1
|
Ulu BU, Hindilerden IY, Yigenoglu TN, Tiryaki TO, Erkurt MA, Korkmaz G, Namdaroglu S, Aksoy E, Korkmaz S, Seyhan M, Yilmaz S, Besisik SK, Dal MS, Ulas T, Altuntas F. Are mesenchymal stem cells still effective in acute GvHD management? Transfus Apher Sci 2025; 64:104051. [PMID: 39721135 DOI: 10.1016/j.transci.2024.104051] [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] [Indexed: 12/28/2024]
Abstract
OBJECTIVE Graft-versus-host disease (GvHD) is a common and serious complication following allogeneic hematopoietic stem cell transplantation (allo-HSCT), significantly impacting transplant efficacy. In the treatment of GvHD, numerous therapeutic approaches have been explored, with mesenchymal stem cells (MSCs) emerging as a prominent immunomodulatory option. We aimed to evaluate efficacy and outcomes of using MSCs for steroid refractory acute GVHD (SR-aGvHD) management. MATERIALS AND METHODS We retrospectively analyzed data from 36 patients' who received MSCs for treatment of SR-aGvHD following allo-HSCT between 2018 and 2024 from nine transplantation centers in Türkiye. The product consisted of umbilical cord-derived allogeneic MSCs, which were administered intravenously. RESULTS Our cohort was at the median age of 39 years (range: 19-61 years), with aGvHD diagnosed at a median of two months after allo-HSCT. More than half of the patients (58.3 %) classified as high-grade aGvHD according to the Minnesota risk scoring. Cord blood-derived MSCs were administered at a median dose of 3.45 (range: 0.8-5) million MSCs/kg, with a median of 3th (range: 2-5) line treatment. The rate of responses exceeding partial response (PR) was approximately 20 % at the first month, increasing to 24 % at the second month. The six-month survival rate was 33 %, with 46 % of mortality attributed to sepsis and 12.5 % related to GvHD. Multivariate analysis indicated that increasing age (≥35 years) and lower platelet counts (≤75 x109/L) were associated with higher mortality (p < 0.05). CONCLUSION MSC therapy has shown promising potential in improving response rates in aGvHD treatment, with efficacy enhanced by younger age and higher platelet counts.
Collapse
Affiliation(s)
- Bahar Uncu Ulu
- University of Health Sciences, Ankara Oncology Training and Research Hospital, Department of Hematology & Apheresis Unit, Ankara, Turkey.
| | - Ipek Yonal Hindilerden
- Istanbul University Istanbul Medical Faculty, Department of Internal Medicine, Division of Hematology, Istanbul, Turkey
| | - Tugce Nur Yigenoglu
- University of Health Sciences, Ankara Oncology Training and Research Hospital, Department of Hematology & Apheresis Unit, Ankara, Turkey
| | - Tarik Onur Tiryaki
- Istanbul University Istanbul Medical Faculty, Department of Internal Medicine, Division of Hematology, Istanbul, Turkey
| | - Mehmet Ali Erkurt
- Inonu University, Faculty of Medicine, Department of Hematology, Malatya, Turkey
| | - Gulten Korkmaz
- Ankara Bilkent City Hospital, Department of Hematology and Bone Marrow Transplantation Unit, Ankara, Turkey
| | - Sinem Namdaroglu
- Dokuz Eylul University, Faculty of Medicine, Department of Hematology, Izmir, Turkey
| | - Elif Aksoy
- University of Health Sciences, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Clinic of Hematology, Istanbul, Turkey
| | - Serdal Korkmaz
- University of Health Sciences, Kayseri Medical Faculty, Department of Hematology and Bone Marrow Transplantation Unit, Kayseri, Turkey
| | - Mert Seyhan
- University of Health Sciences, Ankara Oncology Training and Research Hospital, Department of Hematology & Apheresis Unit, Ankara, Turkey
| | - Seda Yilmaz
- University of Health Sciences, Konya Medical Faculty, Department of Hematology and Bone Marrow Transplantation Unit, Konya, Turkey
| | - Sevgi Kalayoglu Besisik
- Istanbul University Istanbul Medical Faculty, Department of Internal Medicine, Division of Hematology, Istanbul, Turkey
| | - Mehmet Sinan Dal
- University of Health Sciences, Ankara Oncology Training and Research Hospital, Department of Hematology & Apheresis Unit, Ankara, Turkey
| | - Turgay Ulas
- University of Health Sciences, Ankara Oncology Training and Research Hospital, Department of Hematology & Apheresis Unit, Ankara, Turkey
| | - Fevzi Altuntas
- University of Health Sciences, Ankara Oncology Training and Research Hospital, Department of Hematology & Apheresis Unit, Ankara, Turkey; Ankara Yildirim Beyazit University, School of Medicine, Department of Internal Medicine, Division of Hematology, Ankara, Turkey
| |
Collapse
|
2
|
Castilla-Llorente C, Bonnin A, Lansiaux P, Tudesq JJ, Beuvon C, Fabreguettes JR, Pers YM, Pugnet G, Maria ATJ, Puyade M, Urbain F, Terriou L, Poindron V, Jachiet M, Cacciatore C, Lescoat A, Prata PH, Munia I, Madelaine I, Thieblemont C, Tarte K, Yakoub-Agha I, Magro L, Farge D, Marjanovic Z. [Prerequisite and organisation of health-care pathways for Cell and Gene therapies, using Mesenchymal Stromal Cells (MSC) or Chimeric Antigen Receptor (CAR) T cells, in patients with autoimmune systemic diseases]. Bull Cancer 2025; 112:S36-S53. [PMID: 39242251 DOI: 10.1016/j.bulcan.2024.06.008] [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: 01/03/2024] [Revised: 04/30/2024] [Accepted: 06/10/2024] [Indexed: 09/09/2024]
Abstract
First-line treatments of autoimmune systemic diseases (ARD) are based on the use of various types of immunosuppressive or immunomodulatory drugs, either alone or in association, according to standardized reference protocols. Prolonged use of these drugs in severe or refractory ARD is associated with high morbidity and increased mortality. Innovative cell therapies represent a new promising approach for patients with ARDs, with the recent clinical use of: a) mesenchymal stromal cells (MSCs), based on their immunomodulatory, antifibrotic and pro-angiogenic properties and b) Chimeric Antigen Receptors (CAR) T cell therapies T lymphocytes, where genetically modified expression of a chimeric antigen receptor (CAR-T cells). Therapeutic use of MSC or CAR-T cells, remains indications of exception in patients with severe ARDs resistant to prior standard therapies with new prerequisite and organisation of health-care pathways as compared to traditional drugs, not only for the Cell and Gene Therapy (CGT) product definition and delivery process, but also for the patient clinical management before and after administration of the CGT product. The aim of this workshop under the auspices of the French Speaking Society of Bone Marrow and Cell transplantation (SFGM-TC) working group on autoimmune diseases (MATHEC) is to describe: a) the prerequisite for French hospitals to set-up the specific health-care pathways for MSC or CART therapy in ARDs patients, in accordance with regulatory and safety needs to perform academic or industry sponsored clinical trials, and b) the care-pathway for ARD patients treated with CGT, highlighting the importance of working in tandem between the ARD and the CAR-T cell specialist all along the indication, procedures and follow-up of ARDs. Patient safety considerations are central to guidance on patient selection to be validated collectively at the multidisciplinary team meeting (MDTM) based on recent (less than 3 months) thorough patient evaluation. MSC and CAR-T procedural aspects and follow-up are then carried out within appropriately experienced and SFGM-TC accredited centres in close collaboration with the ADs specialist.
Collapse
Affiliation(s)
| | - Agnès Bonnin
- Service d'hématologie clinique et thérapie cellulaire, hôpital Saint-Antoine, AP-HP, 184, rue du Faubourg-Saint-Antoine, 75012 Paris, France
| | - Pauline Lansiaux
- Unité de médecine interne (UF04) : CRMR MATHEC, maladies auto-immunes et thérapie cellulaire, Centre de référence des maladies auto-immunes systémiques rares d'Île-de-France, AP-HP, hôpital St-Louis, 75010 Paris, France; URP3518, IRSL, recherche clinique en hématologie, immunologie et transplantation, université Paris Cité, 75010 Paris, France
| | - Jean-Jacques Tudesq
- Service d'hématologie clinique, CHU de Montpellier, université de Montpellier, 80, avenue Augustin-Fliche, 34295 Montpellier, France
| | - Clément Beuvon
- Service de médecine interne et maladies infectieuses, CHU de Poitiers, 2, rue de la Milétrie, 86000 Poitiers, France
| | - Jean-Roch Fabreguettes
- Agence générale des équipements et produits de santé (AGEPS), Assistance publique-Hôpitaux de Paris (AP-HP), 75005 Paris, France
| | - Yves-Marie Pers
- Inserm UMR 1183, Institute for Regenerative Medicine and Biotherapy, University of Montpellier, 34298 Montpellier, France; Inserm, Clinical immunology and osteoarticular diseases Therapeutic Unit, Lapeyronie University Hospital, CHU Montpellier, IRMB, University of Montpellier, Montpellier, France
| | - Grégory Pugnet
- Service de médecine interne et immunologie clinique, CHU de Toulouse Rangueil, 2, rue Viguerie, 31059 Toulouse, France
| | - Alexandre Thibault Jacques Maria
- Médecine interne et immuno-oncologie (MedI20), CHU de Montpellier, hôpital Saint-Eloi, université de Montpellier, Institute for Regenerative Medicine and Biotherapy (IRMB), 80, avenue Augustin-Fliche, 34295 Montpellier, France
| | - Mathieu Puyade
- Service de médecine interne et maladies infectieuses, CIC-1402, CHU de Poitiers, 2, rue de la Milétrie, 86000 Poitiers, France; Université de Poitiers, 9, rue de la Milétrie, 86000 Poitiers, France
| | - Fanny Urbain
- Service de médecine interne 2, Sorbonne Université, Assistance publique-Hôpitaux de Paris (AP-HP), groupement hospitalier Pitié-Salpêtrière, Centre de référence pour le lupus, le syndrome des anti-phospholipides et autres maladies auto-immunes rares, Paris, France
| | - Louis Terriou
- Département de médecine interne et immunologie clinique, CHU de Lille, 59000 Lille, France; Centre de référence des maladies auto-immunes et auto-inflammatoires rares (CERAINO), 59000 Lille, France
| | - Vincent Poindron
- Service d'immunologie clinique et médecine interne, centre de références des maladies auto-immunes et systémiques rares (CNR RESO), hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France
| | - Marie Jachiet
- Service de dermatologie, hôpital Saint-Louis, Assistance publique-Hôpitaux de Paris, université Paris Cité, Paris, France
| | - Carlotta Cacciatore
- Unité de médecine interne (UF04) : CRMR MATHEC, maladies auto-immunes et thérapie cellulaire, Centre de référence des maladies auto-immunes systémiques rares d'Île-de-France, AP-HP, hôpital St-Louis, 75010 Paris, France; URP3518, IRSL, recherche clinique en hématologie, immunologie et transplantation, université Paris Cité, 75010 Paris, France
| | - Alain Lescoat
- Department of Internal Medicine & Clinical Immunology, Rennes University Hospital, 35000 Rennes, France
| | | | - Ingrid Munia
- Unité de médecine interne (UF04) : CRMR MATHEC, maladies auto-immunes et thérapie cellulaire, Centre de référence des maladies auto-immunes systémiques rares d'Île-de-France, AP-HP, hôpital St-Louis, 75010 Paris, France; URP3518, IRSL, recherche clinique en hématologie, immunologie et transplantation, université Paris Cité, 75010 Paris, France
| | - Isabelle Madelaine
- Pharmacie, hôpital Saint-Louis, AP-HP, Paris, France; Société française de pharmacie oncologique (SFPO), Paris, France
| | | | - Karin Tarte
- Équipe labellisée Ligue, UMR_ S 1236, Inserm, Université de Rennes, EFS Bretagne, Rennes, France
| | | | - Leonardo Magro
- Unité d'allogreffe, maladies du sang, CHRU, 59000 Lille, France
| | - Dominique Farge
- Service d'hématologie clinique et thérapie cellulaire, hôpital Saint-Antoine, AP-HP, 184, rue du Faubourg-Saint-Antoine, 75012 Paris, France; URP3518, IRSL, recherche clinique en hématologie, immunologie et transplantation, université Paris Cité, 75010 Paris, France; Department of Medicine, McGill University, H3A 1A1 Montreal, Canada
| | - Zora Marjanovic
- Service d'hématologie clinique et thérapie cellulaire, hôpital Saint-Antoine, AP-HP, 184, rue du Faubourg-Saint-Antoine, 75012 Paris, France.
| |
Collapse
|
3
|
Pierro M, Thébaud B. Cell-based strategies for the treatment of injury to the developing lung. THE LUNG 2025:403-426. [DOI: 10.1016/b978-0-323-91824-4.00020-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
4
|
Im GI. Clinical updates in mesenchymal stromal cell therapy for osteoarthritis treatment. Expert Opin Biol Ther 2025; 25:187-195. [PMID: 39710894 DOI: 10.1080/14712598.2024.2446612] [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: 07/26/2024] [Revised: 11/21/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION Osteoarthritis (OA) is a common chronic musculoskeletal disease with heterogeneous clinical manifestations and variable responses to different treatments. Unfortunately, there is no effective disease modifying therapy at present that can alter the natural course of the disease. Cell therapy based on mesenchymal stromal cells (MSCs) may offer an attractive therapeutic option for OA with their multiple modes of action, particularly immune-regulatory and regenerative capacities. AREAS COVERED In this narrative review, updates on mode of action based on patient's data, factors that can influence the efficacy of MSC treatment, current status in clinical application of MSCs as seen from randomized, controlled OA trials are introduced as well as the author's perspectives in the future of MSCs as OA therapeutics. EXPERT OPINION Symptomatic relief is not sufficient to justify the high cost associated with culture-expanded stem cells. Its advantages and efficacy over simple and low risk/cost modalities should be seriously reevaluated. Also, as the short-term strategy, efforts should be made to lower the cost of MSC therapy. In the future, multiomics technology may help to predict that subgroup of patients who will favorably respond to stem cell treatment, which would enhance the cost effectiveness and therapeutic benefit of MSC therapy.
Collapse
Affiliation(s)
- Gun-Il Im
- Department of Orthopedics, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| |
Collapse
|
5
|
Ljubić A, Dinić M, Švraka D, Vujović S. Dual-Double Stem Cell Ovarian Therapy: A Comprehensive Approach in Regenerative Medicine. Int J Mol Sci 2024; 26:69. [PMID: 39795929 PMCID: PMC11719681 DOI: 10.3390/ijms26010069] [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: 09/23/2024] [Revised: 11/10/2024] [Accepted: 11/27/2024] [Indexed: 01/13/2025] Open
Abstract
Dual-double stem cell therapy, which integrates mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs), represents a cutting-edge approach in regenerative medicine, particularly for conditions such as ovarian decline, premature ovarian insufficiency (POI), and induced ovarian failure. This therapy leverages the unique properties of MSCs and HSCs, enhancing tissue repair, immune modulation, and overall regenerative outcomes. MSCs, known for their ability to differentiate into various cell types, provide a supportive microenvironment and secrete bioactive molecules that promote angiogenesis and reduce inflammation. HSCs, crucial for hematopoiesis and immune function, further enhance this environment by supporting hematopoietic processes and immune regulation. Clinical evidence increasingly supports the effectiveness of stem cell therapy in ovarian regeneration. Studies have demonstrated improved folliculogenesis, normalization of hormone profiles, and successful pregnancies in patients with POI. Furthermore, recent clinical trials in various medical fields underline the superior potential of dual-double therapy compared to monotherapies involving MSCs or HSCs alone, enhancing tissue repair and functional outcomes. However, despite these benefits, the therapy presents risks that require careful consideration. For autologous MSC therapy involving expanded cell populations, risks include tumorigenic potential, with evidence of sarcoma formation in certain cases of cultured MSCs. In contrast, autologous non-expanded MSC and HSC therapies may be limited by low cell yields, potentially compromising therapeutic efficacy. Additionally, non-expanded HSC therapy poses risks of insufficient cell numbers for successful engraftment and delayed immune reconstitution. These considerations underscore the importance of quality control and rigorous screening to optimize safety and efficacy. This article explores the mechanisms of action, clinical applications, and potential complications of dual-double stem cell therapy, underscoring the need for continued research and optimized protocols to enhance safety and outcomes in ovarian insufficiency and related conditions, offering new hope for affected women.
Collapse
Affiliation(s)
- Aleksandar Ljubić
- Pronatal Hospital, 11000 Belgrade, Serbia;
- Academy of Sciences and Arts of Bosnia and Herzegovina, 71000 Sarajevo, Bosnia and Herzegovina
- Medigroup Health System, Dubrovnik International University, 20000 Dubrovnik, Croatia
| | - Marija Dinić
- Department of Therapeutic Apheresis, University Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | | | - Svetlana Vujović
- Clinic of Endocrinology, Diabetes and Diseases of National Center for Infertility and Endocrinology of Gender, 11000 Belgrade, Serbia;
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
6
|
Farge D, Biard L, Weil B, Girault V, Lansiaux P, Munia I, Loisel S, Charles C, Saout J, Resche-Rigon M, Korganow AS, Beuvon C, Pugnet G, Cacciatore C, Abisror N, Taupin JL, Cras A, Lowdell MW, Tarte K. Allogeneic umbilical cord-derived mesenchymal stromal cells as treatment for systemic lupus erythematosus: a single-centre, open-label, dose-escalation, phase 1 study. THE LANCET. RHEUMATOLOGY 2024:S2665-9913(24)00298-4. [PMID: 39706212 DOI: 10.1016/s2665-9913(24)00298-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Patients with systemic lupus erythematosus (SLE) with inadequate responses to standard therapies have unmet therapeutic needs. The immunomodulatory, proangiogenic, and antifibrotic properties of mesenchymal stromal cells support their use in treating patients with SLE. We aimed to assess the safety of a single intravenous infusion of allogeneic umbilical cord-derived mesenchymal stromal cells in patients with severe SLE. METHODS This prospective, single-centre, open-label, dose-escalation, Bayesian phase 1 study was done at the Saint-Louis University Hospital (Paris, France). Eligible patients were aged 18-70 years, were diagnosed with SLE according to American College of Rheumatology criteria with positive antinuclear antibodies, had a baseline Safety of Estrogens in Lupus Erythematosus National Assessment-SLE Disease Activity Index (SELENA-SLEDAI) score of 6 or more, and had disease that was refractory to first and second line SLE therapies. Patients were to receive a single intravenous infusion of 1 × 106, 2 × 106, or 4 × 106 umbilical cord-derived mesenchymal stromal cells per kg (manufactured from a single umbilical cord) in cohorts of five patients per dose, starting at 2 × 106 cells per kg. The primary endpoint was the rate of treatment-related severe adverse events (grade ≥3) in the first 10 days after infusion of umbilical cord-derived mesenchymal stromal cells. People with lived experience were involved in study design, patient enrolment, and dissemination of the study findings. This study is registered with ClinicalTrials.gov, NCT03562065, and the EU Clinical Trials Register, EudraCT2017-001400-29. FINDINGS From May 14, 2019, to March 6, 2023, 29 patients were screened for eligibility, eight of whom were enrolled in the study. Enrolment was terminated early after inclusion of eight patients and no patients received the 1 × 106 dose of umbilical cord-derived mesenchymal stromal cells. Seven (88%) of eight participants were cisgender women and one (13%) was a cisgender man. The median age was 35 years (range 26-57) and the median SLE disease duration was 12 years (5-19). All patients received at least 2 × 106 cells per kg (range 2 × 106 to 4 × 106). No severe adverse events and three infusion-related adverse events (two grade 1 and one grade 2) occurred in two patients in the first 10 days after infusion. After 12·4 months (range 12-13) of follow-up, no treatment-related severe adverse events and three non-treatment-related severe adverse events occurred in one patient after relapse. INTERPRETATION Our results suggest that a single infusion of 2 × 106 cells per kg or 4 × 106 cells per kg of allogeneic umbilical cord-derived mesenchymal stromal cells was safe in patients with severe SLE. Placebo-controlled trials are needed to confirm clinical efficacy and the role of B-cell modifications in clinical benefit. FUNDING Fondation du Rein, Alliance Maladies Rares AFM-Téléthon, Direction de la Recherche Clinique et de l'Innovation AP-HP, and ANR eCellFrance.
Collapse
Affiliation(s)
- Dominique Farge
- Unité de Médecine Interne (UF 04) CRMR MATHEC, Maladies Auto-Immunes et Thérapie Cellulaire, Centre de Référence des Maladies Auto-Immunes Systémiques Rares d'Ile-de-France MATHEC, AP-HP, Hôpital St-Louis, Paris, France; Université Paris Cité, IRSL, Recherche Clinique en Hématologie, Immunologie et Transplantation, URP3518, Paris, France; Department of Medicine, McGill University, Montreal, QC, Canada.
| | - Lucie Biard
- Université Paris Cité, AP-HP, Hôpital Saint Louis, Service de Biostatistique et Information Médicale (DMU PRISME), INSERM U1153 Team ECSTRRA, Paris, France
| | - Ben Weil
- Royal Free London NHS Foundation Trust, London, UK
| | - Virginie Girault
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France; INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Pauline Lansiaux
- Unité de Médecine Interne (UF 04) CRMR MATHEC, Maladies Auto-Immunes et Thérapie Cellulaire, Centre de Référence des Maladies Auto-Immunes Systémiques Rares d'Ile-de-France MATHEC, AP-HP, Hôpital St-Louis, Paris, France; Université Paris Cité, IRSL, Recherche Clinique en Hématologie, Immunologie et Transplantation, URP3518, Paris, France
| | - Ingrid Munia
- Unité de Médecine Interne (UF 04) CRMR MATHEC, Maladies Auto-Immunes et Thérapie Cellulaire, Centre de Référence des Maladies Auto-Immunes Systémiques Rares d'Ile-de-France MATHEC, AP-HP, Hôpital St-Louis, Paris, France; Université Paris Cité, IRSL, Recherche Clinique en Hématologie, Immunologie et Transplantation, URP3518, Paris, France
| | - Séverine Loisel
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France; INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Catney Charles
- Unité de Médecine Interne (UF 04) CRMR MATHEC, Maladies Auto-Immunes et Thérapie Cellulaire, Centre de Référence des Maladies Auto-Immunes Systémiques Rares d'Ile-de-France MATHEC, AP-HP, Hôpital St-Louis, Paris, France; Université Paris Cité, IRSL, Recherche Clinique en Hématologie, Immunologie et Transplantation, URP3518, Paris, France
| | - Judikael Saout
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France; INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Matthieu Resche-Rigon
- Université Paris Cité, AP-HP, Hôpital Saint Louis, Service de Biostatistique et Information Médicale (DMU PRISME), INSERM U1153 Team ECSTRRA, Paris, France
| | - Anne Sophie Korganow
- Hôpitaux Universitaires de Strasbourg, Département d'Immunologie Clinique, Centre National de Reference pour les Maladies Autoimmunes RESO, Université de Strasbourg, INSERM U1109, Strasbourg, France
| | - Clément Beuvon
- CHU de Poitiers, Service de Médecine Interne, 2, Rue de La Miletrie, Poitiers, France
| | - Grégory Pugnet
- Service de Médecine Interne et Immunologie Clinique Pôle Hospitalo-Universitaire des Maladies Digestives, CHU Rangueil, Toulouse, France
| | - Carlotta Cacciatore
- Unité de Médecine Interne (UF 04) CRMR MATHEC, Maladies Auto-Immunes et Thérapie Cellulaire, Centre de Référence des Maladies Auto-Immunes Systémiques Rares d'Ile-de-France MATHEC, AP-HP, Hôpital St-Louis, Paris, France; Université Paris Cité, IRSL, Recherche Clinique en Hématologie, Immunologie et Transplantation, URP3518, Paris, France
| | - Noémie Abisror
- Sorbonne Université, Service de Médecine Interne, AP-HP, Hôpital Saint Antoine, Paris, France
| | - Jean Luc Taupin
- INSERM U976 HIPI IRSL, Université Paris Cité, Laboratory of Immunology and Histocompatibility Hôpital Saint-Louis APHP, Paris, France
| | - Audrey Cras
- Cell Therapy Unit, AP-HP, Saint Louis Hospital, Paris, France; Université Paris Cité, INSERM UMR1140, Paris, France; INSERM, CIC de Biothérapies CBT501, Paris, France
| | | | - Karin Tarte
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France; INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France.
| |
Collapse
|
7
|
Lombardo G, Lechanteur C, Briquet A, Seidel L, Willems E, Servais S, Baudoux E, Kerre T, Zachee P, Herman J, Janssen A, Muller J, Baron F, Beguin Y. Co-infusion of mesenchymal stromal cells to prevent GVHD after allogeneic hematopoietic cell transplantation from HLA-mismatched unrelated donors after reduced-intensity conditioning: a double-blind randomized study and literature review. Stem Cell Res Ther 2024; 15:461. [PMID: 39627816 PMCID: PMC11613890 DOI: 10.1186/s13287-024-04064-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/14/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSC) have immunomodulatory and hematopoiesis-supporting properties that could potentially benefit hematopoietic stem cell (HSC) engraftment and decrease the incidence and/or severity of graft-versus-host disease (GVHD). METHODS Based on our previous pilot study, we established a multicenter, prospective, randomized, double-blind trial evaluating the efficacy of co-infusing third-party MSC (1.5-3 × 106/kg) versus placebo on the day of HSC transplantation (HCT) to prevent GVHD in recipients of HLA-mismatched unrelated donors after reduced-intensity conditioning. RESULTS The study planned to include 120 patients to improve 1-year overall survival (OS) from 55 to 77% but was stopped after 9 years for low recruitment (n = 38). One-year OS was 74% in the MSC group and 80% in the placebo group. In multivariate analysis, the incidence of grade II-IV acute GVHD was significantly lower in patients receiving MSC (HR 0.332, 95% CI 0.124-0.890, p = 0.0284). No difference was observed in the incidences of chronic GVHD, infection or relapse, overall or progression-free survival at 1 year or long-term, or hematopoietic and immune reconstitution. CONCLUSIONS Despite premature study closure, the suggested beneficial effect of MSC co-transplantation for the prevention of acute GVHD in HLA-mismatched HCT warrants further investigation.
Collapse
Affiliation(s)
- Gérôme Lombardo
- Department of Clinical Hematology, CHU of Liège, 1 Avenue de L'hôpital, 4000, Liège, Belgium
| | - Chantal Lechanteur
- Laboratory of Cell and Gene Therapy, CHU and University of Liège, Liège, Belgium
| | - Alexandra Briquet
- Laboratory of Cell and Gene Therapy, CHU and University of Liège, Liège, Belgium
| | - Laurence Seidel
- Center for Biostatistics and Research Methods, CHU and University of Liège, Liège, Belgium
| | - Evelyne Willems
- Department of Clinical Hematology, CHU of Liège, 1 Avenue de L'hôpital, 4000, Liège, Belgium
| | - Sophie Servais
- Department of Clinical Hematology, CHU of Liège, 1 Avenue de L'hôpital, 4000, Liège, Belgium
| | - Etienne Baudoux
- Laboratory of Cell and Gene Therapy, CHU and University of Liège, Liège, Belgium
| | - Tessa Kerre
- Department of Clinical Hematology, Ghent University Hospital, Ghent, Belgium
| | - Pierre Zachee
- Department of Clinical Hematology, ZNA Stuivenberg, Antwerp, Belgium
| | - Julie Herman
- Department of Clinical Hematology, CHU of Liège, 1 Avenue de L'hôpital, 4000, Liège, Belgium
| | - Audrey Janssen
- Department of Clinical Hematology, CHU of Liège, 1 Avenue de L'hôpital, 4000, Liège, Belgium
| | - Joséphine Muller
- Department of Clinical Hematology, CHU of Liège, 1 Avenue de L'hôpital, 4000, Liège, Belgium
| | - Frédéric Baron
- Department of Clinical Hematology, CHU of Liège, 1 Avenue de L'hôpital, 4000, Liège, Belgium
| | - Yves Beguin
- Department of Clinical Hematology, CHU of Liège, 1 Avenue de L'hôpital, 4000, Liège, Belgium.
- Laboratory of Cell and Gene Therapy, CHU and University of Liège, Liège, Belgium.
| |
Collapse
|
8
|
Wang WL, Lian H, Liang Y, Ye Y, Tam PKH, Chen Y. Molecular Mechanisms of Fibrosis in Cholestatic Liver Diseases and Regenerative Medicine-Based Therapies. Cells 2024; 13:1997. [PMID: 39682745 DOI: 10.3390/cells13231997] [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: 10/14/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
The aim of this review is to explore the potential of new regenerative medicine approaches in the treatment of cholestatic liver fibrosis. Cholestatic liver diseases, such as primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC), and biliary atresia (BA), due to the accumulation of bile, often progress to liver fibrosis, cirrhosis, and liver failure. When the disease becomes severe enough to require liver transplantation. Deeply understanding the disease's progression and fibrosis formation is crucial for better diagnosis and treatment. Current liver fibrosis treatments mainly target the root causes and no direct treatment method in fibrosis itself. Recent advances in regenerative medicine offer a potential approach that may help find the ways to target fibrosis directly, offering hope for improved outcomes. We also summarize, analyze, and discuss the current state and benefits of regenerative medicine therapies such as mesenchymal stem cell (MSC) therapy, induced pluripotent stem cells (iPSCs), and organoid technology, which may help the treatment of cholestatic liver diseases. Focusing on the latest research may reveal new targets and enhance therapeutic efficacy, potentially leading to more effective management and even curative strategies for cholestatic liver diseases.
Collapse
Affiliation(s)
- Wei-Lu Wang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
| | - Haoran Lian
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
| | - Yingyu Liang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
| | - Yongqin Ye
- Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
| | - Paul Kwong Hang Tam
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
- Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
- Precision Regenerative Medicine Research Centre, Medical Sciences Division, Macau University of Science and Technology, Macao SAR, China
| | - Yan Chen
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
- Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China
- Precision Regenerative Medicine Research Centre, Medical Sciences Division, Macau University of Science and Technology, Macao SAR, China
| |
Collapse
|
9
|
Bates SM, Evans KV, Delsing L, Wong R, Cornish G, Bahjat M. Immune safety challenges facing the preclinical assessment and clinical progression of cell therapies. Drug Discov Today 2024; 29:104239. [PMID: 39521331 DOI: 10.1016/j.drudis.2024.104239] [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: 04/08/2024] [Revised: 10/15/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
The promise of curative outcomes for life-limiting diseases using cell therapies is starting to become a reality, not only for patients with end-stage cancer, but also increasingly for regenerative therapies, including dentistry, ocular, neurodegenerative, and cardiac diseases. The introduction of often genetically modified cells into a patient can come with an extensive range of safety considerations. From an immune perspective, cell-based therapies carry inherent consequences and consideration of factors, such as the cell source (donor-derived autologous cells versus allogeneic cells), the intrinsic cellular nature of the therapy, and engineering/manufacturing methods, all of which influence the likelihood of inducing unwanted immune responses. Here, we provide an overview of the potential immune safety risks associated with cell therapies and explore possible mitigation approaches.
Collapse
Affiliation(s)
- Stephanie M Bates
- Safety Innovation, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Kelly V Evans
- Safety Innovation, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Louise Delsing
- Cell and Gene Therapy Safety, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ryan Wong
- Cell and Gene Therapy Safety, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Georgina Cornish
- Oncology Safety, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Mahnoush Bahjat
- Safety Innovation, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
10
|
Niebergall-Roth E, Dieter K, Frank MH, Kluth MA. Systemic treatment of recessive dystrophic epidermolysis bullosa with mesenchymal stromal cells: a scoping review of the literature and conclusions for future clinical research. J DERMATOL TREAT 2024; 35:2419931. [PMID: 39551482 DOI: 10.1080/09546634.2024.2419931] [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: 09/10/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Background: The ability of mesenchymal stromal cells (MSCs) to facilitate regenerative responses in inflamed and injured tissues, coupled with preclinical data suggesting potential to restore defective collagen VII at the dermo-epidermal junction, has raised the hope that MSCs may provide an effective disease-modifying therapy for patients suffering from recessive dystrophic epidermolysis bullosa (RDEB). Methods: We present a descriptive analysis of the clinical research on systemic MSC administration to RDEB patients available in PubMed, including six early-phase studies and one case report, involving 59 patients who received 1-3 intravenous infusions of MSCs from various sources. Results: Based on 133 MSC infusions, a total of 44 mostly mild adverse events were reported as definitely, possibly or likely related to the study treatment, only two of which led to treatment discontinuation. Improvements were seen in skin manifestations, disease activity, pain, pruritus and quality of life, with considerable heterogeneity in reported outcome variables and measurement tools between studies, and large inter-patient variability within studies. Conclusions: Although the current evidence base is limited, reflecting the typical challenges of clinical research in rare diseases, the reported results suggest potential treatment benefits for patients and provide a rationale for continuing to pursue this therapeutic approach.
Collapse
Affiliation(s)
| | | | - Markus H Frank
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
- Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | | |
Collapse
|
11
|
Sagar R, David AL. Fetal therapies - (Stem cell transplantation; enzyme replacement therapy; in utero genetic therapies). Best Pract Res Clin Obstet Gynaecol 2024; 97:102542. [PMID: 39298891 DOI: 10.1016/j.bpobgyn.2024.102542] [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: 08/06/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024]
Abstract
Advances in ultrasound and prenatal diagnosis are leading an expansion in the options for parents whose fetus is identified with a congenital disease. Obstetric diseases such as pre-eclampsia and fetal growth restriction may also be amenable to intervention to improve maternal and neonatal outcomes. Advanced Medicinal Therapeutic Products such as stem cell, gene, enzyme and protein therapies are most commonly being investigated as the trajectory of treatment for severe genetic diseases moves toward earlier intervention. Theoretical benefits include prevention of in utero damage, smaller treatment doses compared to postnatal intervention, use of fetal circulatory shunts and induction of immune tolerance. New systematic terminology can capture adverse maternal and fetal adverse events to improve safe trial conduct. First-in-human clinical trials are now beginning to generate results with a focus on safety first and efficacy second. If successful, these trials will transform the care of fetuses with severe early-onset congenital disease.
Collapse
Affiliation(s)
- Rachel Sagar
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, WC1E 6AU, UK.
| | - Anna L David
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, WC1E 6AU, UK; National Institute for Health and Care Research, University College London Hospitals NHS Foundation Trust Biomedical Research Centre, 149 Tottenham Court Road, London, W1T 7DN, UK.
| |
Collapse
|
12
|
Athiel Y, Cariot L, Jouannic JM, Maillet C, Mauffré V, Adam C, Huet H, Larghero J, Nasone J, Guilbaud L. Safety and efficacy of human umbilical cord-derived mesenchymal stromal cells in fetal ovine myelomeningocele repair. Stem Cell Res Ther 2024; 15:444. [PMID: 39568021 PMCID: PMC11580231 DOI: 10.1186/s13287-024-03991-y] [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: 07/10/2024] [Accepted: 10/09/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND The aim of this study was to assess the safety and efficacy of human umbilical cord mesenchymal stromal cells (hUC-MSCs) patch used as an adjuvant therapy in fetal myelomeningocele (MMC) surgery in the ovine model. METHODS hUC-MSCs were isolated from human umbilical cords (UC) using the explant method, cultured and characterized. hUC-MSCs were then embedded in a fibrin patch. MMC were surgically created at 75 days of gestation and repaired at 89 days of gestation in sheep fetuses. Two groups were compared: the hUC-MSCs group in which MMC was repaired using a cellular patch and the control group, in which MMC was repaired using an acellular patch. Safety was evaluated by clinical ewes' monitoring during gestation, and clinical and histological examinations of lambs after birth. Efficacy was assessed by clinical neurological evaluation at 2 and 24 h of life using the sheep locomotor rating scale and by histological analyses. RESULTS Among the 17 operated lambs, nine were born alive: six in the hUC-MSCs group and three in the control group. Overall fetal loss was 47% (8/17) without differences between the two groups. No fever was reported in ewes. No tumors were detected in clinical and histological examinations in the lambs. At 24 h of life, mean Sheep Locomotor Rating score was higher in the hUC-MSCs group than in the control group: 15.0 versus 2.0 (p = 0.07). Histological analyses showed a higher large neurons density in the hUC-MSCs group in comparison with the control group: 9.9 versus 6.3/mm2 of gray matter (p = 0.04). Lambs in the hUC-MSCs group had lower fibrosis around the spinal cord and at the level of the MMC scar: 70.9 versus 253.7 μm (p = 0.10) and 691.3 versus 1684.4 μm (p = 0,18), respectively. CONCLUSIONS Ovine fetal repair of MMC using human UC-MSCs seems to be an effective and safe procedure.
Collapse
Affiliation(s)
- Yoann Athiel
- Service de médecine foetale, DMU ORIGYN, APHP, Hôpital Trousseau, Sorbonne Université, Paris, France
- Unité de Thérapie Cellulaire, Université Paris Cité, AP-HP, Hôpital Saint-Louis, U976 et CIC de Biothérapies, INSERM, Paris, France
| | - Laura Cariot
- Service de médecine foetale, DMU ORIGYN, APHP, Hôpital Trousseau, Sorbonne Université, Paris, France
- Unité de Thérapie Cellulaire, Université Paris Cité, AP-HP, Hôpital Saint-Louis, U976 et CIC de Biothérapies, INSERM, Paris, France
| | - Jean-Marie Jouannic
- Service de médecine foetale, DMU ORIGYN, APHP, Hôpital Trousseau, Sorbonne Université, Paris, France
- Working Group Spina Bifida and Other Dysraphisms, European Reference Network ITHACA, Paris, France
| | - Corentin Maillet
- Service de médecine foetale, DMU ORIGYN, APHP, Hôpital Trousseau, Sorbonne Université, Paris, France
- Unité de Thérapie Cellulaire, Université Paris Cité, AP-HP, Hôpital Saint-Louis, U976 et CIC de Biothérapies, INSERM, Paris, France
| | - Vincent Mauffré
- École Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Clovis Adam
- Service d'anatomopathologie, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Hélène Huet
- École Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Jérôme Larghero
- Unité de Thérapie Cellulaire, Université Paris Cité, AP-HP, Hôpital Saint-Louis, U976 et CIC de Biothérapies, INSERM, Paris, France
| | - Justine Nasone
- Unité de Thérapie Cellulaire, Université Paris Cité, AP-HP, Hôpital Saint-Louis, U976 et CIC de Biothérapies, INSERM, Paris, France
| | - Lucie Guilbaud
- Service de médecine foetale, DMU ORIGYN, APHP, Hôpital Trousseau, Sorbonne Université, Paris, France.
- Unité de Thérapie Cellulaire, Université Paris Cité, AP-HP, Hôpital Saint-Louis, U976 et CIC de Biothérapies, INSERM, Paris, France.
- Working Group Spina Bifida and Other Dysraphisms, European Reference Network ITHACA, Paris, France.
| |
Collapse
|
13
|
Cridge H, Johnson V. Allogenic Adipose-Derived Mesenchymal Stem Cell Infusion for the Management of Acute-Onset Pancreatitis in Dogs: A Pilot Study. Animals (Basel) 2024; 14:2905. [PMID: 39409854 PMCID: PMC11482583 DOI: 10.3390/ani14192905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have significant anti-inflammatory properties and are beneficial in rodent models of pancreatitis. The safety and efficacy of MSCs is unknown in dogs with acute pancreatitis (AP). Dogs with AP who were treated with MSCs (n = 4) were identified prospectively for this pilot study from an academic hospital. Serum Spec cPL and C-reactive protein (CRP) concentrations were measured on the day of MSC administration and 2 days later. The clinical severity, via the Modified Clinical Activity Index (MCAI), was also calculated. Two dogs received MSCs shortly after AP diagnosis, while the remaining dogs received MSCs due to clinically refractory disease. Changes in Spec cPL, CRP, and MCAI in the MSC-treated dogs were compared to a control population (n = 7) receiving the standard-of-care treatment for AP. No significant differences were noted between the populations for changes in Spec cPL (p = 0.79), CRP (p = 0.67), or MCAI (p = 0.91). However, subjective clinical improvements were noted within 24 h of MSC infusion in the two dogs with previously refractory disease. MSC infusions appear safe in the management of AP in dogs and may be considered in refractory disease. However, given the nature of this pilot study and its limitations, larger randomized controlled clinical trials are needed to truly evaluate the efficacy of MSC infusions in dogs with AP.
Collapse
Affiliation(s)
- Harry Cridge
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 28824, USA
| | | |
Collapse
|
14
|
Liu C, Liu M, Liu X, Li B, Gao L, Wu S, Ji Q, Zhang Z, Zhang S, Xiao P, Lu J, Li J, Hu S. The efficacy and safety of third-party umbilical blood/umbilical cord mesenchymal stem cell assisted related haploid hematopoietic stem cell transplantation in pediatric patients with acute leukemia: an observational study. Ther Adv Hematol 2024; 15:20406207241277549. [PMID: 39372558 PMCID: PMC11452895 DOI: 10.1177/20406207241277549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/25/2024] [Indexed: 10/08/2024] Open
Abstract
Background There is limited data on third-party umbilical cord blood (UCB) or mesenchymal stem cell (MSC) transplantation-assisted haploidentical hematopoietic stem cell transplantation (haplo-HSCT) in pediatric patients. Objective To evaluate the efficacy and safety of UCB and MSC transplantation-assisted haplo-HSCT in pediatric patients with acute leukemia (AL). Design Observational study. Methods Clinical data of 152 children with AL undergoing haplo-HSCT at the Children's Hospital of Soochow University between January 2020 and June 2022 were collected. The patients were divided into the haplo-HSCT + UCB group (n = 76), haplo-HSCT + MSC group (n = 31), and haplo-HSCT group (n = 45). Hematopoietic reconstruction time, complications within 30 days after transplantation, and survival and recurrence at 3 years after transplantation were compared among the groups. Results Multivariate analysis revealed that haplo-HSCT with MSC and human leukocyte antigen (HLA) matching ⩾6/10 were independent factors reducing engraftment syndrome (ES) incidence. There were no significant differences among the groups in the hematopoietic reconstruction time or incidence of complications within 30 days after transplantation (p > 0.05). Overall survival, relapse-free survival, cumulative incidence of relapse, cumulative incidence of hematological relapse, and 3-year transplant-related mortality were not significantly different (p > 0.05). The incidence of adverse reactions in the haplo-HSCT + UCB group was 97.3% within 4 h after UCB infusion, with a particularly high occurrence rate of 94.7% for hypertension. No transfusion-related adverse reactions occurred after the transfusion of umbilical cord MSC in the haplo-HSCT + MSC group. Conclusion MSC-assisted haplo-HSCT can reduce ES incidence after transplantation in pediatric patients with AL. UCB infusion is associated with a high incidence of reversible hypertension. However, no adverse reactions were observed in umbilical cord MSC transfusion.
Collapse
Affiliation(s)
- Chang Liu
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Minyuan Liu
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Xin Liu
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Bohan Li
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Li Gao
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Shuiyan Wu
- Department of Hematological Intensive Care Unit, Children’s Hospital of Soochow University, Suzhou, China
| | - Qi Ji
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Zhiqi Zhang
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Senlin Zhang
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Peifang Xiao
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Jun Lu
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Jie Li
- Department of Hematology, Children’s Hospital of Soochow University, No.92 Zhongnan Street, Industrial Park, Suzhou 215003, China
| | - Shaoyan Hu
- Department of Hematology, Children’s Hospital of Soochow University, No. 92 Zhongnan Street, Industrial Park, Suzhou 215003, China
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, China
| |
Collapse
|
15
|
Cyr-Depauw C, Cook DP, Mižik I, Lesage F, Vadivel A, Renesme L, Deng Y, Zhong S, Bardin P, Xu L, Möbius MA, Marzahn J, Freund D, Stewart DJ, Vanderhyden BC, Rüdiger M, Thébaud B. Single-Cell RNA Sequencing Reveals Repair Features of Human Umbilical Cord Mesenchymal Stromal Cells. Am J Respir Crit Care Med 2024; 210:814-827. [PMID: 38564376 DOI: 10.1164/rccm.202310-1975oc] [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: 10/31/2023] [Accepted: 04/01/2024] [Indexed: 04/04/2024] Open
Abstract
Rationale: The chronic lung disease bronchopulmonary dysplasia (BPD) is the most severe complication of extreme prematurity. BPD results in impaired lung alveolar and vascular development and long-term respiratory morbidity, for which only supportive therapies exist. Umbilical cord-derived mesenchymal stromal cells (UC-MSCs) improve lung structure and function in experimental BPD. Results of clinical trials with MSCs for many disorders do not yet match the promising preclinical studies. A lack of specific criteria to define functionally distinct MSCs persists. Objectives: To determine and correlate single-cell UC-MSC transcriptomic profiles with therapeutic potential. Methods: UC-MSCs from five term donors and human neonatal dermal fibroblasts (HNDFs; control cells of mesenchymal origin) transcriptomes were investigated using single-cell RNA sequencing (scRNA-seq) analysis. The lung-protective effect of UC-MSCs with a distinct transcriptome and control HNDFs was tested in vivo in hyperoxia-induced neonatal lung injury in rats. Measurements and Main Results: UC-MSCs showed limited transcriptomic heterogeneity but were different from HNDFs. Gene Ontology enrichment analysis revealed distinct (progenitor-like and fibroblast-like) UC-MSC subpopulations. Only treatment with progenitor-like UC-MSCs improved lung function and structure and attenuated pulmonary hypertension in hyperoxia-exposed rat pups. Moreover, scRNA-seq identified major histocompatibility complex class I as a molecular marker of nontherapeutic cells and associated with decreased lung retention. Conclusions: UC-MSCs with a progenitor-like transcriptome, but not with a fibroblast-like transcriptome, provide lung protection in experimental BPD. High expression of major histocompatibility complex class I is associated with reduced therapeutic benefit. scRNA-seq may be useful to identify subsets of MSCs with superior repair capacity for clinical application.
Collapse
Affiliation(s)
- Chanèle Cyr-Depauw
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David P Cook
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ivana Mižik
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Flore Lesage
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Laurent Renesme
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Yupu Deng
- Sinclair Centre for Regenerative Medicine and
| | | | - Pauline Bardin
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Liqun Xu
- Sinclair Centre for Regenerative Medicine and
| | - Marius A Möbius
- Neonatology and Pediatric Critical Care Medicine, Department of Pediatrics, University Hospital Carl Gustav Carus, and
- Research Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany
| | - Jenny Marzahn
- Neonatology and Pediatric Critical Care Medicine, Department of Pediatrics, University Hospital Carl Gustav Carus, and
| | - Daniel Freund
- Research Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany
| | - Duncan J Stewart
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Barbara C Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Obstetrics and Gynecology, University of Ottawa/The Ottawa Hospital, Ottawa, Ontario, Canada; and
| | - Mario Rüdiger
- Neonatology and Pediatric Critical Care Medicine, Department of Pediatrics, University Hospital Carl Gustav Carus, and
| | - Bernard Thébaud
- Sinclair Centre for Regenerative Medicine and
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
16
|
Riazuelo L, Planat-Bénard V, Vinel A, Laurencin S, Casteilla L, Kémoun P, Marty M, Monsarrat P. Acceptability of Allogeneic Mesenchymal Stromal Cell-Based Tissue Engineering for the Treatment of Periodontitis: A Qualitative Study in France. Int Dent J 2024:S0020-6539(24)01398-4. [PMID: 39245621 DOI: 10.1016/j.identj.2024.07.1208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/15/2024] [Accepted: 07/20/2024] [Indexed: 09/10/2024] Open
Abstract
INTRODUCTION AND AIMS Periodontitis, the main cause of tooth loss in adults, is a public health concern; its incidence increases with age, and its prevalence increases with increasing life expectancy of the population. Innovative therapies such as cell therapy represent promising future solutions for guided tissue regeneration. However, these therapies may be associated with fears and mistrust from the general public. The aim of this study was to estimate the acceptability of an advanced therapy medicinal product combining allogeneic mesenchymal stromal cells from adipose tissue with a natural fibrin hydrogel in the treatment of periodontitis. METHODS The methodology was based on a qualitative study conducted through semi-structured interviews with patients followed for periodontitis in the Oral Medicine Department of the Toulouse University Hospital, Toulouse, France. Qualitative studies are essential methodologies to understand the patterns of health behaviours, describe illness experiences, and design health interventions in a humanistic and person-centred way of discovering. RESULTS Eleven interviews (with 4 men and 7 women) were required to reach thematic saturation. Analysis allowed 4 main themes to emerge: (1) perception of new treatments, science, and caregivers; (2) conditions that the treatment must meet; (3) patient perception of the disease; and (4) factors related to the content of the treatment. CONCLUSIONS Patients find cell therapy for periodontitis to be acceptable. If they express a need to be informed about the benefit/risk ratio, they are not particularly worried about side effects of the treatment, for either allogeneic or blood-derived products. Periodontitis is a prototypical model of chronic inflammatory pathology and is multitissular, with hard- and soft-tissue lesions. In a patient-centred approach, the success of cell therapy will require a bilateral, informed decision, taking into account potential therapeutic effectiveness and patient expectations for regeneration.
Collapse
Affiliation(s)
- Lucas Riazuelo
- Oral Medicine Department and CHU de Toulouse, Toulouse Institute of Oral Medicine and Science, Toulouse, France
| | - Valérie Planat-Bénard
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Alexia Vinel
- Oral Medicine Department and CHU de Toulouse, Toulouse Institute of Oral Medicine and Science, Toulouse, France; I2MC, INSERM UMR 1297, University of Toulouse III, Toulouse, France
| | - Sara Laurencin
- Oral Medicine Department and CHU de Toulouse, Toulouse Institute of Oral Medicine and Science, Toulouse, France; Center for Epidemiology and Research in POPulation Health (CERPOP), UMR 1295, Paul Sabatier University, Toulouse, France
| | - Louis Casteilla
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Philippe Kémoun
- Oral Medicine Department and CHU de Toulouse, Toulouse Institute of Oral Medicine and Science, Toulouse, France; RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France
| | - Mathieu Marty
- Oral Medicine Department and CHU de Toulouse, Toulouse Institute of Oral Medicine and Science, Toulouse, France; LIRDEF, Faculty of Educational Sciences, Paul Valery University, Montpellier, France
| | - Paul Monsarrat
- Oral Medicine Department and CHU de Toulouse, Toulouse Institute of Oral Medicine and Science, Toulouse, France; RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, Toulouse, France; Artificial and Natural Intelligence Toulouse Institute ANITI, Toulouse, France.
| |
Collapse
|
17
|
Tan Y, Salkhordeh M, Murray ABP, Souza-Moreira L, Stewart DJ, Mei SHJ. Key quality parameter comparison of mesenchymal stem cell product cryopreserved in different cryopreservation solutions for clinical applications. Front Bioeng Biotechnol 2024; 12:1412811. [PMID: 39148941 PMCID: PMC11324487 DOI: 10.3389/fbioe.2024.1412811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/12/2024] [Indexed: 08/17/2024] Open
Abstract
Introduction Cryopreservation is a critical process of cell products for achieving a commercial viability through wide scale adoption. By preserving cells in a lower temperature, cryopreservation enables a product to be off-the-shelf and ready for infusion. An optimized cryopreservation strategy can maintain the viability, phenotype, and potency of thawed mesenchymal stromal/stem cells (MSCs) while being regulatory compliant. We compared three clinical-ready formulations with one research cryopreservation solutions and evaluated key quality parameters of post thawed MSCs. Method and result MSCs were cryopreserved at 3, 6, and 9 million cells/mL (M/mL) in four different cryopreservation solutions: NutriFreez (10% dimethyl sulfoxide [DMSO]), Plasmalyte A (PLA)/5% human albumin (HA)/10% DMSO (PHD10), CryoStor CS5 (5% DMSO), and CryoStor CS10 (10% DMSO). To establish post thaw viability, cells were evaluated with no dilution of DMSO (from 3 M/mL), 1:1 dilution (from 6 M/mL), or 1:2 dilution (from 9 M/mL) with PLA/5% HA, to achieve uniform concentration at 3 M/mL. Cell viability was measured at 0-, 2-, 4-, and 6-h post thaw with Trypan blue exclusion and Annexin V/PI staining. Dilution (1:2) of final cell products from 9M/mL resulted in an improvement of cell viability over 6 h but showed a trend of decreased recovery. MSCs cryopreserved in solutions with 10% DMSO displayed comparable viabilities and recoveries up to 6 h after thawing, whereas a decreasing trend was noted in cell viability and recovery with CS5. Cells from all groups exhibited surface marker characteristics of MSCs. We further evaluated cell proliferation after 6-day recovery in culture. While cells cryopreserved in NutriFreez and PHD10 presented similar cell growth post thaw, MSCs cryopreserved in CS5 and CS10 at 3 M/mL and 6M/mL showed 10-fold less proliferative capacity. No significant differences were observed between MSCs cryopreserved in NutriFreez and PHD10 in their potency to inhibit T cell proliferation and improve monocytic phagocytosis. Conclusion MSCs can be cryopreserved up to 9 M/mL without losing notable viability and recovery, while exhibiting comparable post thaw potency with NutriFreez and PHD10. These results highlight the importance of key parameter testing for selecting the optimal cryopreservation solution for MSC-based therapy.
Collapse
Affiliation(s)
- Yuan Tan
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Mahmoud Salkhordeh
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Aidan B P Murray
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Luciana Souza-Moreira
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Duncan J Stewart
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Shirley H J Mei
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| |
Collapse
|
18
|
Chrościńska-Kawczyk M, Zdolińska-Malinowska I, Boruczkowski D. The Impact of Umbilical Cord Mesenchymal Stem Cells on Motor Function in Children with Cerebral Palsy: Results of a Real-world, Compassionate use Study. Stem Cell Rev Rep 2024; 20:1636-1649. [PMID: 38877284 DOI: 10.1007/s12015-024-10742-2] [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] [Accepted: 05/27/2024] [Indexed: 06/16/2024]
Abstract
The aim of this study was to analyze the impact of human umbilical cord-derived MSCs (hUC-MSCs) on motor function in children with cerebral palsy (CP). The study enrolled 152 children with CP who received up to two courses of five hUC-MSCs injections. Children's motor functions were assessed with the Gross Motor Function Measure (GMFM), 6-Minute Walk Test (6-MWT), Timed Up and Go test (Up&Go test), and Lovett's test, and mental abilities were assessed with the Clinical Global Impression (CGI) scale. Data collected at visit 1 (baseline) and visit 5 (after four injections) were analyzed retrospectively. After four hUC-MSCs administrations, all evaluated parameters improved. The change in GMFM score, by a median of 1.9 points (IQR: 0.0-8.0), correlated with age. This change was observed in all GFMCS groups and was noticed in all assessed GMFM areas. A median increase of 75 m (IQR: 20.0-115.0) was noted on the 6-MWT, and this correlated with GMFM score change. Time on the Up&Go test was reduced by a median of 2 s (IQR: -3 to - 1) and the change correlated with age, GMFM score at baseline, and the difference observed on the 6-MWT. Results of Lovett's test indicated slight changes in muscle strength. According to the CGI, 75.5% (96/151) of children were seriously (level VI) or significantly ill (level V) at the 1st visit, with any improvement observed in 63.6% (96/151) of patients at the 5th visit, 23.8% (36/151) with improvement (level II) or great improvement (level I). In conclusion, the application of hUC-MSCs generally enhanced functional performance, but individual responses varied. The therapy also benefited children with high level of disability but not to the same extent as the initially less disabled children. Although younger patients responded better to the treatment, older children can also benefit. Trial Registration 152/2018/KB/VII and 119/2021/KB/VIII. Retrospective registration in ClinicalTrials: ongoing.
Collapse
|
19
|
Tang J, Chen Y, Wang C, Xia Y, Yu T, Tang M, Meng K, Yin L, Yang Y, Shen L, Xing H, Mao X. The role of mesenchymal stem cells in cancer and prospects for their use in cancer therapeutics. MedComm (Beijing) 2024; 5:e663. [PMID: 39070181 PMCID: PMC11283587 DOI: 10.1002/mco2.663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are recruited by malignant tumor cells to the tumor microenvironment (TME) and play a crucial role in the initiation and progression of malignant tumors. This role encompasses immune evasion, promotion of angiogenesis, stimulation of cancer cell proliferation, correlation with cancer stem cells, multilineage differentiation within the TME, and development of treatment resistance. Simultaneously, extensive research is exploring the homing effect of MSCs and MSC-derived extracellular vesicles (MSCs-EVs) in tumors, aiming to design them as carriers for antitumor substances. These substances are targeted to deliver antitumor drugs to enhance drug efficacy while reducing drug toxicity. This paper provides a review of the supportive role of MSCs in tumor progression and the associated molecular mechanisms. Additionally, we summarize the latest therapeutic strategies involving engineered MSCs and MSCs-EVs in cancer treatment, including their utilization as carriers for gene therapeutic agents, chemotherapeutics, and oncolytic viruses. We also discuss the distribution and clearance of MSCs and MSCs-EVs upon entry into the body to elucidate the potential of targeted therapies based on MSCs and MSCs-EVs in cancer treatment, along with the challenges they face.
Collapse
Affiliation(s)
- Jian Tang
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Yu Chen
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
- Medical Affairs, Xiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Chunhua Wang
- Department of Clinical LaboratoryXiangyang No. 1 People's HospitalHubei University of MedicineXiangyangHubei ProvinceChina
| | - Ying Xia
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Tingyu Yu
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Mengjun Tang
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Kun Meng
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Lijuan Yin
- State Key Laboratory of Food Nutrition and SafetyKey Laboratory of Industrial MicrobiologyMinistry of EducationTianjin Key Laboratory of Industry MicrobiologyNational and Local United Engineering Lab of Metabolic Control Fermentation TechnologyChina International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal ChemistryCollege of BiotechnologyTianjin University of Science & TechnologyTianjinChina
| | - Yang Yang
- Shenzhen Key Laboratory of Pathogen and ImmunityNational Clinical Research Center for Infectious DiseaseState Key Discipline of Infectious DiseaseShenzhen Third People's HospitalSecond Hospital Affiliated to Southern University of Science and TechnologyShenzhenChina
| | - Liang Shen
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
| | - Hui Xing
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
- Department of Obstetrics and GynecologyXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and SciencesXiangyangChina
| | - Xiaogang Mao
- Central LaboratoryXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and ScienceXiangyangChina
- Department of Obstetrics and GynecologyXiangyang Central HospitalAffiliated Hospital of Hubei University of Arts and SciencesXiangyangChina
| |
Collapse
|
20
|
Česnik AB, Švajger U. The issue of heterogeneity of MSC-based advanced therapy medicinal products-a review. Front Cell Dev Biol 2024; 12:1400347. [PMID: 39129786 PMCID: PMC11310176 DOI: 10.3389/fcell.2024.1400347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024] Open
Abstract
Mesenchymal stromal stem cells (MSCs) possess a remarkable potential for numerous clinical applications due to their unique properties including self-renewal, immunomodulation, paracrine actions and multilineage differentiation. However, the translation of MSC-based Advanced Therapy Medicinal Products (ATMPs) into the clinic has frequently met with inconsistent outcomes. One of the suspected reasons for this issue is the inherent and extensive variability that exists among such ATMPs, which makes the interpretation of their clinical efficacy difficult to assess, as well as to compare the results of various studies. This variability stems from numerous reasons including differences in tissue sources, donor attributes, variances in manufacturing protocols, as well as modes of administration. MSCs can be isolated from various tissues including bone marrow, umbilical cord, adipose tissue and others, each with its unique phenotypic and functional characteristics. While MSCs from different sources do share common features, they also exhibit distinct gene expression profiles and functional properites. Donor-specific factors such as age, sex, body mass index, and underlying health conditions can influence MSC phenotype, morphology, differentiation potential and function. Moreover, variations in preparation of MSC products introduces additional heterogeneity as a result of cell culture media composition, presence or absence of added growth factors, use of different serum supplements and culturing techniques. Once MSC products are formulated, storage protocols play a pivotal role in its efficacy. Factors that affect cell viability include cell concentration, delivery solution and importantly, post-thawing protocols where applicable. Ensuing, differences in administration protocols can critically affect the distribution and functionallity of administered cells. As MSC-based therapies continue to advance through numerous clinical trials, implication of strategies to reduce product heterogeneity is imperative. Central to addressing these challenges is the need for precise prediction of clinical responses, which require well-defined MSC populations and harmonized assessment of their specific functions. By addressing these issues by meaningful approaches, such as, e.g., MSC pooling, the field can overcome barriers to advance towards more consistent and effective MSC-based therapies.
Collapse
Affiliation(s)
- Ana Bajc Česnik
- Slovenian Institute for Transfusion Medicine, Department for Therapeutic Services, Ljubljana, Slovenia
| | - Urban Švajger
- Slovenian Institute for Transfusion Medicine, Department for Therapeutic Services, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
21
|
Zhankina R, Zhanbyrbekuly U, Askarov M, Zare A, Jafari N, Saipiyeva D, Sherkhanov R, Akhmetov D, Hashemi A, Farjam M, Tanideh N, Aflatoonian B, Mussin NM, Kaliyev AA, Sultangereyev Y, Baneshi H, Shirazi R, Mahdipour M, Bakhshalizadeh S, Rahmanifar F, Tamadon A. Improving Fertility in Non-obstructive Azoospermia: Results from an Autologous Bone Mar-row-Derived Mesenchymal Stromal/Stem Cell Phase I Clinical Trial. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2024; 18:60-70. [PMID: 39033372 PMCID: PMC11263852 DOI: 10.22074/ijfs.2023.2005045.1480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 10/25/2023] [Accepted: 11/07/2023] [Indexed: 07/23/2024]
Abstract
BACKGROUND In this phase I clinical trial, our primary objective was to develop an innovative therapeutic approach utilizing autologous bone marrow-derived mesenchymal stromal/stem cells (BM-MSCs) for the treatment of nonobstructive azoospermia (NOA). Additionally, we aimed to assess the feasibility and safety of this approach. MATERIALS AND METHODS We recruited 80 participants in this non-randomized, open-label clinical trial, including patients undergoing NOA treatment using autologous BM-MSCs (n=40) and those receiving hormone therapy as a control group (n=40). Detailed participant characteristics, such as age, baseline hormonal profiles, etiology of NOA, and medical history, were thoroughly documented. Autotransplantation of BM-MSCs into the testicular network was achieved using microsurgical testicular sperm extraction (microTESE). Semen analysis and hormonal assessments were performed both before and six months after treatment. Additionally, we conducted an in-silico analysis to explore potential protein-protein interactions between exosomes secreted from BM-MSCs and receptors present in human seminiferous tubule cells. RESULTS Our results revealed significant improvements following treatment, including increased testosterone and inhibin B levels, elevated sperm concentration, and reduced levels of follicle-stimulating hormone (FSH), luteinizing hormone (LH), and prolactin. Notably, in nine patients (22.5%) previously diagnosed with secondary infertility and exhibiting azoospermia before treatment, the proposed approach yielded successful outcomes, as indicated by hormonal profile changes over six months. Importantly, these improvements were achieved without complications. Additionally, our in-silico analysis identified potential binding interactions between the protein content of BM-MSC-derived exosomes and receptors integral to spermatogenesis. CONCLUSION Autotransplantation of BM-MSCs into the testicular network using microTESE in NOA patients led to the regeneration of seminiferous tubules and the regulation of hormonal profiles governing spermatogenesis. Our findings support the safety and effectiveness of autologous BM-MSCs as a promising treatment modality for NOA, with a particular focus on the achieved outcomes in patients with secondary infertility (registration number: IRCT20190519043634N1).
Collapse
Affiliation(s)
- Rano Zhankina
- Department of Urology and Andrology, Astana Medical University, Astana, Kazakhstan
| | | | | | - Afshin Zare
- Department of R&D Research, PerciaVista R&D Co., Shiraz, Iran
| | - Nazanin Jafari
- Department of R&D Research, PerciaVista R&D Co., Shiraz, Iran
| | - Dana Saipiyeva
- Department of Urology and Andrology, Astana Medical University, Astana, Kazakhstan
| | - Ravil Sherkhanov
- Department of Urology and Andrology, Astana Medical University, Astana, Kazakhstan
| | - Daniyar Akhmetov
- Department of Urology and Andrology, Astana Medical University, Astana, Kazakhstan
| | - Alireza Hashemi
- Department of R&D Research, PerciaVista R&D Co., Shiraz, Iran
| | - Mojtaba Farjam
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Nader Tanideh
- Department of R&D Research, PerciaVista R&D Co., Shiraz, Iran
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmacology, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behrouz Aflatoonian
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Nadiar Maratovich Mussin
- Department of General Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Asset Askerovich Kaliyev
- Department of General Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Yerlan Sultangereyev
- Department of General Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
- Department of Surgery and Transplantation, Aktobe Medical Center, Aktobe, Kazakhstan
| | - Hanieh Baneshi
- Department of R&D Research, PerciaVista R&D Co., Shiraz, Iran
| | - Reza Shirazi
- Department of Anatomy, School of Biomedical Sciences, Medicine & Health, UNSW Sydney, Sydney, Australia
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shabnam Bakhshalizadeh
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Farhad Rahmanifar
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Amin Tamadon
- Department of R&D Research, PerciaVista R&D Co., Shiraz, Iran
- Department of Natural Sciences, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan.
| |
Collapse
|
22
|
Nie DQ, Yan GX, Wang ZY, Yan X, Yu GM, Gao JL, Liu D, Li HB. Combination treatment with interferon-γ may be a potential strategy to improve the efficacy of cytotherapy for rheumatoid arthritis: A network meta-analysis. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2024; 29:29. [PMID: 39239074 PMCID: PMC11376714 DOI: 10.4103/jrms.jrms_697_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 03/21/2022] [Accepted: 12/07/2022] [Indexed: 09/07/2024]
Abstract
Background Mesenchymal stem cells (MSCs) are considered a promising therapeutic strategy for rheumatoid arthritis (RA), but the current clinical results are varied. This study is to analyze the therapeutic effect of cell-based strategies on RA. Materials and Methods The searches were performed with public databases from inception to June 17, 2021. Randomized controlled trials researching cell-based therapies in RA patients were included. Results Eight studies, including 480 patients, were included in the analysis. The results showed that compared to the control, MSC treatment significantly reduced the disease activity score (DAS) at the second standardized mean difference (SMD): -0.70; 95% confidence interval (CI): -1.25, -0.15; P = 0.01) and 3rd month (SMD: -1.47; 95% CI: -2.77, -0.18; P < 0.01) and significantly reduced the rheumatoid factor (RF) level at the first (SMD: -0.38; 95% CI: -0.72, -0.05; P = 0.03) and 6th months (SMD: -0.81; 95% CI: -1.32, -0.31; P < 0.01). In the network meta-analysis, MSCs combined with interferon-γ (MSC_IFN) had a significant effect on increasing the American college of rheumatology criteria (ACR) 20, ACR50, and DAS <3.2 populations, had a significant effect on reducing the DAS, and decreased the RF level for a long period. Conclusion MSCs could relieve the DAS of RA patients in the short term and reduce the level of RF. MSC_IFN showed a more obvious effect, which could significantly improve the results of ACR20, ACR50, and DAS <3.2 and reduce the DAS and RF levels.
Collapse
Affiliation(s)
- Da-Qing Nie
- Department of Rheumatism, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Gui-Xiu Yan
- The Innovation Practice Center, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zheng-Yi Wang
- Department of College of Nursing and Health Science, Nanfang Medical College of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xue Yan
- Department of Rheumatism, The Third Clinical Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Gui-Mei Yu
- Department of Rheumatism, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Jin-Liang Gao
- Department of Rheumatism, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Di Liu
- Department of Rheumatism, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hong-Bo Li
- Department of Rheumatism, The Third Clinical Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
23
|
Botelho BF, Barreira AL, Filippo MG, Asensi KD, Faccioli LAP, dos Santos Salgado AB, de Salles EF, Marques CEC, Silva PL, dos Santos Goldenberg RC, Maiolino A, Gutfilen B, de Souza SAL, Junior ML, Morales MM. Safety and Biodistribution of an Autologous Bone Marrow-Derived Mononuclear Cell Infusion into Renal Arteries in Patients with Focal Segmental Glomerulosclerosis: A Phase 1 Study. Stem Cells Int 2024; 2024:2385568. [PMID: 39015674 PMCID: PMC11251782 DOI: 10.1155/2024/2385568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 04/26/2024] [Accepted: 06/12/2024] [Indexed: 07/18/2024] Open
Abstract
Patients with focal segmental glomerulosclerosis (FSGS) who are refractory to drug treatment may present progressive loss of kidney function, leading to chronic kidney disease stage 5 under dialysis treatment. The safety of systemic administration of bone marrow-derived mononuclear cells (BMDMCs) has been shown in different preclinical models of kidney diseases. However, to date, no study has evaluated the safety and biodistribution of BMDMCs after infusion in renal arteries in patients with FSGS. We used a prospective, non-randomized, single-center longitudinal design to investigate this approach. Five patients with refractory FSGS and an estimated glomerular filtration rate (eGFR) between 20 and 40 ml/min/1.73 m2 underwent bone marrow aspiration and received an arterial infusion of autologous BMDMCs (5 × 107) for each kidney. In addition, BMDMCs labeled with technetium-99m (99mTc-BMDMCs) were used to assess the biodistribution by scintigraphy. All patients completed the 270-day follow-up protocol with no serious adverse events. A transient increase in creatinine was observed after the cell therapy, with improvement on day 30. 99mTc-BMDMCs were detected in both kidneys and counts were higher after 2 hr compared with 24 hr. The arterial infusion of BMDMCs in both kidneys of patients with FSGS was considered safe with stable eGFR at the end of follow-up. This trial is registered with NCT02693366.
Collapse
Affiliation(s)
- Bruno Freire Botelho
- Department of NephrologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - André Luis Barreira
- Department of NephrologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcio Gomes Filippo
- Department of Vascular SurgeryClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Karina Dutra Asensi
- Cellular and Molecular Cardiology LaboratoryCarlos Chagas Filho Biophysics InstituteFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lanuza A P Faccioli
- Cellular and Molecular Cardiology LaboratoryCarlos Chagas Filho Biophysics InstituteFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anna Beatriz dos Santos Salgado
- Cellular and Molecular Cardiology LaboratoryCarlos Chagas Filho Biophysics InstituteFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elizabeth Figueiredo de Salles
- Department of RadiologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Eduardo Cruz Marques
- Department of RadiologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Leme Silva
- Laboratory of Pulmonary InvestigationCarlos Chagas Filho Biophysics InstituteFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Regina Coeli dos Santos Goldenberg
- Precision Medicine Research CenterCarlos Chagas Filho Institute of Biophysics e Brazilian Institute of Science and Technology—INCT REGENERA, Rio de Janeiro, Brazil
| | - Angelo Maiolino
- Department of HematologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bianca Gutfilen
- Department of RadiologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sergio Augusto Lopes de Souza
- Department of RadiologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maurilo Leite Junior
- Department of NephrologyClementino Fraga Filho University HospitalFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Marcos Morales
- Laboratory of Cellular and Molecular PhysiologyCarlos Chagas Filho Biophysics InstituteHealth Sciences CenterFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Zamanian MH, Norooznezhad AH, Hosseinkhani Z, Hassaninia D, Mansouri F, Vaziri S, Payandeh M, Heydarpour F, Kiani S, Shirvani M, Rajati M, Bakhtiari M, Esmaili F, Yarani R, Mansouri K. Human placental mesenchymal stromal cell-derived small extracellular vesicles as a treatment for severe COVID-19: A double-blind randomized controlled clinical trial. J Extracell Vesicles 2024; 13:e12492. [PMID: 39051747 PMCID: PMC11270582 DOI: 10.1002/jev2.12492] [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: 10/03/2023] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
The current study aimed to investigate the effects of human placental mesenchymal stromal cell-derived small extracellular vesicles (hPMSC-sEVs) as a treatment for COVID-19. This double-blind, randomized, controlled clinical trial was conducted on two groups of patients with COVID-19-associated acute respiratory distress syndrome. After randomization, the control group received standard treatment and placebo, and the intervention arm received standard treatment plus hPMSC-sEVs. The number of hospital deaths was considered the primary outcome. After meeting the exclusion and inclusion criteria, 21 and 24 patients were allocated to intervention and control arms, respectively. Besides admission SpO2 levels, which were significantly lower in the intervention arm (p = 0.008), all the baseline demo-biographic and laboratory variables were similar between the groups. It was shown that hPMSC-sEVs could significantly (p = 0.015) decrease the mortality ratio in the intervention group (4/21 [19.04%]) compared to the controls (13/24 [54.16%]). The mean time to death in the intervention and control groups was 28.06 and 11.10 days, respectively (p < 0.001). This study showed that hPMSC-sEVs are a possible treatment for critically ill patients with COVID-19.
Collapse
Affiliation(s)
| | - Amir Hossein Norooznezhad
- Medical Biology Research Centre, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| | - Zohreh Hosseinkhani
- Medical Biology Research Centre, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| | - Daryoush Hassaninia
- Infectious Diseases Research CenterKermanshah University of Medical SciencesKermanshahIran
| | - Feizollah Mansouri
- Infectious Diseases Research CenterKermanshah University of Medical SciencesKermanshahIran
| | - Siavash Vaziri
- Infectious Diseases Research CenterKermanshah University of Medical SciencesKermanshahIran
| | - Mehrdad Payandeh
- Bone Marrow Transplantation Department, School of Medicine, KermanshahUniversity of Medical SciencesKermanshahIran
| | - Fatemeh Heydarpour
- Medical Biology Research Centre, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| | - Sara Kiani
- Medical Biology Research Centre, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| | - Maria Shirvani
- Department of Infectious Disease, School of MedicineKermanshah University of Medical SciencesKermanshahIran
| | - Mojgan Rajati
- Motazedi Hospital, Kermanshah University of Medical SciencesKermanshahIran
| | - Mitra Bakhtiari
- Motazedi Hospital, Kermanshah University of Medical SciencesKermanshahIran
| | - Farzaneh Esmaili
- Motazedi Hospital, Kermanshah University of Medical SciencesKermanshahIran
| | - Reza Yarani
- Medical Biology Research Centre, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
- Translational Type 1 Diabetes, Department of Clinical ResearchSteno Diabetes Center CopenhagenHerlevDenmark
| | - Kamran Mansouri
- Medical Biology Research Centre, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| |
Collapse
|
25
|
Sagar RL, Åström E, Chitty LS, Crowe B, David AL, DeVile C, Forsmark A, Franzen V, Hermeren G, Hill M, Johansson M, Lindemans C, Lindgren P, Nijhuis W, Oepkes D, Rehberg M, Sahlin NE, Sakkers R, Semler O, Sundin M, Walther-Jallow L, Verweij EJTJ, Westgren M, Götherström C. An exploratory open-label multicentre phase I/II trial evaluating the safety and efficacy of postnatal or prenatal and postnatal administration of allogeneic expanded fetal mesenchymal stem cells for the treatment of severe osteogenesis imperfecta in infants and fetuses: the BOOSTB4 trial protocol. BMJ Open 2024; 14:e079767. [PMID: 38834319 PMCID: PMC11163617 DOI: 10.1136/bmjopen-2023-079767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 05/22/2024] [Indexed: 06/06/2024] Open
Abstract
INTRODUCTION Severe osteogenesis imperfecta (OI) is a debilitating disease with no cure or sufficiently effective treatment. Mesenchymal stem cells (MSCs) have good safety profile, show promising effects and can form bone. The Boost Brittle Bones Before Birth (BOOSTB4) trial evaluates administration of allogeneic expanded human first trimester fetal liver MSCs (BOOST cells) for OI type 3 or severe type 4. METHODS AND ANALYSIS BOOSTB4 is an exploratory, open-label, multiple dose, phase I/II clinical trial evaluating safety and efficacy of postnatal (n=15) or prenatal and postnatal (n=3, originally n=15) administration of BOOST cells for the treatment of severe OI compared with a combination of historical (1-5/subject) and untreated prospective controls (≤30). Infants<18 months of age (originally<12 months) and singleton pregnant women whose fetus has severe OI with confirmed glycine substitution in COL1A1 or COL1A2 can be included in the trial.Each subject receives four intravenous doses of 3×106/kg BOOST cells at 4 month intervals, with 48 (doses 1-2) or 24 (doses 3-4) hours in-patient follow-up, primary follow-up at 6 and 12 months after the last dose and long-term follow-up yearly until 10 years after the first dose. Prenatal subjects receive the first dose via ultrasound-guided injection into the umbilical vein within the fetal liver (16+0 to 35+6 weeks), and three doses postnatally.The primary outcome measures are safety and tolerability of repeated BOOST cell administration. The secondary outcome measures are number of fractures from baseline to primary and long-term follow-up, growth, change in bone mineral density, clinical OI status and biochemical bone turnover. ETHICS AND DISSEMINATION The trial is approved by Competent Authorities in Sweden, the UK and the Netherlands (postnatal only). Results from the trial will be disseminated via CTIS, ClinicalTrials.gov and in scientific open-access scientific journals. TRIAL REGISTRATION NUMBERS EudraCT 2015-003699-60, EUCT: 2023-504593-38-00, NCT03706482.
Collapse
Affiliation(s)
- Rachel L Sagar
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK
- NIHR University College London Hospitals Biomedical Research Centre, London, UK
| | - Eva Åström
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Lyn S Chitty
- North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- Genetics and Genomics, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Belinda Crowe
- Department of Neurosciences, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Anna L David
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK
- NIHR University College London Hospitals Biomedical Research Centre, London, UK
| | - Catherine DeVile
- Department of Neurosciences, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | | | | | - Göran Hermeren
- Department of Clinical Sciences, Lund University Faculty of Medicine, Lund, Sweden
| | - Melissa Hill
- North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- Genetics and Genomics, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Mats Johansson
- Department of Clinical Sciences, Lund University Faculty of Medicine, Lund, Sweden
| | - Caroline Lindemans
- Department of Pediatrics, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Peter Lindgren
- Center for Fetal Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Wouter Nijhuis
- Department of Orthopedic Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Dick Oepkes
- Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Mirko Rehberg
- Department of Pediatrics, University Hospital Cologne, Koln, Nordrhein-Westfalen, Germany
| | - Nils-Eric Sahlin
- Department of Clinical Sciences, Lund University Faculty of Medicine, Lund, Sweden
| | - Ralph Sakkers
- Department of Orthopedic Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - O Semler
- Department of Pediatrics, University Hospital Cologne, Koln, Nordrhein-Westfalen, Germany
| | - Mikael Sundin
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Section of Pediatic Hematology, Immunology and HCT, Karolinska University Hospital, Stockholm, Sweden
| | - Lilian Walther-Jallow
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - E J T Joanne Verweij
- Department of Obstetrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Magnus Westgren
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Cecilia Götherström
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
26
|
Marquez-Curtis LA, Elliott JAW. Mesenchymal stromal cells derived from various tissues: Biological, clinical and cryopreservation aspects: Update from 2015 review. Cryobiology 2024; 115:104856. [PMID: 38340887 DOI: 10.1016/j.cryobiol.2024.104856] [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: 11/28/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Mesenchymal stromal cells (MSCs) have become one of the most investigated and applied cells for cellular therapy and regenerative medicine. In this update of our review published in 2015, we show that studies continue to abound regarding the characterization of MSCs to distinguish them from other similar cell types, the discovery of new tissue sources of MSCs, and the confirmation of their properties and functions that render them suitable as a therapeutic. Because cryopreservation is widely recognized as the only technology that would enable the on-demand availability of MSCs, here we show that although the traditional method of cryopreserving cells by slow cooling in the presence of 10% dimethyl sulfoxide (Me2SO) continues to be used by many, several novel MSC cryopreservation approaches have emerged. As in our previous review, we conclude from these recent reports that viable and functional MSCs from diverse tissues can be recovered after cryopreservation using a variety of cryoprotectants, freezing protocols, storage temperatures, and periods of storage. We also show that for logistical reasons there are now more studies devoted to the cryopreservation of tissues from which MSCs are derived. A new topic included in this review covers the application in COVID-19 of MSCs arising from their immunomodulatory and antiviral properties. Due to the inherent heterogeneity in MSC populations from different sources there is still no standardized procedure for their isolation, identification, functional characterization, cryopreservation, and route of administration, and not likely to be a "one-size-fits-all" approach in their applications in cell-based therapy and regenerative medicine.
Collapse
Affiliation(s)
- Leah A Marquez-Curtis
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada, T6G 1H9; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada, T6G 1C9
| | - Janet A W Elliott
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada, T6G 1H9; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada, T6G 1C9.
| |
Collapse
|
27
|
Mello DB, Mesquita FCP, Silva dos Santos D, Asensi KD, Dias ML, Campos de Carvalho AC, Goldenberg RCDS, Kasai-Brunswick TH. Mesenchymal Stromal Cell-Based Products: Challenges and Clinical Therapeutic Options. Int J Mol Sci 2024; 25:6063. [PMID: 38892249 PMCID: PMC11173248 DOI: 10.3390/ijms25116063] [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: 05/02/2024] [Revised: 05/24/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Mesenchymal stromal cell (MSC)-based advanced therapy medicinal products (ATMPs) are being tried in a vast range of clinical applications. These cells can be isolated from different donor tissues by using several methods, or they can even be derived from induced pluripotent stem cells or embryonic stem cells. However, ATMP heterogeneity may impact product identity and potency, and, consequently, clinical trial outcomes. In this review, we discuss these topics and the need to establish minimal criteria regarding the manufacturing of MSCs so that these innovative therapeutics may be better positioned to contribute to the advancement of regenerative medicine.
Collapse
Affiliation(s)
- Debora B. Mello
- National Center of Structural Biology and Bioimaging, CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.B.M.); (A.C.C.d.C.)
| | | | - Danúbia Silva dos Santos
- Center of Cellular Technology, National Institute of Cardiology, INC, Rio de Janeiro 22240-002, Brazil;
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
| | - Karina Dutra Asensi
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Marlon Lemos Dias
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Antonio Carlos Campos de Carvalho
- National Center of Structural Biology and Bioimaging, CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.B.M.); (A.C.C.d.C.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Regina Coeli dos Santos Goldenberg
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Tais Hanae Kasai-Brunswick
- National Center of Structural Biology and Bioimaging, CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (D.B.M.); (A.C.C.d.C.)
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (K.D.A.); (R.C.d.S.G.)
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| |
Collapse
|
28
|
Ma W, Che J, Chen W, Wang D, Zhang H, Zhao Y. Dexamethasone-Integrated Mesenchymal Stem Cells for Systemic Lupus Erythematosus Treatment via Multiple Immunomodulatory Mechanisms. ACS NANO 2024; 18:13249-13265. [PMID: 38720584 DOI: 10.1021/acsnano.4c02420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The therapeutic application of mesenchymal stem cells (MSCs) has good potential as a treatment strategy for systemic lupus erythematosus (SLE), but traditional MSC therapy still has limitations in effectively modulating immune cells. Herein, we present a promising strategy based on dexamethasone liposome-integrated MSCs (Dexlip-MSCs) for treating SLE via multiple immunomodulatory pathways. This therapeutic strategy prolonged the circulation time of dexamethasone liposomes in vivo, restrained CD4+T-cell proliferation, and inhibited the release of proinflammatory mediators (IFN-γ and TNF-α) by CD4+T cells. In addition, Dexlip-MSCs initiated cellular reprogramming by activating the glucocorticoid receptor (GR) signaling pathway to upregulate the expression of anti-inflammatory factors such as cysteine-rich secretory protein LCCL-containing domain 2 (CRISPLD2) and downregulate the expression of proinflammatory factors. In addition, Dexlip-MSCs synergistically increased the anti-inflammatory inhibitory effect of CD4+T cells through the release of dexamethasone liposomes or Dex-integrated MSC-derived exosomes (Dex-MSC-EXOs). Based on these synergistic biological effects, we demonstrated that Dexlip-MSCs alleviated disease progression in MRL/lpr mice more effectively than Dexlip or MSCs alone. These features indicate that our stem cell delivery strategy is a promising therapeutic approach for clinical SLE treatment.
Collapse
Affiliation(s)
- Wenjuan Ma
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
- Department of Pharmacy, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Junyi Che
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Weiwei Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Dandan Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Huayong Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
29
|
Aliakbari F, Marzookian K, Parsafar S, Hourfar H, Nayeri Z, Fattahi A, Raeiji M, Boroujeni NN, Otzen DE, Morshedi D. The impact of hUC MSC-derived exosome-nanoliposome hybrids on α-synuclein fibrillation and neurotoxicity. SCIENCE ADVANCES 2024; 10:eadl3406. [PMID: 38569030 PMCID: PMC10990263 DOI: 10.1126/sciadv.adl3406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 02/28/2024] [Indexed: 04/05/2024]
Abstract
Amyloid aggregation of α-synuclein (αSN) protein amplifies the pathogenesis of neurodegenerative diseases (NDs) such as Parkinson's disease (PD). Consequently, blocking aggregation or redirecting self-assembly to less toxic aggregates could be therapeutic. Here, we improve brain-specific nanocarriers using a hybrid of exosomes (Ex) from human umbilical cord mesenchymal stem cells (hUC MSCs) and nanoliposomes containing baicalein (Ex-NLP-Ba) and oleuropein (Ex-NLP-Ole). The hybrids contained both lipid membranes, Ex proteins, and baicalein or oleuropein. Fluorescence resonance energy transfer analysis confirmed their proper integration. The hybrids reduced the extent of αSN fibrillation and interfered with secondary nucleation and disaggregation. They not only reduced αSN pathogenicity but also enhanced drug internalization into cells, surpassing the efficacy of NLP alone, and also crossed the blood-brain barrier in a cellular model. We conclude that Ex can be successfully extracted and efficiently merged with NLPs while retaining its original properties, demonstrating great potential as a theranostic drug delivery vehicle against NDs like PD.
Collapse
Affiliation(s)
- Farhang Aliakbari
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Kimia Marzookian
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Soha Parsafar
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Hamdam Hourfar
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Zahra Nayeri
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Arghavan Fattahi
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mohammad Raeiji
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Narges Nasrollahi Boroujeni
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Daniel E. Otzen
- Interdisciplinary Nanoscience Centre (iNANO) and Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark
| | - Dina Morshedi
- Bioprocess Engineering Department, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
30
|
Curley GF, O’Kane CM, McAuley DF, Matthay MA, Laffey JG. Cell-based Therapies for Acute Respiratory Distress Syndrome: Where Are We Now? Am J Respir Crit Care Med 2024; 209:789-797. [PMID: 38324017 PMCID: PMC10995569 DOI: 10.1164/rccm.202311-2046cp] [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: 11/08/2023] [Accepted: 02/07/2024] [Indexed: 02/08/2024] Open
Abstract
There is considerable interest in the potential for cell-based therapies, particularly mesenchymal stromal cells (MSCs) and their products, as a therapy for acute respiratory distress syndrome (ARDS). MSCs exert effects via diverse mechanisms including reducing excessive inflammation by modulating neutrophil, macrophage and T-cell function, decreasing pulmonary permeability and lung edema, and promoting tissue repair. Clinical studies indicate that MSCs are safe and well tolerated, with promising therapeutic benefits in specific clinical settings, leading to regulatory approvals of MSCs for specific indications in some countries.This perspective reassesses the therapeutic potential of MSC-based therapies for ARDS given insights from recent cell therapy trials in both COVID-19 and in 'classic' ARDS, and discusses studies in graft-vs.-host disease, one of the few licensed indications for MSC therapies. We identify important unknowns in the current literature, address challenges to clinical translation, and propose an approach to facilitate assessment of the therapeutic promise of MSC-based therapies for ARDS.
Collapse
Affiliation(s)
- Gerard F. Curley
- Department of Anaesthesia, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Cecilia M. O’Kane
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Daniel F. McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
- Department of Critical Care, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Michael A. Matthay
- Department of Medicine and Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California
| | - John G. Laffey
- Department of Anaesthesia and Intensive Care Medicine, Galway University Hospitals, Saolta University Healthcare System, Galway, Ireland; and
- Anaesthesia, School of Medicine, College of Medicine, Nursing and Health Sciences, and CÚRAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| |
Collapse
|
31
|
Greco R, Alexander T, Del Papa N, Müller F, Saccardi R, Sanchez-Guijo F, Schett G, Sharrack B, Snowden JA, Tarte K, Onida F, Sánchez-Ortega I, Burman J, Castilla Llorente C, Cervera R, Ciceri F, Doria A, Henes J, Lindsay J, Mackensen A, Muraro PA, Ricart E, Rovira M, Zuckerman T, Yakoub-Agha I, Farge D. Innovative cellular therapies for autoimmune diseases: expert-based position statement and clinical practice recommendations from the EBMT practice harmonization and guidelines committee. EClinicalMedicine 2024; 69:102476. [PMID: 38361991 PMCID: PMC10867419 DOI: 10.1016/j.eclinm.2024.102476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/05/2024] [Accepted: 01/24/2024] [Indexed: 02/17/2024] Open
Abstract
Autoimmune diseases (ADs) are characterized by loss of immune tolerance, high chronicity, with substantial morbidity and mortality, despite conventional immunosuppression (IS) or targeted disease modifying therapies (DMTs), which usually require repeated administration. Recently, novel cellular therapies (CT), including mesenchymal stromal cells (MSC), Chimeric Antigen Receptors T cells (CART) and regulatory T cells (Tregs), have been successfully adopted in ADs. An international expert panel of the European Society for Blood and Marrow Transplantation and the International Society for the Cell and Gene Therapy, reviewed all available evidence, based on the current literature and expert practices, on use of MSC, CART and Tregs, in AD patients with rheumatological, neurological, and gastroenterological indications. Expert-based consensus and recommendations for best practice and quality of patient care were developed to support clinicians, scientists, and their multidisciplinary teams, as well as patients and care providers and will be regularly updated.
Collapse
Affiliation(s)
- Raffaella Greco
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
- Co-Chair of the Practice Harmonization and Guidelines Committee of EBMT and Chair of the ADWP of the EBMT, Barcelona, Spain
| | - Tobias Alexander
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany
| | - Nicoletta Del Papa
- Scleroderma Clinic, Rheumatology Department, ASST G. Pini-CTO, Università degli Studi di Milano, Milano, Italy
| | - Fabian Müller
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen, Erlangen, Germany
- Bayrisches Zentrum für Krebsforschung (BZKF) Erlangen, Germany
| | - Riccardo Saccardi
- Cellular Therapies and Transfusion Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Fermin Sanchez-Guijo
- Department of Hematology, IBSAL-University Hospital of Salamanca and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Friedrich-Alexander University (FAU) Erlangen- Nürnberg, Erlangen, Germany
| | - Basil Sharrack
- Department of Neuroscience and Sheffield NIHR Translational Neuroscience BRC, Sheffield Teaching Hospitals NHS Foundation Trust & University of Sheffield, Sheffield, England, United Kingdom
| | - John A. Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Karin Tarte
- SITI Lab, CHU Rennes, EFS Bretagne, University Rennes, Rennes, France
| | - Francesco Onida
- Hematology & ASCT Unit, ASST Fatebenefratelli-Sacco, University of Milan, Italy
- Co-Chair of the Practice Harmonization and Guidelines Committee of EBMT, Spain
| | - Isabel Sánchez-Ortega
- Secretary of the Practice Harmonization and Guidelines Committee of EBMT, Barcelona, Spain
- EBMT Medical Officer, Executive Office, Barcelona, Spain
| | - Joachim Burman
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Ricard Cervera
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC, CSUR) of the Catalan and Spanish Health Systems/Member of ERN-ReCONNET, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Fabio Ciceri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine (DiMED), University of Padua, Padua, Italy
| | - Jörg Henes
- Center for Interdisciplinary Rheumatology, Immunology and Autoimmune diseases and Department of Internal Medicine II (Haematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Germany
| | - James Lindsay
- Department of Gastroenterology, The Royal London Hospital, Barts Health NHS Trust, London, UK
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Andreas Mackensen
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen, Erlangen, Germany
- Bayrisches Zentrum für Krebsforschung (BZKF) Erlangen, Germany
| | - Paolo A. Muraro
- Department of Brain Sciences, Imperial College London, London, UK
| | - Elena Ricart
- Gastroenterology Department. Hospital Clínic Barcelona. Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Montserrat Rovira
- BMT Unit, Haematology Department, Institute of Haematology and Oncology, IDIBAPS, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Foundation, Spain
| | - Tsila Zuckerman
- Rambam Health Care Campus and Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ibrahim Yakoub-Agha
- CHU de Lille, University Lille, INSERM U1286, Infinite, 59000, Lille, France
- Chair of the Practice Harmonization and Guidelines Committee of EBMT, Spain
| | - Dominique Farge
- Internal Medicine Unit (04): CRMR MATHEC, Maladies Auto-immunes et Thérapie Cellulaire, Centre de Référence des Maladies auto-immunes systémiques Rares d’Ile-de-France, AP-HP, St-Louis Hospital Paris-Cite University, France
- Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
32
|
Yue D, Wang F, Han Y, Xiong C, Yang R. Exosomes derived from umbilical cord mesenchymal stem cells ameliorate male infertility caused by busulfan in vivo and in vitro. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116063. [PMID: 38306818 DOI: 10.1016/j.ecoenv.2024.116063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
Environmental pollution has emerged as a global concern due to its detrimental effects on human health. One of the critical aspects of this concern is the impact of environmental pollution on sperm quality in males. Male factor infertility accounts for approximately 40%- 50% of all infertility cases. Nonobstructive azoospermia (NOA) is the most severe type of male infertility. Human umbilical cord mesenchymal stem cell (hUCMSC) exosomes enhance proliferation and migration, playing crucial roles in tissue and organ injury repair. However, whether hUCMSC exosomes impacting on NOA caused by chemotherapeutic agents remains unknown. This study aimed to explore the functional restoration and mechanism of hUCMSC exosomes on busulfan-induced injury in GC-1 spg cells and ICR mouse testes. Our results revealed that hUCMSC exosomes effectively promoted the proliferation and migration of busulfan-treated GC-1 spg cells. Additionally, oxidative stress and apoptosis were significantly reduced when hUCMSC exosomes were treated. Furthermore, the injection of hUCMSC exosomes into the testes of ICR mice treated with busulfan upregulated the expression of mouse germ cell-specific genes, such as vasa, miwi, Stra8 and Dazl. Moreover, the expression of cellular junction- and cytoskeleton-related genes, including connexin 43, ICAM-1, β-catenin and androgen receptor (AR), was increased in the testicular tissues treated with exosomes. Western blot analysis demonstrated significant downregulation of apoptosis-associated proteins, such as bax and caspase-3, and upregulation of bcl-2 in the mouse testicular tissues injected with hUCMSC exosomes. Further, the spermatogenesis in the experimental group of mice injected with exosomes showed partial restoration of spermatogenesis compared to the busulfan-treated group. Collectively, these findings provide evidence for the potential clinical applications of hUCMSC exosomes in cell repair and open up new avenues for the clinical treatment of NOA.
Collapse
Affiliation(s)
- Dezhi Yue
- Reproductive Medicine Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Fang Wang
- Laboratory Animal Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Ying Han
- Laboratory Animal Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Chengliang Xiong
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ruifeng Yang
- Reproductive Medicine Center, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China.
| |
Collapse
|
33
|
Cavaliere F, Allegri M, Apan A, Brazzi L, Carassiti M, Cohen E, DI Marco P, Langeron O, Rossi M, Spieth P, Turnbull D, Weber F. A year in review in Minerva Anestesiologica 2023: anesthesia, analgesia, and perioperative medicine. Minerva Anestesiol 2024; 90:222-234. [PMID: 38535972 DOI: 10.23736/s0375-9393.24.18067-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Affiliation(s)
- Franco Cavaliere
- IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy -
| | - Massimo Allegri
- Lemanic Center of Analgesia and Neuromodulation EHC, Morges, Switzerland
| | - Alparslan Apan
- Department of Anesthesiology and Intensive Care, Faculty of Medicine, University of Giresun, Giresun, Türkiye
| | - Luca Brazzi
- Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Massimiliano Carassiti
- Unit of Anesthesia, Intensive Care and Pain Management, Campus Bio-Medico University Hospital, Rome, Italy
| | - Edmond Cohen
- Department of Anesthesiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pierangelo DI Marco
- Department of Cardiovascular, Respiratory, Nephrologic, Anesthesiologic, and Geriatric Sciences, Faculty of Medicine, Sapienza University, Rome, Italy
| | - Olivier Langeron
- Department of Anesthesia and Intensive Care, Henri Mondor University Hospital, Assistance Publique - Hôpitaux de Paris (APHP), University Paris-Est Créteil (UPEC), Paris, France
| | - Marco Rossi
- IRCCS A. Gemelli University Polyclinic Foundation, Sacred Heart Catholic University, Rome, Italy
| | - Peter Spieth
- Department of Anesthesiology and Critical Care Medicine, University Hospital of Dresden, Dresden, Germany
| | - David Turnbull
- Department of Anesthetics and Neuro Critical Care, Royal Hallamshire Hospital, Sheffield, UK
| | - Frank Weber
- Department of Anesthesiology, Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
34
|
Bellon A. Comparing stem cells, transdifferentiation and brain organoids as tools for psychiatric research. Transl Psychiatry 2024; 14:127. [PMID: 38418498 PMCID: PMC10901833 DOI: 10.1038/s41398-024-02780-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 03/01/2024] Open
Abstract
The inaccessibility of neurons coming directly from patients has hindered our understanding of mental illnesses at the cellular level. To overcome this obstacle, six different cellular approaches that carry the genetic vulnerability to psychiatric disorders are currently available: Olfactory Neuroepithelial Cells, Mesenchymal Stem Cells, Pluripotent Monocytes, Induced Pluripotent Stem Cells, Induced Neuronal cells and more recently Brain Organoids. Here we contrast advantages and disadvantages of each of these six cell-based methodologies. Neuronal-like cells derived from pluripotent monocytes are presented in more detail as this technique was recently used in psychiatry for the first time. Among the parameters used for comparison are; accessibility, need for reprograming, time to deliver differentiated cells, differentiation efficiency, reproducibility of results and cost. We provide a timeline on the discovery of these cell-based methodologies, but, our main goal is to assist researchers selecting which cellular approach is best suited for any given project. This manuscript also aims to help readers better interpret results from the published literature. With this goal in mind, we end our work with a discussion about the differences and similarities between cell-based techniques and postmortem research, the only currently available tools that allow the study of mental illness in neurons or neuronal-like cells coming directly from patients.
Collapse
Affiliation(s)
- Alfredo Bellon
- Penn State Hershey Medical Center, Department of Psychiatry and Behavioral Health, Hershey, PA, USA.
- Penn State Hershey Medical Center, Department of Pharmacology, Hershey, PA, USA.
| |
Collapse
|
35
|
Lammi MJ, Qu C. Regulation of Oxygen Tension as a Strategy to Control Chondrocytic Phenotype for Cartilage Tissue Engineering and Regeneration. Bioengineering (Basel) 2024; 11:211. [PMID: 38534484 DOI: 10.3390/bioengineering11030211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 02/18/2024] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
Cartilage defects and osteoarthritis are health problems which are major burdens on health care systems globally, especially in aging populations. Cartilage is a vulnerable tissue, which generally faces a progressive degenerative process when injured. This makes it the 11th most common cause of global disability. Conservative methods are used to treat the initial phases of the illness, while orthopedic management is the method used for more progressed phases. These include, for instance, arthroscopic shaving, microfracturing and mosaicplasty, and joint replacement as the final treatment. Cell-based implantation methods have also been developed. Despite reports of successful treatments, they often suffer from the non-optimal nature of chondrocyte phenotype in the repair tissue. Thus, improved strategies to control the phenotype of the regenerating cells are needed. Avascular tissue cartilage relies on diffusion for nutrients acquisition and the removal of metabolic waste products. A low oxygen content is also present in cartilage, and the chondrocytes are, in fact, well adapted to it. Therefore, this raises an idea that the regulation of oxygen tension could be a strategy to control the chondrocyte phenotype expression, important in cartilage tissue for regenerative purposes. This narrative review discusses the aspects related to oxygen tension in the metabolism and regulation of articular and growth plate chondrocytes and progenitor cell phenotypes, and the role of some microenvironmental factors as regulators of chondrocytes.
Collapse
Affiliation(s)
- Mikko J Lammi
- Department of Medical and Translational Biology, Umeå University, SE-90187 Umeå, Sweden
| | - Chengjuan Qu
- Department of Odontology, Umeå University, SE-90187 Umeå, Sweden
| |
Collapse
|
36
|
Vo QD, Saito Y, Nakamura K, Iida T, Yuasa S. Induced Pluripotent Stem Cell-Derived Cardiomyocytes Therapy for Ischemic Heart Disease in Animal Model: A Meta-Analysis. Int J Mol Sci 2024; 25:987. [PMID: 38256060 PMCID: PMC10815661 DOI: 10.3390/ijms25020987] [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: 12/08/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Ischemic heart disease (IHD) poses a significant challenge in cardiovascular health, with current treatments showing limited success. Induced pluripotent derived-cardiomyocyte (iPSC-CM) therapy within regenerative medicine offers potential for IHD patients, although its clinical impacts remain uncertain. This study utilizes meta-analysis to assess iPSC-CM outcomes in terms of efficacy and safety in IHD animal model studies. A meta-analysis encompassing PUBMED, ScienceDirect, Web of Science, and the Cochrane Library databases, from inception until October 2023, investigated iPSC therapy effects on cardiac function and safety outcomes. Among 51 eligible studies involving 1012 animals, despite substantial heterogeneity, the iPSC-CM transplantation improved left ventricular ejection fraction (LVEF) by 8.23% (95% CI, 7.15 to 9.32%; p < 0.001) compared to control groups. Additionally, cell-based treatment reduced the left ventricle fibrosis area and showed a tendency to reduce left ventricular end-systolic volume (LVESV) and end-diastolic volume (LVEDV). No significant differences emerged in mortality and arrhythmia risk between iPSC-CM treatment and control groups. In conclusion, this meta-analysis indicates iPSC-CM therapy's promise as a safe and beneficial intervention for enhancing heart function in IHD. However, due to observed heterogeneity, the efficacy of this treatment must be further explored through large randomized controlled trials based on rigorous research design.
Collapse
Affiliation(s)
- Quan Duy Vo
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (Q.D.V.); (T.I.); (S.Y.)
| | - Yukihiro Saito
- Department of Cardiovascular Medicine, Okayama University Hospital, Okayama 700-8558, Japan
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (Q.D.V.); (T.I.); (S.Y.)
| | - Toshihiro Iida
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (Q.D.V.); (T.I.); (S.Y.)
| | - Shinsuke Yuasa
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (Q.D.V.); (T.I.); (S.Y.)
| |
Collapse
|
37
|
dos Santos CC, Lopes-Pacheco M, English K, Rolandsson Enes S, Krasnodembskaya A, Rocco PRM. The MSC-EV-microRNAome: A Perspective on Therapeutic Mechanisms of Action in Sepsis and ARDS. Cells 2024; 13:122. [PMID: 38247814 PMCID: PMC10813908 DOI: 10.3390/cells13020122] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) and MSC-derived extracellular vesicles (EVs) have emerged as innovative therapeutic agents for the treatment of sepsis and acute respiratory distress syndrome (ARDS). Although their potential remains undisputed in pre-clinical models, this has yet to be translated to the clinic. In this review, we focused on the role of microRNAs contained in MSC-derived EVs, the EV microRNAome, and their potential contribution to therapeutic mechanisms of action. The evidence that miRNA transfer in MSC-derived EVs has a role in the overall therapeutic effects is compelling. However, several questions remain regarding how to reconcile the stochiometric issue of the low copy numbers of the miRNAs present in the EV particles, how different miRNAs delivered simultaneously interact with their targets within recipient cells, and the best miRNA or combination of miRNAs to use as therapy, potency markers, and biomarkers of efficacy in the clinic. Here, we offer a molecular genetics and systems biology perspective on the function of EV microRNAs, their contribution to mechanisms of action, and their therapeutic potential.
Collapse
Affiliation(s)
- Claudia C. dos Santos
- Institute of Medical Sciences and Interdepartmental Division of Critical Care, Department of Medicine, University of Toronto, Toronto, ON M5B 1T8, Canada
- Keenan Center for Biomedical Research, Unity Health Toronto, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal;
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Karen English
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Ireland;
- Department of Biology, Maynooth University, W23 F2H6 Maynooth, Ireland
| | - Sara Rolandsson Enes
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22184 Lund, Sweden;
| | - Anna Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT9 7BL, UK;
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-599, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro 20020-000, Brazil
| |
Collapse
|
38
|
Şişli HB, Şenkal Turhan S, Bulut E, Şahin F, Doğan A. The Role of Aplnr Signaling in the Developmental Regulation of Mesenchymal Stem Cell Differentiation from Human Pluripotent Stem Cells. Adv Biol (Weinh) 2024; 8:e2300217. [PMID: 37840394 DOI: 10.1002/adbi.202300217] [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: 06/12/2023] [Revised: 09/01/2023] [Indexed: 10/17/2023]
Abstract
Stem cells are invaluable resources for personalized medicine. Mesenchymal stem cells (MSCs) have received great attention as therapeutic tools due to being a safe, ethical, and accessible option with immunomodulatory and controlled differentiation properties. Apelin receptor (Aplnr) signaling is reported to be involved in biological events, including gastrulation, mesoderm migration, proliferation of MSCs. However, the knowledge about the exact role and mechanism of Aplnr signaling during mesoderm and MSCs differentiation is still primitive. The current study aims to unveil the role of Aplnr signaling during mesoderm and MSC differentiation from pluripotent stem cells (PSCs) through peptide/small molecule activation, overexpression, knock down or CRISPR/Cas9 mediated knock out of the pathway components. Morphological changes, gene and protein expression analysis, including antibody array, LC/MS, mRNA/miRNA sequencing, reveal that Aplnr signaling promotes mesoderm commitment possibly via EGFR and TGF-beta signaling pathways and enhances migration of cells during mesoderm differentiation. Moreover, Aplnr signaling positively regulates MSCs differentiation from hPSCs and increases MSC characteristics and differentiation capacity by regulating pathways, such as EGFR, TGFβ, Wnt, PDGF, and FGF. Osteogenic, chondrogenic, adipogenic, and myogenic differentiations are significantly enhanced with Aplnr signaling activity. This study generates an important foundation to generate high potential MSCs from PSCs to be used in personalized cell therapy.
Collapse
Affiliation(s)
- Hatice Burcu Şişli
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Selinay Şenkal Turhan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Ezgi Bulut
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Fikrettin Şahin
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Ayşegül Doğan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| |
Collapse
|
39
|
Kresse JC, Gregersen E, Atay JCL, Eijken M, Nørregaard R. Does the route matter? A preclinical review of mesenchymal stromal cell delivery to the kidney. APMIS 2023; 131:687-697. [PMID: 37750005 DOI: 10.1111/apm.13352] [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: 09/01/2023] [Accepted: 09/08/2023] [Indexed: 09/27/2023]
Abstract
Mesenchymal stromal/stem cell (MSC) therapy has been thoroughly tested in preclinical animal models and holds great promise for the treatment of kidney diseases. It is becoming increasingly evident that the efficacy of MSC therapy is dependent on several factors including dosage, the tissue source of MSCs, the route of delivery and timing of administration. In a time where MSC therapy is moving from preclinical research to clinically therapeutic use, the importance of choice of delivery method, modality, and administration route increases. In this review, we provide an overview of the different MSC delivery routes used in preclinical kidney disease models, highlight the recent advances in the field, and summarize studies comparing delivery routes of MSCs to the kidney.
Collapse
Affiliation(s)
| | - Emil Gregersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Marco Eijken
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
40
|
Lu B, Lerman LO. MSC therapy for diabetic kidney disease and nephrotic syndrome. Nat Rev Nephrol 2023; 19:754-755. [PMID: 37783947 DOI: 10.1038/s41581-023-00776-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Affiliation(s)
- Bo Lu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
41
|
Zaripova LN, Midgley A, Christmas SE, Beresford MW, Pain C, Baildam EM, Oldershaw RA. Mesenchymal Stem Cells in the Pathogenesis and Therapy of Autoimmune and Autoinflammatory Diseases. Int J Mol Sci 2023; 24:16040. [PMID: 38003230 PMCID: PMC10671211 DOI: 10.3390/ijms242216040] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Mesenchymal stem cells (MSCs) modulate immune responses and maintain self-tolerance. Their trophic activities and regenerative properties make them potential immunosuppressants for treating autoimmune and autoinflammatory diseases. MSCs are drawn to sites of injury and inflammation where they can both reduce inflammation and contribute to tissue regeneration. An increased understanding of the role of MSCs in the development and progression of autoimmune disorders has revealed that MSCs are passive targets in the inflammatory process, becoming impaired by it and exhibiting loss of immunomodulatory activity. MSCs have been considered as potential novel cell therapies for severe autoimmune and autoinflammatory diseases, which at present have only disease modifying rather than curative treatment options. MSCs are emerging as potential therapies for severe autoimmune and autoinflammatory diseases. Clinical application of MSCs in rare cases of severe disease in which other existing treatment modalities have failed, have demonstrated potential use in treating multiple diseases, including rheumatoid arthritis, systemic lupus erythematosus, myocardial infarction, liver cirrhosis, spinal cord injury, multiple sclerosis, and COVID-19 pneumonia. This review explores the biological mechanisms behind the role of MSCs in autoimmune and autoinflammatory diseases. It also covers their immunomodulatory capabilities, potential therapeutic applications, and the challenges and risks associated with MSC therapy.
Collapse
Affiliation(s)
- Lina N. Zaripova
- Institute of Fundamental and Applied Medicine, National Scientific Medical Center, 42 Abylai Khan Avenue, Astana 010000, Kazakhstan;
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| | - Angela Midgley
- Department of Women and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Institute in the Park, Alder Hey Children’s NHS Foundation Trust, Liverpool L14 5AB, UK; (A.M.); (M.W.B.); (C.P.)
| | - Stephen E. Christmas
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, Faculty of Health and Life Sciences, University of Liverpool, The Ronald Ross Building, 8 West Derby Street, Liverpool L69 7BE, UK;
| | - Michael W. Beresford
- Department of Women and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Institute in the Park, Alder Hey Children’s NHS Foundation Trust, Liverpool L14 5AB, UK; (A.M.); (M.W.B.); (C.P.)
- Department of Paediatric Rheumatology, Alder Hey Children’s NHS Foundation Trust, East Prescott Road, Liverpool L14 5AB, UK
| | - Clare Pain
- Department of Women and Children’s Health, Institute of Life Course and Medical Sciences, University of Liverpool, Institute in the Park, Alder Hey Children’s NHS Foundation Trust, Liverpool L14 5AB, UK; (A.M.); (M.W.B.); (C.P.)
- Department of Paediatric Rheumatology, Alder Hey Children’s NHS Foundation Trust, East Prescott Road, Liverpool L14 5AB, UK
| | - Eileen M. Baildam
- Department of Paediatric Rheumatology, The Alexandra Hospital, Mill Lane, Cheadle SK8 2PX, UK;
| | - Rachel A. Oldershaw
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK
| |
Collapse
|
42
|
Zeiser R, Ringden O, Sadeghi B, Gonen-Yaacovi G, Segurado OG. Novel therapies for graft versus host disease with a focus on cell therapies. Front Immunol 2023; 14:1241068. [PMID: 37868964 PMCID: PMC10585098 DOI: 10.3389/fimmu.2023.1241068] [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: 06/16/2023] [Accepted: 09/11/2023] [Indexed: 10/24/2023] Open
Abstract
Graft versus host disease (GVHD) can occur at any period post allogeneic hematopoietic stem cell transplantation as a common clinical complication contributing to significant morbidity and mortality. Acute GVHD develops in approximately 30-50% of patients receiving transplants from matched related donors. High doses of steroids are used as first-line treatment, but are unsuccessful in around 40% of patients, resulting in the diagnosis of steroid-refractory acute GVHD. Consensus has yet to develop for the management of steroid-refractory acute GVHD, and prognosis at six months has been estimated at around 50%. Thus, it is critical to find effective treatments that increase survival of steroid-refractory acute GVHD. This article describes the currently known characteristics, pathophysiology, and treatments for GVHD, with a special focus on recent advances in cell therapies. In particular, a novel cell therapy using decidua stromal cells (DSCs) was recently shown to have promising results for acute GVHD, with improved effectiveness over previous treatments including mesenchymal stromal cells. At the Karolinska Institute, severe acute GVHD patients treated with placenta-derived DSCs supplemented with either 5% albumin or 10% AB plasma displayed a one-year survival rate of 76% and 47% respectively. Furthermore, patients with steroid-refractory acute GVHD, displayed survival rates of 73% with albumin and 31% with AB plasma-supplemented DSCs, compared to the 20% survival rate in the mesenchymal stromal cell control group. Adverse events and deaths were found to be attributed only to complications of hematopoietic stem cell transplant and GVHD, not to the study intervention. ASC Therapeutics, Inc, in collaboration with the Karolinska Institute, will soon initiate a phase 2 multicenter, open-label study to further assess the efficacy and safety of intravenous DSC treatment in sixty patients with Grade II-IV steroid-refractory acute GVHD. This novel cell therapy represents a promising treatment to combat the poor prognosis that steroid-refractory acute GVHD patients currently face.
Collapse
Affiliation(s)
- Robert Zeiser
- Department of Medicine at the University of Freiburg, Freiburg, Germany
| | - Olle Ringden
- Department of Clinical Sciences, Karolinska Institute, Stockholm, Sweden
| | - Behnam Sadeghi
- Department of Clinical Sciences, Karolinska Institute, Stockholm, Sweden
| | | | | |
Collapse
|
43
|
Perico N, Remuzzi G, Griffin MD, Cockwell P, Maxwell AP, Casiraghi F, Rubis N, Peracchi T, Villa A, Todeschini M, Carrara F, Magee BA, Ruggenenti PL, Rota S, Cappelletti L, McInerney V, Griffin TP, Islam MN, Introna M, Pedrini O, Golay J, Finnerty AA, Smythe J, Fibbe WE, Elliman SJ, O'Brien T. Safety and Preliminary Efficacy of Mesenchymal Stromal Cell (ORBCEL-M) Therapy in Diabetic Kidney Disease: A Randomized Clinical Trial (NEPHSTROM). J Am Soc Nephrol 2023; 34:1733-1751. [PMID: 37560967 PMCID: PMC10561817 DOI: 10.1681/asn.0000000000000189] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/26/2023] [Indexed: 08/11/2023] Open
Abstract
SIGNIFICANCE STATEMENT Mesenchymal stromal cells (MSCs) may offer a novel therapy for diabetic kidney disease (DKD), although clinical translation of this approach has been limited. The authors present findings from the first, lowest dose cohort of 16 adults with type 2 diabetes and progressive DKD participating in a randomized, placebo-controlled, dose-escalation phase 1b/2a trial of next-generation bone marrow-derived, anti-CD362 antibody-selected allogeneic MSCs (ORBCEL-M). A single intravenous (iv) infusion of 80×10 6 cells was safe and well-tolerated, with one quickly resolved infusion reaction in the placebo group and no subsequent treatment-related serious adverse events (SAEs). Compared with placebo, the median annual rate of decline in eGFR was significantly lower with ORBCEL-M, although mGFR did not differ. The results support further investigation of ORBCEL-M in this patient population in an appropriately sized phase 2b study. BACKGROUND Systemic therapy with mesenchymal stromal cells may target maladaptive processes involved in diabetic kidney disease progression. However, clinical translation of this approach has been limited. METHODS The Novel Stromal Cell Therapy for Diabetic Kidney Disease (NEPHSTROM) study, a randomized, placebo-controlled phase 1b/2a trial, assesses safety, tolerability, and preliminary efficacy of next-generation bone marrow-derived, anti-CD362-selected, allogeneic mesenchymal stromal cells (ORBCEL-M) in adults with type 2 diabetes and progressive diabetic kidney disease. This first, lowest dose cohort of 16 participants at three European sites was randomized (3:1) to receive intravenous infusion of ORBCEL-M (80×10 6 cells, n =12) or placebo ( n =4) and was followed for 18 months. RESULTS At baseline, all participants were negative for anti-HLA antibodies and the measured GFR (mGFR) and estimated GFR were comparable between groups. The intervention was safe and well-tolerated. One placebo-treated participant had a quickly resolved infusion reaction (bronchospasm), with no subsequent treatment-related serious adverse events. Two ORBCEL-M recipients died during follow-up of causes deemed unrelated to the trial intervention; one recipient developed low-level anti-HLA antibodies. The median annual rate of kidney function decline after ORBCEL-M therapy compared with placebo did not differ by mGFR, but was significantly lower by eGFR estimated by the Chronic Kidney Disease Epidemiology Collaboration and Modification of Diet in Renal Disease equations. Immunologic profiling provided evidence of preservation of circulating regulatory T cells, lower natural killer T cells, and stabilization of inflammatory monocyte subsets in those receiving the cell therapy compared with placebo. CONCLUSIONS Findings indicate safety and tolerability of intravenous ORBCEL-M cell therapy in the trial's lowest dose cohort. The rate of decline in eGFR (but not mGFR) over 18 months was significantly lower among those receiving cell therapy compared with placebo. Further studies will be needed to determine the therapy's effect on CKD progression. CLINICAL TRIAL REGISTRATION NUMBER ClinicalTrial.gov NCT02585622 .
Collapse
Affiliation(s)
- Norberto Perico
- Centro di Ricerche Cliniche per le Malattie Rare “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Centro di Ricerche Cliniche per le Malattie Rare “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Matthew D. Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland
| | - Paul Cockwell
- Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Institute of Ageing and Immunity, University of Birmingham, Birmingham, United Kingdom
| | | | - Federica Casiraghi
- Centro di Ricerche Cliniche per le Malattie Rare “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Nadia Rubis
- Centro di Ricerche Cliniche per le Malattie Rare “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Tobia Peracchi
- Centro di Ricerche Cliniche per le Malattie Rare “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Alessandro Villa
- Centro di Ricerche Cliniche per le Malattie Rare “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Marta Todeschini
- Centro di Ricerche Cliniche per le Malattie Rare “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Fabiola Carrara
- Centro di Ricerche Cliniche per le Malattie Rare “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Bernadette A. Magee
- Northern Ireland Histocompatibility and Immunogenetics Laboratory, Belfast City Hospital, Belfast, Northern Ireland
| | - Piero L. Ruggenenti
- Centro di Ricerche Cliniche per le Malattie Rare “Aldo e Cele Daccò”, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
- Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Stefano Rota
- Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Laura Cappelletti
- Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Veronica McInerney
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland
- HRB Clinical Research Facility, University of Galway, Galway, Ireland
| | - Tomás P. Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland
| | - Md Nahidul Islam
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland
| | - Martino Introna
- Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Olga Pedrini
- Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- Fondazione per la Ricerca Ospedale di Bergamo (FROM), Bergamo, Italy
| | - Josée Golay
- Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Andrew A. Finnerty
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland
- HRB Clinical Research Facility, University of Galway, Galway, Ireland
- Centre for Cell Manufacturing Ireland, University of Galway, Galway, Ireland
| | - Jon Smythe
- NHS Blood and Transplant Oxford Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | | | | | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre for Medical Devices, School of Medicine, University of Galway, Galway, Ireland
| |
Collapse
|
44
|
Russo E, Alberti G, Corrao S, Borlongan CV, Miceli V, Conaldi PG, Di Gaudio F, La Rocca G. The Truth Is Out There: Biological Features and Clinical Indications of Extracellular Vesicles from Human Perinatal Stem Cells. Cells 2023; 12:2347. [PMID: 37830562 PMCID: PMC10571796 DOI: 10.3390/cells12192347] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023] Open
Abstract
The potential of perinatal tissues to provide cellular populations to be used in different applications of regenerative medicine is well established. Recently, the efforts of researchers are being addressed regarding the evaluation of cell products (secreted molecules or extracellular vesicles, EVs) to be used as an alternative to cellular infusion. The data regarding the effective recapitulation of most perinatal cells' properties by their secreted complement point in this direction. EVs secreted from perinatal cells exhibit key therapeutic effects such as tissue repair and regeneration, the suppression of inflammatory responses, immune system modulation, and a variety of other functions. Although the properties of EVs from perinatal derivatives and their significant potential for therapeutic success are amply recognized, several challenges still remain that need to be addressed. In the present review, we provide an up-to-date analysis of the most recent results in the field, which can be addressed in future research in order to overcome the challenges that are still present in the characterization and utilization of the secreted complement of perinatal cells and, in particular, mesenchymal stromal cells.
Collapse
Affiliation(s)
- Eleonora Russo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| | - Giusi Alberti
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| | - Simona Corrao
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | - Cesar V. Borlongan
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA;
| | - Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (S.C.); (V.M.); (P.G.C.)
| | - Francesca Di Gaudio
- Department of Health Promotion, Maternal-Infantile Care, Excellence Internal and Specialist Medicine “G. D’Alessandro” (PROMISE), University of Palermo, 90127 Palermo, Italy;
| | - Giampiero La Rocca
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (E.R.); (G.A.)
| |
Collapse
|
45
|
Araldi RP, Delvalle DA, da Costa VR, Alievi AL, Teixeira MR, Dias Pinto JR, Kerkis I. Exosomes as a Nano-Carrier for Chemotherapeutics: A New Era of Oncology. Cells 2023; 12:2144. [PMID: 37681875 PMCID: PMC10486723 DOI: 10.3390/cells12172144] [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: 07/31/2023] [Revised: 08/08/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
Despite the considerable advancements in oncology, cancer remains one of the leading causes of death worldwide. Drug resistance mechanisms acquired by cancer cells and inefficient drug delivery limit the therapeutic efficacy of available chemotherapeutics drugs. However, studies have demonstrated that nano-drug carriers (NDCs) can overcome these limitations. In this sense, exosomes emerge as potential candidates for NDCs. This is because exosomes have better organotropism, homing capacity, cellular uptake, and cargo release ability than synthetic NDCs. In addition, exosomes can serve as NDCs for both hydrophilic and hydrophobic chemotherapeutic drugs. Thus, this review aimed to summarize the latest advances in cell-free therapy, describing how the exosomes can contribute to each step of the carcinogenesis process and discussing how these nanosized vesicles could be explored as nano-drug carriers for chemotherapeutics.
Collapse
Affiliation(s)
- Rodrigo Pinheiro Araldi
- Genetics Laboratory, Butantan Institute, São Paulo 05503-900, SP, Brazil; (D.A.D.); (V.R.d.C.); (A.L.A.); (M.R.T.)
- Structural and Functional Biology Post-Graduation Program, Paulista School of Medicine, São Paulo Federal University (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil
- BioDecision Analytics Ltd.a., São Paulo 13271-650, SP, Brazil;
| | - Denis Adrián Delvalle
- Genetics Laboratory, Butantan Institute, São Paulo 05503-900, SP, Brazil; (D.A.D.); (V.R.d.C.); (A.L.A.); (M.R.T.)
- Structural and Functional Biology Post-Graduation Program, Paulista School of Medicine, São Paulo Federal University (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil
| | - Vitor Rodrigues da Costa
- Genetics Laboratory, Butantan Institute, São Paulo 05503-900, SP, Brazil; (D.A.D.); (V.R.d.C.); (A.L.A.); (M.R.T.)
- Structural and Functional Biology Post-Graduation Program, Paulista School of Medicine, São Paulo Federal University (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil
| | - Anderson Lucas Alievi
- Genetics Laboratory, Butantan Institute, São Paulo 05503-900, SP, Brazil; (D.A.D.); (V.R.d.C.); (A.L.A.); (M.R.T.)
- Endocrinology and Metabology Post-Graduation Program, Paulista School of Medicine, São Paulo Federal University (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil
| | - Michelli Ramires Teixeira
- Genetics Laboratory, Butantan Institute, São Paulo 05503-900, SP, Brazil; (D.A.D.); (V.R.d.C.); (A.L.A.); (M.R.T.)
- Endocrinology and Metabology Post-Graduation Program, Paulista School of Medicine, São Paulo Federal University (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil
| | | | - Irina Kerkis
- Genetics Laboratory, Butantan Institute, São Paulo 05503-900, SP, Brazil; (D.A.D.); (V.R.d.C.); (A.L.A.); (M.R.T.)
| |
Collapse
|
46
|
Zanier ER, Pischiutta F, Rulli E, Vargiolu A, Elli F, Gritti P, Gaipa G, Belotti D, Basso G, Zoerle T, Stocchetti N, Citerio G. MesenchymAl stromal cells for Traumatic bRain Injury (MATRIx): a study protocol for a multicenter, double-blind, randomised, placebo-controlled phase II trial. Intensive Care Med Exp 2023; 11:56. [PMID: 37620640 PMCID: PMC10449745 DOI: 10.1186/s40635-023-00535-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/07/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a significant cause of death and disability, with no effective neuroprotective drugs currently available for its treatment. Mesenchymal stromal cell (MSC)-based therapy shows promise as MSCs release various soluble factors that can enhance the injury microenvironment through processes, such as immunomodulation, neuroprotection, and brain repair. Preclinical studies across different TBI models and severities have demonstrated that MSCs can improve functional and structural outcomes. Moreover, clinical evidence supports the safety of third-party donor bank-stored MSCs in adult subjects. Building on this preclinical and clinical data, we present the protocol for an academic, investigator-initiated, multicenter, double-blind, randomised, placebo-controlled, adaptive phase II dose-finding study aiming to evaluate the safety and efficacy of intravenous administration of allogeneic bone marrow-derived MSCs to severe TBI patients within 48 h of injury. METHODS/DESIGN The study will be conducted in two steps. Step 1 will enrol 42 patients, randomised in a 1:1:1 ratio to receive 80 million MSCs, 160 million MSCs or a placebo to establish safety and identify the most promising dose. Step 2 will enrol an additional 36 patients, randomised in a 1:1 ratio to receive the selected dose of MSCs or placebo. The activity of MSCs will be assessed by quantifying the plasmatic levels of neurofilament light (NfL) at 14 days as a biomarker of neuronal damage. It could be a significant breakthrough if the study demonstrates the safety and efficacy of MSC-based therapy for severe TBI patients. The results of this trial could inform the design of a phase III clinical trial aimed at establishing the efficacy of the first neurorestorative therapy for TBI. DISCUSSION Overall, the MATRIx trial is a critical step towards developing an effective treatment for TBI, which could significantly improve the lives of millions worldwide affected by this debilitating condition. Trial Registration EudraCT: 2022-000680-49.
Collapse
Affiliation(s)
- Elisa R Zanier
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Francesca Pischiutta
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Eliana Rulli
- Department of Clinical Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Alessia Vargiolu
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Francesca Elli
- Neurological Intensive Care Unit, Department of Neurosciences, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Paolo Gritti
- Department of Anesthesia, Emergency and Critical Care Medicine, ASST Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | - Giuseppe Gaipa
- M. Tettamanti Research Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Daniela Belotti
- M. Tettamanti Research Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Gianpaolo Basso
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
- Department of Neurosciences, Neuroradiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Tommaso Zoerle
- Neuroscience Intensive Care Unit, Department of Anaesthesia and Critical Care, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplants, University of Milan, Milan, Italy
| | - Nino Stocchetti
- Neuroscience Intensive Care Unit, Department of Anaesthesia and Critical Care, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplants, University of Milan, Milan, Italy
| | - Giuseppe Citerio
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy.
- Neurological Intensive Care Unit, Department of Neurosciences, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
| |
Collapse
|
47
|
Rebelatto CLK, Boldrini-Leite LM, Daga DR, Marsaro DB, Vaz IM, Jamur VR, de Aguiar AM, Vieira TB, Furman BP, Aguiar CO, Brofman PRS. Quality Control Optimization for Minimizing Security Risks Associated with Mesenchymal Stromal Cell-Based Product Development. Int J Mol Sci 2023; 24:12955. [PMID: 37629136 PMCID: PMC10455270 DOI: 10.3390/ijms241612955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have been considered a therapeutic strategy in regenerative medicine because of their regenerative and immunomodulatory properties. The translation of MSC-based products has some challenges, such as regulatory and scientific issues. Quality control should be standardized and optimized to guarantee the reproducibility, safety, and efficacy of MSC-based products to be administered to patients. The aim of this study was to develop MSC-based products for use in clinical practice. Quality control assays include cell characterization, cell viability, immunogenicity, and cell differentiation; safety tests such as procoagulant tissue factor (TF), microbiological, mycoplasma, endotoxin, genomic stability, and tumorigenicity tests; and potency tests. The results confirm that the cells express MSC markers; an average cell viability of 96.9%; a low expression of HLA-DR and costimulatory molecules; differentiation potential; a high expression of TF/CD142; an absence of pathogenic microorganisms; negative endotoxins; an absence of chromosomal abnormalities; an absence of genotoxicity and tumorigenicity; and T-lymphocyte proliferation inhibition potential. This study shows the relevance of standardizing the manufacturing process and quality controls to reduce variability due to the heterogeneity between donors. The results might also be useful for the implementation and optimization of new analytical techniques and automated methods to improve safety, which are the major concerns related to MSC-based therapy.
Collapse
Affiliation(s)
- Carmen Lúcia Kuniyoshi Rebelatto
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| | - Lidiane Maria Boldrini-Leite
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| | - Debora Regina Daga
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| | - Daniela Boscaro Marsaro
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| | - Isadora May Vaz
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| | - Valderez Ravaglio Jamur
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| | - Alessandra Melo de Aguiar
- Laboratory of Basic Biology of Stem Cells, Carlos Chagas Institute—Fiocruz-Paraná, Curitiba 81350-010, Brazil;
| | - Thalita Bastida Vieira
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
| | - Bianca Polak Furman
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
| | - Cecília Oliveira Aguiar
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
| | - Paulo Roberto Slud Brofman
- Core for Cell Technology, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Brazil; (L.M.B.-L.); (D.R.D.); (D.B.M.); (I.M.V.); (V.R.J.); (T.B.V.); (B.P.F.); (C.O.A.); (P.R.S.B.)
- National Institute of Science and Technology for Regenerative Medicine—INCT-REGENERA, Rio de Janeiro 21941-599, Brazil
| |
Collapse
|
48
|
Viswanathan S, Blanc KL, Ciccocioppo R, Dagher G, Filiano AJ, Galipeau J, Krampera M, Krieger L, Lalu MM, Nolta J, Rodriguez Pardo VM, Shi Y, Tarte K, Weiss DJ, Martin I. An International Society for Cell and Gene Therapy Mesenchymal Stromal Cells (MSC) Committee perspectives on International Standards Organization/Technical Committee 276 Biobanking Standards for bone marrow-MSCs and umbilical cord tissue-derived MSCs for research purposes. Cytotherapy 2023; 25:803-807. [PMID: 37149800 DOI: 10.1016/j.jcyt.2023.04.005] [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: 10/31/2022] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 05/08/2023]
Abstract
The rapidly growing field of mesenchymal stromal cell (MSC) basic and translational research requires standardization of terminology and functional characterization. The International Standards Organization's (ISO) Technical Committee (TC) on Biotechnology, working with extensive input from the International Society for Cells and Gene Therapy (ISCT), has recently published ISO standardization documents that are focused on biobanking of MSCs from two tissue sources, Wharton's Jelly, MSC(WJ) and Bone Marrow, MSC(M)), for research and development purposes and development. This manuscript explains the path towards the consensus on the following two documents: the Technical Standard ISO/TS 22859 for MSC(WJ) and the full ISO Standard 24651 for MSC(M) biobanking. The ISO standardization documents are aligned with ISCT's MSC committee position and recommendations on nomenclature because there was active input and incorporation of ISCT MSC committee recommendations in the development of these standards. The ISO standardization documents contain both requirements and recommendations for functional characterization of MSC(WJ) and MSC(M) using a matrix of assays. Importantly, the ISO standardization documents have a carefully defined scope and are meant for research use of culture expanded MSC(WJ) and MSC(M). The ISO standardization documents can be updated in a revision process and will be systematically reviewed after 3-5 years as scientific insights grow. They represent international consensus on MSC identity, definition, and characterization; are rigorous in detailing multivariate characterization of MSCs and represent an evolving-but-important first step in standardization of MSC biobanking and characterization for research use and development.
Collapse
Affiliation(s)
- Sowmya Viswanathan
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, Division of Hematology, University of Toronto, Toronto, Ontario, Canada.
| | - Katarina Le Blanc
- Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rachele Ciccocioppo
- Department of Medicine, AOUI Policlinico GB Rossi & University of Verona, Verona, Italy
| | - Georges Dagher
- Inserm UMR-S 1124, Paris-Descartes University, Paris, France
| | - Anthony J Filiano
- Department of Neurosurgery, Duke University, Durham, North Carolina, USA; Marcus Center for Cellular Cures, Duke University Medical Center, Durham, North Carolina, USA
| | - Jacques Galipeau
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Mauro Krampera
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Lena Krieger
- DIN - German Institute for Standardization, Berlin, Germany
| | - Manoj M Lalu
- Clinical Epidemiology Program, Blueprint Translational Group, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada; Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Jan Nolta
- Department of Internal Medicine, Stem Cell Program and Institute for Regenerative Cures, University of California Davis, Sacramento, California, USA
| | - Viviana Marcela Rodriguez Pardo
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana. Bogotá, Colombia; Biotechnology National Committee Convenor, National Standars of Colombia - ICONTEC. Bogotá, Colombia
| | - Yufang Shi
- The First Affiliated Hospital, Soochow University Institutes for Translational Medicine, Suzhou, China; Institute of Health Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Karin Tarte
- UMR U1236-MICMAC, Immunology and Cell Therapy Lab, Rennes University Hospital, Rennes, France
| | - Daniel J Weiss
- University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
49
|
Gorman EA, Rynne J, Gardiner HJ, Rostron AJ, Bannard-Smith J, Bentley AM, Brealey D, Campbell C, Curley G, Clarke M, Dushianthan A, Hopkins P, Jackson C, Kefela K, Krasnodembskaya A, Laffey JG, McDowell C, McFarland M, McFerran J, McGuigan P, Perkins GD, Silversides J, Smythe J, Thompson J, Tunnicliffe WS, Welters IDM, Amado-Rodríguez L, Albaiceta G, Williams B, Shankar-Hari M, McAuley DF, O'Kane CM. Repair of Acute Respiratory Distress Syndrome in COVID-19 by Stromal Cells (REALIST-COVID Trial): A Multicenter, Randomized, Controlled Clinical Trial. Am J Respir Crit Care Med 2023; 208:256-269. [PMID: 37154608 DOI: 10.1164/rccm.202302-0297oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023] Open
Abstract
Rationale: Mesenchymal stromal cells (MSCs) may modulate inflammation, promoting repair in coronavirus disease (COVID-19)-related acute respiratory distress syndrome (ARDS). Objectives: We investigated the safety and efficacy of ORBCEL-C (CD362 [cluster of differentiation 362]-enriched, umbilical cord-derived MSCs) in COVID-19-related ARDS. Methods: In this multicenter, randomized, double-blind, allocation-concealed, placebo-controlled trial (NCT03042143), patients with moderate to severe COVID-19-related ARDS were randomized to receive ORBCEL-C (400 million cells) or placebo (Plasma-Lyte 148). The primary safety and efficacy outcomes were the incidence of serious adverse events and oxygenation index at Day 7, respectively. Secondary outcomes included respiratory compliance, driving pressure, PaO2:FiO2 ratio, and Sequential Organ Failure Assessment score. Clinical outcomes relating to duration of ventilation, lengths of ICU and hospital stays, and mortality were collected. Long-term follow-up included diagnosis of interstitial lung disease at 1 year and significant medical events and mortality at 2 years. Transcriptomic analysis was performed on whole blood at Days 0, 4, and 7. Measurements and Main Results: Sixty participants were recruited (final analysis: n = 30 received ORBCEL-C, n = 29 received placebo; 1 participant in the placebo group withdrew consent). Six serious adverse events occurred in the ORBCEL-C group and three in the placebo group (risk ratio, 2.9 [95% confidence interval, 0.6-13.2]; P = 0.25). Day 7 mean (SD) oxygenation index did not differ (ORBCEL-C, 98.3 [57.2] cm H2O/kPa; placebo, 96.6 [67.3] cm H2O/kPa). There were no differences in secondary surrogate outcomes or in mortality at Day 28, Day 90, 1 year, or 2 years. There was no difference in the prevalence of interstitial lung disease at 1 year or significant medical events up to 2 years. ORBCEL-C modulated the peripheral blood transcriptome. Conclusion: ORBCEL-C MSCs were safe in subjects with moderate to severe COVID-19-related ARDS but did not improve surrogates of pulmonary organ dysfunction.
Collapse
Affiliation(s)
- Ellen A Gorman
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Jennifer Rynne
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Hannah J Gardiner
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Anthony J Rostron
- Sunderland Royal Hospital, South Tyneside and Sunderland National Health Service Foundation Trust, Sunderland, United Kingdom
- Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Andrew M Bentley
- Acute Intensive Care Unit, Wythenshawe Hospital, Manchester, United Kingdom
| | - David Brealey
- University College Hospital London, London, United Kingdom
| | | | - Gerard Curley
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mike Clarke
- Northern Ireland Clinical Trials Unit, Belfast, United Kingdom
| | - Ahilanadan Dushianthan
- University Hospital Southampton, Southampton, United Kingdom
- National Institute for Health and Care Research Southampton Biomedical Research Centre, University of Southampton, Southampton, United Kingdom
| | - Phillip Hopkins
- King's Trauma Centre, King's College Hospital, London, United Kingdom
| | - Colette Jackson
- Northern Ireland Clinical Trials Unit, Belfast, United Kingdom
| | - Kallirroi Kefela
- Department of Critical Care, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Anna Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - John G Laffey
- Regenerative Medicine Institute at CÚRAM Centre for Research in Medical Devices, University of Galway, Galway, Ireland
| | - Cliona McDowell
- Northern Ireland Clinical Trials Unit, Belfast, United Kingdom
| | - Margaret McFarland
- Department of Critical Care, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Jamie McFerran
- Northern Ireland Clinical Trials Unit, Belfast, United Kingdom
| | - Peter McGuigan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
- Department of Critical Care, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Gavin D Perkins
- Critical Care Unit, University Hospitals Birmingham, Birmingham, United Kingdom
- Warwick Clinical Trials Unit, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Jonathan Silversides
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
- Department of Critical Care, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Jon Smythe
- National Health Service Blood and Transplant, Oxford, United Kingdom
| | - Jacqui Thompson
- National Health Service Blood and Transplant, Birmingham, United Kingdom
| | | | - Ingeborg D M Welters
- Intensive Care Unit, Royal Liverpool University Hospital, Liverpool, United Kingdom
- Institute of Life Course Medical Sciences, University of Liverpool, Liverpool Centre for Cardiovascular Science, Liverpool, United Kingdom
| | - Laura Amado-Rodríguez
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Guillermo Albaiceta
- Centro de Investigación Biomédica en Red-Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
- Unidad de Cuidados Intensivos Cardiológicos, Hospital Universitario Central de Asturias, Oviedo, Spain
- Departamento de Biología Funcional, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain; and
| | - Barry Williams
- Independent Patient and Public Representative, Sherborne, United Kingdom
| | - Manu Shankar-Hari
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Daniel F McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
- Department of Critical Care, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Cecilia M O'Kane
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
50
|
Sitbon A, Delmotte PR, Goumard C, Turco C, Gautheron J, Conti F, Aoudjehane L, Scatton O, Monsel A. Therapeutic potentials of mesenchymal stromal cells-derived extracellular vesicles in liver failure and marginal liver graft rehabilitation: a scoping review. Minerva Anestesiol 2023; 89:690-706. [PMID: 37079286 DOI: 10.23736/s0375-9393.23.17265-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
Liver failure includes distinct subgroups of diseases: Acute liver failure (ALF) without preexisting cirrhosis, acute-on-chronic liver failure (ACLF) (severe form of cirrhosis associated with organ failures and excess mortality), and liver fibrosis (LF). Inflammation plays a key role in ALF, LF, and more specifically in ACLF for which we have currently no treatment other than liver transplantation (LT). The increasing incidence of marginal liver grafts and the shortage of liver grafts require us to consider strategies to increase the quantity and quality of available liver grafts. Mesenchymal stromal cells (MSCs) have shown beneficial pleiotropic properties with limited translational potential due to the pitfalls associated with their cellular nature. MSC-derived extracellular vesicles (MSC-EVs) are innovative cell-free therapeutics for immunomodulation and regenerative purposes. MSC-EVs encompass further advantages: pleiotropic effects, low immunogenicity, storage stability, good safety profile, and possibility of bioengineering. Currently, no human studies explored the impact of MSC-EVs on liver disease, but several preclinical studies highlighted their beneficial effects. In ALF and ACLF, data showed that MSC-EVs attenuate hepatic stellate cells activation, exert antioxidant, anti-inflammatory, anti-apoptosis, anti-ferroptosis properties, and promote regeneration of the liver, autophagy, and improve metabolism through mitochondrial function recovery. In LF, MSC-EVs demonstrated anti-fibrotic properties associated with liver tissue regeneration. Normothermic-machine perfusion (NMP) combined with MSC-EVs represents an attractive therapy to improve liver regeneration before LT. Our review suggests a growing interest in MSC-EVs in liver failure and gives an appealing insight into their development to rehabilitate marginal liver grafts through NMP.
Collapse
Affiliation(s)
- Alexandre Sitbon
- Multidisciplinary Intensive Care Unit, Department of Anesthesiology and Critical Care, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne University, Paris, France -
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France -
| | - Pierre-Romain Delmotte
- Multidisciplinary Intensive Care Unit, Department of Anesthesiology and Critical Care, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne University, Paris, France
| | - Claire Goumard
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France
- Department of Digestive, Hepatobiliary Surgery and Liver Transplantation, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne University, Paris, France
| | - Célia Turco
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France
- Liver Transplantation Unit, Department of Digestive and Oncologic Surgery, University Hospital of Besançon, Besançon, France
| | - Jérémie Gautheron
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France
| | - Filomena Conti
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France
- Department of Digestive, Hepatobiliary Surgery and Liver Transplantation, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne University, Paris, France
- IHU-Innovation of Cardiometabolism and Nutrition (ICAN), INSERM, Sorbonne University, Paris, France
| | - Lynda Aoudjehane
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France
- IHU-Innovation of Cardiometabolism and Nutrition (ICAN), INSERM, Sorbonne University, Paris, France
| | - Olivier Scatton
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France
- Department of Digestive, Hepatobiliary Surgery and Liver Transplantation, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne University, Paris, France
| | - Antoine Monsel
- Multidisciplinary Intensive Care Unit, Department of Anesthesiology and Critical Care, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne University, Paris, France
- INSERM UMRS-959 Immunology-Immunopathology-Immunotherapy (I3), Sorbonne University, Paris, France
| |
Collapse
|