Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Virol ; 97(6): e0068923, 2023 Jun 29.
Article in English | MEDLINE | ID: covidwho-20245290

ABSTRACT

Goblet cells and their secreted mucus are important elements of the intestinal mucosal barrier, which allows host cells to resist invasion by intestinal pathogens. Porcine deltacoronavirus (PDCoV) is an emerging swine enteric virus that causes severe diarrhea in pigs and causes large economic losses to pork producers worldwide. To date, the molecular mechanisms by which PDCoV regulates the function and differentiation of goblet cells and disrupts the intestinal mucosal barrier remain to be determined. Here, we report that in newborn piglets, PDCoV infection disrupts the intestinal barrier: specifically, there is intestinal villus atrophy, crypt depth increases, and tight junctions are disrupted. There is also a significant reduction in the number of goblet cells and the expression of MUC-2. In vitro, using intestinal monolayer organoids, we found that PDCoV infection activates the Notch signaling pathway, resulting in upregulated expression of HES-1 and downregulated expression of ATOH-1 and thereby inhibiting the differentiation of intestinal stem cells into goblet cells. Our study shows that PDCoV infection activates the Notch signaling pathway to inhibit the differentiation of goblet cells and their mucus secretion, resulting in disruption of the intestinal mucosal barrier. IMPORTANCE The intestinal mucosal barrier, mainly secreted by the intestinal goblet cells, is a crucial first line of defense against pathogenic microorganisms. PDCoV regulates the function and differentiation of goblet cells, thereby disrupting the mucosal barrier; however, the mechanism by which PDCoV disrupts the barrier is not known. Here, we report that in vivo, PDCoV infection decreases villus length, increases crypt depth, and disrupts tight junctions. Moreover, PDCoV activates the Notch signaling pathway, inhibiting goblet cell differentiation and mucus secretion in vivo and in vitro. Thus, our results provide a novel insight into the mechanism underlying intestinal mucosal barrier dysfunction caused by coronavirus infection.


Subject(s)
Coronavirus Infections , Goblet Cells , Receptors, Notch , Swine Diseases , Animals , Coronavirus , Coronavirus Infections/pathology , Coronavirus Infections/veterinary , Goblet Cells/cytology , Signal Transduction , Swine , Swine Diseases/pathology , Swine Diseases/virology , Stem Cells/cytology , Cell Differentiation , Receptors, Notch/metabolism
2.
J Virol ; 96(18): e0096222, 2022 09 28.
Article in English | MEDLINE | ID: covidwho-2137410

ABSTRACT

Intestinal stem cells (ISCs) play an important role in tissue repair after injury. A recent report delineates the effect of transmissible gastroenteritis virus (TGEV) infection on the small intestine of recovered pigs. However, the mechanism behind the epithelium regeneration upon TGEV infection remains unclear. To address this, we established a TGEV infection model based on the porcine intestinal organoid monolayer. The results illustrated that the porcine intestinal organoid monolayer was susceptible to TGEV. In addition, the TGEV infection initiated the interferon and inflammatory responses following the loss of absorptive enterocytes and goblet cells. However, TGEV infection did not disturb epithelial integrity but induced the proliferation of ISCs. Furthermore, TGEV infection activated the Wnt/ß-catenin pathway by upregulating the accumulation and nuclear translocation of ß-catenin, as well as promoting the expression of Wnt target genes, such as C-myc, Cyclin D1, Mmp7, Lgr5, and Sox9, which were associated with the self-renewal of ISCs. Collectively, these data demonstrated that the TGEV infection activated the Wnt/ß-catenin pathway to promote the self-renewal of ISCs and resulted in intestinal epithelium regeneration. IMPORTANCE The intestinal epithelium is a physical barrier to enteric viruses and commensal bacteria. It plays an essential role in maintaining the balance between the host and intestinal microenvironment. In addition, intestinal stem cells (ISCs) are responsible for tissue repair after injury. Therefore, prompt self-renewal of intestinal epithelium will facilitate the rebuilding of the physical barrier and maintain gut health. In the manuscript, we found that the transmissible gastroenteritis virus (TGEV) infection did not disturb epithelial integrity but induced the proliferation of ISCs and facilitated epithelium regeneration. Detailed mechanism investigations revealed that the TGEV infection activated the Wnt/ß-catenin pathway to promote the self-renewal of ISCs and resulted in intestinal epithelium regeneration. These findings will contribute to understanding the mechanism of intestinal epithelial regeneration and reparation upon viral infection.


Subject(s)
Stem Cells , Transmissible gastroenteritis virus , Animals , Cyclin D1/metabolism , Interferons/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/virology , Matrix Metalloproteinase 7 , Stem Cells/cytology , Stem Cells/virology , Swine , Transmissible gastroenteritis virus/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
3.
Regen Med ; 15(7): 1833-1840, 2020 07.
Article in English | MEDLINE | ID: covidwho-1910922

ABSTRACT

Latest developments in the field of stem cell research and regenerative medicine compiled from publicly available information and press releases from nonacademic institutions in March 2020.


Subject(s)
Drug Industry , Regenerative Medicine/trends , Stem Cell Research , Stem Cells/cytology , Clinical Trials as Topic , Humans
4.
Cells ; 10(11)2021 10 25.
Article in English | MEDLINE | ID: covidwho-1480601

ABSTRACT

As the number of confirmed cases and deaths occurring from Coronavirus disease 2019 (COVID-19) surges worldwide, health experts are striving hard to fully comprehend the extent of damage caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although COVID-19 primarily manifests itself in the form of severe respiratory distress, it is also known to cause systemic damage to almost all major organs and organ systems within the body. In this review, we discuss the molecular mechanisms leading to multi-organ failure seen in COVID-19 patients. We also examine the potential of stem cell therapy in treating COVID-19 multi-organ failure cases.


Subject(s)
COVID-19/complications , COVID-19/therapy , Multiple Organ Failure/etiology , Multiple Organ Failure/therapy , Stem Cell Transplantation , COVID-19/immunology , Clinical Trials as Topic , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/therapy , Humans , Immunomodulation , Multiple Organ Failure/immunology , Regenerative Medicine , SARS-CoV-2/pathogenicity , Stem Cells/cytology , Stem Cells/immunology
5.
Biol Direct ; 16(1): 18, 2021 10 20.
Article in English | MEDLINE | ID: covidwho-1477451

ABSTRACT

Skeletal muscle has an extraordinary regenerative capacity reflecting the rapid activation and effective differentiation of muscle stem cells (MuSCs). In the course of muscle regeneration, MuSCs are reprogrammed by immune cells. In turn, MuSCs confer immune cells anti-inflammatory properties to resolve inflammation and facilitate tissue repair. Indeed, MuSCs can exert therapeutic effects on various degenerative and inflammatory disorders based on their immunoregulatory ability, including effects primed by interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α). At the molecular level, the tryptophan metabolites, kynurenine or kynurenic acid, produced by indoleamine 2,3-dioxygenase (IDO), augment the expression of TNF-stimulated gene 6 (TSG6) through the activation of the aryl hydrocarbon receptor (AHR). In addition, insulin growth factor 2 (IGF2) produced by MuSCs can endow maturing macrophages oxidative phosphorylation (OXPHOS)-dependent anti-inflammatory functions. Herein, we summarize the current understanding of the immunomodulatory characteristics of MuSCs and the issues related to their potential applications in pathological conditions, including COVID-19.


Subject(s)
COVID-19/therapy , Immune System/physiology , Muscles/physiology , Regeneration/physiology , Stem Cells/cytology , Animals , COVID-19/immunology , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cell Proliferation , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation , Insulin-Like Growth Factor II/metabolism , Interferon-gamma/metabolism , Kynurenic Acid/metabolism , Kynurenine/metabolism , Macrophages/metabolism , Mice , Muscles/metabolism , Oxidative Phosphorylation , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan/chemistry , Tumor Necrosis Factor-alpha/metabolism
6.
Cells ; 10(10)2021 09 24.
Article in English | MEDLINE | ID: covidwho-1438525

ABSTRACT

The objective of this review is to describe the evolution of lung tissue-derived diploid progenitor cell applications, ranging from historical biotechnological substrate functions for vaccine production and testing to current investigations around potential therapeutic use in respiratory tract regenerative medicine. Such cell types (e.g., MRC-5 or WI-38 sources) were extensively studied since the 1960s and have been continuously used over five decades as safe and sustainable industrial vaccine substrates. Recent research and development efforts around diploid progenitor lung cells (e.g., FE002-Lu or Walvax-2 sources) consist in qualification for potential use as optimal and renewed vaccine production substrates and, alternatively, for potential therapeutic applications in respiratory tract regenerative medicine. Potentially effective, safe, and sustainable cell therapy approaches for the management of inflammatory lung diseases or affections and related symptoms (e.g., COVID-19 patients and burn patient severe inhalation syndrome) using local homologous allogeneic cell-based or cell-derived product administrations are considered. Overall, lung tissue-derived progenitor cells isolated and produced under good manufacturing practices (GMP) may be used with high versatility. They can either act as key industrial platforms optimally conforming to specific pharmacopoeial requirements or as active pharmaceutical ingredients (API) for potentially effective promotion of lung tissue repair or regeneration.


Subject(s)
Biotechnology/methods , Diploidy , Lung/cytology , Regenerative Medicine/methods , Respiratory Tract Infections/therapy , Animals , Biological Specimen Banks , COVID-19 Vaccines , Cell Line , Cell- and Tissue-Based Therapy , History, 20th Century , History, 21st Century , Humans , Lung/physiology , Regeneration , Regenerative Medicine/history , SARS-CoV-2 , Stem Cell Transplantation , Stem Cells/cytology , Transplantation, Homologous
7.
Biomed Res Int ; 2021: 9915927, 2021.
Article in English | MEDLINE | ID: covidwho-1376538

ABSTRACT

BACKGROUND: The SARS-CoV-2 virus is the cause of the latest pandemic of the 21st century; it is responsible for the development of COVID-19. Within the multiple study models for both the biology and the treatment of SARS-CoV-2, the use of stem cells has been proposed because of their ability to increase the immune response and to repair tissue. Therefore, the objective of this review is to evaluate the role of stem cells against SARS-CoV-2 and COVID-19 in order to identify their potential as a study model and as a possible therapeutic source against tissue damage caused by this virus. Therefore, the following research question was established: What is the role of stem cells in the study of SARS-CoV-2 and the treatment of COVID-19? MATERIALS AND METHODS: A search was carried out in the electronic databases of PUBMED, Scopus, and ScienceDirect. The following keywords were used: "SARS-CoV-2," "COVID-19," and "STEM CELL," plus independent search strategies with the Boolean operators "OR" and "AND." The identified reports were those whose main objective was the study of stem cells in relation to SARS-CoV-2 or COVID-19. For the development of this study, the following inclusion criteria were taken into account: studies whose main objective was the study of stem cells in relation to SARS-CoV-2 or COVID-19 and clinical case studies, case reports, clinical trials, pilot studies, in vitro, or in vivo studies. For assessment of the risk of bias for in vitro studies, the SciRAP tool was used. The data collected for each type of study, clinical or in vitro, were analyzed with descriptive statistics using the SPSS V.22 program. RESULTS: Of the total of studies included (n = 39), 22 corresponded to in vitro investigations and 17 to human studies (clinical cases (n = 9), case series (n = 2), pilot clinical trials (n = 5), clinical trials (n = 1)). In vitro studies that induced pluripotent stem cells were the most used (n = 12), and in clinical studies, the umbilical stem cells derived were the most reported (n = 11). The mean age of the study subjects was 58.3 years. After the application of stem cell therapy, the follow-up period was 8 days minimum and 90 days maximum. Discussion. The mechanism by which the virus enters the cell is through protein "S," located on the surface of the membrane, by recognizing the ACE2 receptor located on the target cell. The evidence that the expression of ACE2 and TMPRSS2 in stem cells indicates that stem cells from bone marrow and amniotic fluid have very little expression. This shows that stem cell has a low risk of infection with SARS-CoV-2. CONCLUSION: The use of stem cells is a highly relevant therapeutic option. It has been shown in both in vitro studies and clinical trials that it counteracts the excessive secretion of cytokines. There are even more studies that focus on long-term follow-up; thus, the potential for major side effects can be analyzed more clearly. Finally, the ethical use of stem cells from fetal or infant origin needs to be regulated. The study was registered in PROSPERO (no. CRD42021229038). The limitations of the study were because of the methodology employed, the sample was not very large, and the follow-up period of the clinical studies was relatively short.


Subject(s)
COVID-19/pathology , COVID-19/therapy , SARS-CoV-2/isolation & purification , Stem Cell Transplantation/methods , Stem Cells/cytology , COVID-19/metabolism , COVID-19/virology , Clinical Trials as Topic , Humans , Stem Cells/pathology
9.
Stem Cell Res Ther ; 12(1): 155, 2021 03 01.
Article in English | MEDLINE | ID: covidwho-1112452

ABSTRACT

A new coronavirus respiratory disease (COVID-19) caused by the SARS-CoV-2 virus, surprised the entire world, producing social, economic, and health problems. The COVID-19 triggers a lung infection with a multiple proinflammatory cytokine storm in severe patients. Without effective and safe treatments, COVID-19 has killed thousands of people, becoming a pandemic. Stem cells have been suggested as a therapy for lung-related diseases. In particular, mesenchymal stem cells (MSCs) have been successfully tested in some clinical trials in patients with COVID-19. The encouraging results positioned MSCs as a possible cell therapy for COVID-19. The amniotic membrane from the human placenta at term is a valuable stem cell source, including human amniotic epithelial cells (hAECs) and human mesenchymal stromal cells (hAMSCs). Interestingly, amnion cells have immunoregulatory, regenerative, and anti-inflammatory properties. Moreover, hAECs and hAMSCs have been used both in preclinical studies and in clinical trials against respiratory diseases. They have reduced the inflammatory response and restored the pulmonary tissue architecture in lung injury in vivo models. Here, we review the existing data about the stem cells use for COVID-19 treatment, including the ongoing clinical trials. We also consider the non-cellular therapies that are being applied. Finally, we discuss the human amniotic membrane cells use in patients who suffer from immune/inflammatory lung diseases and hypothesize their possible use as a successful treatment against COVID-19.


Subject(s)
Amnion/cytology , COVID-19/therapy , Mesenchymal Stem Cell Transplantation , Stem Cells/cytology , Clinical Trials as Topic , Female , Humans , Inflammation , Mesenchymal Stem Cells/cytology , Placenta/cytology , Pregnancy , Risk
10.
Stem Cell Res Ther ; 12(1): 1, 2021 01 04.
Article in English | MEDLINE | ID: covidwho-1007143

ABSTRACT

Adipose-derived stem cells (ADSCs) have raised big interest in therapeutic applications in regenerative medicine and appear to fulfill the criteria for a successful cell therapy. Their low immunogenicity and their ability to self-renew, to differentiate into different tissue-specific progenitors, to migrate into damaged sites, and to act through autocrine and paracrine pathways have been altogether testified as the main mechanisms whereby cell repair and regeneration occur. The absence of standardization protocols in cell management within laboratories or facilities added to the new technologies improved at patient's bedside and the discrepancies in cell outcomes and engraftment increase the limitations on their widespread use by balancing their real benefit versus the patient safety and security. Also, comparisons across pooled patients are particularly difficult in the fact that multiple medical devices are used and there is absence of harmonized assessment assays despite meeting regulations agencies and efficient GMP protocols. Moreover, the emergence of the COVID-19 breakdown added to the complexity of implementing standardization. Cell- and tissue-based therapies are completely dependent on the biological manifestations and parameters associated to and induced by this virus where the scope is still unknown. The initial flow chart identified for stem cell therapies should be reformulated and updated to overcome patient infection and avoid significant variability, thus enabling more patient safety and therapeutic efficiency. The aim of this work is to highlight the major guidelines and differences in ADSC processing meeting the current good manufacturing practices (cGMP) and the cellular therapy-related policies. Specific insights on standardization of ADSCs proceeding at different check points are also presented as a setup for the cord blood and bone marrow.


Subject(s)
Adipose Tissue/cytology , COVID-19 , Cell Separation/standards , Stem Cell Transplantation/standards , Stem Cells/cytology , Humans
11.
Genes (Basel) ; 12(1)2020 12 23.
Article in English | MEDLINE | ID: covidwho-1000248

ABSTRACT

The placenta is a temporary organ that is discarded after birth and is one of the most promising sources of various cells and tissues for use in regenerative medicine and tissue engineering, both in experimental and clinical settings. The placenta has unique, intrinsic features because it plays many roles during gestation: it is formed by cells from two individuals (mother and fetus), contributes to the development and growth of an allogeneic fetus, and has two independent and interacting circulatory systems. Different stem and progenitor cell types can be isolated from the different perinatal tissues making them particularly interesting candidates for use in cell therapy and regenerative medicine. The primary source of perinatal stem cells is cord blood. Cord blood has been a well-known source of hematopoietic stem/progenitor cells since 1974. Biobanked cord blood has been used to treat different hematological and immunological disorders for over 30 years. Other perinatal tissues that are routinely discarded as medical waste contain non-hematopoietic cells with potential therapeutic value. Indeed, in advanced perinatal cell therapy trials, mesenchymal stromal cells are the most commonly used. Here, we review one by one the different perinatal tissues and the different perinatal stem cells isolated with their phenotypical characteristics and the preclinical uses of these cells in numerous pathologies. An overview of clinical applications of perinatal derived cells is also described with special emphasis on the clinical trials being carried out to treat COVID19 pneumonia. Furthermore, we describe the use of new technologies in the field of perinatal stem cells and the future directions and challenges of this fascinating and rapidly progressing field of perinatal cells and regenerative medicine.


Subject(s)
COVID-19/therapy , Placenta/cytology , SARS-CoV-2 , Stem Cell Transplantation/trends , Stem Cells/cytology , Amniotic Fluid/cytology , Clinical Trials as Topic , Cord Blood Stem Cell Transplantation/methods , Cord Blood Stem Cell Transplantation/trends , Cytokine Release Syndrome/therapy , Drug Carriers , Extraembryonic Membranes/cytology , Female , Forecasting , Hematopoietic Stem Cells/cytology , Humans , Lung/pathology , Macrophage Activation , Mesenchymal Stem Cells/cytology , Nanoparticles , Pregnancy , Preservation, Biological , Regenerative Medicine/methods , Stem Cell Transplantation/methods , Stem Cells/immunology
12.
Biochem Biophys Res Commun ; 533(4): 1276-1282, 2020 12 17.
Article in English | MEDLINE | ID: covidwho-885206

ABSTRACT

BACKGROUND: The whole world was hit hard by the coronavirus disease-19 (COVID-19). Given that angiotensin I converting enzyme 2 (ACE2) is the viral entry molecule, understanding ACE2 has become a major focus of current COVID-19 research. ACE2 is highly expressed in the gut, but its role has not been fully understood and thus COVID-19 treatments intending to downregulate ACE2 level may cause untoward side effects. Gaining insight into the functions of ACE2 in gut homeostasis therefore merits closer examination, and is beneficial to find potential therapeutic alternatives for COVID-19. METHODS: We took advantage of Ace2 knockout out mice and isolated intestinal organoids to examine the role of ACE2 in intestinal stemness. Inflammatory bowel disease (IBD) mouse model was established by 4% dextran sodium sulfate. LGR5 and KI67 levels were quantitated to reflect the virtue of intestinal stem cells (ISCs). FITC-dextran 4 (FD-4) assay was used to assess intestinal barrier function. RESULTS: Western blotting identified the expression of ACE2 in colon, which was consistent with the results of immunofluorescence and RT-PCR. Moreover, Ace2-/- organoids showed decreased LRG5 and KI67 levels, and elevated calcium concentration. Furthermore, the permeability of ace2-/- organoids was markedly increased compared with ace2+/+ organoids. Collectively, ace2-/- mice were more susceptible than ace2+/+ mice to IBD, including earlier bloody stool, undermined intestinal architecture and more pronounced weight loss. CONCLUSIONS: Our data reveal that ACE2 contributes to the proliferation of intestinal stem cells and hence orchestrates the mucosal homeostasis.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Epithelium/metabolism , Angiotensin-Converting Enzyme 2/deficiency , Animals , Calcium/metabolism , Cell Membrane Permeability , Inflammatory Bowel Diseases/enzymology , Inflammatory Bowel Diseases/pathology , Intestines/pathology , Mice, Inbred C57BL , Mice, Knockout , Organoids/metabolism , Stem Cells/cytology , Stem Cells/metabolism
13.
Sci Rep ; 10(1): 17772, 2020 10 20.
Article in English | MEDLINE | ID: covidwho-882925

ABSTRACT

SARS-CoV-2 can infiltrate the lower respiratory tract, resulting in severe respiratory failure and a high death rate. Normally, the airway and alveolar epithelium can be rapidly reconstituted by multipotent stem cells after episodes of infection. Here, we analyzed published RNA-seq datasets and demonstrated that cells of four different lung epithelial stem cell types express SARS-CoV-2 entry factors, including Ace2. Thus, stem cells can be potentially infected by SARS-CoV-2, which may lead to defects in regeneration capacity partially accounting for the severity of SARS-CoV-2 infection and its consequences.


Subject(s)
Betacoronavirus/physiology , Viral Proteins/metabolism , Virus Internalization , Angiotensin-Converting Enzyme 2 , Betacoronavirus/isolation & purification , COVID-19 , Cell Differentiation , Cell Line , Coronavirus Infections/pathology , Coronavirus Infections/virology , Humans , Lung/cytology , Pandemics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , SARS-CoV-2 , Serine Endopeptidases/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/virology , Viral Proteins/genetics
14.
Stem Cell Reports ; 15(5): 1015-1025, 2020 11 10.
Article in English | MEDLINE | ID: covidwho-864993

ABSTRACT

Despite the central importance of the respiratory system, the exact mechanisms governing lung repair after severe injury remain unclear. The notion that alveolar type 2 cells (AT2s) self-renew and differentiate into alveolar type 1 cells (AT1s) does not fully encompass scenarios where these progenitors are severely affected by disease, e.g., H1N1 influenza or SARS-CoV-2 (COVID-19). Intrapulmonary p63+ progenitor cells, a rare cell type in mice but potentially encompassing more numerous classic basal cells in humans, are activated in such severe injury settings, proliferating and migrating into the injured alveolar parenchyma, providing a short-term "emergency" benefit. While the fate of these cells is controversial, most studies indicate that they represent a maladaptive repair pathway with a fate restriction toward airway cell types, rarely differentiating into AT2 or AT1 cells. Here, we discuss the role of intrapulmonary basal-like p63+ cells in alveolar regeneration and suggest a unified model to guide future studies.


Subject(s)
Lung/physiology , Regeneration , Stem Cells/metabolism , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/metabolism , COVID-19/pathology , COVID-19/virology , Cell Differentiation , Humans , Lung/metabolism , Lung Diseases/pathology , Lung Diseases/therapy , Lung Diseases/virology , SARS-CoV-2/isolation & purification , Stem Cell Transplantation , Stem Cells/cytology
15.
Basic Res Cardiol ; 115(4): 36, 2020 05 12.
Article in English | MEDLINE | ID: covidwho-245247

ABSTRACT

There are no definitive therapies for patients with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. Therefore, new therapeutic strategies are needed to improve clinical outcomes, particularly in patients with severe disease. This case series explores the safety and effectiveness of intravenous allogeneic cardiosphere-derived cells (CDCs), formulated as CAP-1002, in critically ill patients with confirmed coronavirus disease 2019 (COVID-19). Adverse reactions to CAP-1002, clinical status on the World Health Organization (WHO) ordinal scale, and changes in pro-inflammatory biomarkers and leukocyte counts were analyzed. All patients (n = 6; age range 19-75 years, 1 female) required ventilatory support (invasive mechanical ventilation, n = 5) with PaO2/FiO2 ranging from 69 to 198. No adverse events related to CAP-1002 administration were observed. Four patients (67%) were weaned from respiratory support and discharged from the hospital. One patient remains mechanically ventilated as of April 28th, 2020; all survive. A contemporaneous control group of critically ill COVID-19 patients (n = 34) at our institution showed 18% overall mortality at a similar stage of hospitalization. Ferritin was elevated in all patients at baseline (range of all patients 605.43-2991.52 ng/ml) and decreased in 5/6 patients (range of all patients 252.89-1029.90 ng/ml). Absolute lymphocyte counts were low in 5/6 patients at baseline (range 0.26-0.82 × 103/µl) but had increased in three of these five patients at last follow-up (range 0.23-1.02 × 103/µl). In this series of six critically ill COVID-19 patients, intravenous infusion of CAP-1002 was well tolerated and associated with resolution of critical illness in 4 patients. This series demonstrates the apparent safety of CAP-1002 in COVID-19. While this initial experience is promising, efficacy will need to be further assessed in a randomized controlled trial.


Subject(s)
Cell- and Tissue-Based Therapy , Compassionate Use Trials , Coronavirus Infections/therapy , Myocardium/cytology , Pneumonia, Viral/therapy , Stem Cells/cytology , Aged , Betacoronavirus , Biomarkers/blood , COVID-19 , Critical Illness/therapy , Female , Ferritins/blood , Humans , Infusions, Intravenous , Los Angeles , Lymphocyte Count , Male , Middle Aged , Pandemics , SARS-CoV-2 , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL