04371393MSCs in COVID-19 ARDSPhase 3Randomized/parallel assignment/masking: TripleRemestemcel-L2 × 106/kg2IV223Active, not recruitingUnited States
57NCT04333368Cell therapy using UC-derived mesenchymal stromal cells in SARS-CoV-2-related ARDSPhase 1/phase 2Randomized/parallel assignment/masking: TripleWJ-UC-MSCs1 × 106/kg3IV47Active, not recruitingFrance
58NCT04447833Mesenchymal stromal cell therapy for the treatment of ARDSPhase 1Single group assignment/open labelKI-MSC-PL-205 (BM-MSCs)1-2 × 106/kg1IV9RecruitingSweden
59NCT04491240Evaluation of safety and efficiency of method of exosome inhalation in SARS-CoV-2 associated pneumoniaPhase 1/phase 2Randomized/parallel assignment/masking: DoubleDrug: EXO-1 and EXO- (exosomes from MSCs)0.5-2 × 1010 nanoparticles20 (2/d for 20 d)Aerosol inhalation30CompletedRussia
60NCT04299152Stem cell educator therapy treat the viral inflammation in COVID-19Phase 2Randomized/parallel assignment/masking: Single (care provider)Stem Cell Educator (from UC-MSCs)Not reported1IV20Not yet recruitingUnited States
61NCT04466098Multiple dosing of mesenchymal stromal cells in patients with ARDS (COVID-19)Phase 2Randomized/parallel assignment/masking: TripleMSCs3 × 1083IV30RecruitingUnited States
62NCT04524962Study of descartes-30 in ARDSPhase 1/phase 2Single group assignment/open labelDescartes 30 (MSCs RNA-engineered to secrete a combination of DNases)Not reportedNot reportedNot reported30RecruitingUnited States
63NCT04445220A study of cell therapy in COVID-19 subjects with acute kidney injury who are receiving renal replacement therapyPhase 1/phase 2Randomized/parallel assignment/masking: QuadrupleAllogeneic MSCs (SBI-101)G1: 2.5 × 108; G2: 7.5 × 108Not reportedNot reported22RecruitingUnited States
64NCT04400032Cellular immuno-therapy for COVID-19 ARDS vanguardPhase 1Non-randomized/sequential assignment/open labelBM-MSCsG1: 2.5 × 107; G2: 5 × 107; G3: 9 × 1073IV9RecruitingCanada
65NCT04615429Clinical trial to assess the efficacy of MSC in patients with ARDS due to COVID-19Phase 2Randomized/Parallel Assignment/Double-blindMSCs1 × 106/kg1IV20RecruitingSpain
66NCT04525378MSC-based therapy in COVID-19-associated ARDSPhase 1Randomized/parallel assignment/open labelMSCsG1: 2.5 × 107 G2: 5 × 107 G3: 10 × 107G1 and G2: 2 G3: 1IV20RecruitingBrazil
67NCT04399889hCT-MSCs for COVID19 ARDSPhase 1/phase 2Randomized/single group assignment/masking: QuadrupleHuman cord tissue MSCs1 × 106/kg3IV30RecruitingUnited States
68NCT04445454Mesenchymal stromal cell therapy for severe COVID-19 infectionPhase 1/phase 2Single group assignment/open labelBM-MSCs1.5-3.0 × 106/kg3IV20RecruitingBelgium
69NCT04276987A pilot clinical study on inhalation of MSCs exosomes treating severe novel coronavirus pneumoniaPhase 1Single group assignment/open labelAD-MSCs-derived exosomes2 × 108 nanovesicles5Aerosol inhalation24CompletedChina
70NCT04482699RAPA-501-Allo Off-the-shelf therapy of COVID-19Phase 1/phase 2Randomized/sequential assignment/masking: DoubleRAPA-501-ALLOG1: 4 × 107 G2: 1.6 × 108Not reportedNot reported88RecruitingUnited States
71NCT04614025Open-label multicenter study to evaluate the efficacy of PLX-PAD for the treatment of COVID-19Phase 2Randomized/parallel assignment/open labelPLX-PAD (placental MSC-like)2 × 10715IM40RecruitingIsrael

Other studies have addressed the therapeutic use of MSCs-derived components, such as exosomes, in COVID-19 patients. Sengupta and colleagues run a non-randomized open-label cohort study to assess the safety and efficacy of exosomes derived from allogeneic BM-MSCs for the treatment of severe COVID-19 in 24 patients. No adverse effects related to infusion were reported. Treatment was associated with better clinical status and oxygenation, improvement in laboratory absolute neutrophils count, and a decline in acute phase reactants[46]. The above-mentioned results indicate that MSCs may play a pivotal role in the treatment of COVID-19 related respiratory complications due to their excellent safety profile and a wide range of therapeutic outcomes as improvement in the clinical, radiological and immunological status of COVID-19 concomitant pneumonia.

COVID-19-ARDS and MSCs

COVID-19, that is concomitant with ARDS, emerged as a primary concern all over the world. ARDS is a life-threatening severe lung condition that causes damage to the pulmonary capillary endothelium and alveolar epithelium. The ARDS, which is caused by infection and other mechanical stimulation, allows fluid to leak into the lungs. As a result, breathing becomes difficult as the lungs turn non-functional and cannot supply the body's vital organs with enough oxygen. Most people who get ARDS are already at the hospital for trauma or illness[47]. The Spike proteins (S proteins) of SARS-CoV-bind the ACE2 in the alveolar cells which is catalyzed by a transmembrane protease serine 2 (TMPRSS2) protease in type II alveolar epithelial cells[48,49]. The high binding affinity of SARS-CoV-2 Spike proteins for ACE2 is the main determinant of the replication rate of SARS-CoV-2 and the cause of the severity of COVID-19 disease[50].

Various animal studies and early Phase I/II clinical trials have espoused the tremendous potential of MSCs therapy in treating the ARDS. SARS-CoV-2 infects type II alveolar epithelial cells or other target cells that express ACE2. The keratinocyte growth factor (KGF), which is secreted by MSCs, can decrease injury and improve the proliferation and regeneration of alveolar epithelial cells by rising surface-active substances, including MMP-9, IL-1Ra, and granulocyte-macrophage colony-stimulating factor (GM-CSF)[51]. In addition, it has been reported that MSCs can improve gas exchange and reduce the levels of bronchoalveolar lavage fluid’ chemokines and cytokines, including GM-CSF, MIG, IL-1α, IFN-γ, IL-6, and TNF-α[18]. Researching experts elucidated that MSCs treatment expresses vital anti-inflammatory cytokines like IL-1RA, IL-8, and IL-10 and an array of bioactive molecules that stimulate local tissue regeneration by buffering the cytokine and chemokine storm provoked by COVID-19 virus[52]. A published clinical study conducted in Beijing Youan Hospital in China, showed that the transplantation of MSCs improved the clinical outcomes in all recruited COVID-19 elderly patients at day 4 post-MSCs injection without serious adverse effects[17]. Cytokine-secreting immune cells, including CXCR3+CD4+ T cells, CXCR3+CD8+ T cells, and CXCR3+ NK cells, eradicated within 1 wk of MSCs transplantation with a significant reduction in the inflammation biomarkers[17]. MSCs treatment in COVID-19 patients having ARDS were able to subside the over-activated immune system and promote endogenous repair by improving the lung microenvironment[53]. Taking into regard the positive results that have been revealed in some published phase I clinical trials, MSCs will be probably effective in reducing the risk of cytokine storms which cause ARDS and organs failure in patients with severe COVID-19 disease.

COVID-19 pulmonary fibrosis and MSCs

Many studies interestingly revealed that COVID-19 patients who develop ARDS can survive and might eventually be discharged. On the other hand, a large proportion of COVID-19 patients cannot survive once they develop idiopathic pulmonary fibrosis[54]. Idiopathic pulmonary fibrosis is a progressive lung disease manifested by compromised lung functions with extensive damage to the alveolar cells and capillaries. Patients with severe idiopathic pulmonary fibrosis need lung transplantation to improve pulmonary functions and save their lives[55]. Cytokine storm, which causes oxygen stress and the hyperactive inflammation presenting in the severe stages of COVID-19 infection, is probably the main cause of pulmonary fibrosis. Elevated levels of cytokines such as IL1-β, IL-7, IL-8, IL-9, IL-10, GM-CSF, IFN-γ, MCP1, TNF-α and VEGF have been reported in COVID-19 patients[56-58]. The levels of above-mentioned cytokines with high neutrophil counts are typically more pronounced in patients with severe stages of COVID-19 disease. The development of pulmonary fibrosis in severe cases of COVID-19 is due to the induction of oxidative stress, excessive production of reactive oxygen species (ROS), and the presence of high levels of fibrotic factors mainly TGF-b, FGF, and PDGF[59,60].

MSCs have been introduced as one of the therapeutic approaches in the treatment of COVID-19 pulmonary fibrosis[61]. Golchin et al[61] reported that using MSCs immunomodulatory and differentiation abilities can counteract the cytokine storm and prevent lung tissues death and pulmonary fibrosis by regenerating and reconstructing damaged tissues. Similarly, Yang et al[62] showed that MSCs transplantation in ALI mouse model significantly reduced lung permeability, attenuated lung epithelium apoptosis, facilitated vascular endothelium (VE)-cadherin recovery, and reduced lung injury. These beneficial outcomes were mediated by VEGF released from the transplanted MSCs. Moreover, Jung et al[63] reported that injecting MSCs in a LPS-induced ARDS mouse model resulted in the diminution of neutrophil infiltration, collagen deposition and lung fibrosis. MSCs have specific cytokines counteracting potentials to oppose the viral infection and reduce the spread of pulmonary fibrosis. MSCs secrete anti-inflammatory chemokines and cytokines such as IL-10, and PGE2 which help in preventing pulmonary fibrosis[64]. Preclinical studies on animal models with COVID-19 indicated that MSCs considerably improve the disease symptoms, including pulmonary fibrosis. MSCs can secrete a variety of growth factors, including epidermal growth factor (EGF), KGF, PDGF, fibroblast growth factor (FGF), HGF, and VEGF[65]. These factors can recuperate the inflammatory microenvironment of the lung tissues and promote the endogenous lung repair mechanisms and differentiation. Furthermore, MSCs prevent myofibroblast growth by reducing the levels of TGF-β, TNF-α, type I collagen, type III collagen, hydroxyproline, and serum ceruloplasmin in lung tissues, thereby alleviating pulmonary fibrosis[66]. Based on the findings of previous studies, the administration of MSCs in severe cases of COVID-19 appears to be beneficial in resolving and reducing the mortality rate of life-threatening pulmonary fibrosis.

COVID-19 RENAL COMPLICATIONS AND MSCs

Various renal complications have been reported in hospitalized COVID-19 patients, mainly the electrolyte disturbance and acute kidney injury (AKI)[67]. The incidence of renal complications in COVID-19 patients increases if there is an existing kidney diseases, e.g., chronic kidney disease, or a risk factors for kidney complications, including diabetes mellitus[68]. The presence of AKI in COVID-19 patients is associated with worse prognosis and high death rate[69]. In Europe, the death rate in COVID-19 older than 60 years patients was found to be higher when renal complications existed[70]. In severe cases of COVID-19, the rate of continuous renal replacement therapy was significantly higher in North America and Europe compared to other regions[71]. The proposed mechanism of how COVID-19 can induce renal injury is supposed to be through ACE2[72]. It is already known that SARS-CoV-2 (COVID-19) virus attacks the host cells and binds via spike proteins (S-protein) to the extracellular enzymatic domain of ACE2 causing its internalization and down-regulation. Renal tissues contain ample amount of Ang (1-7) peptides that are produced by ACE2 mediated hydrolysis of AngII[73]. A significant decrease in ACE2 and Ang (1-7) has been found in many kidney diseases, including AKI[73]. Additionally, it has been reported that deficiency of ACE2 by either pharmacological inhibition or genetic knocking out of ACE2 is correlated to the onset of diabetic nephropathy which is manifested by albuminuria and glomerulosclerosis. SARS-CoV-2 mediated down-regulation of ACE2 causes the loss of the beneficial and protective Ang (1-7) peptides and accumulates the AngII[73]. This molecular disturbance in the RAS system by SARS-CoV-2 induces many pathological changes in the kidney, including tubular necrosis and multiple inflammatory infiltrates, endothelial injury and dysfunction, and erythrocytes aggregation which obstructs the capillary lumina (microthrombus formation)[73]. In addition to direct SARS-CoV-2 mediated pathological changes in the kidneys, SARS-CoV-2 invades the alveolar cells and enkindles the activation of many immune cells. These recruited immune cells secrete massive amount of cytokines that circulate systematically inducing endothelial dysfunction and promoting the coagulation and micro thrombus formation resulting in multiple organ failure and septic shock[68]. AKI can occur in severe cases of COVID-19 as a result of this cytokines storm. Interestingly, a study examined SARS-CoV-2 viral load in 22 patients who died from COVID-19. It has been reported that 17 out of 22 autopsies showed high load of SARS-CoV-2 in the kidney indicating high mortality rate when COVID-19 was complicated with AKI[74]. Moreover, the co-expression of ACE2 and protease TMPRSS2 in the podocytes and proximal convoluted tubules increases the amplification rate of SARS-CoV-2 in the kidney cells[74]. MSCs have been corroborated by many studies as an effective treatment option for AKI, especially when they are injected as early as possible. Zhou et al[75] found that the transplantation of erythropoietin pretreated BM MSCs in AKI rat model significantly reduced the inflammation and improved the kidney function parameters. Similarly, Zhou et al[75] reported that the infusion of human UC MSC-derived exosomes (HucMSC-Ex) in a mouse model of sepsis-associated AKI was able to restore the renal function parameters and decrease inflammation via the inhibition of NF-κB activity. HucMSC-Ex transplantation was effective in ameliorating the apoptosis in renal tubular cells and improving survival in mice with sepsis. Cao et al[76] reported that the extracellular vesicles derived from MSCs were capable of stimulating mitochondrial antioxidant defense and ATP production in renal ischemia-reperfusion (I/R) mouse model resulting in recovery of kidney function via activating the Keap1-Nrf2 signaling pathway. Ko et al[77] used a hyperbaric oxygen and autologous adipose-derived MSC combined therapy in SD rat model of renal I/R injury. They found that this combination was effective in reducing the expression of inflammatory markers (MMP-9/TNF-α/NF-κB/ICAM-1) and apoptotic markers (mitochondrial-Bax/cleaved-caspase3/PARP), ameliorating oxidative damage and promoting angiogenesis in renal tubular cells which ultimately improved the kidney injury score and restored many renal function parameters, including the blood urea nitrogen and creatinine. Based on the proved potential of MSCs in alleviating AKI and renal I/R injury, we can anticipate that they might exert comparable therapeutic effects in COVID-19 patients with AKI and other renal complications[77]. Future studies should investigate the effectiveness of MSCs-based cell therapy in COVID-19 patients who developed renal complications. Such therapy could be a worthwhile approach in restoring their kidney functions and decreasing the death rate in this group of patients.

COVID-19 CARDIOVASCULAR COMPLICATIONS AND MSCS

Many patients with COVID-19 were reported to develop acute cardiac injury during the course of the illness. Concomitant presence of cardiac events and COVID-19 increases the mortality rate in COVID-19 patients. COVID-19 cardiovascular complications (CV) include acute myocardial injury, heart failure, pericardial effusion with or without tamponade, arrhythmias, sudden cardiac death, and thrombosis of small and large blood vessels. The clinical reports regarding the severity of COVID-19 were noticeable in patients with previous medical history of underlying CV diseases. Furthermore, many patients infected with COVID-19 developed severe myocardial injury[78], including myocardial dysfunction, cardiomyopathy, arrhythmias and heart failure during the course of critical illness[79-84]. Clinical reports revealed that 38% of patients testing positive for COVID-19 display high levels of cardiac damage biomarkers, such as cardiac troponin and creatinine kinase with significant electrocardiographic changes. Data acquired from multiple clinical centers showed conspicuous correlation between plasma lactate dehydrogenase and chronic kidney levels with COVID-19 severity and the rate of ICU[85].

The proposed mechanisms of the deleterious effect of COVID-19 on the heart are intriguing many clinicians and researchers. Acute myocardial injury caused by SARS-CoV-2 infection could be due to its high affinity for ACE2 which is extensively expressed in the heart, and is associated with direct myocardial injury and remodeling[22,86]. Another proposed mechanism is linked to cytokine storm. Cytokine storm is triggered by an excessive response of type 1 and type 2 T-helper cells, sympathetic hyperactivity, anemia, and hypoxemic myocardial cells damage caused by respiratory dysfunction [type 2 myocardial infarction (MI)][86]. Myocardial oxygen supply/ demand mismatch as a result of severe pneumonia or ARDS can lead to inadequate supply of oxygen and subsequent myocardial damage[87]. The current treatment regimen, including antiviral medications, corticosteroids, and immunological agents; can also potentiate the risk of developing CV complications. The disturbance of electrolytes can occur in any critical systemic illness, including COVID-19 and can trigger life-threatening arrhythmias. There is a particular concern about the risk of hypokalemia in COVID-19 patients as a result of the interaction between SARS-CoV2 with the RAS system. Hypokalemia is a well-known electrolyte disturbance that is associated with serious arrhythmias[87]. Several studies have demonstrated the possible role of MSCs in CV diseases in non-COVID-19 patients. MSCs can offer the potential as regenerative cells for the CV system where through a paracrine mechanism, they activate endogenous repair mechanisms leading to blood vessel growth via angiogenesis, improve cardiomyocytes survival, and reduce cardiomyocytes reactive hypertrophy and fibrosis[88]. Treatment with MSCs may give a clinical benefit to patients due to their regenerative and reparative potential if there are significant myocardial injury and myocardial cell death.

The infusion and transplantation of MSCs have been deemed safe for treating MI patients[89,90]. A study was carried out to evaluate the safety and feasibility of intra-myocardial MSCs injection in patients, shortly after AMI during short-term and 5-year follow-up, a safety analysis demonstrated one transient ischemic attack without any adverse events related to MSCs treatment[90].

Furthermore, MSCs injection successfully improves some cardiac functional measures post-MI[91,92]. MSCs were shown to restore endothelial function by increasing endothelial progenitor cells function and regain flow-mediated vasodilation (FMD which is clinically significant for heart failure patients[93].

There is study that has suggested a potential effect for in situ myocardial regeneration in ischemic heart failure by delivering allogeneic mesenchymal precursor cells called immunomodulatory progenitor cells (iMP) via intramyocardial route[94]. Intramyocardial implantation of iMP cells with CABG was safe with preliminary evidence of efficacy of improved myocardial contractility and perfusion of non-revascularized territories resulting in a significant reduction in left ventricular scar area at 12 mo after treatment. Clinical improvement was associated with a significant improvement in quality of life at 6 mo post-treatment in all patients[94].

A study focused on dose comparison of allogeneic MSCs in patients with ischemic cardiomyopathy demonstrated that the optimal dose of MSCs was 100 million cells, and this optimal dose increases the ejection fraction in ischemic patients[95].

A meta-analysis of 52 pre-clinical animal studies performed on different animal species such as pig, dog and sheep investigated the effect of cardiac stem cells therapy in ischemic cardiomyopathy (52 studies; n = 888 animals). This study reported that MSCs therapy is safe and associated with significant approximately 7.5% improvements in left ventricular ejection fraction (LVEF), without any increased mortality due to MSCs administration[96].

The use of MSCs to treat CV dysfunction and damage in COVID-19 patients has yet to be fully elucidated. The studies over the past decade provide good preliminary evidence for researchers and clinicians alike to further investigate the use of this MSCs therapy in COVID-19 patient cohorts.

CONCLUSION

As the number of COVID-19 cases upsurges rampantly with many patients suffering from life-threatening multiple organ failure, finding a curative way to save the life of critically-ill patients and improve the clinical outcomes is urgently needed. As mentioned above, there are no specific therapeutic medications for COVID-19 serious symptoms and complications, and the applied therapeutic guidelines in the clinical settings are non-specific and based on recommendations. At present, cell therapy is a highly sophisticated approach for treating many diseases, and a large number of studies have been carried out in recent months to treat the SARS-CoV-2 virus using stem cells, particularly MSCs. The safety and therapeutic role of MSCs for ARDS and organ dysfunction that are caused by SARS-CoV-2 infection have been validated by many studies. MSCs can secrete a cocktail of factors that modulate the lung microenvironment, subdue the immune system over-activation, potentiate tissue repair machinery, invigorate alveolar epithelial cells, and inhibit pulmonary remodeling and fibrosis that result in massive improvement in the lung functions. MSCs can regenerate other damaged organs which are associated with COVID-19. Many issues related to the application of MSCs, including the ideal dose and optimum timing of MSCs delivery should be further explored with the objective of enhancing the clinical outcomes in COVID-19 critically-ill patients.

ACKNOWLEDGEMENTS

We would like to thank Mr. Abu-El-Rub M for the time he spent in proof reading and editing of the review manuscript.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Cell and tissue engineering

Country/Territory of origin: Canada

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Jia Z, Yassa M S-Editor: Liu M L-Editor: A P-Editor: Ma YJ

References
1.  Gavriatopoulou M, Korompoki E, Fotiou D, Ntanasis-Stathopoulos I, Psaltopoulou T, Kastritis E, Terpos E, Dimopoulos MA. Organ-specific manifestations of COVID-19 infection. Clin Exp Med. 2020;20:493-506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 282]  [Article Influence: 70.5]  [Reference Citation Analysis (0)]
2.  Neupane K, Ahmed Z, Pervez H, Ashraf R, Majeed A. Potential Treatment Options for COVID-19: A Comprehensive Review of Global Pharmacological Development Efforts. Cureus. 2020;12:e8845.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 12]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
3.  Sarkar C, Mondal M, Torequl Islam M, Martorell M, Docea AO, Maroyi A, Sharifi-Rad J, Calina D. Potential Therapeutic Options for COVID-19: Current Status, Challenges, and Future Perspectives. Front Pharmacol. 2020;11:572870.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 57]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
4.  Coelho A, Alvites RD, Branquinho MV, Guerreiro SG, Maurício AC. Mesenchymal Stem Cells (MSCs) as a Potential Therapeutic Strategy in COVID-19 Patients: Literature Research. Front Cell Dev Biol. 2020;8:602647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
5.  Ankrum JA, Ong JF, Karp JM. Mesenchymal stem cells: immune evasive, not immune privileged. Nat Biotechnol. 2014;32:252-260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 872]  [Cited by in F6Publishing: 975]  [Article Influence: 97.5]  [Reference Citation Analysis (0)]
6.  Klinker MW, Wei CH. Mesenchymal stem cells in the treatment of inflammatory and autoimmune diseases in experimental animal models. World J Stem Cells. 2015;7:556-567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 67]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
7.  Wang LT, Ting CH, Yen ML, Liu KJ, Sytwu HK, Wu KK, Yen BL. Human mesenchymal stem cells (MSCs) for treatment towards immune- and inflammation-mediated diseases: review of current clinical trials. J Biomed Sci. 2016;23:76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 225]  [Article Influence: 28.1]  [Reference Citation Analysis (0)]
8.  Tao H, Han Z, Han ZC, Li Z. Proangiogenic Features of Mesenchymal Stem Cells and Their Therapeutic Applications. Stem Cells Int. 2016;2016:1314709.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 165]  [Article Influence: 20.6]  [Reference Citation Analysis (0)]
9.  Ullah I, Subbarao RB, Rho GJ. Human mesenchymal stem cells - current trends and future prospective. Biosci Rep. 2015;35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 715]  [Cited by in F6Publishing: 813]  [Article Influence: 90.3]  [Reference Citation Analysis (0)]
10.  Aguilar RB, Hardigan P, Mayi B, Sider D, Piotrkowski J, Mehta JP, Dev J, Seijo Y, Camargo AL, Andux L, Hagen K, Hernandez MB. Current Understanding of COVID-19 Clinical Course and Investigational Treatments. Front Med (Lausanne). 2020;7:555301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
11.  Yadav P, Vats R, Bano A, Bhardwaj R. Mesenchymal stem cell immunomodulation and regeneration therapeutics as an ameliorative approach for COVID-19 pandemics. Life Sci. 2020;263:118588.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
12.  Chen W, Pan JY. Anatomical and Pathological Observation and Analysis of SARS and COVID-19: Microthrombosis Is the Main Cause of Death. Biol Proced Online. 2021;23:4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 53]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
13.  Tang Y, Liu J, Zhang D, Xu Z, Ji J, Wen C. Cytokine Storm in COVID-19: The Current Evidence and Treatment Strategies. Front Immunol. 2020;11:1708.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 768]  [Cited by in F6Publishing: 647]  [Article Influence: 161.8]  [Reference Citation Analysis (0)]
14.  Weiss ARR, Dahlke MH. Immunomodulation by Mesenchymal Stem Cells (MSCs): Mechanisms of Action of Living, Apoptotic, and Dead MSCs. Front Immunol. 2019;10:1191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 287]  [Cited by in F6Publishing: 401]  [Article Influence: 80.2]  [Reference Citation Analysis (0)]
15.  Xu Z, Shi L, Wang Y, Zhang J, Huang L, Zhang C, Liu S, Zhao P, Liu H, Zhu L, Tai Y, Bai C, Gao T, Song J, Xia P, Dong J, Zhao J, Wang FS. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med. 2020;8:420-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5228]  [Cited by in F6Publishing: 5530]  [Article Influence: 1382.5]  [Reference Citation Analysis (2)]
16.  Lin L, Lu L, Cao W, Li T. Hypothesis for potential pathogenesis of SARS-CoV-2 infection-a review of immune changes in patients with viral pneumonia. Emerg Microbes Infect. 2020;9:727-732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 582]  [Cited by in F6Publishing: 527]  [Article Influence: 131.8]  [Reference Citation Analysis (0)]
17.  Leng Z, Zhu R, Hou W, Feng Y, Yang Y, Han Q, Shan G, Meng F, Du D, Wang S, Fan J, Wang W, Deng L, Shi H, Li H, Hu Z, Zhang F, Gao J, Liu H, Li X, Zhao Y, Yin K, He X, Gao Z, Wang Y, Yang B, Jin R, Stambler I, Lim LW, Su H, Moskalev A, Cano A, Chakrabarti S, Min KJ, Ellison-Hughes G, Caruso C, Jin K, Zhao RC. Transplantation of ACE2- Mesenchymal Stem Cells Improves the Outcome of Patients with COVID-19 Pneumonia. Aging Dis. 2020;11:216-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 730]  [Cited by in F6Publishing: 798]  [Article Influence: 199.5]  [Reference Citation Analysis (0)]
18.  Li Y, Xu J, Shi W, Chen C, Shao Y, Zhu L, Lu W, Han X. Mesenchymal stromal cell treatment prevents H9N2 avian influenza virus-induced acute lung injury in mice. Stem Cell Res Ther. 2016;7:159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 87]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
19.  Zhang Y, Ding J, Ren S, Wang W, Yang Y, Li S, Meng M, Wu T, Liu D, Tian S, Tian H, Chen S, Zhou C. Intravenous infusion of human umbilical cord Wharton's jelly-derived mesenchymal stem cells as a potential treatment for patients with COVID-19 pneumonia. Stem Cell Res Ther. 2020;11:207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 86]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
20.  Liang B, Chen J, Li T, Wu H, Yang W, Li Y, Li J, Yu C, Nie F, Ma Z, Yang M, Xiao M, Nie P, Gao Y, Qian C, Hu M. Clinical remission of a critically ill COVID-19 patient treated by human umbilical cord mesenchymal stem cells: A case report. Medicine (Baltimore). 2020;99:e21429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 212]  [Article Influence: 53.0]  [Reference Citation Analysis (0)]
21.  Meng F, Xu R, Wang S, Xu Z, Zhang C, Li Y, Yang T, Shi L, Fu J, Jiang T, Huang L, Zhao P, Yuan X, Fan X, Zhang JY, Song J, Zhang D, Jiao Y, Liu L, Zhou C, Maeurer M, Zumla A, Shi M, Wang FS. Human umbilical cord-derived mesenchymal stem cell therapy in patients with COVID-19: a phase 1 clinical trial. Signal Transduct Target Ther. 2020;5:172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 203]  [Article Influence: 50.8]  [Reference Citation Analysis (0)]
22.  Wu Z, McGoogan JM. Characteristics of and Important Lessons From the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary of a Report of 72 314 Cases From the Chinese Center for Disease Control and Prevention. JAMA. 2020;323:1239-1242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11409]  [Cited by in F6Publishing: 10972]  [Article Influence: 2743.0]  [Reference Citation Analysis (0)]
23.  Rathi H, Burman V, Datta SK, Rana SV, Mirza AA, Saha S, Kumar R, Naithani M. Review on COVID-19 Etiopathogenesis, Clinical Presentation and Treatment Available with Emphasis on ACE2. Indian J Clin Biochem. 2021;1-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 4]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
24.  Al Adem K, Shanti A, Stefanini C, Lee S. Inhibition of SARS-CoV-2 Entry into Host Cells Using Small Molecules. Pharmaceuticals (Basel). 2020;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
25.  Li X, Ma X. Acute respiratory failure in COVID-19: is it "typical" ARDS? Crit Care. 2020;24:198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 440]  [Cited by in F6Publishing: 412]  [Article Influence: 103.0]  [Reference Citation Analysis (0)]
26.  ARDS Definition Task Force. , Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, Fan E, Camporota L, Slutsky AS. Acute respiratory distress syndrome: the Berlin Definition. JAMA. 2012;307:2526-2533.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1812]  [Cited by in F6Publishing: 4025]  [Article Influence: 335.4]  [Reference Citation Analysis (0)]
27.  Torres Acosta MA, Singer BD. Pathogenesis of COVID-19-induced ARDS: implications for an ageing population. Eur Respir J. 2020;56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 129]  [Article Influence: 32.3]  [Reference Citation Analysis (0)]
28.  Chen G, Wu D, Guo W, Cao Y, Huang D, Wang H, Wang T, Zhang X, Chen H, Yu H, Zhang M, Wu S, Song J, Chen T, Han M, Li S, Luo X, Zhao J, Ning Q. Clinical and immunological features of severe and moderate coronavirus disease 2019. J Clin Invest. 2020;130:2620-2629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2835]  [Cited by in F6Publishing: 3176]  [Article Influence: 794.0]  [Reference Citation Analysis (0)]
29.  Blanco-Melo D, Nilsson-Payant BE, Liu WC, Uhl S, Hoagland D, Møller R, Jordan TX, Oishi K, Panis M, Sachs D, Wang TT, Schwartz RE, Lim JK, Albrecht RA, tenOever BR. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell. 2020;181:1036-1045.e9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3305]  [Cited by in F6Publishing: 2886]  [Article Influence: 721.5]  [Reference Citation Analysis (0)]
30.  Liu S, Peng D, Qiu H, Yang K, Fu Z, Zou L. Mesenchymal stem cells as a potential therapy for COVID-19. Stem Cell Res Ther. 2020;11:169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 56]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
31.  Fengyun W, LiXin Z, Xinhua Q, Bin F. Mesenchymal Stromal Cells Attenuate Infection-Induced Acute Respiratory Distress Syndrome in Animal Experiments: A Meta-Analysis. Cell Transplant. 2020;29:963689720969186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
32.  Chen J, Hu C, Chen L, Tang L, Zhu Y, Xu X, Gao H, Lu X, Yu L, Dai X, Xiang C, Li L. Clinical Study of Mesenchymal Stem Cell Treatment for Acute Respiratory Distress Syndrome Induced by Epidemic Influenza A (H7N9) Infection: A Hint for COVID-19 Treatment. Engineering (Beijing). 2020;6:1153-1161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 157]  [Article Influence: 39.3]  [Reference Citation Analysis (0)]
33.  Zheng G, Huang L, Tong H, Shu Q, Hu Y, Ge M, Deng K, Zhang L, Zou B, Cheng B, Xu J. Treatment of acute respiratory distress syndrome with allogeneic adipose-derived mesenchymal stem cells: a randomized, placebo-controlled pilot study. Respir Res. 2014;15:39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 308]  [Article Influence: 30.8]  [Reference Citation Analysis (0)]
34.  Wilson JG, Liu KD, Zhuo H, Caballero L, McMillan M, Fang X, Cosgrove K, Vojnik R, Calfee CS, Lee JW, Rogers AJ, Levitt J, Wiener-Kronish J, Bajwa EK, Leavitt A, McKenna D, Thompson BT, Matthay MA. Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. Lancet Respir Med. 2015;3:24-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 492]  [Cited by in F6Publishing: 542]  [Article Influence: 54.2]  [Reference Citation Analysis (0)]
35.  Matthay MA, Calfee CS, Zhuo H, Thompson BT, Wilson JG, Levitt JE, Rogers AJ, Gotts JE, Wiener-Kronish JP, Bajwa EK, Donahoe MP, McVerry BJ, Ortiz LA, Exline M, Christman JW, Abbott J, Delucchi KL, Caballero L, McMillan M, McKenna DH, Liu KD. 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;7:154-162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 392]  [Article Influence: 65.3]  [Reference Citation Analysis (0)]
36.  Yilmaz R, Adas G, Cukurova Z, Kart Yasar K, Isiksacan N, Oztel ON, Karaoz E. Mesenchymal stem cells treatment in COVID-19 patient with multi-organ involvement. Bratisl Lek Listy. 2020;121:847-852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
37.  Tao J, Nie Y, Wu H, Cheng L, Qiu Y, Fu J, Jiang X. Umbilical cord blood-derived mesenchymal stem cells in treating a critically ill COVID-19 patient. J Infect Dev Ctries. 2020;14:1138-1145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
38.  Soler Rich R, Rius Tarruella J, Melgosa Camarero MT. [Expanded Mesenchymal Stem Cells: a novel therapeutic approach of SARS-CoV-2 pneumonia (COVID-19). Concepts regarding a first case in Spain]. Med Clin (Barc). 2020;155:318-319.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
39.  Peng H, Gong T, Huang X, Sun X, Luo H, Wang W, Luo J, Luo B, Chen Y, Wang X, Long H, Mei H, Li C, Dai Y, Li H. A synergistic role of convalescent plasma and mesenchymal stem cells in the treatment of severely ill COVID-19 patients: a clinical case report. Stem Cell Res Ther. 2020;11:291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
40.  Zengin R, Beyaz O, Koc ES, Akinci IO, Kocagoz S, Sagcan G, Ovali E, Cuhadaroglu C. Mesenchymal stem cell treatment in a critically ill COVID-19 patient: a case report. Stem Cell Investig. 2020;7:17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
41.  Zhu Y, Zhu R, Liu K, Li X, Chen D, Bai D, Luo J, Liu Y, Zhang Y, Li L, Hu J, Xu D, Zhao RC. Human Umbilical Cord Mesenchymal Stem Cells for Adjuvant Treatment of a Critically Ill COVID-19 Patient: A Case Report. Infect Drug Resist. 2020;13:3295-3300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
42.  Tyumina O  Evaluation of Safety and Efficiency of Method of Exosome Inhalation in SARS-CoV-2 Associated Pneumonia. (COVID-19EXO). [2021 Mar 10]. In: ClinicalTrials.gov [Internet]. Samara: U.S. National Library of Medicine. Available from: https://clinicaltrials.gov/ct2/show/results/NCT04491240?term=mesenchymal+cells&recrs=abdef&type=Intr&cond=covid-19&draw=2&rank=59 ClinicalTrials.gov Identifier: NCT04491240.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Lanzoni G, Linetsky E, Correa D, Messinger Cayetano S, Alvarez RA, Kouroupis D, Alvarez Gil A, Poggioli R, Ruiz P, Marttos AC, Hirani K, Bell CA, Kusack H, Rafkin L, Baidal D, Pastewski A, Gawri K, Leñero C, Mantero AMA, Metalonis SW, Wang X, Roque L, Masters B, Kenyon NS, Ginzburg E, Xu X, Tan J, Caplan AI, Glassberg MK, Alejandro R, Ricordi C. Umbilical cord mesenchymal stem cells for COVID-19 acute respiratory distress syndrome: A double-blind, phase 1/2a, randomized controlled trial. Stem Cells Transl Med. 2021;10:660-673.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 266]  [Cited by in F6Publishing: 230]  [Article Influence: 76.7]  [Reference Citation Analysis (0)]
44.  Shu L, Niu C, Li R, Huang T, Wang Y, Huang M, Ji N, Zheng Y, Chen X, Shi L, Wu M, Deng K, Wei J, Wang X, Cao Y, Yan J, Feng G. Treatment of severe COVID-19 with human umbilical cord mesenchymal stem cells. Stem Cell Res Ther. 2020;11:361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 193]  [Article Influence: 48.3]  [Reference Citation Analysis (0)]
45.  Sánchez-Guijo F, García-Arranz M, López-Parra M, Monedero P, Mata-Martínez C, Santos A, Sagredo V, Álvarez-Avello JM, Guerrero JE, Pérez-Calvo C, Sánchez-Hernández MV, Del-Pozo JL, Andreu EJ, Fernández-Santos ME, Soria-Juan B, Hernández-Blasco LM, Andreu E, Sempere JM, Zapata AG, Moraleda JM, Soria B, Fernández-Avilés F, García-Olmo D, Prósper F. Adipose-derived mesenchymal stromal cells for the treatment of patients with severe SARS-CoV-2 pneumonia requiring mechanical ventilation. A proof of concept study. EClinicalMedicine. 2020;25:100454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 109]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
46.  Sengupta V, Sengupta S, Lazo A, Woods P, Nolan A, Bremer N. Exosomes Derived from Bone Marrow Mesenchymal Stem Cells as Treatment for Severe COVID-19. Stem Cells Dev. 2020;29:747-754.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 449]  [Cited by in F6Publishing: 409]  [Article Influence: 102.3]  [Reference Citation Analysis (0)]
47.  Thompson BT, Chambers RC, Liu KD. Acute Respiratory Distress Syndrome. N Engl J Med. 2017;377:562-572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 921]  [Cited by in F6Publishing: 997]  [Article Influence: 142.4]  [Reference Citation Analysis (0)]
48.  Li W, Moore MJ, Vasilieva N, Sui J, Wong SK, Berne MA, Somasundaran M, Sullivan JL, Luzuriaga K, Greenough TC, Choe H, Farzan M. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature. 2003;426:450-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4113]  [Cited by in F6Publishing: 4360]  [Article Influence: 207.6]  [Reference Citation Analysis (0)]
49.  Matsuyama S, Nagata N, Shirato K, Kawase M, Takeda M, Taguchi F. Efficient activation of the severe acute respiratory syndrome coronavirus spike protein by the transmembrane protease TMPRSS2. J Virol. 2010;84:12658-12664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 551]  [Cited by in F6Publishing: 578]  [Article Influence: 41.3]  [Reference Citation Analysis (0)]
50.  Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, Müller MA, Drosten C, Pöhlmann S. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell. 2020;181:271-280.e8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11946]  [Cited by in F6Publishing: 13083]  [Article Influence: 3270.8]  [Reference Citation Analysis (0)]
51.  Shyamsundar M, McAuley DF, Ingram RJ, Gibson DS, O'Kane D, McKeown ST, Edwards A, Taggart C, Elborn JS, Calfee CS, Matthay MA, O'Kane CM. Keratinocyte growth factor promotes epithelial survival and resolution in a human model of lung injury. Am J Respir Crit Care Med. 2014;189:1520-1529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 85]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
52.  Caplan AI, Correa D. The MSC: an injury drugstore. Cell Stem Cell. 2011;9:11-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1127]  [Cited by in F6Publishing: 1134]  [Article Influence: 94.5]  [Reference Citation Analysis (0)]
53.  Shetty AK. Mesenchymal Stem Cell Infusion Shows Promise for Combating Coronavirus (COVID-19)- Induced Pneumonia. Aging Dis. 2020;11:462-464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 92]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
54.  George PM, Wells AU, Jenkins RG. Pulmonary fibrosis and COVID-19: the potential role for antifibrotic therapy. Lancet Respir Med. 2020;8:807-815.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 595]  [Cited by in F6Publishing: 665]  [Article Influence: 166.3]  [Reference Citation Analysis (0)]
55.  George PM, Patterson CM, Reed AK, Thillai M. Lung transplantation for idiopathic pulmonary fibrosis. Lancet Respir Med. 2019;7:271-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 137]  [Article Influence: 27.4]  [Reference Citation Analysis (0)]
56.  Durand N, Mallea J, Zubair AC. Insights into the use of mesenchymal stem cells in COVID-19 mediated acute respiratory failure. NPJ Regen Med. 2020;5:17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 33]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
57.  Vitiello A, Pelliccia C, Ferrara F. COVID-19 Patients with Pulmonary Fibrotic Tissue: Clinical Pharmacological Rational of Antifibrotic Therapy. SN Compr Clin Med. 2020;1-4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
58.  Qin C, Zhou L, Hu Z, Zhang S, Yang S, Tao Y, Xie C, Ma K, Shang K, Wang W, Tian DS. Dysregulation of Immune Response in Patients With Coronavirus 2019 (COVID-19) in Wuhan, China. Clin Infect Dis. 2020;71:762-768.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2495]  [Cited by in F6Publishing: 3143]  [Article Influence: 785.8]  [Reference Citation Analysis (0)]
59.  Gonzalez-Gonzalez FJ, Chandel NS, Jain M, Budinger GRS. Reactive oxygen species as signaling molecules in the development of lung fibrosis. Transl Res. 2017;190:61-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 59]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
60.  Nile SH, Nile A, Qiu J, Li L, Jia X, Kai G. COVID-19: Pathogenesis, cytokine storm and therapeutic potential of interferons. Cytokine Growth Factor Rev. 2020;53:66-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 273]  [Article Influence: 68.3]  [Reference Citation Analysis (0)]
61.  Golchin A, Seyedjafari E, Ardeshirylajimi A. Mesenchymal Stem Cell Therapy for COVID-19: Present or Future. Stem Cell Rev Rep. 2020;16:427-433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 186]  [Cited by in F6Publishing: 207]  [Article Influence: 51.8]  [Reference Citation Analysis (1)]
62.  Yang Y, Hu S, Xu X, Li J, Liu A, Han J, Liu S, Liu L, Qiu H. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 42]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
63.  Jung YJ, Park YY, Huh JW, Hong SB. The effect of human adipose-derived stem cells on lipopolysaccharide-induced acute respiratory distress syndrome in mice. Ann Transl Med. 2019;7:674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
64.  Chuang HM, Shih TE, Lu KY, Tsai SF, Harn HJ, Ho LI. Mesenchymal Stem Cell Therapy of Pulmonary Fibrosis: Improvement with Target Combination. Cell Transplant. 2018;27:1581-1587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
65.  Kuraitis D, Giordano C, Ruel M, Musarò A, Suuronen EJ. Exploiting extracellular matrix-stem cell interactions: a review of natural materials for therapeutic muscle regeneration. Biomaterials. 2012;33:428-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 76]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
66.  Moodley Y, Atienza D, Manuelpillai U, Samuel CS, Tchongue J, Ilancheran S, Boyd R, Trounson A. Human umbilical cord mesenchymal stem cells reduce fibrosis of bleomycin-induced lung injury. Am J Pathol. 2009;175:303-313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 263]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
67.  Carriazo S, Kanbay M, Ortiz A. Kidney disease and electrolytes in COVID-19: more than meets the eye. Clin Kidney J. 2020;13:274-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 26]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
68.  Damtie S, Biadgo B, Baynes HW, Ambachew S, Melak T, Asmelash D, Abebe M. Chronic Kidney Disease and Associated Risk Factors Assessment among Diabetes Mellitus Patients at A Tertiary Hospital, Northwest Ethiopia. Ethiop J Health Sci. 2018;28:691-700.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 25]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
69.  Zamoner W, Santos CADS, Magalhães LE, de Oliveira PGS, Balbi AL, Ponce D. Acute Kidney Injury in COVID-19: 90 Days of the Pandemic in a Brazilian Public Hospital. Front Med (Lausanne). 2021;8:622577.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 19]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
70.  Russo E, Esposito P, Taramasso L, Magnasco L, Saio M, Briano F, Russo C, Dettori S, Vena A, Di Biagio A, Garibotto G, Bassetti M, Viazzi F; GECOVID working group. Kidney disease and all-cause mortality in patients with COVID-19 hospitalized in Genoa, Northern Italy. J Nephrol. 2021;34:173-183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
71.  Lin L, Wang X, Ren J, Sun Y, Yu R, Li K, Zheng L, Yang J. Risk factors and prognosis for COVID-19-induced acute kidney injury: a meta-analysis. BMJ Open. 2020;10:e042573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 70]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
72.  Ni W, Yang X, Yang D, Bao J, Li R, Xiao Y, Hou C, Wang H, Liu J, Xu Y, Cao Z, Gao Z. Role of angiotensin-converting enzyme 2 (ACE2) in COVID-19. Crit Care. 2020;24:422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 587]  [Cited by in F6Publishing: 609]  [Article Influence: 152.3]  [Reference Citation Analysis (1)]
73.  Varagic J, Ahmad S, Nagata S, Ferrario CM. ACE2: angiotensin II/angiotensin-(1-7) balance in cardiac and renal injury. Curr Hypertens Rep. 2014;16:420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 75]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
74.  Hardenberg JB, Luft FC. Covid-19, ACE2 and the kidney. Acta Physiol (Oxf). 2020;230:e13539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
75.  Zhou S, Qiao YM, Liu YG, Liu D, Hu JM, Liao J, Li M, Guo Y, Fan LP, Li LY, Zhao M. Bone marrow derived mesenchymal stem cells pretreated with erythropoietin accelerate the repair of acute kidney injury. Cell Biosci. 2020;10:130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
76.  Cao H, Cheng Y, Gao H, Zhuang J, Zhang W, Bian Q, Wang F, Du Y, Li Z, Kong D, Ding D, Wang Y. In Vivo Tracking of Mesenchymal Stem Cell-Derived Extracellular Vesicles Improving Mitochondrial Function in Renal Ischemia-Reperfusion Injury. ACS Nano. 2020;14:4014-4026.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 113]  [Article Influence: 28.3]  [Reference Citation Analysis (0)]
77.  Ko SF, Chen KH, Wallace CG, Yang CC, Sung PH, Shao PL, Li YC, Chen YT, Yip HK. Protective effect of combined therapy with hyperbaric oxygen and autologous adipose-derived mesenchymal stem cells on renal function in rodent after acute ischemia-reperfusion injury. Am J Transl Res. 2020;12:3272-3287.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Guo T, Fan Y, Chen M, Wu X, Zhang L, He T, Wang H, Wan J, Wang X, Lu Z. Cardiovascular Implications of Fatal Outcomes of Patients With Coronavirus Disease 2019 (COVID-19). JAMA Cardiol. 2020;5:811-818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2516]  [Cited by in F6Publishing: 2688]  [Article Influence: 672.0]  [Reference Citation Analysis (0)]
79.  Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, Xiang J, Wang Y, Song B, Gu X, Guan L, Wei Y, Li H, Wu X, Xu J, Tu S, Zhang Y, Chen H, Cao B. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054-1062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17476]  [Cited by in F6Publishing: 17250]  [Article Influence: 4312.5]  [Reference Citation Analysis (0)]
80.  Shi S, Qin M, Shen B, Cai Y, Liu T, Yang F, Gong W, Liu X, Liang J, Zhao Q, Huang H, Yang B, Huang C. Association of Cardiac Injury With Mortality in Hospitalized Patients With COVID-19 in Wuhan, China. JAMA Cardiol. 2020;5:802-810.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2428]  [Cited by in F6Publishing: 2858]  [Article Influence: 714.5]  [Reference Citation Analysis (0)]
81.  Ruan Q, Yang K, Wang W, Jiang L, Song J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020;46:846-848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2604]  [Cited by in F6Publishing: 3048]  [Article Influence: 762.0]  [Reference Citation Analysis (0)]
82.  Arentz M, Yim E, Klaff L, Lokhandwala S, Riedo FX, Chong M, Lee M. Characteristics and Outcomes of 21 Critically Ill Patients With COVID-19 in Washington State. JAMA. 2020;323:1612-1614.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1511]  [Cited by in F6Publishing: 1572]  [Article Influence: 393.0]  [Reference Citation Analysis (0)]
83.  Wang CJ, Ng CY, Brook RH. Response to COVID-19 in Taiwan: Big Data Analytics, New Technology, and Proactive Testing. JAMA. 2020;323:1341-1342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 974]  [Cited by in F6Publishing: 758]  [Article Influence: 189.5]  [Reference Citation Analysis (0)]
84.  Vaduganathan M, Vardeny O, Michel T, McMurray JJV, Pfeffer MA, Solomon SD. Renin-Angiotensin-Aldosterone System Inhibitors in Patients with Covid-19. N Engl J Med. 2020;382:1653-1659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1484]  [Cited by in F6Publishing: 1491]  [Article Influence: 372.8]  [Reference Citation Analysis (0)]
85.  Bats ML, Rucheton B, Fleur T, Orieux A, Chemin C, Rubin S, Colombies B, Desclaux A, Rivoisy C, Mériglier E, Rivière E, Boyer A, Gruson D, Pellegrin I, Trimoulet P, Garrigue I, Alkouri R, Dupin C, Moreau-Gaudry F, Bedel A, Dabernat S. Covichem: A biochemical severity risk score of COVID-19 upon hospital admission. PLoS One. 2021;16:e0250956.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
86.  Zheng YY, Ma YT, Zhang JY, Xie X. COVID-19 and the cardiovascular system. Nat Rev Cardiol. 2020;17:259-260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1825]  [Cited by in F6Publishing: 1965]  [Article Influence: 491.3]  [Reference Citation Analysis (0)]
87.  Xiong TY, Redwood S, Prendergast B, Chen M. Coronaviruses and the cardiovascular system: acute and long-term implications. Eur Heart J. 2020;41:1798-1800.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 464]  [Cited by in F6Publishing: 466]  [Article Influence: 155.3]  [Reference Citation Analysis (0)]
88.  Ellison-Hughes GM, Colley L, O'Brien KA, Roberts KA, Agbaedeng TA, Ross MD. The Role of MSC Therapy in Attenuating the Damaging Effects of the Cytokine Storm Induced by COVID-19 on the Heart and Cardiovascular System. Front Cardiovasc Med. 2020;7:602183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
89.  Lee JW, Lee SH, Youn YJ, Ahn MS, Kim JY, Yoo BS, Yoon J, Kwon W, Hong IS, Lee K, Kwan J, Park KS, Choi D, Jang YS, Hong MK. A randomized, open-label, multicenter trial for the safety and efficacy of adult mesenchymal stem cells after acute myocardial infarction. J Korean Med Sci. 2014;29:23-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 114]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
90.  Rodrigo SF, van Ramshorst J, Hoogslag GE, Boden H, Velders MA, Cannegieter SC, Roelofs H, Al Younis I, Dibbets-Schneider P, Fibbe WE, Zwaginga JJ, Bax JJ, Schalij MJ, Beeres SL, Atsma DE. Intramyocardial injection of autologous bone marrow-derived ex vivo expanded mesenchymal stem cells in acute myocardial infarction patients is feasible and safe up to 5 years of follow-up. J Cardiovasc Transl Res. 2013;6:816-825.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 75]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
91.  Qi Z, Duan F, Liu S, Lv X, Wang H, Gao Y, Wang J. Effects of Bone Marrow Mononuclear Cells Delivered through a Graft Vessel for Patients with Previous Myocardial Infarction and Chronic Heart Failure: An Echocardiographic Study of Left Ventricular Function. Echocardiography. 2015;32:937-946.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
92.  Kim SH, Cho JH, Lee YH, Lee JH, Kim SS, Kim MY, Lee MG, Kang WY, Lee KS, Ahn YK, Jeong MH, Kim HS. Improvement in Left Ventricular Function with Intracoronary Mesenchymal Stem Cell Therapy in a Patient with Anterior Wall ST-Segment Elevation Myocardial Infarction. Cardiovasc Drugs Ther. 2018;32:329-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 53]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
93.  Premer C, Blum A, Bellio MA, Schulman IH, Hurwitz BE, Parker M, Dermarkarian CR, DiFede DL, Balkan W, Khan A, Hare JM. Allogeneic Mesenchymal Stem Cells Restore Endothelial Function in Heart Failure by Stimulating Endothelial Progenitor Cells. EBioMedicine. 2015;2:467-475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 88]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
94.  Anastasiadis K, Antonitsis P, Westaby S, Reginald A, Sultan S, Doumas A, Efthimiadis G, Evans MJ. Implantation of a Novel Allogeneic Mesenchymal Precursor Cell Type in Patients with Ischemic Cardiomyopathy Undergoing Coronary Artery Bypass Grafting: an Open Label Phase IIa Trial. J Cardiovasc Transl Res. 2016;9:202-213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
95.  Florea V, Rieger AC, DiFede DL, El-Khorazaty J, Natsumeda M, Banerjee MN, Tompkins BA, Khan A, Schulman IH, Landin AM, Mushtaq M, Golpanian S, Lowery MH, Byrnes JJ, Hendel RC, Cohen MG, Valasaki K, Pujol MV, Ghersin E, Miki R, Delgado C, Abuzeid F, Vidro-Casiano M, Saltzman RG, DaFonseca D, Caceres LV, Ramdas KN, Mendizabal A, Heldman AW, Mitrani RD, Hare JM. Dose Comparison Study of Allogeneic Mesenchymal Stem Cells in Patients With Ischemic Cardiomyopathy (The TRIDENT Study). Circ Res. 2017;121:1279-1290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 132]  [Article Influence: 18.9]  [Reference Citation Analysis (0)]
96.  van der Spoel TI, Jansen of Lorkeers SJ, Agostoni P, van Belle E, Gyöngyösi M, Sluijter JP, Cramer MJ, Doevendans PA, Chamuleau SA. Human relevance of pre-clinical studies in stem cell therapy: systematic review and meta-analysis of large animal models of ischaemic heart disease. Cardiovasc Res. 2011;91:649-658.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 185]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]