Revistas
Autores:
Soria-Juan, B.; García-Arranz, M.; Jiménez, L. L.; et al.
Revista:
TRIALS
ISSN:
1745-6215
Año:
2021
Vol.:
22
N°:
1
Págs.:
595
Background: Chronic lower limb ischemia develops earlier and more frequently in patients with type 2 diabetes mellitus. Diabetes remains the main cause of lower-extremity non-traumatic amputations. Current medical treatment, based on antiplatelet therapy and statins, has demonstrated deficient improvement of the disease. In recent years, research has shown that it is possible to improve tissue perfusion through therapeutic angiogenesis. Both in animal models and humans, it has been shown that cell therapy can induce therapeutic angiogenesis, making mesenchymal stromal cell-based therapy one of the most promising therapeutic alternatives. The aim of this study is to evaluate the feasibility, safety, and efficacy of cell therapy based on mesenchymal stromal cells derived from adipose tissue intramuscular administration to patients with type 2 diabetes mellitus with critical limb ischemia and without possibility of revascularization. Methods: A multicenter, randomized double-blind, placebo-controlled trial has been designed. Ninety eligible patients will be randomly assigned at a ratio 1:1:1 to one of the following: control group (n = 30), low-cell dose treatment group (n = 30), and high-cell dose treatment group (n = 30). Treatment will be administered in a single-dose way and patients will be followed for 12 months. Primary outcome (safety) will be evaluated by measuring the rate of adverse events within the study period. Secondary outcomes (efficacy) will be measured by assessing clinical, analytical, and imaging-test parameters. Tertiary outcome (quality of life) will be evaluated with SF-12 and VascuQol-6 scales. Discussion: Chronic lower limb ischemia has limited therapeutic options and constitutes a public health problem in both developed and underdeveloped countries. Given that the current treatment is not established in daily clinical practice, it is essential to provide evidence-based data that allow taking a step forward in its clinical development. Also, the multidisciplinary coordination exercise needed to develop this clinical trial protocol will undoubtfully be useful to conduct academic clinical trials in the field of cell therapy in the near future.
Revista:
ERJ OPEN RESEARCH
ISSN:
2312-0541
Año:
2021
Vol.:
7
N°:
2
Págs.:
00773 - 2020
Rationale: Idiopathic pulmonary fibrosis (IPF) has a dismal prognosis. Mesenchymal stromal cells (MSCs) have shown benefit in other inflammatory diseases. Objectives: To evaluate the safety and feasibility of endobronchial administration of bone marrow autologous MSCs (BM-MSC) in patients with mild-to-moderate IPF. Methods: A phase I multicentre clinical trial (ClinicalTrials.gov NCT01919827) with a single endobronchial administration of autologous adult BM-MSCs in patients diagnosed with mild-to-moderate IPF. In a first escalating-dose phase, three patients were included sequentially in three dose cohorts (10×106, 50×106 and 100×106 cells). In a second phase, nine patients received the highest tolerated dose. Follow-up with pulmonary function testing, 6-min walk test and St George¿s Respiratory Questionnaire was done at 1, 2, 3, 6 and 12 months, and with computed tomography at 3, 6 and 12 months. Results: 21 bone marrow samples were obtained from 17 patients. Three patients were excluded from treatment due to chromosome aberrations detected in MSCs after culture, and one patient died before treatment. Finally, 13 patients received the BM-MSC infusion. No treatment-related severe adverse events were observed during follow-up. Compared to baseline, the mean forced vital capacity showed an initial decline of 8.1% at 3 months. The number of patients without functional progression was six (46%) at 3 months and three (23%) at 12 months. Conclusions: The endobronchial infusion of BM-MSCs did not cause immediate serious adverse events in IPF patients, but a relevant proportion of patients suffered clinical and/or functional progression. Genomic instability of BM-MSCs during culture found in three patients may be troublesome for the use of autologous MSCs in IPF patients
Revista:
PHARMACEUTICS
ISSN:
1999-4923
Año:
2021
Vol.:
13
N°:
8
Págs.:
1269
The use of allogeneic adipose-derived mesenchymal stromal cells (alloADSCs) represents an attractive approach for treating myocardial infarction (MI). Furthermore, adding a natural support improves alloADSCs engraftment and survival in heart tissues, leading to a greater therapeutic effect. We aimed to examine the safety and immunological reaction induced by epicardial implantation of a clinical-grade collagen scaffold (CS) seeded with alloADSCs for its future application in humans. Thus, cellularized scaffolds were myocardially or subcutaneously implanted in immunosuppressed rodent models. The toxicological parameters were not significantly altered, and tumor formation was not found over the short or long term. Furthermore, biodistribution analyses in the infarcted immunocompetent rats displayed cell engraftment in the myocardium but no migration to other organs. The immunogenicity of alloADSC-CS was also evaluated in a preclinical porcine model of chronic MI; no significant humoral or cellular alloreactive responses were found. Moreover, CS cellularized with human ADSCs cocultured with human allogeneic immune cells produced no alloreactive response. Interestingly, alloADSC-CS significantly inhibited lymphocyte responses, confirming its immunomodulatory action. Thus, alloADSC-CS is likely safe and does not elicit any alloreactive immunological response in the host. Moreover, it exerts an immunomodulatory action, which supports its translation to a clinical setting.
Revista:
JOURNAL OF TRANSLATIONAL MEDICINE
ISSN:
1479-5876
Año:
2020
Vol.:
18
N°:
1
Págs.:
356
Background Mesenchymal stromal cells are a safe and promising option to treat knee osteoarthritis as previously demonstrated in different clinical trials. However, their efficacy, optimal dose and addition of adjuvants must be determined. Here, we evaluated the clinical effects of a dose of 100 x 10(6)bone marrow mesenchymal stromal cells (BM-MSCs) in combination with Platelet Rich Plasma (PRGF (R)) as adjuvant in a randomized clinical trial. Methods A phase II, multicenter, randomized clinical trial with active control was conducted. Sixty patients diagnosed with knee OA were randomly assigned to 3 weekly doses of PRGF (R) or intraarticular administration of 100 x 10(6)cultured autologous BM-MSCs plus PRGF (R). Patients were followed up for 12 months, and pain and function were assessed using VAS and WOMAC and by measuring the knee range of motion range. X-ray and magnetic resonance imaging analyses were performed to analyze joint damage. Results No adverse effects were reported after BM-MSC administration or during follow-up. According to VAS, the mean value (SD) for PRGF (R) and BM-MSC with PRGF (R) went from 5 (1.8) to 4.5 (2.2) (p = 0.389) and from 5.3 (1.9) to 3.5 (2.5) (p = 0.01), respectively at 12 months. In WOMAC, the mean (SD) baseline and 12-month overall WOMAC scores in patients treated with PRGF (R) was 31.9 (16.2) and 22.3 (15.8) respectively (p = 0.002) while that for patients treated with BM-MSC plus PRGF (R) was 33.4 (18.7) and 23.0 (16.6) (p = 0.053). Although statistical significances between groups have been not detected, only patients being treated with BM-MSC plus PRGF (R) could be considered as a OA treatment responders following OARSI criteria. X-ray and MRI (WORMS protocol) revealed no changes in knee joint space width or joint damage. Conclusions Treatment with BM-MSC associated with PRGF (R) was shown to be a viable therapeutic option for osteoarthritis of the knee, with clinical improvement at the end of follow-up. Further phase III clinical trials would be necessary to confirm the efficacy. Trial registrationClinical Trials.gov identifier NCT02365142. No EudraCT: 2011-006036-23
Revista:
JOURNAL OF TRANSLATIONAL MEDICINE
ISSN:
1479-5876
Año:
2018
Vol.:
16
Págs.:
213
Background: Mesenchymal stromal cells (MSCs) are a promising option to treat knee osteoarthritis (OA). Their safety and usefulness have been reported in several short-term clinical trials but less information is available on the long-term effects of MSC in patients with osteoarthritis. We have evaluated patients included in our previous randomized clinical trial (CMM-ART, NCT02123368) to determine their long-term clinical effect. Materials: A phase I/II multicenter randomized clinical trial with active control was conducted between 2012 and 2014. Thirty patients diagnosed with knee OA were randomly assigned to Control group, intraarticularly administered hyaluronic acid alone, or to two treatment groups, hyaluronic acid together with 10 x 10(6) or 100 x 10(6) cultured autologous bone marrow-derived MSCs (BM-MSCs), and followed up for 12 months. After a follow up of 4 years adverse effects and clinical evolution, assessed using VAS and WOMAC scorings are reported. Results: No adverse effects were reported after BM-MSCs administration or during the follow-up. BM-MSCs-administered patients improved according to VAS, median value (IQR) for Control, Low-dose and High-dose groups changed from 5 (3, 7), 7 (5, 8) and 6 (4, 8) to 7 (6, 7), 2 (2, 5) and 3 (3, 4), respectively at the end of follow up (Low-dose vs Control group, p = 0.01; High-dose vs Control group, p = 0.004). Patients receiving BM-MSCs also improved clinically according to WOMAC. Control group showed an increase median value of 4 points (-11;10) while Low-dose and High-dose groups exhibited values of -18 (-28;-9) and -10 (-21;-3) points, respectively (Low-dose vs Control group p = 0.043). No clinical differences between the BM-MSCs receiving groups were found. Conclusions: Single intraarticular injection of in vitro expanded autologous BM-MSCs is a safe and feasible procedure that results in long-term clinical and functional improvement of knee OA.
Revista:
JOURNAL OF TRANSLATIONAL MEDICINE
ISSN:
1479-5876
Año:
2017
Vol.:
15
N°:
1
Págs.:
Article number 104
Background: Prognosis of patients with glioblastoma multiforme (GBM) remains dismal, with median overall survival (OS) of about 15 months. It is therefore crucial to search alternative strategies that improve these results obtained with conventional treatments. In this context, immunotherapy seems to be a promising therapeutic option. We hypothesized that the addition of tumor lysate-pulsed autologous dendritic cells (DCs) vaccination to maximal safe resection followed by radiotherapy and concomitant and adjuvant temozolomide could improve patients' survival.
Methods: We conducted a phase-II clinical trial of autologous DCs vaccination in patients with newly diagnosed patients GBM who were candidates to complete or near complete resection. Candidates were finally included if residual tumor volume was lower than 1 cc on postoperative radiological examination. Autologous DCs were generated from peripheral blood monocytes and pulsed with autologous whole tumor lysate. The vaccination calendar started before radiotherapy and was continued during adjuvant chemotherapy. Progression free survival (PFS) and OS were analyzed with the Kaplan-Meier method. Immune response were assessed in blood samples obtained before each vaccines.
Results: Thirty-two consecutive patients were screened, one of which was a screening failure due to insufficient resection. Median age was 61 years (range 42-70). Karnofsky performance score (KPS) was 90-100 in 29%, 80 in 35.5% and 60-70 in 35.5% of cases. MGMT (O6-methylguanine-DNA-methyltransferase) promoter was methylated in 45.2% of patients. No severe adverse effects related to immunotherapy were registered. Median PFS was 12.7 months (CI 95% 7-16) and median OS was 23.4 months (95% CI 16-33.1). Increase in post-vaccination tumor specific immune response after vaccines (proliferation or cytokine production) was detected in 11/27 evaluated patients. No correlation between immune response and survival was found.
Conclusions: Our results suggest that the addition of tumor lysate-pulsed autologous DCs vaccination to tumor resection and combined radio-chemotherapy is feasible and safe. A multicenter randomized clinical trial is warranted to evaluate the potential survival benefit of this therapeutic approach. Trial registration This phase-II trial was registered as EudraCT: 2009-009879-35 and ClinicalTrials.gov Identifier: NCT01006044 retrospectively registered.
Revista:
INVESTIGATIVE OPHTHALMOLOGY AND VISUAL SCIENCE
ISSN:
0146-0404
Año:
2017
Vol.:
58
N°:
2
Págs.:
745-754
We demonstrated that the combined activation of PI3K/Akt and Smad2 results in in vitro expansion of phenotypic and functional CEC. Expanded cells were able to contribute to restoration of corneal endothelium in a rabbit model. These findings may represent a new therapeutic approach for treating corneal endothelial diseases
Revista:
TRANSLATIONAL RESEARCH
ISSN:
1931-5244
Año:
2017
Vol.:
188
Págs.:
80 - 91.e2
The aim of this nonrandomized, open label, phase 1 clinical trial was to evaluate the safety and the feasibility of the treatment with autologous bone marrow-derived endothelial progenitor cells (EPC) in decompensated liver cirrhosis. In addition, the changes in liver function and hepatic venous pressure gradient (HVPG) and their relation with the characteristics of the cellular product were analyzed. Twelve patients with Child-Pugh ¿8 liver cirrhosis underwent bone marrow harvest for ex vivo differentiation of EPC. The final product was administered through the hepatic artery in a single administration. Patients underwent clinical and radiologic follow-up for 12 months. The phenotype and the ability to produce cytokines and growth factors of the final cellular suspension were analyzed. Eleven patients were treated (feasibility 91%). No treatment-related severe adverse events were observed as consequence of any study procedure or treatment. Model for end-stage liver disease score improved significantly (P 0.042) in the first 90 days after cells administration and 5 of the 9 patients alive at 90 days showed a decreased of HVPG. There was a direct correlation between the expression of acetylated-low density lipoprotein and von Willebrand factor in the cellular product and the improvement in liver function and HVPG. The treatment with EPCs in patients with decompensated liver cirrhosis is safe and feasible and might have therapeutic potential. Patients receiving a higher amount
Revista:
STEM CELL RESEARCH
ISSN:
1873-5061
Año:
2016
Vol.:
16
N°:
1
Págs.:
20 - 23
In this work, mesenchymal stem cells derived from adipose tissue (ADSCs) were used for the generation of the human-induced pluripotent stem cell line G15.AO. Cell reprogramming was performed using retroviral vectors containing the Yamanaka factors, and the generated G15.AO hiPSC line showed normal karyotype, silencing of the exogenous reprogramming factors, induction of the typical pluripotency-associated markers, alkaline phosphatase enzymatic activity, and in vivo and in vitro differentiation ability to the three germ layers.
Revista:
JOURNAL OF TRANSLATIONAL MEDICINE
ISSN:
1479-5876
Año:
2016
Vol.:
14
N°:
1
Págs.:
246
The single intraarticular injection of in vitro expanded autologous BM-MSCs together with HA is a safe and feasible procedure that results in a clinical and functional improvement of knee OA, especially when 100 × 10(6) cells are administered. These results pave the way for a future phase III clinical trial.
Revista:
JAMA DERMATOLOGY
ISSN:
2168-6068
Año:
2015
Vol.:
151
N°:
8
Págs.:
897 - 899
Revista:
INTERNATIONAL JOURNAL OF OPHTHALMOLOGY
ISSN:
2222-3959
Año:
2014
Vol.:
7
N°:
6
Págs.:
988 - 995
AIM: To assess cultured limbal epithelial stem cell transplantation in patients with limbal stem cell deficiency by analyzing and quantifying corneal neovascularization.
METHODS: This retrospective, interventional case series included eight eyes with total limbal stem cell deficiency. Ex vivo limbal epithelial stem cells were cultured on human amniotic membrane using an animal-free culture method. The clinical parameters of limbal stem cell deficiency, impression cytology, and quantification of corneal neovascularization were evaluated before and after cultured limbal stem cell transplantation. The area of corneal neovascularization, vessel caliber (VC), and invasive area (IA) were analyzed before and after stem cell transplantation by image analysis software. Best-corrected visual acuity (BCVA), epithelial transparency, and impression cytology were also measured.
RESULTS: One year after surgery, successful cases showed a reduction (improvement) of all three parameters of corneal neovascularization [neovascular area (NA), VC, IA], while failed cases did not. NA decreased a mean of 32.31% (P=0.035), invasion area 29.37% (P=0.018) and VC 14.29% (P=0.072). BCVA improved in all eyes (mean follow-up, 76±21mo). Epithelial transparency improved significantly from 2.00±0.93 to 0.88±1.25 (P=0.014). Impression cytology showed that three cases failed after limbal epithelial stem cell therapy before 1y of follow-up.
CONCLUSION: This method of analyzing and monitoring surface vessels is useful for evaluating the epithelial status during follow-up, as successful cases showed a bigger reduction in corneal neovascularization parameters than failed cases. Using this method, successful cases could be differentiated from failed cases.
Autores:
Sara Llufriu; María Sepúlveda; Yolanda Blanco; et al.
Revista:
PLOS ONE
ISSN:
1932-6203
Año:
2014
Vol.:
9
N°:
12
Págs.:
e113936
At baseline 9 patients were randomized to receive MSCs (n¿=¿5) or placebo (n¿=¿4). One patient on placebo withdrew after having 3 relapses in the first 5 months. We did not identify any serious adverse events. At 6 months, patients treated with MSCs had a trend to lower mean cumulative number of GEL (3.1, 95% CI¿=¿1.1-8.8 vs 12.3, 95% CI¿=¿4.4-34.5, p¿=¿0.064), and at the end of study to reduced mean GEL (-2.8±5.9 vs 3±5.4, p¿=¿0.075). No significant treatment differences were detected in the secondary endpoints. We observed a non-significant decrease of the frequency of Th1 (CD4+ IFN-¿+) cells in blood of MSCs treated patients.
CONCLUSION:
Bone-marrow-MSCs are safe and may reduce inflammatory MRI parameters supporting their immunomodulatory properties. ClinicalTrials.gov NCT01228266.
Autores:
Fermín Sánchez-Guijo; Teresa Caballero-Velázquez; Olga López-Villar; et al.
Revista:
BIOLOGY OF BLOOD AND MARROW TRANSPLANTATION
ISSN:
1083-8791
Año:
2014
Vol.:
20
N°:
10
Págs.:
1580 - 1585
We evaluated the feasibility, safety, and efficacy of the administration of 4 sequential doses (intravenously administered on days 1, 4, 11, and 18) of cryopreserved bone marrow-derived mesenchymal stromal cells (MSC) expanded with platelet lysate and obtained from third-party donors as a second-line treatment for steroid-refractory acute graft-versus-host (aGVHD) disease in a series of 25 patients. All patients received at least 2 doses of MSC, whereas 21 received 3 doses and 18 received the initially planned 4 doses. Because of the achievement of partial response, 4 patients received additional doses of MSC. Median single cell dose administered was 1.1 × 10(6) MSC/kg of recipient body weight. There were no adverse events related to the MSC infusion in the 99 procedures performed, with the exception of a cardiac ischemic event that occurred twice in a patient with prior history of cardiac ischemia. Response to MSC at 60 days after the first dose was evaluable in 24 patients. Seventeen patients (71%) responded (11 complete and 6 partial responses), with a median time to response of 28 days after the first MSC dose, whereas 7 patients did not respond. In summary, we can conclude that sequential cryopreserved third-party MSC therapy administered on days 1, 4, 11, and 18 is a safe procedure for patients with steroid-refractory aGVHD. This strategy may provide a high rate of overall responses of aGVHD with a low toxicity profile.
Autores:
Iñigo Izal; Pablo Aranda; Patricia Sanz-Ramos; et al.
Revista:
KNEE SURGERY SPORTS TRAUMATOLOGY ARTHROSCOPY
ISSN:
0942-2056
Biocompatible PLLA scaffolds have been developed that can be efficiently loaded with MSCs. The scaffold supports chondrogenic differentiation and ECM deposition that improves the mechanics of the scaffold. Although this improvement does not met the expectations of a hyaline-like cartilage ECM, in part due to the lack of a mechanical stimulation, their potential use in the treatment of cartilage pathologies encourages to improve the mechanical component.
Revista:
WORLD JOURNAL OF CLINICAL ONCOLOGY
ISSN:
2218-4333
Año:
2012
Vol.:
3
N°:
11
Págs.:
142-149
Active immunotherapy with tumor lysate-pulsed, autologous dendritic cells is feasible, safe, well tolerated and biologically efficacious. A phase-II study is ongoing to possibly improve further on our very encouraging clinical results.
Autores:
Redondo, P. ; Giménez de Azcárate Trívez, A.; Marqués Martín, L.; et al.
Revista:
Dermatology Research and Practice
ISSN:
1687-6105
Año:
2011
Vol.:
Article ID 532139, 6 pages
Págs.:
532139-
Patients were followed for up to 6 months using clinical assessment of achromic lesions. Treated areas ranged between 4¿cm(2) and 210.6¿cm(2). Response to treatment was excellent in all patients with 90-95% repigmentation success rate.
Revista:
CELL TRANSPLANTATION
ISSN:
0963-6897
Año:
2011
Vol.:
20
N°:
2
Págs.:
259 - 269
There is a need for comparative studies to determine which cell types are better candidates to remedy ischemia. Here, we compared human AC133(+) cells and multipotent adult progenitor cells (hMAPC) in a mouse model reminiscent of critical limb ischemia. hMAPC or hAC133(+) cell transplantation induced a significant improvement in tissue perfusion (measured by microPET) 15 days posttransplantation compared to controls. This improvement persisted for 30 days in hMAPC-treated but not in hAC133(+)-injected animals. While transplantation of hAC133(+) cells promoted capillary growth, hMAPC transplantation also induced collateral expansion, decreased muscle necrosis/fibrosis, and improved muscle regeneration. Incorporation of differentiated MC 133(+) or hMAPC progeny into new vessels was limited; however, a paracrine angio/arteriogenic effect was demonstrated in animals treated with hMAPC. Accordingly, hMAPC-conditioned, but not hAC133(+)-conditioned, media stimulated vascular cell proliferation and prevented myoblast, endothelial, and smooth muscle cell apoptosis in vitro. Our study suggests that although hAC133(+) cell and hMAPC transplantation both contribute to vascular regeneration in ischemic limbs, hMAPC exert a more robust effect through trophic mechanisms, which translated into collateral and muscle fiber regeneration. This, in turn, conferred tissue protection and regeneration with longer term functional improvement.
Revista:
JOURNAL OF BIOMEDICAL MATERIALS RESEARCH PART A
ISSN:
1549-3296
Año:
2011
Vol.:
96
N°:
2
Págs.:
341 - 348
A better cell adhesion rate was observed in the cross-linked meshes. An increase in the mechanical properties after cell seeding was observed with a direct relation with the degree of cross-linking. All meshes analyzed showed a marked anisotropy that should be taken into account during the surgical procedure. The cross-linking treatment increased cell adhesion and the mechanical properties of the collagen meshes after seeding. These results suggest that the mechanical properties of this type of collagen mesh could be useful as scaffolds for repair of pelvic organ prolapse.
Revista:
HAEMATOLOGICA
ISSN:
1138-0381
Año:
2011
Vol.:
96
N°:
7
Págs.:
1072-1076
This trial evaluated the feasibility and efficacy of the infusion of mesenchymal stem cells expanded using human serum for the treatment of refractory acute or chronic graft-versus-host disease. Twenty-eight expansions were started. In 22, a minimum of more than 1x106 mesenchymal stem cells/kg were obtained after a median of 26 days; this threshold was not obtained in the remaining cases. Ten patients received cells for the treatment of refractory or relapsed acute graft-versus-host disease and 8 for chronic disease. One patient treated for acute graft-versus-host disease obtained a complete response, 6 had a partial response and 3 did not respond. One of the chronic patients achieved complete remision, 3 a partial response, and 4 did not respond. The current study supports the use of this approach in less heavily treated patients for both acute and chronic graft-versus-host disease.
Revista:
Oncogene
ISSN:
0950-9232
Año:
2010
Vol.:
29
N°:
22
Págs.:
3276 - 3286
Nacionales y Regionales
Título:
Desarrollo clínico de una terapia celular para reparación cardíaca basada en ingeniería de tejidos-CARDIOMESH
Código de expediente:
RTC-2016-4911-1
Investigador principal:
Felipe Luis Prósper Cardoso
Financiador:
MINISTERIO DE CIENCIA E INNOVACIÓN
Convocatoria:
2016 MINECO RETOS-COLABORACION
Fecha de inicio:
09/03/2016
Fecha fin:
28/02/2019
Importe concedido:
222.982,00€
Otros fondos:
-
Título:
Ensayo Clínico de fase I/II para el tratamiento de la enfermedad del injerto contra el huésped con una nueva generación de células estromales mesenquimales que expresan ectópicamente CXCR4 e IL10
Código de expediente:
ICI22/00067
Investigador principal:
Felipe Luis Prósper Cardoso
Financiador:
INSTITUTO DE SALUD CARLOS III
Convocatoria:
2022 AES Investigación Clínica Independiente
Fecha de inicio:
01/01/2023
Fecha fin:
31/12/2026
Importe concedido:
1.207.250,00€
Otros fondos:
-
Título:
Plataformas automáticas de producción de células CAR T para el tratamiento de leucemia B y linfoma B
Código de expediente:
RTC-2017-6578-1
Investigador principal:
Felipe Luis Prósper Cardoso
Financiador:
MINISTERIO DE CIENCIA E INNOVACIÓN
Convocatoria:
2017 MINECO RETOS COLABORACIÓN
Fecha de inicio:
01/01/2018
Fecha fin:
31/10/2021
Importe concedido:
100.985,84€
Otros fondos:
Fondos FEDER
Internacionales y Europeos
Título:
LGMed: La tecnología a servicio de la salud: desarrollo de dispositivos médicos de última generación
Código de expediente:
EFA313/19
Investigador principal:
Felipe Luis Prósper Cardoso
Financiador:
POCTEFA
Convocatoria:
3ª convocatoria de proyectos POCTEFA 2014-2020
Fecha de inicio:
01/11/2019
Fecha fin:
31/05/2022
Importe concedido:
144.469,00€
Otros fondos:
Fondos FEDER
Título:
Desarrollo de actividades innovadoras de carácter transfronterizas en el sector salud
Investigador principal:
Felipe Luis Prósper Cardoso
Financiador:
POCTEFA
Convocatoria:
INTERREG POCTEFA 2016
Fecha de inicio:
01/06/2016
Fecha fin:
31/05/2019
Importe concedido:
307.665,40€
Otros fondos:
Fondos FEDER
Título:
REgenerative therapy of intervertebral disc: a double blind phase 2b trial of intradiscal injection of mesenchymal stromal cells in degenerative disc disease unresponsive to conventional therapy.
Investigador principal:
Felipe Luis Prósper Cardoso
Financiador:
COMISIÓN EUROPEA
Convocatoria:
H2020-EC-SC1-PM-11-2016
Fecha de inicio:
01/01/2017
Fecha fin:
31/12/2022
Importe concedido:
373.000,00€
Otros fondos:
-
Otros (PIUNA, fundaciones, contratos…)
Título:
Fabricación Pei 15-103 para NOMA
Investigador principal:
Enrique José Andreu Oltra
Fecha de inicio:
17/04/2020
Fecha fin:
17/04/2023
Importe:
58.400,00€
Otros fondos:
-
Título:
Fabriación Células Mesenquimales Troncales Adultas Alógenica
Investigador principal:
Felipe Luis Prósper Cardoso, Enrique José Andreu Oltra
Fecha de inicio:
14/04/2021
Fecha fin:
14/04/2024
Importe:
0,00€
Otros fondos:
-
Título:
Efectos terapéuticos de la administración de células madre m
Investigador principal:
Felipe Luis Prósper Cardoso
Fecha de inicio:
11/07/2014
Fecha fin:
11/07/2024
Importe:
45.000,00€
Otros fondos:
Título:
Contrato para fabricación de producto celular para ensayo cl
Investigador principal:
Felipe Luis Prósper Cardoso
Fecha de inicio:
09/12/2020
Fecha fin:
09/12/2023
Importe:
196.897,91€
Otros fondos:
-
Título:
Fabricación PEI 15-103 para Ensayo Clínico FJD-MEIC-21-01
Investigador principal:
Enrique José Andreu Oltra
Fecha de inicio:
06/06/2022
Fecha fin:
06/06/2025
Importe:
94.840,00€
Otros fondos:
-