1
|
Trigo CM, Rodrigues JS, Camões SP, Solá S, Miranda JP. Mesenchymal stem cell secretome for regenerative medicine: Where do we stand? J Adv Res 2025; 70:103-124. [PMID: 38729561 PMCID: PMC11976416 DOI: 10.1016/j.jare.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/27/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC)-based therapies have yielded beneficial effects in a broad range of preclinical models and clinical trials for human diseases. In the context of MSC transplantation, it is widely recognized that the main mechanism for the regenerative potential of MSCs is not their differentiation, with in vivo data revealing transient and low engraftment rates. Instead, MSCs therapeutic effects are mainly attributed to its secretome, i.e., paracrine factors secreted by these cells, further offering a more attractive and innovative approach due to the effectiveness and safety of a cell-free product. AIM OF REVIEW In this review, we will discuss the potential benefits of MSC-derived secretome in regenerative medicine with particular focus on respiratory, hepatic, and neurological diseases. Both free and vesicular factors of MSC secretome will be detailed. We will also address novel potential strategies capable of improving their healing potential, namely by delivering important regenerative molecules according to specific diseases and tissue needs, as well as non-clinical and clinical studies that allow us to dissect their mechanisms of action. KEY SCIENTIFIC CONCEPTS OF REVIEW MSC-derived secretome includes both soluble and non-soluble factors, organized in extracellular vesicles (EVs). Importantly, besides depending on the cell origin, the characteristics and therapeutic potential of MSC secretome is deeply influenced by external stimuli, highlighting the possibility of optimizing their characteristics through preconditioning approaches. Nevertheless, the clarity around their mechanisms of action remains ambiguous, whereas the need for standardized procedures for the successful translation of those products to the clinics urges.
Collapse
Affiliation(s)
- Catarina M Trigo
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana S Rodrigues
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sérgio P Camões
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Susana Solá
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana P Miranda
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
2
|
Ghanem R, Youf R, Haute T, Buin X, Riool M, Pourchez J, Montier T. The (re)emergence of aerosol delivery: Treatment of pulmonary diseases and its clinical challenges. J Control Release 2025; 379:421-439. [PMID: 39800241 DOI: 10.1016/j.jconrel.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Aerosol delivery represents a rapid and non-invasive way to directly reach the lungs while escaping the hepatic first-pass effect. The development of pulmonary drugs for respiratory diseases such as cystic fibrosis, lung infections, pulmonary fibrosis or lung cancer requires an enhanced understanding of the relationships between the natural physiology of the respiratory system and the pathophysiology of these conditions. This knowledge is crucial to better predict and thereby control drug deposition. Moreover, aerosol administration faces several challenges, including the pulmonary tract, immune system, mucociliary clearance, the presence of fluid on the airway surfaces, and, in some cases, bacterial colonisation. Each of them directly influences on the bioavailability of the active molecule. In addition to these challenges, particle size and the device used to administer the treatment are critical factors that can significantly impact the biodistribution of the drugs. Nanoparticles are very promising in the development of new formulations for aerosol drug delivery, as they can be fine-tuned to reach the entire pulmonary tract and overcome the difficulties encountered along the way. However, to properly assess drug delivery, preclinical studies need to be more thorough to efficiently enhance drug delivery.
Collapse
Affiliation(s)
- Rosy Ghanem
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200, Brest, France; CHU de Brest, Service de Génétique Médicale et de Biologie de la Reproduction, F-29200 Brest, France
| | - Raphaëlle Youf
- Department of Trauma Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Tanguy Haute
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - Xavier Buin
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - Martijn Riool
- Department of Trauma Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Jérémie Pourchez
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, F - 42023 Saint-Etienne, France
| | - Tristan Montier
- Univ Brest, Inserm, EFS, UMR 1078, GGB, F-29200, Brest, France; CHU de Brest, Service de Génétique Médicale et de Biologie de la Reproduction, F-29200 Brest, France.
| |
Collapse
|
3
|
Wang Z, Zhang D, Liu N, Wang J, Zhang Q, Zheng S, Zhang Z, Zhang W. A review on recent advances in polymeric microneedle loading cells: Design strategies, fabrication technologies, transdermal application and challenges. Int J Biol Macromol 2025; 297:138885. [PMID: 39719236 DOI: 10.1016/j.ijbiomac.2024.138885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
Microneedle systems (MNs) loading living cells are a powerful platform to treat various previously incurable diseases in the era of precision medicine. Herein, an overview of recent advances in MN-based strategies for cell delivery is summarized, including material selection, design of morphological structures, and processing methods. We also systematically outlined the law of microstructural design relative to the structure-effective/function relationship in transdermal delivery or precision medicine and the design principles of cell microneedle (CMN). Furthermore, the representative works of precision treatments focusing on inflammatory skin diseases were tracked and discussed using CMN. Indeed, it highlights a practical path to solving the dilemma of cell therapy and raising the hope of precision medicine. However, there are still some challenges in developing CMN since they need multi-dimensional comprehensive properties, including mechanical properties, cell viability preservation, release, therapeutic effect, etc. The manuscript could provide insights into developing an innovative fit-to-purpose vehicle in cell therapy for interested researchers.
Collapse
Affiliation(s)
- Zixin Wang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Dongmei Zhang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China; Collaborative Innovation Center of Fragrance Flavour and Cosmetics, School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China.
| | - Ningning Liu
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Jiayi Wang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Qianjie Zhang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China; Collaborative Innovation Center of Fragrance Flavour and Cosmetics, School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Shilian Zheng
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Zijia Zhang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Wanping Zhang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China; Collaborative Innovation Center of Fragrance Flavour and Cosmetics, School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China.
| |
Collapse
|
4
|
Tessier S, Ploszay VK, Robitaille C, Vaghasiya J, Halayko AJ, Chartrand L. Optimization of Albuterol Delivery via Anesthesia Bag in Pediatric Critical Care. Pharmaceutics 2025; 17:218. [PMID: 40006585 PMCID: PMC11858898 DOI: 10.3390/pharmaceutics17020218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Aerosolized medications are common practice for mechanically ventilated pediatric patients. Infants often receive nebulized medications via hand ventilation using an anesthesia bag, but evidence on optimal aerosol delivery with this method is limited. For this study, various configurations of the Mapleson breathing circuit were tested to optimize albuterol delivery to a simulated pediatric model. Methods: Using a simulated pediatric lung model (ASL 5000) with the semi-open Mapleson anesthesia circuit, 2.5 mg/3 mL of albuterol sulfate solution was nebulized to a viral/bacterial filter (Respiguard 202). Four models were compared with varying fresh gas flows (FGFs), small-volume nebulizer (SVN) placements, and adjusting dead space. Five Registered Respiratory Therapists (RRTs) bagged the aerosol into a collection filter following defined ventilation parameters. Each model was tested in random order to avoid fatigue bias. Albuterol concentrations eluted from in-line filters were measured by spectrophotometry (absorbance at 276 nm). Results: No inter-user variability was observed among the RRTs. Significant differences in albuterol recovered were noted between models (One Way ANOVA, Tukey's post hoc, n = 5). Model 4, with the nebulizer closest to the collecting filter, recovered 21.77 ± 1.89% of albuterol. The standard clinical model was the least effective, with only 0.10 ± 0.17% albuterol recovery. Conclusions: Modifying the anesthesia breathing circuit significantly improved aerosol drug delivery efficiency. Our findings suggest that current clinical practices for nebulized drug delivery are inefficient and can be markedly improved with simple adjustments in nebulizer positioning and gas flow within the circuit.
Collapse
Affiliation(s)
- Sébastien Tessier
- Children’s Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P4, Canada;
| | | | | | - Jigneshkumar Vaghasiya
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada; (J.V.); (A.J.H.)
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Biology of Breathing Group, Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Andrew J. Halayko
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada; (J.V.); (A.J.H.)
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Biology of Breathing Group, Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Louise Chartrand
- Department of Respiratory Therapy, College of Rehabilitation Sciences, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| |
Collapse
|
5
|
Wan R, Liu Y, Yan J, Lin J. Cell therapy: A beacon of hope in the battle against pulmonary fibrosis. FASEB J 2025; 39:e70356. [PMID: 39873972 DOI: 10.1096/fj.202402790r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/28/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
Pulmonary fibrosis (PF) is a chronic and progressive interstitial lung disease characterized by abnormal activation of myofibroblasts and pathological remodeling of the extracellular matrix, with a poor prognosis and limited treatment options. Lung transplantation is currently the only approach that can extend the life expectancy of patients; however, its applicability is severely restricted due to donor shortages and patient-specific limitations. Therefore, the search for novel therapeutic strategies is imperative. In recent years, stem cells have shown great promise in the field of regenerative medicine due to their self-renewal capacity and multidirectional differentiation potential, and a growing body of literature supports the efficacy of stem cell therapy in PF treatment. This paper systematically summarizes the research progress of various stem cell types in the treatment of PF. Furthermore, it discusses the primary methods and clinical outcomes of stem cell therapy in PF, based on both preclinical and clinical data. Finally, the current challenges and key factors to consider in stem cell therapy for PF are objectively analyzed, and future directions for improving this therapy are proposed, providing new insights and references for the clinical treatment of PF patients.
Collapse
Affiliation(s)
- Ruyan Wan
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Yanli Liu
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Jingwen Yan
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
6
|
Chen T, Ellman DG, Fang S, Bak ST, Nørgård MØ, Svenningsen P, Andersen DC. Transfer of cardiomyocyte-derived extracellular vesicles to neighboring cardiac cells requires tunneling nanotubes during heart development. Theranostics 2024; 14:3843-3858. [PMID: 38994028 PMCID: PMC11234280 DOI: 10.7150/thno.91604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/26/2024] [Indexed: 07/13/2024] Open
Abstract
Rationale: Extracellular vesicles (EVs) are thought to mediate intercellular communication during development and disease. Yet, biological insight to intercellular EV transfer remains elusive, also in the heart, and is technically challenging to demonstrate. Here, we aimed to investigate biological transfer of cardiomyocyte-derived EVs in the neonatal heart. Methods: We exploited CD9 as a marker of EVs, and generated two lines of cardiomyocyte specific EV reporter mice: Tnnt2-Cre; double-floxed inverted CD9/EGFP and αMHC-MerCreMer; double-floxed inverted CD9/EGFP. The two mouse lines were utilized to determine whether developing cardiomyocytes transfer EVs to other cardiac cells (non-myocytes and cardiomyocytes) in vitro and in vivo and investigate the intercellular transport pathway of cardiomyocyte-derived EVs. Results: Genetic tagging of cardiomyocytes was confirmed in both reporter mouse lines and proof of concept in the postnatal heart showed that, a fraction of EGFP+/MYH1- non-myocytes exist firmly demonstrating in vivo cardiomyocyte-derived EV transfer. However, two sets of direct and indirect EGFP +/- cardiac cell co-cultures showed that cardiomyocyte-derived EGFP+ EV transfer requires cell-cell contact and that uptake of EGFP+ EVs from the medium is limited. The same was observed when co-cultiring with mouse macrophages. Further mechanistic insight showed that cardiomyocyte EV transfer occurs through type I tunneling nanotubes. Conclusion: While the current notion assumes that EVs are transferred through secretion to the surroundings, our data show that cardiomyocyte-derived EV transfer in the developing heart occurs through nanotubes between neighboring cells. Whether these data are fundamental and relate to adult hearts and other organs remains to be determined, but they imply that the normal developmental process of EV transfer goes through cell-cell contact rather than through the extracellular compartment.
Collapse
Affiliation(s)
- Ting Chen
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Urology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ditte Gry Ellman
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Shu Fang
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Sara Thornby Bak
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mikkel Ørnfeldt Nørgård
- Department of Molecular Medicine, Unit of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Per Svenningsen
- Department of Molecular Medicine, Unit of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Ditte Caroline Andersen
- Andersen Group, Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
7
|
Grigoropoulos I, Tsioulos G, Kastrissianakis A, Shapira S, Green O, Rapti V, Tsakona M, Konstantinos T, Savva A, Kavatha D, Boumpas D, Syrigos K, Xynogalas I, Leontis K, Ntousopoulos V, Sakka V, Sardelis Z, Fotiadis A, Vlassi L, Kontogianni C, Levounets A, Poulakou G, Gaga M, MacLoughlin R, Stebbing J, Arber N, Antoniadou A, Tsiodras S. The safety and potential efficacy of exosomes overexpressing CD24 (EXO-CD24) in mild-moderate COVID-19 related ARDS. Respir Res 2024; 25:151. [PMID: 38561798 PMCID: PMC10983648 DOI: 10.1186/s12931-024-02759-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION EXO-CD24 are exosomes genetically manipulated to over-express Cluster of Differentiation (CD) 24. It consists of two breakthrough technologies: CD24, the drug, as a novel immunomodulator that is smarter than steroids without any side effects, and exosomes as the ideal natural drug carrier. METHODS A randomized, single blind, dose-finding phase IIb trial in hospitalized patients with mild to moderate Coronavirus disease 2019 (COVID-19) related Acute Respiratory Distress Syndrome (ARDS) was carried out in two medical centers in Athens. Patients received either 109 or 1010 exosome particles of EXO-CD24, daily, for five consecutive days and monitored for 28 days. Efficacy was assessed at day 7 among 91 patients who underwent randomization. The outcome was also compared in a post-hoc analysis with an income control group (n = 202) that fit the inclusion and exclusion criteria. RESULTS The mean age was 49.4 (± 13.2) years and 74.4% were male. By day 7, 83.7% showed improved respiratory signs and 64% had better oxygen saturation (SpO2) (p < 0.05). There were significant reductions in all inflammatory markers, most notably in C-reactive protein (CRP), lactate dehydrogenase (LDH), ferritin, fibrinogen and an array of cytokines. Conversely, levels of the anti-inflammatory cytokine Interleukin-10 (IL-10) were increased (p < 0.05). Of all the documented adverse events, none were considered treatment related. No drug-drug interactions were noted. Two patients succumbed to COVID-19. Post-hoc analysis revealed that EXO-CD24 patients exhibited greater improvements in clinical and laboratory outcomes compared to an observational income control group. CONCLUSIONS EXO-CD24 presents a promising therapeutic approach for hyper-inflammatory state and in particular ARDS. Its unique combination of exosomes, as a drug carrier, and CD24, as an immunomodulator, coupled with inhalation administration, warrants further investigation in a larger, international, randomized, quadri-blind trial against a placebo.
Collapse
Affiliation(s)
- Ioannis Grigoropoulos
- 4, Department of Internal Medicine, University General Hospital Attikon, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Georgios Tsioulos
- 4, Department of Internal Medicine, University General Hospital Attikon, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Artemis Kastrissianakis
- 4, Department of Internal Medicine, University General Hospital Attikon, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Shiran Shapira
- Integrated Cancer Prevention Center, Tel-Aviv Sourasky Medical Center, 6 Weizmann St., 6423906, Tel Aviv, Israel
- Department of Molecular Genetic and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orr Green
- Integrated Cancer Prevention Center, Tel-Aviv Sourasky Medical Center, 6 Weizmann St., 6423906, Tel Aviv, Israel
| | - Vasiliki Rapti
- 3, Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, "Sotiria" General Hospital, 11527, Athens, Greece
| | - Maria Tsakona
- 4, Department of Internal Medicine, University General Hospital Attikon, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Thomas Konstantinos
- 4, Department of Internal Medicine, University General Hospital Attikon, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Athina Savva
- 4, Department of Internal Medicine, University General Hospital Attikon, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Dimitra Kavatha
- 4, Department of Internal Medicine, University General Hospital Attikon, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Dimitrios Boumpas
- 4, Department of Internal Medicine, University General Hospital Attikon, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Konstantinos Syrigos
- 3, Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, "Sotiria" General Hospital, 11527, Athens, Greece
| | - Ioannis Xynogalas
- 3, Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, "Sotiria" General Hospital, 11527, Athens, Greece
| | - Konstantinos Leontis
- 3, Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, "Sotiria" General Hospital, 11527, Athens, Greece
| | - Vasileios Ntousopoulos
- 3, Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, "Sotiria" General Hospital, 11527, Athens, Greece
| | - Vissaria Sakka
- 3, Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, "Sotiria" General Hospital, 11527, Athens, Greece
| | - Zafeiris Sardelis
- 7, Respiratory Medicine Department "Sotiria" General Hospital, 11527, Athens, Greece
| | - Andreas Fotiadis
- 7, Respiratory Medicine Department "Sotiria" General Hospital, 11527, Athens, Greece
| | - Lamprini Vlassi
- 7, Respiratory Medicine Department "Sotiria" General Hospital, 11527, Athens, Greece
| | - Chrysoula Kontogianni
- 7, Respiratory Medicine Department "Sotiria" General Hospital, 11527, Athens, Greece
| | - Anastasia Levounets
- 7, Respiratory Medicine Department "Sotiria" General Hospital, 11527, Athens, Greece
| | - Garyfalia Poulakou
- 3, Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, "Sotiria" General Hospital, 11527, Athens, Greece
| | - Mina Gaga
- 7, Respiratory Medicine Department "Sotiria" General Hospital, 11527, Athens, Greece
| | - Ronan MacLoughlin
- R&D Science & Emerging Technologies, Aerogen Ltd., IDA Business Park, Dangan, Galway, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons, Dublin, Ireland
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, Dublin, Ireland
| | - Justin Stebbing
- Department of Surgery and Cancer, Anglia Ruskin University, London, UK
- Department of Life Sciences, ARU, Cambridge, UK
| | - Nadir Arber
- Integrated Cancer Prevention Center, Tel-Aviv Sourasky Medical Center, 6 Weizmann St., 6423906, Tel Aviv, Israel.
- Department of Molecular Genetic and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Anastasia Antoniadou
- 4, Department of Internal Medicine, University General Hospital Attikon, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| | - Sotirios Tsiodras
- 4, Department of Internal Medicine, University General Hospital Attikon, Medical School, National and Kapodistrian University of Athens, 12462, Athens, Greece
| |
Collapse
|
8
|
Shapira S, Schwartz R, Tsiodras S, Bar-Shai A, Melloul A, Borsekofsky S, Peer M, Adi N, MacLoughlin R, Arber N. Inhaled CD24-Enriched Exosomes (EXO-CD24) as a Novel Immune Modulator in Respiratory Disease. Int J Mol Sci 2023; 25:77. [PMID: 38203250 PMCID: PMC10779124 DOI: 10.3390/ijms25010077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Acute Respiratory Distress Syndrome (ARDS) is a major health concern with urgent unmet need for treatment options. There are three million new ARDS cases annually, and the disease's mortality rate is high (35-46%). Cluster of differentiation 24 (CD24), a long-known protein with multifaceted functions, is a small, heavily glycosylated, membrane-anchored protein which functions as an immune checkpoint control. CD24 allows for immune discrimination between Damage-Associated Molecular Patterns and Pathogen-Associated Molecular Patterns derived from pathogens. Exosomes are intraluminal vesicles which play an important role in intercellular communication. Exosomes offer the advantage of targeted delivery, which improves safety and efficacy. The safety and efficacy of EXO-CD24 is promising, as was shown in >180 ARDS patients in phase 1b/2a, phase 2b, and compassionate use. CD24 binds Damage-associated molecular patterns (DAMPs) and inhibits the activation of the NF-ĸB pathway, a pivotal mediator of inflammatory responses. In contrast to anti-inflammatory therapies that are cytokine-specific or steroids that shut down the entire immune system, EXO-CD24 acts upstream, reverting the immune system back to normal activity. Herein, the safety and efficacy of mEXO-CD24 is shown in murine models of several pulmonary diseases (sepsis, allergic asthma, Chronic Obstructive Pulmonary Disease(COPD), fibrosis). EXO CD24 can suppress the hyperinflammatory response in the lungs in several pulmonary diseases with a significant unmet need for treatment options.
Collapse
Affiliation(s)
- Shiran Shapira
- Health Promotion Center and Integrated Cancer Prevention Center, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (S.S.); (A.B.-S.); (A.M.)
- Department of Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Reut Schwartz
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (R.S.); (N.A.)
- Anesthesia and Intensive Care Unit, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Sotirios Tsiodras
- 4th Department of Internal Medicine, University General Hospital Attikon, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Amir Bar-Shai
- Health Promotion Center and Integrated Cancer Prevention Center, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (S.S.); (A.B.-S.); (A.M.)
| | - Ariel Melloul
- Health Promotion Center and Integrated Cancer Prevention Center, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (S.S.); (A.B.-S.); (A.M.)
| | - Sarah Borsekofsky
- Department of Pathology, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel;
| | - Michael Peer
- Department of Chest Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel;
| | - Nimrod Adi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (R.S.); (N.A.)
- Anesthesia and Intensive Care Unit, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Ronan MacLoughlin
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons, D02 YN77 Dublin, Ireland;
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
| | - Nadir Arber
- Health Promotion Center and Integrated Cancer Prevention Center, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (S.S.); (A.B.-S.); (A.M.)
- Department of Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (R.S.); (N.A.)
| |
Collapse
|
9
|
Amirzadeh Gougheri K, Ahmadi A, Ahmadabadi MG, Babajani A, Yazdanpanah G, Bahrami S, Hassani M, Niknejad H. Exosomal Cargo: Pro-angiogeneic, anti-inflammatory, and regenerative effects in ischemic and non-ischemic heart diseases - A comprehensive review. Biomed Pharmacother 2023; 168:115801. [PMID: 37918257 DOI: 10.1016/j.biopha.2023.115801] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/04/2023] Open
Abstract
Heart diseases are the primary cause of mortality and morbidity worldwide which inflict a heavy social and economic burden. Among heart diseases, most deaths are due to myocardial infarction (MI) or heart attack, which occurs when a decrement in blood flow to the heart causes injury to cardiac tissue. Despite several available diagnostic, therapeutic, and prognostic approaches, heart disease remains a significant concern. Exosomes are a kind of small extracellular vesicles released by different types of cells that play a part in intercellular communication by transferring bioactive molecules important in regenerative medicine. Many studies have reported the diagnostic, therapeutic, and prognostic role of exosomes in various heart diseases. Herein, we reviewed the roles of exosomes as new emerging agents in various types of heart diseases, including ischemic heart disease, cardiomyopathy, arrhythmia, and valvular disease, focusing on pathogenesis, therapeutic, diagnostic, and prognostic roles in different areas. We have also mentioned different routes of exosome delivery to target tissues, the effects of preconditioning and modification on exosome's capability, exosome production in compliance with good manufacturing practice (GMP), and their ongoing clinical applications in various medical contexts to shed light on possible clinical translation.
Collapse
Affiliation(s)
- Kowsar Amirzadeh Gougheri
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Armin Ahmadi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Amirhesam Babajani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghasem Yazdanpanah
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, 1855 W. Taylor Street, MC 648, Chicago, IL 60612, USA
| | - Soheyl Bahrami
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Vienna, Austria
| | - Mohammad Hassani
- Department of Vascular and Endovascular Surgery, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Sababathy M, Ramanathan G, Abd Rahaman NY, Ramasamy R, Biau FJ, Qi Hao DL, Hamid NFS. A 'one stone, two birds' approach with mesenchymal stem cells for acute respiratory distress syndrome and Type II diabetes mellitus. Regen Med 2023; 18:913-934. [PMID: 38111999 DOI: 10.2217/rme-2023-0193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023] Open
Abstract
This review explores the intricate relationship between acute respiratory distress syndrome (ARDS) and Type II diabetes mellitus (T2DM). It covers ARDS epidemiology, etiology and pathophysiology, along with current treatment trends and challenges. The lipopolysaccharides (LPS) role in ARDS and its association between non-communicable diseases and COVID-19 are discussed. The review highlights the therapeutic potential of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) for ARDS and T2DM, emphasizing their immunomodulatory effects. This review also underlines how T2DM exacerbates ARDS pathophysiology and discusses the potential of hUC-MSCs in modulating immune responses. In conclusion, the review highlights the multidisciplinary approach to managing ARDS and T2DM, focusing on inflammation, oxidative stress and potential therapy of hUC-MSCs in the future.
Collapse
Affiliation(s)
- Mogesh Sababathy
- Department of Veterinary Pathology & Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Ghayathri Ramanathan
- Faculty of Computer Science & Information Technology, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Nor Yasmin Abd Rahaman
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Laboratory of Vaccines & Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Rajesh Ramasamy
- Department of Pathology, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Foo Jhi Biau
- Centre for Drug Discovery & Molecular Pharmacology (CDDMP), Faculty of Health & Medical Sciences, Taylor's University, Selangor, Subang Jaya, 47500, Malaysia
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, Selangor, Subang Jaya, 47500, Malaysia
| | - Daniel Looi Qi Hao
- My Cytohealth Sdn. Bhd., 18-2, Jalan Radin Bagus 1, Bandar Seri Petaling, Kuala Lumpur, 57000, Malaysia
| | - Nur-Fazila Saulol Hamid
- Department of Veterinary Pathology & Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Laboratory of Vaccines & Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
11
|
Niimi Y, Baljinnyam T, Fukuda S, Andersen CR, Salsbury JR, Lee JO, Prough DS, Enkhbaatar P. Effects of nebulized adipose-derived mesenchymal stem cells on acute lung injury following smoke inhalation in sheep. Int Immunopharmacol 2023; 123:110638. [PMID: 37494838 DOI: 10.1016/j.intimp.2023.110638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/07/2023] [Accepted: 07/09/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION Treatment of ARDS caused by smoke inhalation is challenging with no specific therapies available. The aim of this study was to test the efficacy of nebulized adipose-derived mesenchymal stem cells (ASCs) in a well-characterized, clinically relevant ovine model of smoke inhalation injury. MATERIAL AND METHODS Fourteen female Merino sheep were surgically instrumented 5-7 days prior to study. After induction of acute lung injury (ALI) by cooled cotton smoke insufflation into the lungs (under anesthesia and analgesia), sheep were placed on a mechanical ventilator for 48 hrs and monitored for cardiopulmonary hemodynamics in a conscious state. ASCs were isolated from ovine adipose tissue. Sheep were randomly allocated to two groups after smoke injury: 1) ASCs group (n = 6): 10 million ASCs were nebulized into the airway at 1 hr post-injury; and 2) Control group (n = 8): Nebulized with saline into the airways at 1 hr post-injury. ASCs were labeled with green fluorescent protein (GFP) to trace cells within the lung. ASCs viability was determined in bronchoalveolar lavage fluid (BALF). RESULTS PaO2/FiO2 in the ASCs group was significantly higher than in the control group (p = 0.001) at 24 hrs. Oxygenation index: (mean airway pressure × FiO2/PaO2) was significantly lower in the ASCs group at 36 hr (p = 0.003). Pulmonary shunt fraction tended to be lower in the ASCs group as compared to the control group. GFP-labelled ASCs were found on the surface of trachea epithelium 48 hrs after injury. The viability of ASCs in BALF was significantly lower than those exposed to the control vehicle solution. CONCLUSION Nebulized ASCs moderately improved pulmonary function and delayed the onset of ARDS.
Collapse
Affiliation(s)
- Yosuke Niimi
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555-1102, USA
| | - Tuvshintugs Baljinnyam
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555-1102, USA; Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555-1102, USA
| | - Satoshi Fukuda
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555-1102, USA
| | - Clark R Andersen
- Department of Biostatistics, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555-1102, USA
| | - John R Salsbury
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555-1102, USA
| | - Jong O Lee
- Department of Surgery, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555-1102, USA
| | - Donald S Prough
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555-1102, USA
| | - Perenlei Enkhbaatar
- Department of Anesthesiology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555-1102, USA.
| |
Collapse
|
12
|
Green O, Shenberg G, Baruch R, Argaman L, Levin T, Michelson I, Hadary R, Isakovich B, Golos M, Schwartz R, MacLoughlin R, Adi N, Arber N, Shapira S. Inhaled Exosomes Genetically Manipulated to Overexpress CD24 (EXO-CD24) as a Compassionate Use in Severe ARDS Patients. Biomedicines 2023; 11:2523. [PMID: 37760963 PMCID: PMC10525844 DOI: 10.3390/biomedicines11092523] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/01/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
RATIONALE Acute respiratory distress syndrome (ARDS) is a major global health concern with a significant unmet need. EXO-CD24 is delivered via inhalation-reduced cytokines and chemokine secretion and lung injury in ARDS and improved survival in mice models of ARDS, influenza, and sepsis. OBJECTIVES This clinical paper aims to evaluate the potential of EXO-CD24, a novel immunomodulatory treatment, in the compassionate care of critically ill, intubated patients with post-infection-induced acute respiratory distress syndrome (ARDS). METHODS Eleven critically ill patients diagnosed with post-infection ARDS (10 with COVID-19 and one with an adenovirus-associated infection) were administered EXO-CD24 in four medical centers across Israel. The patients had multiple co-morbidities, including cancer, hypertension, diabetes, and ischemic heart disease, and met the criteria for severe ARDS according to the Berlin classification. EXO-CD24 was administered via inhalation, and adverse events related to its use were carefully monitored. MEASUREMENTS AND MAIN RESULTS The administration of EXO-CD24 did not result in any recorded adverse events. The median hospitalization duration was 11.5 days, and the overall mortality rate was 36%. Notably, patients treated at the Tel Aviv Medical Center (TASMC) showed a lower mortality rate of 12.5%. The WBC and CRP levels decreased in comparison to baseline levels at hospitalization, and rapid responses occurred even in patients with kidney transplants who were off the ventilator within a few days and discharged shortly thereafter. The production of cytokines and chemokines was significantly suppressed in all patients, including those who died. Among the patients at TASMC, four had kidney transplants and were on immunosuppressive drugs, and all of them fully recovered and were discharged from the hospital. CONCLUSIONS EXO-CD24 holds promise as a potential therapeutic agent for all stages of ARDS, even in severe intubated cases. Importantly, EXO-CD24 demonstrated a favorable safety profile without any apparent side effects with promising efficacy. Furthermore, the potential of EXO-CD24 as a platform for addressing hyper-inflammatory states warrants exploration. Further research and larger-scale clinical trials are warranted to validate these preliminary findings.
Collapse
Affiliation(s)
- Orr Green
- Health Promotion Center and Integrated Cancer Prevention Center, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (O.G.); (G.S.); (L.A.); (I.M.); (N.A.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6423906, Israel; (R.B.); (R.S.); (N.A.)
| | - Gil Shenberg
- Health Promotion Center and Integrated Cancer Prevention Center, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (O.G.); (G.S.); (L.A.); (I.M.); (N.A.)
| | - Roni Baruch
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6423906, Israel; (R.B.); (R.S.); (N.A.)
- Department of Kidney Transplantation, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Lihi Argaman
- Health Promotion Center and Integrated Cancer Prevention Center, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (O.G.); (G.S.); (L.A.); (I.M.); (N.A.)
| | - Talya Levin
- Health Promotion Center and Integrated Cancer Prevention Center, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (O.G.); (G.S.); (L.A.); (I.M.); (N.A.)
| | - Ian Michelson
- Health Promotion Center and Integrated Cancer Prevention Center, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (O.G.); (G.S.); (L.A.); (I.M.); (N.A.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6423906, Israel; (R.B.); (R.S.); (N.A.)
| | - Ruthy Hadary
- Department of Internal Medicine C, Meir Medical Center, Kefar-Saba 4428164, Israel;
| | - Boris Isakovich
- Intensive Care Unit, Hillel Yaffe Medical Center, Hadera 3820302, Israel;
| | - Miri Golos
- Carmel Medical Center, Haifa 3436212, Israel;
| | - Reut Schwartz
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6423906, Israel; (R.B.); (R.S.); (N.A.)
- Anesthesia and Intensive Care Unit, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Ronan MacLoughlin
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons, D02 YN77 Dublin, Ireland;
- School of Pharmacy and Pharmaceutical Sciences, Trinity College, D02 PN40 Dublin, Ireland
| | - Nimrod Adi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6423906, Israel; (R.B.); (R.S.); (N.A.)
- Anesthesia and Intensive Care Unit, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Nadir Arber
- Health Promotion Center and Integrated Cancer Prevention Center, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (O.G.); (G.S.); (L.A.); (I.M.); (N.A.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6423906, Israel; (R.B.); (R.S.); (N.A.)
| | - Shiran Shapira
- Health Promotion Center and Integrated Cancer Prevention Center, Tel-Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel; (O.G.); (G.S.); (L.A.); (I.M.); (N.A.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6423906, Israel; (R.B.); (R.S.); (N.A.)
| |
Collapse
|
13
|
Soetjahjo B, Malueka RG, Nurudhin A, Purwoko, Sumardi, Wisaksana R, Adhiputri A, Sudadi, Soeroto AY, Sidharta BRA, Thobari JA, Murni TW, Soewondo W, Herningtyas EH, Sudjud RW, Trisnawati I, Ananda NR, Faried A. Effectiveness and safety of normoxic allogenic umbilical cord mesenchymal stem cells administered as adjunctive treatment in patients with severe COVID-19. Sci Rep 2023; 13:12520. [PMID: 37532730 PMCID: PMC10397314 DOI: 10.1038/s41598-023-39268-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 07/22/2023] [Indexed: 08/04/2023] Open
Abstract
Inflammatory response in COVID-19 contributes greatly to disease severity. Mesenchymal Stem Cells (MSCs) have the potential to alleviate inflammation and reduce mortality and length of stay in COVID-19 patients. We investigated the safety and effectiveness of normoxic-allogenic umbilical cord (NA-UC)-MSCs as an adjunctive treatment in severe COVID-19 patients. A double-blind, multicentric, randomized, placebo-controlled trial involving severe COVID-19 patients was performed from January to June 2021 in three major hospitals across Java, Indonesia. Eligible participants (n = 42) were randomly assigned to two groups (1:1), namely the intervention (n = 21) and control (n = 21) groups. UC-MSCs dose was 1 × 106 /kg body weight on day D0, D3, and D6. The primary outcome was the duration of hospitalization. Meanwhile, the secondary outcomes were radiographical progression (Brixia score), respiratory and oxygenation parameters, and inflammatory markers, in addition to the safety profile of NA-UC-MSCs. NA-UC-MSCs administration did not affect the length of hospital stay of severe COVID-19 patients, nor did it improve the Brixia score or mMRC dyspnoea scale better than placebo. Nevertheless, NA-UC-MSCs led to a better recuperation in oxygenation index (120.80 ± 72.70 baseline vs. 309.63 ± 319.30 D + 22, p = 0.038) and oxygen saturation (97.24 ± 4.10% vs. 96.19 ± 3.75% in placebo, p = 0.028). Additionally, compared to the placebo group, the treatment group had a significantly smaller increase in PCT level at D + 22 (1.43 vs. 12.76, p = 0.011). No adverse effects, including serious ones, were recorded until D + 91. NA-UC-MSCs therapy is a very safe adjunct for COVID-19 patients. It improves the oxygenation profile and carries potential to suppress inflammation.
Collapse
Affiliation(s)
- Bintang Soetjahjo
- Department of Orthopaedics and Traumatology, Universitas Sebelas Maret-Dr. Moewardi Hospital, Solo, Indonesia
| | - Rusdy Ghazali Malueka
- Department of Neurology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada-Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Arief Nurudhin
- Department of Internal Medicine, Universitas Sebelas Maret-Dr. Moewardi Hospital, Solo, Indonesia
| | - Purwoko
- Department of Anesthesiology and Intensive Therapy, Universitas Sebelas Maret-Dr. Moewardi Hospital, Solo, Indonesia
| | - Sumardi
- Department of Internal Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada-Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Rudi Wisaksana
- Department of Internal Medicine, Universitas Padjadjaran-Dr. Hasan Sadikin Hospital, Bandung, Indonesia
| | - Artrien Adhiputri
- Department of Pulmonology and Respiratory Medicine, Universitas Sebelas Maret-Dr. Moewardi Hospital, Solo, Indonesia
| | - Sudadi
- Department of Anesthesiology and Intensive Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada-Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Arto Yuwono Soeroto
- Department of Internal Medicine, Universitas Padjadjaran-Dr. Hasan Sadikin Hospital, Bandung, Indonesia
| | | | - Jarir At Thobari
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Tri Wahyu Murni
- Department of Surgery, Universitas Padjadjaran-Dr. Hasan Sadikin Hospital, Bandung, Indonesia
| | - Widiastuti Soewondo
- Department of Radiology, Universitas Sebelas Maret-Dr. Moewardi Hospital, Solo, Indonesia
| | - Elizabeth Henny Herningtyas
- Department of Clinical Pathology, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada-Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Reza Widianto Sudjud
- Department of Anesthesiology-Intensive Therapy, Universitas Padjadjaran-Dr. Hasan Sadikin Hospital, Bandung, Indonesia
| | - Ika Trisnawati
- Department of Internal Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada-Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Nur Rahmi Ananda
- Department of Internal Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada-Dr. Sardjito General Hospital, Yogyakarta, Indonesia
| | - Ahmad Faried
- Department of Neurosurgery, Universitas Padjadjaran - Dr. Hasan Sadikin Hospital, Bandung, 40161, Indonesia.
| |
Collapse
|
14
|
Uskoković V. Lessons from the history of inorganic nanoparticles for inhalable diagnostics and therapeutics. Adv Colloid Interface Sci 2023; 315:102903. [PMID: 37084546 DOI: 10.1016/j.cis.2023.102903] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 04/04/2023] [Accepted: 04/10/2023] [Indexed: 04/23/2023]
Abstract
The respiratory tract is one of the most accessible ones to exogenous nanoparticles, yet drug delivery by their means to it is made extraordinarily challenging because of the plexus of aerodynamic, hemodynamic and biomolecular factors at cellular and extracellular levels that synergistically define the safety and efficacy of this process. Here, the use of inorganic nanoparticles (INPs) for inhalable diagnostics and therapies of the lung is viewed through the prism of the history of studies on the interaction of INPs with the lower respiratory tract. The most conceptually and methodologically innovative and illuminative studies are referred to in the chronological order, as they were reported in the literature, and the trends in the progress of understanding this interaction of immense therapeutic and toxicological significance are being deduced from it. The most outstanding actual trends delineated include the diminishment of toxicity via surface functionalization, cell targeting, tagging and tracking via controlled binding and uptake, hybrid INP treatments, magnetic guidance, combined drug and gene delivery, use as adjuvants in inhalable vaccines, and other. Many of the understudied research directions, which have been accomplished by the nanostructured organic polymers in the pulmonary niche, are discussed. The progress in the use of INPs as inhalable diagnostics or therapeutics has been hampered by their well-recognized inflammatory potential and toxicity in the respiratory tract. However, the annual numbers of methodologically innovative studies have been on the rise throughout the past two decades, suggesting that this is a prolific direction of research, its comparatively poor commercial takings notwithstanding. Still, the lack of consensus on the effects of many INP compositions at low but therapeutically effective doses, the plethora of contradictory reports on ostensibly identical chemical compositions and NP properties, and the many cases of antagonism in combinatorial NP treatments imply that the rational design of inhalable medical devices based on INPs must rely on qualitative principles for the most part and embrace a partially stochastic approach as well. At the same time, the fact that the most studied INPs for pulmonary applications have been those with some of the thickest records of pulmonary toxicity, e.g., carbon, silver, gold, silica and iron oxide, is a silent call for the expansion of the search for new inorganic compositions for use in inhalable therapies to new territories.
Collapse
Affiliation(s)
- Vuk Uskoković
- Advanced Materials and Nanobiotechnology Laboratory, TardigradeNano LLC, 7 Park Vista, Irvine, CA 92604, USA; Department of Mechanical Engineering, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA.
| |
Collapse
|
15
|
Haghighitalab A, Dominici M, Matin MM, Shekari F, Ebrahimi Warkiani M, Lim R, Ahmadiankia N, Mirahmadi M, Bahrami AR, Bidkhori HR. Extracellular vesicles and their cells of origin: Open issues in autoimmune diseases. Front Immunol 2023; 14:1090416. [PMID: 36969255 PMCID: PMC10031021 DOI: 10.3389/fimmu.2023.1090416] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
The conventional therapeutic approaches to treat autoimmune diseases through suppressing the immune system, such as steroidal and non-steroidal anti-inflammatory drugs, are not adequately practical. Moreover, these regimens are associated with considerable complications. Designing tolerogenic therapeutic strategies based on stem cells, immune cells, and their extracellular vesicles (EVs) seems to open a promising path to managing autoimmune diseases' vast burden. Mesenchymal stem/stromal cells (MSCs), dendritic cells, and regulatory T cells (Tregs) are the main cell types applied to restore a tolerogenic immune status; MSCs play a more beneficial role due to their amenable properties and extensive cross-talks with different immune cells. With existing concerns about the employment of cells, new cell-free therapeutic paradigms, such as EV-based therapies, are gaining attention in this field. Additionally, EVs' unique properties have made them to be known as smart immunomodulators and are considered as a potential substitute for cell therapy. This review provides an overview of the advantages and disadvantages of cell-based and EV-based methods for treating autoimmune diseases. The study also presents an outlook on the future of EVs to be implemented in clinics for autoimmune patients.
Collapse
Affiliation(s)
- Azadeh Haghighitalab
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Maryam M. Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Rebecca Lim
- Department of Obstetrics and Gynaecology, Monash University, Clayton VIC, Australia
| | - Naghmeh Ahmadiankia
- Cancer Prevention Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mahdi Mirahmadi
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hamid Reza Bidkhori
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR)-Khorasan Razavi, Mashhad, Iran
| |
Collapse
|
16
|
Man F, Tang J, Swedrowska M, Forbes B, T M de Rosales R. Imaging drug delivery to the lungs: Methods and applications in oncology. Adv Drug Deliv Rev 2023; 192:114641. [PMID: 36509173 PMCID: PMC10227194 DOI: 10.1016/j.addr.2022.114641] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/14/2022]
Abstract
Direct delivery to the lung via inhalation is arguably one of the most logical approaches to treat lung cancer using drugs. However, despite significant efforts and investment in this area, this strategy has not progressed in clinical trials. Imaging drug delivery is a powerful tool to understand and develop novel drug delivery strategies. In this review we focus on imaging studies of drug delivery by the inhalation route, to provide a broad overview of the field to date and attempt to better understand the complexities of this route of administration and the significant barriers that it faces, as well as its advantages. We start with a discussion of the specific challenges for drug delivery to the lung via inhalation. We focus on the barriers that have prevented progress of this approach in oncology, as well as the most recent developments in this area. This is followed by a comprehensive overview of the different imaging modalities that are relevant to lung drug delivery, including nuclear imaging, X-ray imaging, magnetic resonance imaging, optical imaging and mass spectrometry imaging. For each of these modalities, examples from the literature where these techniques have been explored are provided. Finally the different applications of these technologies in oncology are discussed, focusing separately on small molecules and nanomedicines. We hope that this comprehensive review will be informative to the field and will guide the future preclinical and clinical development of this promising drug delivery strategy to maximise its therapeutic potential.
Collapse
Affiliation(s)
- Francis Man
- School of Cancer & Pharmaceutical Sciences, King's College London, London, SE1 9NH, United Kingdom
| | - Jie Tang
- School of Biomedical Engineering & Imaging Sciences, King's College London, London SE1 7EH, United Kingdom
| | - Magda Swedrowska
- School of Cancer & Pharmaceutical Sciences, King's College London, London, SE1 9NH, United Kingdom
| | - Ben Forbes
- School of Cancer & Pharmaceutical Sciences, King's College London, London, SE1 9NH, United Kingdom
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London, London SE1 7EH, United Kingdom.
| |
Collapse
|
17
|
Zhang Y, Yin P, Huang J, Yang L, Liu Z, Fu D, Hu Z, Huang W, Miao Y. Scalable and high-throughput production of an injectable platelet-rich plasma (PRP)/cell-laden microcarrier/hydrogel composite system for hair follicle tissue engineering. J Nanobiotechnology 2022; 20:465. [PMID: 36329527 PMCID: PMC9632161 DOI: 10.1186/s12951-022-01671-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Tissue engineering of hair follicles (HFs) has enormous potential for hair loss treatment. However, certain challenges remain, including weakening of the dermal papilla cell (DPC) viability, proliferation, and HF inducibility, as well as the associated inefficient and tedious preparation process required to generate extracellular matrix (ECM)-mimicking substrates for biomolecules or cells. Herein, we utilized gelatin methacryloyl (GelMA) and chitosan hydrogels to prepare scalable, monodispersed, and diameter-controllable interpenetrating network GelMA/chitosan-microcarriers (IGMs) loaded with platelet-rich plasma (PRP) and seeded with DPCs, on a high-throughput microfluidic chip. RESULTS The ECM-mimicking hydrogels used for IGMs exhibited surface nano-topography and high porosity. Mass production of IGMs with distinct and precise diameters was achieved by adjusting the oil and aqueous phase flow rate ratio. Moreover, IGMs exhibited appropriate swelling and sustained growth factor release to facilitate a relatively long hair growth phase. DPCs seeded on PRP-loaded IGMs exhibited good viability (> 90%), adhesion, spreading, and proliferative properties (1.2-fold greater than control group). Importantly, PRP-loaded IGMs presented a higher hair inducibility of DPCs in vitro compared to the control and IGMs group (p < 0.05). Furthermore, DPC/PRP-laden IGMs were effectively mixed with epidermal cell (EPC)-laden GelMA to form a PRP-loaded DPC/EPC co-cultured hydrogel system (DECHS), which was subcutaneously injected into the hypodermis of nude mice. The PRP-loaded DECHS generated significantly more HFs (~ 35 per site) and novel vessels (~ 12 per site) than the other groups (p < 0.05 for each). CONCLUSION Taken together, these results illustrate that, based on high-throughput microfluidics, we obtained scalable and controllable production of ECM-mimicking IGMs and DECHS, which simulate an effective micro- and macro-environment to promote DPC bioactivity and hair regeneration, thus representing a potential new strategy for HF tissue engineering.
Collapse
Affiliation(s)
- Yufan Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China
| | - Panjing Yin
- Department of Joint Surgery, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Junfei Huang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China
| | - Lunan Yang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China
| | - Zhen Liu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China
| | - Danlan Fu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China.
| | - Wenhua Huang
- Department of Joint Surgery, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China.
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, 510515, Guangzhou, PR China.
| | - Yong Miao
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, 510515, Guangzhou, Guangdong Province, China.
| |
Collapse
|
18
|
Krishnan A, Muthusamy S, Fernandez FB, Kasoju N. Mesenchymal Stem Cell-Derived Extracellular Vesicles in the Management of COVID19-Associated Lung Injury: A Review on Publications, Clinical Trials and Patent Landscape. Tissue Eng Regen Med 2022; 19:659-673. [PMID: 35384633 PMCID: PMC8985390 DOI: 10.1007/s13770-022-00441-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 02/07/2023] Open
Abstract
The unprecedented COVID-19 pandemic situation forced the scientific community to explore all the possibilities from various fields, and so far we have seen a lot of surprises, eureka moments and disappointments. One of the approaches from the cellular therapists was exploiting the immunomodulatory and regenerative potential of mesenchymal stromal cells (MSCs), more so of MSC-derived extracellular vesicles (EVs)-particularly exosomes, in order to alleviate the cytokine storm and regenerate the damaged lung tissues. Unlike MSCs, the EVs are easier to store, deliver, and are previously shown to be as effective as MSCs, yet less immunogenic. These features attracted the attention of many and thus led to a tremendous increase in publications, clinical trials and patent applications. This review presents the current landscape of the field and highlights some interesting findings on MSC-derived EVs in the context of COVID-19, including in silico, in vitro, in vivo and case reports. The data strongly suggests the potential of MSC-derived EVs as a therapeutic regime for the management of acute lung injury and associated complications in COVID-19 and beyond.
Collapse
Affiliation(s)
- Anand Krishnan
- Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Science and Technology, Thiruvananthapuram, 695012, Kerala, India
| | - Senthilkumar Muthusamy
- Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Science and Technology, Thiruvananthapuram, 695012, Kerala, India
| | - Francis B Fernandez
- Department of Biomaterial Science and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Science and Technology, Thiruvananthapuram, 695012, Kerala, India
| | - Naresh Kasoju
- Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Science and Technology, Thiruvananthapuram, 695012, Kerala, India.
| |
Collapse
|
19
|
Mesenchymal stem cell treatment improves outcome of COVID-19 patients via multiple immunomodulatory mechanisms. Cell Res 2021; 31:1244-1262. [PMID: 34702946 PMCID: PMC8546390 DOI: 10.1038/s41422-021-00573-y] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 09/09/2021] [Indexed: 12/24/2022] Open
Abstract
The infusion of coronavirus disease 2019 (COVID-19) patients with mesenchymal stem cells (MSCs) potentially improves clinical symptoms, but the underlying mechanism remains unclear. We conducted a randomized, single-blind, placebo-controlled (29 patients/group) phase II clinical trial to validate previous findings and explore the potential mechanisms. Patients treated with umbilical cord-derived MSCs exhibited a shorter hospital stay (P = 0.0198) and less time required for symptoms remission (P = 0.0194) than those who received placebo. Based on chest images, both severe and critical patients treated with MSCs showed improvement by day 7 (P = 0.0099) and day 21 (P = 0.0084). MSC-treated patients had fewer adverse events. MSC infusion reduced the levels of C-reactive protein, proinflammatory cytokines, and neutrophil extracellular traps (NETs) and promoted the maintenance of SARS-CoV-2-specific antibodies. To explore how MSCs modulate the immune system, we employed single-cell RNA sequencing analysis on peripheral blood. Our analysis identified a novel subpopulation of VNN2+ hematopoietic stem/progenitor-like (HSPC-like) cells expressing CSF3R and PTPRE that were mobilized following MSC infusion. Genes encoding chemotaxis factors - CX3CR1 and L-selectin - were upregulated in various immune cells. MSC treatment also regulated B cell subsets and increased the expression of costimulatory CD28 in T cells in vivo and in vitro. In addition, an in vivo mouse study confirmed that MSCs suppressed NET release and reduced venous thrombosis by upregulating kindlin-3 signaling. Together, our results underscore the role of MSCs in improving COVID-19 patient outcomes via maintenance of immune homeostasis.
Collapse
|
20
|
Acute Respiratory Distress Syndrome: Focus on Viral Origin and Role of Pulmonary Lymphatics. Biomedicines 2021; 9:biomedicines9111732. [PMID: 34829961 PMCID: PMC8615541 DOI: 10.3390/biomedicines9111732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/01/2021] [Accepted: 11/17/2021] [Indexed: 11/30/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a serious affection of the lung caused by a variety of pathologies. Great interest is currently focused on ARDS induced by viruses (pandemic influenza and corona viruses). The review describes pulmonary changes in ARDS and specific effects of the pandemic viruses in ARDS, and summarizes treatment options. Because the known pathogenic mechanisms cannot explain all aspects of the syndrome, the contribution of pulmonary lymphatics to the pathology is discussed. Organization and function of lymphatics in a healthy lung and in resorption of pulmonary edema are described. A future clinical trial may provide more insight into the role of hyaluronan in ARDS but the development of promising pharmacological treatments is unlikely because drugs play no important role in lymphedema therapy.
Collapse
|
21
|
Laurent A, Abdel-Sayed P, Hirt-Burri N, Scaletta C, Michetti M, de Buys Roessingh A, Raffoul W, Applegate LA. Evolution of Diploid Progenitor Lung Cell Applications: From Optimized Biotechnological Substrates to Potential Active Pharmaceutical Ingredients in Respiratory Tract Regenerative Medicine. Cells 2021; 10:2526. [PMID: 34685505 PMCID: PMC8533713 DOI: 10.3390/cells10102526] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/14/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
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.
Collapse
Affiliation(s)
- Alexis Laurent
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
- TEC-PHARMA SA, Manufacturing Department, CH-1038 Bercher, Switzerland
- LAM Biotechnologies SA, Manufacturing Department, CH-1066 Épalinges, Switzerland
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
| | - Corinne Scaletta
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
| | - Murielle Michetti
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
| | - Anthony de Buys Roessingh
- Children and Adolescent Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Romand Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
| | - Wassim Raffoul
- Romand Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Plastic, Reconstructive, and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
- Romand Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| |
Collapse
|
22
|
Mesenchymal Stem Cell-Based Therapy as an Alternative to the Treatment of Acute Respiratory Distress Syndrome: Current Evidence and Future Perspectives. Int J Mol Sci 2021; 22:ijms22157850. [PMID: 34360616 PMCID: PMC8346146 DOI: 10.3390/ijms22157850] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/17/2021] [Accepted: 07/19/2021] [Indexed: 12/25/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) represents a current challenge for medicine due to its incidence, morbidity and mortality and, also, the absence of an optimal treatment. The COVID-19 outbreak only increased the urgent demand for an affordable, safe and effective treatment for this process. Early clinical trials suggest the therapeutic usefulness of mesenchymal stem cells (MSCs) in acute lung injury (ALI) and ARDS. MSC-based therapies show antimicrobial, anti-inflammatory, regenerative, angiogenic, antifibrotic, anti-oxidative stress and anti-apoptotic actions, which can thwart the physiopathological mechanisms engaged in ARDS. In addition, MSC secretome and their derived products, especially exosomes, may reproduce the therapeutic effects of MSC in lung injury. This last strategy of treatment could avoid several safety issues potentially associated with the transplantation of living and proliferative cell populations and may be formulated in different forms. However, the following diverse limitations must be addressed: (i) selection of the optimal MSC, bearing in mind both the heterogeneity among donors and across different histological origins, (ii) massive obtention of these biological products through genetic manipulations of the most appropriate MSC, (iii) bioreactors that allow their growth in 3D, (iv) ideal culture conditions and (v) adequate functional testing of these obtaining biological products before their clinical application.
Collapse
|
23
|
Buschmann D, Mussack V, Byrd JB. Separation, characterization, and standardization of extracellular vesicles for drug delivery applications. Adv Drug Deliv Rev 2021; 174:348-368. [PMID: 33964356 PMCID: PMC8217305 DOI: 10.1016/j.addr.2021.04.027] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/25/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) are membranous nanovesicles secreted from living cells, shuttling macromolecules in intercellular communication and potentially possessing intrinsic therapeutic activity. Due to their stability, low immunogenicity, and inherent interaction with recipient cells, EVs also hold great promise as drug delivery vehicles. Indeed, they have been used to deliver nucleic acids, proteins, and small molecules in preclinical investigations. Furthermore, EV-based drugs have entered early clinical trials for cancer or neurodegenerative diseases. Despite their appeal as delivery vectors, however, EV-based drug delivery progress has been hampered by heterogeneity of sample types and methods as well as a persistent lack of standardization, validation, and comprehensive reporting. This review highlights specific requirements for EVs in drug delivery and describes the most pertinent approaches for separation and characterization. Despite residual uncertainties related to pharmacodynamics, pharmacokinetics, and potential off-target effects, clinical-grade, high-potency EV drugs might be achievable through GMP-compliant workflows in a highly standardized environment.
Collapse
Affiliation(s)
- Dominik Buschmann
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Veronika Mussack
- Department of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - James Brian Byrd
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
24
|
Singh N, Villoutreix BO. Resources and computational strategies to advance small molecule SARS-CoV-2 discovery: Lessons from the pandemic and preparing for future health crises. Comput Struct Biotechnol J 2021; 19:2537-2548. [PMID: 33936562 PMCID: PMC8074526 DOI: 10.1016/j.csbj.2021.04.059] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 12/11/2022] Open
Abstract
There is an urgent need to identify new therapies that prevent SARS-CoV-2 infection and improve the outcome of COVID-19 patients. This pandemic has thus spurred intensive research in most scientific areas and in a short period of time, several vaccines have been developed. But, while the race to find vaccines for COVID-19 has dominated the headlines, other types of therapeutic agents are being developed. In this mini-review, we report several databases and online tools that could assist the discovery of anti-SARS-CoV-2 small chemical compounds and peptides. We then give examples of studies that combined in silico and in vitro screening, either for drug repositioning purposes or to search for novel bioactive compounds. Finally, we question the overall lack of discussion and plan observed in academic research in many countries during this crisis and suggest that there is room for improvement.
Collapse
Affiliation(s)
- Natesh Singh
- Université de Paris, Inserm UMR 1141 NeuroDiderot, Robert-Debré Hospital, 75019 Paris, France
| | - Bruno O. Villoutreix
- Université de Paris, Inserm UMR 1141 NeuroDiderot, Robert-Debré Hospital, 75019 Paris, France
| |
Collapse
|