MSB 2.26% 90.5¢ mesoblast limited

[IMG] Enabling Technologies For Cell‐Based Clinical Translation...

  1. 6,121 Posts.
    lightbulb Created with Sketch. 1055

    Enabling Technologies For Cell‐Based Clinical Translation
    Current status of mesenchymal stem cell therapy for immune/inflammatory lung disorders: Gleaning insights for possible use in COVID ‐19


    B. Linju Yen

    Men‐Luh Yen

    Li‐Tzu Wang

    Ko‐Jiunn Liu

    Huey‐Kang Sytwu

    First published:11 June 2020

    https://doi.org/10.1002/sctm.20-0186

    1 INTRODUCTION

    Human mesenchymal stem/stromal cells (MSCs) are multilineage somatic progenitors with broad immunomodulatory properties. Since initial isolation from the bone marrow (BM), MSCs have been found in numerous adult and fetal‐derived organs/tissues such as adipose tissue, dental pulp, umbilical cord, and placenta.1 In addition to trilineage paraxial mesodermal differentiation capacity toward bone, cartilage, and fat, the immunomodulatory properties of MSCs not only allow for expansion of therapeutic use from regenerative medicine to immune‐ and inflammation‐related diseases, but also for third party allogeneic use.2
    The first published full report on clinical use of MSCs for immune/inflammatory disease was in 2004, in which allogeneic haploidentical bone marrow mesenchymal stem cell (BMMSC) infusions were given for a pediatric patient with acute refractory graft‐vs‐host‐disease (GVHD).3 Of note, median survival at that institution was a mere 2 months for the other 24 patients with similarly severe GVHD, while this patient remained well 1 year after MSC treatment. Surprisingly, prior to this clinical case report, there were only a handful of studies demonstrating MSC immunomodulation, with only one study showing in vivo data of prolonged skin engraftment.4-6 Since then, MSC immunomodulation has shown to be broad‐based, best detailed for CD4 lymphocytes but also for dendritic cells and natural killer cells.7, 8 The immunomodulatory properties are clinically relevant, as evidenced by the increasing proportions of MSC trials focusing on immune/inflammatory diseases which in recent years has accounted for approximately one‐third of the trials.9
    One of the earliest reports demonstrating MSC immunomodulation was in reduction of bleomycin‐induced pulmonary inflammation in mice.10 It comes as somewhat of a surprise that clinical use of MSCs for lung diseases has been relatively slow to start, with most trials initiated in 2015. Moreover, it had been known for over a decade that intravenous delivery of MSCs—the most typical method of intervention for any cell therapy—results in the overwhelming majority of cells (80%~90%) lodging in the lungs which is further increased with inflammation.10, 11 Hence, there is discussion that MSC therapy (MSCT) may be particularly useful in immune/inflammatory pulmonary conditions.12 However, clinical trials for these diseases were still relatively few until this year: as of 17 May 2020, out of 68 MSC trials for lung immune/inflammatory diseases, 31 trials are specifically for COVID‐19 as registered on the NIH Clinical Trial Database (https://ClinicalTrials.gov/) (Figure 1). Due to the rapid global spread of COVID‐19, the high mortality rate of those with severe disease, and no proven effective therapies as of yet, a desperate search for possible treatments is ongoing.13 MSCT is clearly one such attempt, with new trials being added almost daily despite the lack of COVID‐19‐related preclinical data. In this review, we will examine the mechanistic evidence for clinical use of MSCs in pulmonary immune/inflammatory disorders, and survey the ongoing clinical trials—including for COVID‐19—of MSCT for these diseases, with some perspectives and comments on MSCT for COVID‐19.

    FIGURE 1
    Open in figure viewerPowerPoint
    Current disease distribution of clinical trials using human MSC for immune/inflammatory pulmonary disorders. Numbers of MSC clinical trials for various immune/inflammatory pulmonary disorders as registered on the NIH Clinical Trial Registry website (https://ClinicalTrials.gov/) as accessed on May 2020. ARDS, acute respiratory distress syndrome; COPD, chronic obstructive pulmonary disease; COVID‐19, coronavirus disease 2019; IPF, idiopathic pulmonary fibrosis; MSC, mesenchymal stem cell
    2 PRECLINICAL DATA OF MSCT FOR NONINFECTIOUS PULMONARY IMMUNE/INFLAMMATORY DISORDERS

    The lungs are in direct exposure to the external environment, requiring constant immune surveillance by the native epithelial cells and resident alveolar macrophages for homeostasis and health.14 Immune dysregulation and inflammation, therefore, are common components in both infectious and many noninfectious pulmonary diseases, such as obstructive diseases including chronic obstructive pulmonary diseases (COPDs), in which injury is mainly mediated by cytotoxic T cells and neutrophils, and asthma, where type 2 helper T (Th2) lymphocytes and eosinophils are more predominant.15 In restrictive diseases such as idiopathic pulmonary fibrosis (IPF), resident alveolar macrophages appear critical in mediating the fibrosis.16 Despite differences in underlying processes for these diseases, MSCs have generally been shown to have beneficial effects in preclinical studies. Numerous rodent studies on COPD demonstrated that MSC infusion decrease inflammation and parenchymal damage17 with a number of reports showing MSC paracrine factors including epidermal growth factor, hepatocyte growth factor (HGF),18-21 vascular endothelial growth factor,17, 22, 23 and keratinocyte growth factor (KGF)24 to be involved (Figure 2, left box). Rodent studies of asthma demonstrate that MSC induction of CD4 regulatory T cells (Tregs), which are immunomodulatory CD4 cells, is critical in decreasing Th2 responses25, 26 and Th2 cytokines interleukin‐4 (IL‐4), IL‐5, and IL‐13 as well as immunoglobulin E levels to ameliorate disease severity27, 28; one recent study implicated MSC transfer of mitochondria in this process.29 Surprisingly, in these studies on asthma, no specific MSC paracrine factor was identified, but more recent reports implicate that MSC‐expressed microRNA and exosomes can improve disease outcome.30, 31

    FIGURE 2
    Open in figure viewerPowerPoint
    Mechanisms involved in MSC therapy for immune/inflammatory pulmonary disorders. Mechanisms reported in in vivo preclinical studies of MSC therapy for immune/inflammatory lung diseases of non‐infectious etiology—including asthma, IPF, and COPDs—and infectious etiology—including bacterial and/or LPS and viral infection and related ARDS. Detailed descriptions can be found in the text. Ang‐1, angiopoietin‐1; ARDS, acute respiratory distress syndrome; COPD, chronic obstructive lung disease; EGF, epidermal growth factor; EV, extracellular vesicles; HGF, hepatocyte growth factor; IL‐1RA, interleukin‐1 receptor antagonist; IPF, idiopathic pulmonary fibrosis; KGF, keratinocyte growth factor; LPS, lipopolysaccharide; MΦ, macrophage; miRs, microRNAs; mitoch, mitochondria; MSC, mesenchymal stem cell; OCR, oxygen consumption rate; PMNs, polymorphonuclear leukocytes/neutrophils; Th2, T helper type 2 lymphocytes; TNF‐α, tumor necrosis factor‐α; Treg, regulatory T lymphocytes; TSF‐6, TNF‐stimulated gene 6 protein; VEGF, vascular endothelial growth factor; WBCs, white blood cells
    Despite transforming growth factor‐β being a prominent paracrine factor of MSCs7, 32 and also known for strongly inducing fibrosis, MSCT appears to be efficacious even for fibrotic pulmonary conditions, as evidenced by the early study of MSC efficacy for bleomycin‐induced lung fibrosis, a preclinical disease model of IPF.10 MSC‐secreted IL‐1 receptor antagonist (IL‐1RA) was subsequently shown by the same group to be the paracrine factor involved.33 Moreover, MSCs may enhance resident lung bronchioalveolar stem cells to repair and regenerate healthy lung parenchyma.34 There has also been a growing number of reports using MSCs other than BMMSCs including umbilical cord MSCs (UCMSCs),35, 36 adipose‐derived MSCs (AdMSCs),37, 38 and placental MSCs.39 More recent studies also implicate MSC‐secreted factors of HGF and exosomes in mediating the antifibrotic effects.40-42
    3 PRECLINICAL DATA ON MSCT FOR BACTERIAL PNEUMONIA AND COMPLICATIONS

    The immunomodulatory effects of MSCs may lead one reasonably to avoid using these cells in infectious diseases, especially bacterial infectious since a strong effector response is required for clearance of these rapidly growing microorganisms. But surprisingly, the preclinical data have been rather consistent on MSCs actually enhancing antibacterial processes and decreasing overexuberant immune responses leading to pulmonary injury and acute respiratory distress syndrome (ARDS), a complication which is still associated with high morbidity and mortality.43 In rodent models of lung injury using either lipopolysaccharide, a component of Gram‐negative bacterial cell wall, or pneumonia induced by live bacteria (mainly Escherichia coli ), numerous studies have shown that MSCs, MSC‐conditioned medium, or MSC‐exosomes suppress inflammatory cell infiltration, decrease pro‐inflammatory cytokine levels including tumor necrosis factor‐α (TNF‐α and IL‐6, reverse pulmonary tissue damage, and improve survival through numerous paracrine factors including TNF‐stimulated gene 6 protein (TSG‐6),44 angiopoietin‐1,45, 46 LL‐37,47 lipocalin‐2,48 KGF,46, 48, 49 and microRNAs (Figure 2, middle box).50 Beneficial effects of MSCT in ex vivo human lung injury/bacterial infection models were seen as well.51 Similar to a report for asthma, mitochondrial transfer from MSCT—either directly or through exosomes—decreased pulmonary injury and improved macrophage energetics and antibacterial functions.52, 53 Other studies have also found that MSCs modulate macrophages from an M1 inflammatory phenotype to a more immunomodulatory M2 phenotype,54-56 as has been shown in nonpulmonary in vivo models.57, 58 It is surprising, however, that no in‐depth investigation of MSCs with neutrophils, the first‐line and critical leukocyte involved in bacterial clearance, was carried out any of these animal studies, since in vitro reports and one in vivo sepsis model have shown that MSCs preserve neutrophil viability and antibacterial functions.59-61 But overall, these preclinical studies of bacterial‐related lung injury/pneumonia consistently demonstrate that MSCT improves bacterial clearance and pulmonary tissue repair to impact survival.
    4 PRECLINICAL DATA OF MSCT FOR VIRAL PNEUMONIA AND COMPLICATIONS

    Overall, reports on MSCT for viral infections are relatively scarce. Most of the in vitro studies have been on the H1N1 influenza virus using MSCs from many different organisms, finding that MSCs can be infected with resultant cell lysis and death.62, 63 For in vivo studies, there are currently only six reports which have examined intravenous MSCT for viral pneumonitis/pneumonia, all focusing on influenza. In the first two studies on the subject, the outcome was negative. Both studies evaluated syngeneic murine as well as allogeneic human BMMSC treatment in mice infected with pulmonary mouse‐adapted H1N1 and/or swine H1N1, with no improvement in pulmonary inflammation or survival seen.64, 65 In the four other more recent reports, however, pulmonary inflammation was improved overall, with survival seen to improve in two out of the three studies which evaluated this endpoint; no specific factor was shown to be responsible (Figure 2, right box). Interestingly, all three reports which evaluated survival used non‐H1N1 subtypes. The only one beneficial report using H1N1 was a porcine study in which in vivo infection with swine H1N1 in 8‐week‐old pigs improved lung inflammation after intratracheal administration of syngeneic BMMSC extracellular vesicles; survival was not evaluated.66 A report using H9N2 found syngeneic BMMSC treatment suppressed infection and improved survival in infected mice,67 whereas another study using H5N1 found that conditioned medium and exosomes from human UCMSCs but not BMMSCs improved lung injury in infected mice partly due to two paracrine factors, angiopoietin‐1 and HGF, but survival was only minimally improved.68 A more elaborate murine study found that human BMMSCs reduce H5N1‐induced lung injury and survival but only in aged mice, partially through the paracrine factors of angiopoietin‐1 and KGF; the improved response in aged mice (8‐12 months old) but not young mice (6‐8 weeks old) to MSCT was in part attributed to more severe disease in these aged hosts, which may allow for exogenous MSCT to exert a more obvious benefit.69 This report also further discussed that the tissue reparative properties of MSCs may only be apparent with the severe damage caused by highly pathogenic influenza subtypes including H5N1, which is not seen with the less pathogenic H1N1 subtype. The collective results of these in vivo studies, while few, would seem to support this viewpoint. Such differences in the infecting viral subtype and host conditions are unfortunately rarely tackled in preclinical studies, but in clinical practice, differences in patient profiles, including age and sex, are known to highly influence disease progression and outcome—as is strikingly evident with COVID‐19, with higher positivity rates and worse outcome in men, and significantly higher mortality in the elderly and those with underlying chronic diseases.70, 71 Preclinical studies clearly should pay attention to such parameters for improved clinical use and outcome.
    One important point to keep in mind in interpreting these few in vivo MSCT‐virus reports is the existing data on MSC interactions with CD8 cells, which have a more critical role in viral infections.72 Surprisingly, very few reports have studied MSC‐CD8 interactions, in contrast to the several hundred reports on MSC‐CD4 interactions. While some reports show inconsistent MSC regulation of effector CD8 cell types),73, 74 most studies including data from our lab found that MSCs suppress CD8 T proliferation and cytotoxicity.75-77 Interestingly, one in vitro study found MSCs to inhibit proliferation of virus‐specific CD8 cells, leading the authors to comment that the use of MSCs may therefore compromise viral T‐cell immunity.78 However, in reports of clinical MSCT in GVHD patients, MSCT generally did not suppress viral‐specific T‐cell responses in patients, despite demonstrating strong in vitro immunomodulatory effects across CD4 and CD8 lymphocytes.79, 80 While existing in vitro and in vivo patient‐derived reports demonstrate discrepant information, the clinical studies offer some reassurance on the safety of MSCT but also reveal the gap in understanding the efficacious mechanisms—if any—of MSCT in patients. Clearly, preclinical in vivo studies with focus on elucidating specific populations of leukocyte‐MSC interactions during all steps of viral pneumonia/lung injury are urgently needed to provide better insight for clinical use.
    5 CURRENT CLINICAL TRIALS OF MSCT FOR PULMONARY IMMUNE/INFLAMMATORY DISEASES

    To date, 68 clinical studies using MSCs for pulmonary immune/inflammatory disorders have been registered (Figure 1 and Table S1 for detailed information on each trial). The most commonly targeted disease is COVID‐19 with 31 trials. MSCT for ARDS other than for COVID‐19 and COPD are the next two most commonly targeted diseases, with 10 trials each. There are six trials for IPF, and two trials for asthma; the rest of the nine trials include trials for cystic fibrosis, lung transplantation, pneumoconiosis, radiation‐caused injury, and unspecified lung injury.
    MSC sources used are broad (Table 1 and Figure 3), with the two most common types being BMMSCs (22 trials) and UCMSCs (20 trials), then AdMSCs with 12 trials. A few trials use either dental pulp MSCs (two trials), placenta‐MSCs (one trial), and olfactory mucosa MSCs (one trial); eight trials did not specify MSC source. As testament to the strong evidence for MSC immunomodulation, the majority of trials use allogeneic MSCs (49 trials, includes two trials using conditioned medium and exosomes), with all UCMSC trials being allogeneic; 10 trials use autologous sources which are either BMMSCs or AdMSCs, and nine trials were unspecified. There are only two trials using MSC‐derived products such as conditioned medium or exosomes rather than the cells themselves, and these products are derived from allogeneic AdMSCs or UCMSCs. Trials tend to be at early phases, with 30 being phase 1 trials, 17 being combined phase 1/2 trials, 14 trials in phase 2, 2 trials being a combined phase 2/3 trials, and 1 trial in phase 3; 3 trials are unspecified. As expected, the overwhelming majority of trials deliver MSCs intravenously (60 trials) but surprisingly, except for two trials which did not specified delivery method, the remaining six trials deliver MSCs to the lungs more directly, either through intratracheal/endobronchial delivery (three trials), intranasal delivery (one trial using UCMSC‐conditioned medium), aerosolized inhalation (one trial using AdMSC‐derived exosomes), or bronchial lavage (one trial) (Figure 4).
    TABLE 1. Cell source and trial phase of MSC clinical trials for immune/inflammatory lung diseases
    MSC source Total % Total no. No. of clinical trial phases
    1 ? 1 1&2 2 2&3 3
    2 Unspecified 11.8 8   5   2 1  
    3 Bone marrow 32.4 22   11 2 6 2 1
    4 Umbilical cord 29.4 20 3 7 8 2    
    5 Adipose tissue 17.6 12   3 5 4    
    6 Deciduous dental pulp 2.9 2   1 1      
    7 Placenta 1.5 1   1        
    8 Olfactory mucosa 1.5 1     1      
    9 MSC‐derived productsa 2.9 2   2        
    10 Total no. of clinical trial phases   68 3 30 17 14 3 1
    11 Total % of clinical trial phases     4.4 44.1 25.0 20.6 4.4 1.5
    • Abbreviation: MSC, mesenchymal stem cell.
    • a Exosomes or trophic factors collected from conditioned medium.


    FIGURE 3
    Open in figure viewerPowerPoint
    Sources of human MSCs used in immune/inflammatory lung disease clinical trials. Number of trials using different sources of human MSCs is shown, and whether sources are autologous (AUTO‐MSC), allogeneic (ALLO‐MSC), unspecified (UNSP‐MSC), and/or noncell exosomes/conditioned medium (MSC‐derived products). Data accessed on May 2020 from the NIH Clinical Trial website (https://ClinicalTrials.gov/). MSC, mesenchymal stem cell

    FIGURE 4
    Open in figure viewerPowerPoint
    Immune/inflammatory lung disease clinical trials. Number of trials using various methods of MSC administration. Data accessed on May 2020 from the NIH Clinical Trial website (https://ClinicalTrials.gov/). MSC, mesenchymal stem cell
    As most of these trials are just starting or still ongoing, there are very few published results available. An important publication was of the earliest MSCT trial for lung diseases which was started in 2008: a large phase 2 multicenter trial of 62 patients evaluating allogenic BMMSCs for COPD, where safety was demonstrated but efficacy was less clear.81 Several phase 1 trials using various tissue‐source allogeneic MSC infusions have published results: two trials on ARDS, one using AdMSCs82 and one using BMMSCs83; and two trials on IPF, one using placental‐derived MSCs84 and one using BMMSCs.85 All four reports demonstrated safety, but efficacy was weak at best. The strong evidence shown in preclinical animal studies has not yet been seen in these published human trials, and this may be a consequence of the very small patient numbers (approximately 8‐12 patients) in the majority of the trials, and also because many of these are just phase 1 trials, which are focused on safety rather than efficacy. Interestingly, a case report of allogeneic third party BMMSC treatment (multiple infusions) in two patients with severe ARDS from different causes demonstrated very good outcome and recovery for both patients, with inflammatory markers IL‐6 and interferon‐γ decreasing the day after initial MSC infusion in less critically ill patient.86 One setback occurred in this patient who had H1N1‐related ARDS, with development of bacterial pneumonia 5 days after MSC administration which responded to antibiotics treatment. While causation cannot be concluded from just this one case, the risk of infection in MSCT with its strong immunomodulation is nevertheless a possibility, as the authors also cautioned. Close follow‐up of patients undergoing MSC treatment especially in the critically ill—such as patients with ARDS—is clearly warranted.
    Very recently, an open‐label trial from China on MSCT for H7N9‐induced ARDS was published in February 2020,87 in which 17 critically ill patients were given multiple infusions of allogeneic (single‐donor) menstrual blood‐derived MSCs, with 44 patients in the control, nontreated group. Within these two groups, 14 patients (82%) in the MSC‐group were mechanically ventilated, compared to 31 patients (66%) in the control group. No adverse reaction to MSCT was reported, and results showed a lower mortality in the MSCT group with three deaths (82% survival) compared to 24 deaths (46% survival) in the control group. No immunological analyses were performed, but clinical lab data showed no difference in total white blood cell count, neutrophil count, or lymphocyte count at discharge between MSCT and control groups; however, procalcitonin, an inflammatory index marker, was significantly decreased in MSCT patients compared to control patients at discharge. While this data is encouraging, more detailed information on patient parameters, as well as the rationale for multiple infusions—not similarly done across MSCT patients—and immunological status at the time of these of the infusions should have been evaluated for a better understanding of MSC effects during ARDS and cytokine storm. Moreover, there is no information regarding the MSCs—which are from a less commonly used source—used in this trial: no references were cited and no characterization was performed. There is growing preclinical data suggesting that “not all MSCs are equal,” with different tissue sources of MSCs expressing different factors at varying levels as well as having functional differences, with a recent review finding BM and UCMSCs more effective than AdMSCs at reducing mortality in preclinical acute lung injury models.88 There has also been much discussion on whether the use of fresh vs cryopreserved MSCs would have therapeutic implications.89 In the six published clinical reports which all used allogeneic sources, two studies used freshly cultured MSCs84, 85 whereas the other four studies used previously cryopreserved MSCs81-83; no clear difference in efficacy could be easily discerned. In addition, the delivery method and dose of cells given, and whether multiple doses should be given, as well as cell numbers used are also critical parameters that likely impact efficacy, but all are difficult to test in human studies. Further accumulation of preclinical data investigating these parameters is urgently needed for better tailoring of specific tissue‐source MSCs, MSC preparation, as well as dosing regimens in clinical use to improve outcome.

    7 PERSPECTIVE ON MSCT FOR COVID ‐19


    Given the consistent beneficial outcomes of MSCT in the many preclinical in vivo studies of bacteria‐induced ARDS, tempered by the smaller data set of MSC‐virus interactions which include two clinical reports, any consideration of MSCT for COVID‐19 should be steered toward very severe cases where ARDS and an exuberant immune response is seen and not during the early period of viral infection or in mild cases. Like the previous two other novel corona viruses which have caused severe disease, COVID‐19‐ARDS is accompanied by cytokine storm and severe inflammation.94 The preclinical data on MSCT decreasing TNF‐α and IL‐6 levels, two pro‐inflammatory cytokines highly expressed during cytokine storm,95 have been quite consistent, and clinical MSCT data from the other immune/inflammatory diseases—particularly GVHD which have had long follow‐up periods—as well as the two recently published viral pneumonia trials, support the relative safety of MSCT even if efficacy may be more difficult to interpret. In light of the continued increase of COVID‐19 cases and deaths, there has been an avalanche of false information exploiting patients and the public during these uncertain times. The leading stem cell and cell therapy academic societies have all issued statements of caution against unproven stem cell treatments, emphasizing the importance of testing new possible therapies in clinical trials first.96, 97 The prospect of MSCT for COVID‐19, therefore, must be tempered with strict evaluation of patient inclusion/exclusion criteria as well as stringent ethical consideration to foremost protect patient safety.
 
watchlist Created with Sketch. Add MSB (ASX) to my watchlist
(20min delay)
Last
90.5¢
Change
0.020(2.26%)
Mkt cap ! $1.033B
Open High Low Value Volume
88.5¢ 95.0¢ 88.0¢ $5.610M 6.115M

Buyers (Bids)

No. Vol. Price($)
12 317774 90.0¢
 

Sellers (Offers)

Price($) Vol. No.
91.5¢ 23500 2
View Market Depth
Last trade - 16.10pm 06/08/2024 (20 minute delay) ?
MSB (ASX) Chart
arrow-down-2 Created with Sketch. arrow-down-2 Created with Sketch.