Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 63
1.
Cells ; 13(9)2024 May 01.
Article En | MEDLINE | ID: mdl-38727312

We investigated the impact of mesenchymal stem cell (MSC) therapy on treating bilateral human hip osteonecrosis, analyzing 908 cases. This study assesses factors such as tissue source and cell count, comparing core decompression with various cell therapies. This research emphasizes bone repair according to pre-treatment conditions and the specificities of cell therapy in osteonecrosis repair, indicating a potential for improved bone repair strategies in hips without femoral head collapse. This study utilized a single-center retrospective analysis to investigate the efficacy of cellular approaches in the bone repair of osteonecrosis. It examined the impact on bone repair of tissue source (autologous bone marrow concentrate, allogeneic expanded, autologous expanded), cell quantity (from none in core decompression alone to millions in cell therapy), and osteonecrosis stage and volume. Excluding hips with femoral head collapse, it focused on patients who had bilateral hip osteonecrosis, both pre-operative and post-operative MRIs, and a follow-up of over five years. The analysis divided these patients into seven groups based on match control treatment variations in bilateral hip osteonecrosis, primarily investigating the outcomes between core decompression, washing effect, and different tissue sources of MSCs. Younger patients (<30 years) demonstrated significantly better repair volumes, particularly in stage II lesions, than older counterparts. Additionally, bone repair volume increased with the number of implanted MSCs up to 1,000,000, beyond which no additional benefits were observed. No significant difference was observed in repair outcomes between different sources of MSCs (BMAC, allogenic, or expanded cells). The study also highlighted that a 'washing effect' was beneficial, particularly for larger-volume osteonecrosis when combined with core decompression. Partial bone repair was the more frequent event observed, while total bone repair of osteonecrosis was rare. The volume and stage of osteonecrosis, alongside the number of injected cells, significantly affected treatment outcomes. In summary, this study provides comprehensive insights into the effectiveness and variables influencing the use of mesenchymal stem cells in treating human hip osteonecrosis. It emphasizes the potential of cell therapy while acknowledging the complexity and variability of results based on factors such as age, cell count, and disease stage.


Femur Head Necrosis , Mesenchymal Stem Cell Transplantation , Humans , Mesenchymal Stem Cell Transplantation/methods , Male , Female , Adult , Middle Aged , Femur Head Necrosis/therapy , Femur Head Necrosis/pathology , Retrospective Studies , Mesenchymal Stem Cells/cytology , Cell Count , Young Adult , Aged , Treatment Outcome , Adolescent , Magnetic Resonance Imaging
4.
Int Orthop ; 47(7): 1689-1705, 2023 07.
Article En | MEDLINE | ID: mdl-37036496

PURPOSE: Several reports have identified prognostic factors for hip osteonecrosis treated with cell therapy, but no study investigated the accuracy of artificial intelligence method such as machine learning and artificial neural network (ANN) to predict the efficiency of the treatment. We determined the benefit of cell therapy compared with core decompression or natural evolution, and developed machine-learning algorithms for predicting ten year collapse-free survival in hip osteonecrosis treated with cell therapy. Using the best algorithm, we propose a calculator for "prognosis hip osteonecrosis cell therapy (PHOCT)" accessible for clinical use. METHODS: A total of 3145 patients with 5261 osteonecroses without collapses were included in this study, comprising 1321 (42%) men and 1824 (58%) women, with a median age of 34 (12-62) years. Cell therapy was the treatment for 3021 hips, core decompression alone for 1374 hips, while absence of treatment was the control group of 764 hips. First, logistic regression and binary logistic regression analysis were performed to compare results of the three groups at ten years. Then an artificial neural network model was developed for ten year collapse-free survival after cell therapy. The models' performances were compared. The algorithms were assessed by calibration, and performance, and with c-statistic as measure of discrimination. It ranges from 0.5 to 1.0, with 1.0 being perfect discrimination and 0.5 poor (no better than chance at making a prediction). RESULTS: Among the 3021 hips with cell therapy, 1964 hips (65%) were collapse-free survival at ten years, versus 453 (33%) among those 1374 treated with core decompression alone, and versus 115 (15%) among 764 hips with natural evolution. We analyzed factors influencing the prediction of collapse-free period with classical statistics and artificial intelligence among hips with cell therapy. After selecting variables, a machine learning algorithm created a prognosis osteonecrosis cell therapy calculator (POCT). This calculator proved to have good accuracy on validation in these series of 3021 hip osteonecroses treated with cell therapy. The algorithm had a c-statistic of 0.871 suggesting good-to-excellent discrimination when all the osteonecroses were mixed. The c-statistics were calculated separately for subpopulations of categorical osteonecroses. It retained good accuracy, but underestimated ten year survival in some subgroups, suggesting that specific calculators could be useful for some subgroups. This study highlights the importance of multimodal evaluation of patient parameters and shows the degree to which the outcome is modified by some decisions that are within a surgeon's control, as the number of cells to aspirate, the choice of injecting in both the osteonecrosis and the healthy bone, the choice between unilateral or bilateral injection, and the possibility to do a repeat injection. CONCLUSION: Many disease conditions and the heterogeneities of patients are causes of variation of outcome after cell therapy for osteonecrosis. Predicting therapeutic effectiveness with a calculator allows a good discrimination to target patients who are most likely to benefit from this intervention.


Arthroplasty, Replacement, Hip , Femur Head Necrosis , Osteonecrosis , Male , Humans , Female , Adult , Middle Aged , Artificial Intelligence , Prognosis , Osteonecrosis/therapy , Osteonecrosis/surgery , Hip/surgery , Femur Head Necrosis/therapy , Femur Head Necrosis/surgery , Treatment Outcome
5.
Am J Transplant ; 22(12): 2961-2970, 2022 12.
Article En | MEDLINE | ID: mdl-35778956

Over the past 25 years, we have demonstrated the feasibility of airway bioengineering using stented aortic matrices experimentally then in a first-in-human trial (n = 13). The present TRITON-01 study analyzed all the patients who had airway replacement at our center to confirm that this innovative approach can be now used as usual care. For each patient, the following data were prospectively collected: postoperative mortality and morbidity, late airway complications, stent removal and status at last follow-up on November 2, 2021. From October 2009 to October 2021, 35 patients had airway replacement for malignant (n = 29) or benign (n = 6) lesions. The 30-day postoperative mortality and morbidity rates were 2.9% (n = 1/35) and 22.9% (n = 8/35) respectively. At a median follow-up of 29.5 months (range 1-133 months), 27 patients were alive. There have been no deaths directly related to the implanted bioprosthesis. Eighteen patients (52.9%) had stent-related granulomas requiring a bronchoscopic treatment. Ten among 35 patients (28.6%) achieved a stent free survival. The actuarial 2- and 5-year survival rates (Kaplan-Meier estimates) were respectively 88% and 75%. The TRITON-01 study confirmed that airway replacement using stented aortic matrices can be proposed as usual care at our center. Clinicaltrials.gov Identifier: NCT04263129.


Aortic Valve Stenosis , Bioprosthesis , Heart Valve Prosthesis , Adult , Humans , Aortic Valve Stenosis/surgery , Follow-Up Studies , Postoperative Complications , Stents , Treatment Outcome
6.
Injury ; 53 Suppl 2: S26-S33, 2022 Oct.
Article En | MEDLINE | ID: mdl-33423770

INTRODUCTION: Open fractures are at risk of nonunion; surgeons are reluctant to propose early standard bone grafting after open fractures, preferring to wait in order to adequately assess the facture status of infection. Bone marrow contains mesenchymal stem cells (MSCs) and granulocyte and macrophage precursors identified in vitro as colony forming units-granulocyte macrophage (CFU-GM), both of which have a prophylactic action against infection. We therefore tested the hypothesis that early injection of bone marrow concentrate would be useful in these fractures. METHODS: We evaluated a series of 231 patients who had received early percutaneous implantation of bone marrow concentrate (BMC) to treat open fractures (with gap less than 10 mm) that were Gustilo-Anderson Type II or III. The results were compared with those of 67 control (no early graft) patients and with those of 76 patients treated with an early, standard of care, iliac bone graft. All patients were treated with external fixation and were considered to have an aseptic fracture at the time of early grafting, but the actual status of infection was re-assessed at the time of grafting by histology and/or analysis of the aspirate. The bone marrow graft contained after concentration 49,758 ±â€¯21,642 CFU-GM-derived colonies/cc and 9400 ±â€¯1435 MSCs/cc which represents an important increase compared to the level of CFU-GM cells and MSCs present in a standard auto-graft. Healing was evaluated at 9 months. RESULTS: The rate of unsuspected infections was higher than 15% in the 3 groups. Bone union and removal of external fixation was achieved at 9 months by 50.7% of patients in the Control Group, by 86.8% of patients in the group with a standard bone graft, and by 87.4% of patients in the bone marrow group. A 90% risk reduction (p = 0.005) in the need for an invasive standard bone graft to treat a nonunion and in the risk of infection was observed when bone marrow was proposed as early injection to the treatment of type II or type-III tibial fractures. CONCLUSION: Bone marrow concentrate for early grafting in open fractures with limited gap was efficient for healing while decreasing infection.


Fractures, Open , Tibial Fractures , Bone Marrow , Bone Transplantation/methods , Fracture Healing , Fractures, Open/surgery , Humans , Tibia , Tibial Fractures/surgery , Treatment Outcome
8.
Cells ; 10(4)2021 03 28.
Article En | MEDLINE | ID: mdl-33800595

Background: Skeletal muscle is one of the only mammalian tissues capable of rapid and efficient regeneration after trauma or in pathological conditions. Skeletal muscle regeneration is driven by the muscle satellite cells, the stem cell population in interaction with their niche. Upon injury, muscle fibers undergo necrosis and muscle stem cells activate, proliferate and fuse to form new myofibers. In addition to myogenic cell populations, interaction with other cell types such as inflammatory cells, mesenchymal (fibroadipogenic progenitors-FAPs, pericytes) and vascular (endothelial) lineages are important for efficient muscle repair. While the role of the distinct populations involved in skeletal muscle regeneration is well characterized, the quantitative changes in the muscle stem cell and niche during the regeneration process remain poorly characterized. Methods: We have used mass cytometry to follow the main muscle cell types (muscle stem cells, vascular, mesenchymal and immune cell lineages) during early activation and over the course of muscle regeneration at D0, D2, D5 and D7 compared with uninjured muscles. Results: Early activation induces a number of rapid changes in the proteome of multiple cell types. Following the induction of damage, we observe a drastic loss of myogenic, vascular and mesenchymal cell lineages while immune cells invade the damaged tissue to clear debris and promote muscle repair. Immune cells constitute up to 80% of the mononuclear cells 5 days post-injury. We show that muscle stem cells are quickly activated in order to form new myofibers and reconstitute the quiescent muscle stem cell pool. In addition, our study provides a quantitative analysis of the various myogenic populations during muscle repair. Conclusions: We have developed a mass cytometry panel to investigate the dynamic nature of muscle regeneration at a single-cell level. Using our panel, we have identified early changes in the proteome of stressed satellite and niche cells. We have also quantified changes in the major cell types of skeletal muscle during regeneration and analyzed myogenic transcription factor expression in satellite cells throughout this process. Our results highlight the progressive dynamic shifts in cell populations and the distinct states of muscle stem cells adopted during skeletal muscle regeneration. Our findings give a deeper understanding of the cellular and molecular aspects of muscle regeneration.


Muscle, Skeletal/pathology , Proteomics , Single-Cell Analysis , Wound Healing , Animals , Cell Lineage , Mice , Muscle Development , Muscle, Skeletal/physiopathology , Proteome/metabolism , Regeneration , Stem Cells/cytology
9.
Int Orthop ; 45(9): 2383-2393, 2021 09.
Article En | MEDLINE | ID: mdl-33893522

PURPOSE: Corticoid treatment associated with haematologic treatments can lead to ankle osteonecrosis in children's survivors of acute leukemia (ALL). Based on the efficiency of mesenchymal stem cells (MSCs) in hip osteonecrosis, we performed an evaluation of this treatment in 51 children and adolescents who had symptomatic ankle osteonecrosis after therapy for haematologic cancer. MATERIAL AND METHODS: The 51 patients had a total of 79 osteonecrosis sites on MRI, with 29 talus sites, 18 metaphyseal tibia sites, 12 epiphyseal tibia sites, eight calcaneus sites, six fibula sites, four navicular sites, and two cuboid sites. In this prospective randomized trial, 37 ankles were addressed for cell therapy, 37 others for core decompression alone, and 20 were considered as a control group without treatment. We analyzed the outcome of this treatment osteonecrosis, the number and characteristics of bone marrow mesenchymal cells (MSCs) that could be transplanted, and the risks of tumorigenesis in these patients with haematologic cancers. The patients were operated on over a period of ten years from 2000 to 2010 and were monitored through December 31, 2019. RESULTS: Despite a normal systemic blood cells count, MSCs in the iliac crest (counted as CFU-F) were in low number (1021 MSCs/mL; range 314-3015) and were of host origin after even allogeneic bone marrow transplantation. Better clinical outcomes (pain, foot and ankle deformity) and osteonecrosis repair on MRI with absence of collapse were obtained in ankles that received cell therapy as compared with those with core decompression alone or those without initial surgery. No tumour was found on MRI at the sites of injection and this study found no increased risk of recurrence or of new cancer in this population after an average follow-up of 15 years. CONCLUSIONS: These results suggest that autologous MSCs can improve the quality of life of leukemia survivors with ankle osteonecrosis.


Leukemia , Mesenchymal Stem Cells , Osteonecrosis , Adolescent , Ankle , Child , Humans , Osteonecrosis/diagnostic imaging , Osteonecrosis/epidemiology , Osteonecrosis/etiology , Prospective Studies , Quality of Life , Survivors , Treatment Outcome
10.
J Clin Med ; 10(3)2021 Feb 01.
Article En | MEDLINE | ID: mdl-33535589

Background: Osteonecrosis (ON) of the femoral head represents a potentially severe disease of the hip where the lack of bone regeneration may lead to femoral head collapse and secondary osteoarthritis, with serious pain and disability. The aim of this European, multicentric clinical trial was to prove safety and early efficacy to heal early femoral head ON in patients through minimally invasive surgical implantation of autologous mesenchymal stromal cells (MSC) expanded from bone marrow (BM) under good manufacturing practices (GMP). Methods: Twenty-two patients with femoral head ON (up to ARCO 2C) were recruited and surgically treated in France, Germany, Italy and Spain with BM-derived, expanded autologous MSC (total dose 140 million MSC in 7 mL). The investigational advanced therapy medicinal product (ATMP) was expanded from BM under the same protocol in all four countries and approved by each National Competent Authority. Patients were followed during two years for safety, based on adverse events, and for efficacy, based on clinical assessment (pain and hip score) and imaging (X-rays and MRIs). Patients were also reviewed after 5 to 6 years at latest follow-up for final outcome. Results: No severe adverse event was recalled as related to the ATMP. At 12 months, 16/20 per protocol and 16/22 under intention-to-treat (2 drop-out at 3 and 5 months) maintained head sphericity and showed bone regeneration. Of the 4 hips with ON progression, 3 required total hip replacement (THR). At 5 years, one patient (healed at 2 years visit) was not located, and 16/21 showed no progression or THR, 4/21 had received THR (all in the first year) and 1 had progressed one stage without THR. Conclusions: Expanded MSCs implantation was safe. Early efficacy was confirmed in 80% of cases under protocol at 2 years. At 5 years, the overall results were maintained and 19% converted to THR, all in the first year.

11.
Int Orthop ; 45(2): 391-399, 2021 02.
Article En | MEDLINE | ID: mdl-32617651

PURPOSE: There is an increasing number of reports on the treatment of knee osteoarthritis (OA) using mesenchymal stem cells (MSCs). However, it is not known what would better drive osteoarthritis stabilization to postpone total knee arthroplasty (TKA): targeting the synovial fluid by injection or targeting on the subchondral bone with MSCs implantation. METHODS: A prospective randomized controlled clinical trial was carried out between 2000 and 2005 in 120 knees of 60 patients with painful bilateral knee osteoarthritis with a similar osteoarthritis grade. During the same anaesthesia, a bone marrow concentrate of 40 mL containing an average 5727 MSCs/mL (range 2740 to 7540) was divided in two equal parts: after randomization, one part (20 mL) was delivered to the subchondral bone of femur and tibia of one knee (subchondral group) and the other part was injected in the joint for the contralateral knee (intra-articular group). MSCs were counted as CFU-F (colony fibroblastic unit forming). Clinical outcomes of the patient (Knee Society score) were obtained along with radiological imaging outcomes (including MRIs) at two year follow-up. Subsequent revision surgeries were identified until the most recent follow-up (average of 15 years, range 13 to 18 years). RESULTS: At two year follow-up, clinical and imaging (MRI) improvement was higher on the side that received cells in the subchondral bone. At the most recent follow-up (15 years), among the 60 knees treated with subchondral cell therapy, the yearly arthroplasty incidence was 1.3% per knee-year; for the 60 knees with intra-articular cell therapy, the yearly arthroplasty incidence was higher (p = 0.01) with an incidence of 4.6% per knee-year. For the side with subchondral cell therapy, 12 (20%) of 60 knees underwent TKA, while 42 (70%) of 60 knees underwent TKA on the side with intra-articular cell therapy. Among the 18 patients who had no subsequent surgery on both sides, all preferred the knee with subchondral cell therapy. CONCLUSIONS: Implantation of MSCs in the subchondral bone of an osteoarthritic knee is more effective to postpone TKA than injection of the same intra-articular dose in the contralateral knee with the same grade of osteoarthritis.


Arthroplasty, Replacement, Knee , Cartilage, Articular , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Osteoarthritis, Knee , Arthroplasty, Replacement, Knee/adverse effects , Bone Marrow , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/surgery , Humans , Injections, Intra-Articular , Knee Joint/diagnostic imaging , Knee Joint/surgery , Osteoarthritis, Knee/surgery
12.
Stem Cell Reports ; 15(3): 597-611, 2020 09 08.
Article En | MEDLINE | ID: mdl-32763161

Muscle stem cells (or muscle satellite cells [MuSCs]) are required for postnatal growth. Yet, the detailed characterization of myogenic progression and establishment of quiescence during this process remains poorly documented. Here, we provide an overview of myogenic cells heterogeneity and dynamic from birth to adulthood using flow cytometry. We demonstrated that PAX7+ cells acquire an increasing ability to progress in the myogenic program from birth to adulthood. We then simultaneously analyzed the cycling state (KI67 expression) of the MuSCs and progenitors (PAX7+) and their progression into myogenic precursors (PAX7-MYOD+) and differentiating cells (MYOG+) in vivo. We identified two distinct peaks of myogenic differentiation between P7-P10 and P21-P28, and showed that the quiescent MuSC pool is established between 7 and 8 weeks of age. Overall our study provides a comprehensive in vivo characterization of myogenic heterogeneity and demonstrates the highly dynamic nature of skeletal muscle postnatal growth process.


Cell Cycle , Muscle Development , Muscle, Skeletal/cytology , Stem Cells/cytology , Animals , Animals, Newborn , Antigens, CD/metabolism , Antigens, CD34/metabolism , Cell Differentiation , Cells, Cultured , Integrin alpha Chains/metabolism , Mice, Inbred C57BL , Stem Cells/metabolism
13.
Injury ; 51 Suppl 1: S63-S73, 2020 Apr.
Article En | MEDLINE | ID: mdl-32139130

BACKGROUND AND STUDY AIM: Advanced therapy medicinal products (ATMP) frequently lack of clinical data on efficacy to substantiate a future clinical use. This study aims to evaluate the efficacy to heal long bone delayed unions and non-unions, as secondary objective of the EudraCT 2011-005441-13 clinical trial, through clinical and radiological bone consolidation at 3, 6 and 12 months of follow-up, with subgroup analysis of affected bone, gender, tobacco use, and time since the original fracture. PATIENTS AND METHODS: Twenty-eight patients were recruited and surgically treated with autologous bone marrow derived mesenchymal stromal cells expanded under Good Manufacturing Practices, combined to bioceramics in the surgical room before implantation. Mean age was 39 ± 13 years, 57% were males, and mean Body Mass Index 27 ± 7. Thirteen (46%) were active smokers. There were 11 femoral, 4 humeral, and 13 tibial non-unions. Initial fracture occurred at a mean ± SD of 27.9 ± 31.2 months before recruitment. Efficacy results were expressed by clinical consolidation (no or mild pain if values under 30 in VAS scale), and by radiological consolidation with a REBORNE score over 11/16 points (value of or above 0.6875). Means were statistically compared and mixed models for repeated measurements estimated the mean and confidence intervals (95%) of the REBORNE Bone Healing scale. Clinical and radiological consolidation were analyzed in the subgroups with Spearman correlation tests (adjusted by Bonferroni). RESULTS: Clinical consolidation was earlier confirmed, while radiological consolidation at 3 months was 25.0% (7/28 cases), at 6 months 67.8% (19/28 cases), and at 12 months, 92.8% (26/28 cases including the drop-out extrapolation of two failures). Bone biopsies confirmed bone formation surrounding the bioceramic granules. All locations showed similar consolidation, although this was delayed in tibial non-unions. No significant gender difference was found in 12-month consolidation (95% confidence). Higher consolidation scale values were seen in non-smoking patients at 6 (p = 0.012, t-test) and 12 months (p = 0.011, t-test). Longer time elapsed after the initial fracture did not preclude the occurrence of consolidation. CONCLUSION: Bone consolidation was efficaciously obtained with the studied expanded hBM-MSCs combined to biomaterials, by clinical and radiological evaluation, and confirmed by bone biopsies, with lower consolidation scores in smokers.


Biocompatible Materials/pharmacology , Fracture Healing/physiology , Fractures, Bone/therapy , Fractures, Ununited/therapy , Mesenchymal Stem Cell Transplantation/methods , Adult , Europe , Female , Femur/pathology , Humans , Humerus/pathology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Middle Aged , Osteogenesis , Radiography , Tibia/pathology , Transplantation, Autologous , Treatment Outcome
14.
Stem Cells Transl Med ; 8(12): 1230-1241, 2019 12.
Article En | MEDLINE | ID: mdl-31486585

We aimed to evaluate efficiency and safety of transplantation of limbal stem cells (LSC) cultured on human amniotic membrane with no feeders and to compare cultured LSC with limbal tissue transplantation. Thirty eyes with stage III LSC deficiency were treated with autologous (autoLSC) or allogeneic (alloLSC) cultured LSC transplantation (prospective phase II clinical trial; average follow-up time, 72 months) or autologous (autoLT) or allogeneic (alloLT) limbal tissue transplantation (retrospective control group; average follow-up time, 132 months) between 1993 and 2014. The 5-year graft survival defined by absence of recurrence of the clinical signs of limbal deficiency was 71% for autoLSC, 0% for alloLSC, 75% for autoLT, and 33% for alloLT. Visual acuity improved by 9.2 lines for autoLSC and 3.3 lines for autoLT. It decreased by 0.7 lines for alloLSC and 1.9 lines for alloLT. Adverse events were recorded in 1/7 autoLSC, 7/7 alloLSC, 6/8 autoLT, and 8/8 alloLT patients. Corneal epithelial defect was the only adverse event recorded after autoLSC, whereas severe sight-threatening adverse events were recorded in the remaining three groups. Compared with failed grafts, successful grafts featured greater decrease in fluorescein staining, greater superficial vascularization-free corneal area, lower variability of the corneal epithelial thickness, and higher corneal epithelial basal cell density. Autologous cultured LSC transplantation was associated with high long-term survival and dramatic improvement in vision and was very safe. Autologous limbal tissue transplantation resulted in similar efficiency but was less safe. Cadaver allogeneic grafts resulted in low long-term success rate and high prevalence of serious adverse events. Stem Cells Translational Medicine 2019;8:1230&1241.


Corneal Diseases/therapy , Epithelium, Corneal/transplantation , Eye Burns/therapy , Graft Survival , Limbus Corneae/cytology , Stem Cell Transplantation/methods , Stem Cells/cytology , Adolescent , Adult , Aged , Corneal Diseases/pathology , Epithelium, Corneal/cytology , Eye Burns/pathology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Prospective Studies , Retrospective Studies , Transplantation, Autologous , Visual Acuity , Young Adult
15.
Stem Cells Transl Med ; 8(8): 822-832, 2019 08.
Article En | MEDLINE | ID: mdl-31037857

We previously demonstrated that intracardiac delivery of autologous peripheral blood-derived CD34+ stem cells (SCs), mobilized by granulocyte-colony stimulating factor (G-CSF) and collected by leukapheresis after myocardial infarction, structurally and functionally repaired the damaged myocardial area. When used for cardiac indication, CD34+ cells are now considered as Advanced Therapy Medicinal Products (ATMPs). We have industrialized their production by developing an automated device for ex vivo CD34+ -SC expansion, starting from a whole blood (WB) sample. Blood samples were collected from healthy donors after G-CSF mobilization. Manufacturing procedures included: (a) isolation of total nuclear cells, (b) CD34+ immunoselection, (c) expansion and cell culture recovery in the device, and (d) expanded CD34+ cell immunoselection and formulation. The assessment of CD34+ cell counts, viability, and immunophenotype and sterility tests were performed as quality tests. We established graft acceptance criteria and performed validation processes in three cell therapy centers. 59.4 × 106 ± 36.8 × 106 viable CD34+ cells were reproducibly generated as the final product from 220 ml WB containing 17.1 × 106 ± 8.1 × 106 viable CD34+ cells. CD34+ identity, genetic stability, and telomere length were consistent with those of basal CD34+ cells. Gram staining and mycoplasma and endotoxin analyses were negative in all cases. We confirmed the therapeutic efficacy of both CD34+ -cell categories in experimental acute myocardial infarct (AMI) in immunodeficient rats during preclinical studies. This reproducible, automated, and standardized expansion process produces high numbers of CD34+ cells corresponding to the approved ATMP and paves the way for a phase I/IIb study in AMI, which is currently recruiting patients. Stem Cells Translational Medicine 2019;8:822&832.


Antigens, CD34/genetics , Automation, Laboratory/methods , Flow Cytometry/methods , Myocardial Infarction/therapy , Peripheral Blood Stem Cell Transplantation/methods , Peripheral Blood Stem Cells/cytology , Adult , Animals , Antigens, CD34/metabolism , Cells, Cultured , Clinical Trials as Topic , Humans , Immunophenotyping/methods , Male , Middle Aged , Peripheral Blood Stem Cells/metabolism , Primary Cell Culture/methods , Rats
16.
Biomaterials ; 196: 100-108, 2019 03.
Article En | MEDLINE | ID: mdl-29598897

BACKGROUND: ORTHO-1 is a European, multicentric, first in human clinical trial to prove safety and feasibility after surgical implantation of commercially available biphasic calcium phosphate bioceramic granules associated during surgery with autologous mesenchymal stromal cells expanded from bone marrow (BM-hMSC) under good manufacturing practices, in patients with long bone pseudarthrosis. METHODS: Twenty-eight patients with femur, tibia or humerus diaphyseal or metaphyso-diaphyseal non-unions were recruited and surgically treated in France, Germany, Italy and Spain with 100 or 200 million BM-hMSC/mL associated with 5-10 cc of bioceramic granules. Patients were followed up during one year. The investigational advanced therapy medicinal product (ATMP) was expanded under the same protocol in all four countries, and approved by each National Competent Authority. FINDINGS: With safety as primary end-point, no severe adverse event was reported as related to the BM-hMSC. With feasibility as secondary end-point, the participating production centres manufactured the BM-hMSC as planned. The ATMP combined to the bioceramic was surgically delivered to the non-unions, and 26/28 treated patients were found radiologically healed at one year (3 out of 4 cortices with bone bridging). INTERPRETATION: Safety and feasibility were clinically proven for surgical implantation of expanded autologous BM-hMSC with bioceramic. FUNDING: EU-FP7-HEALTH-2009, REBORNE Project (GA: 241876).


Biocompatible Materials/pharmacology , Calcium Phosphates/pharmacology , Femur/pathology , Fractures, Bone/therapy , Fractures, Ununited/therapy , Humerus/pathology , Mesenchymal Stem Cell Transplantation/adverse effects , Tibia/pathology , Cell Proliferation/drug effects , Feasibility Studies , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Transplantation, Autologous
17.
Front Immunol ; 10: 2908, 2019.
Article En | MEDLINE | ID: mdl-31921167

The induction of specific and sustainable tolerance is a challenging issue in organ transplantation. The discovery of the immunosuppressive properties of apoptotic cells in animal models has paved the way for their use in human transplantation. In this work, we aimed to define a stable, reproducible, and clinically compatible production procedure of human apoptotic cells (Apo-cells). Using a clinically approved extracorporeal photopheresis technique, we have produced and characterized phenotypically and functionally human apoptotic cells. These Apo-cells have immunosuppressive properties proved in vitro and in vivo in NOD/SCID/γC mice by their capacity to modulate an allogeneic response following both a direct and an indirect antigen presentation. These results brought the rationale for the use of Apo-cells in tolerance induction protocol for organ transplantation.


Antigen Presentation , Apoptosis/immunology , Immune Tolerance , Photopheresis , Animals , Humans , Mice , Mice, Inbred NOD , Mice, SCID
18.
Int Orthop ; 42(7): 1639-1649, 2018 07.
Article En | MEDLINE | ID: mdl-29744647

PURPOSE: Symptomatic osteonecrosis related to corticosteroids has a high risk of progression to collapse in absence of treatment. The purposes of this study were to evaluate the results of autologous bone marrow grafting of the symptomatic hip in adult patients with osteonecrosis and to compare the results with core decompression alone in the contralateral symptomatic hip. MATERIALS AND METHODS: A total of 125 consecutive patients (78 males and 47 females) with bilateral osteonecrosis (ON) and who had both hips symptomatic and at the same stage on each side (stage I or II) were included in this study from 1988 to 1998. The volume of osteonecrosis was measured with MRI in both hips; the smaller size ON was treated with core decompression, and the contralateral hip with the larger ON was treated with percutaneous mesenchymal cell (MSC) injection obtained from bone marrow concentration. The average total number of MSCs (counted as number of colony forming units-fibroblast) injected in each hip was 90,000 ± 25,000 cells (range 45,000 to 180,000 cells). RESULTS: At the most recent FU (average 25 years after the first surgery, range 20 to 30 years), among the 250 hips included in the study, 35 hips (28%) had collapsed at the most recent follow-up after bone marrow grafting, and 90 (72%) after core decompression (CD). Ninety-five hips (76%) in the CD group underwent total hip replacement and 30 hips (24%) in the bone marrow graft group (p < 0.0001). Hips undergoing only CD were approximately three times more likely to undergo a primary THA (odds ratio: 10.0278; 95% CI: 5.6117 to 17.9190; p < 0.0001) as compared with hips undergoing an initial bone marrow grafting. For the 90 hips treated with bone marrow injection and without collapse, the mean volume of repair evaluated by MRI at the most recent follow-up was 16.4 cm3 (range 12 to 21 cm3) corresponding to a decrease of the pre-operative average volume from 22.4 cm3 (range 35-15 cm3) to 6 cm3 (range 12-0 cm3); as percentage of the volume of the femoral head, the decrease moved from 44.8 to 12%. CONCLUSION: Core decompression with bone marrow injection improved the outcome of the disease as compared with core decompression alone in the same patient.


Decompression, Surgical/methods , Femur Head Necrosis/therapy , Glucocorticoids/adverse effects , Mesenchymal Stem Cell Transplantation/methods , Adolescent , Adult , Arthroplasty, Replacement, Hip/statistics & numerical data , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Decompression, Surgical/adverse effects , Female , Femur Head Necrosis/chemically induced , Follow-Up Studies , Hip Joint/pathology , Hip Joint/surgery , Humans , Magnetic Resonance Imaging , Male , Mesenchymal Stem Cell Transplantation/adverse effects , Middle Aged , Prospective Studies , Reoperation/statistics & numerical data , Transplantation, Autologous , Treatment Outcome , Young Adult
19.
Int Orthop ; 42(9): 2263-2272, 2018 09.
Article En | MEDLINE | ID: mdl-29744650

PURPOSE: Degenerative disc disease involves sequential events that lead to the loss of cells, a decrease in disc matrix production, disc dehydration, and alteration of its biomechanical properties. The aim of this study was to determine whether cryoinjury of the nucleus pulposus performed through endplate perforation contributes to disc degeneration and to compare this technique with standard methods. METHOD: Under general anesthesia, the lumbar discs of six pigs were exposed and randomly submitted to needle puncture of the annulus fibrosus (NeP), isolated endplate injury (EP), or cryoinjury using a 2.5-J Thompson cryoprobe applied through a single endplate perforation (EP+cryo). The remaining discs served as controls. Animals were sacrificed at two months and the harvested lumbar spines were submitted to CT scan and MRI investigations. Histologic analysis was performed to assess the degree of disc degeneration. RESULTS: CT scan showed that decrease in average disc height was more important after cryoinjury (49.3%) than after endplate perforation (16.9%) (P < 0.0001) or needle puncture (19.4%) (P < 0.0001). On MRI, the dehydration ratio was significantly more important after EP+cryo (60%) than after NP (40%) or EP (30%) (P < 0.0001). After cryoinjury, the histologic score developed for this study was significantly higher than after needle puncture or endplate perforation (P < 0.0001). CONCLUSIONS: Imaging and histological analysis showed that disc cryoinjury applied through endplate perforation was superior to the classical NeP and EP models to induce experimental disc degeneration. This model appears suitable for testing safety and efficacy of novel treatments of intervertebral disc degeneration.


Cryosurgery/methods , Intervertebral Disc Degeneration/etiology , Intervertebral Disc/injuries , Animals , Cryosurgery/veterinary , Disease Models, Animal , Female , Intervertebral Disc/pathology , Intervertebral Disc Degeneration/veterinary , Lumbar Vertebrae/pathology , Magnetic Resonance Imaging , Needles , Random Allocation , Swine , Tomography, X-Ray Computed
20.
Int Orthop ; 42(7): 1739-1745, 2018 07.
Article En | MEDLINE | ID: mdl-29796764

PURPOSE: Despite multiple possible treatments, the risk of collapse remains the main problem of osteonecrosis. Heart failure (HF). In an effort to address the reverse this issue, curative strategies with regenerative medicine are increasingly being considered. The aim of this technology is to halt or reverse progression of the disease to collapse. MATERIAL AND METHODS: The pioneering report by Hernigou published in 2002 was the first pilot study suggesting that injection of bone marrow stem cells was a safe approach able to improve osteonecrosis in patients with early stages. Since then, an impressive number of studies and trials employing unselected BM-derived cells (1000 the last 2 years) showed that delivery of those cells to the site of osteonecrosis during core decompression was somehow able to ameliorate the patient with osteonecrosis. In order to translate the promise of this cell therapy into better clinical benefit, many questions need to be addressed. In this review, we therefore analyzed current clinical experience of the literature and our experience of 4000 cases to address these questions and particularly the number of cells that should be injected. RESULTS: After almost 20 years of clinical research in this field, we are still far from having drawn conclusions on the number of cells we should inject in regenerating hip osteonecrosis. Findings are difficult to interpret due to heterogeneity of causes of osteonecrosis, as well as differences in the cells count, sample quality, and stages of osteonecrosis. The authors address specific issues, as cell quality, cell numbers, volume of osteonecrosis, concentration of cells, and ex vivo expansion. Bone marrow mesenchymal stem cells are supposed to be "functionally competent," but are collected from the bon, marrow of patients with diseases and risk factors of osteonecrosis. The recipient organ (bone osteonecrosis) is a tissue where several alterations have already occurred. These questions are addressed in this review. CONCLUSION: In this review, we analyzed current clinical experience regarding cell therapy and address issues that should be a guide for future cell-based therapeutic application in osteonecrosis.


Femur Head Necrosis/therapy , Mesenchymal Stem Cell Transplantation/methods , Decompression, Surgical/methods , Disease Progression , Hip Joint/surgery , Humans , Transplantation, Autologous
...