advanced therapy medicinal product
Recently Published Documents


TOTAL DOCUMENTS

54
(FIVE YEARS 39)

H-INDEX

6
(FIVE YEARS 4)

2022 ◽  
Vol 20 (1) ◽  
Author(s):  
Cristiana Lavazza ◽  
Silvia Budelli ◽  
Elisa Montelatici ◽  
Mariele Viganò ◽  
Francesca Ulbar ◽  
...  

Abstract Background A growing number of clinical trials have shown that regulatory T (Treg) cell transfer may have a favorable effect on the maintenance of self-tolerance and immune homeostasis in different conditions such as graft-versus-host disease (GvHD), solid organ transplantation, type 1 diabetes, and others. In this context, the availability of a robust manufacturing protocol that is able to produce a sufficient number of functional Treg cells represents a fundamental prerequisite for the success of a cell therapy clinical protocol. However, extended workflow guidelines for nonprofit manufacturers are currently lacking. Despite the fact that different successful manufacturing procedures and cell products with excellent safety profiles have been reported from early clinical trials, the selection and expansion protocols for Treg cells vary a lot. The objective of this study was to validate a Good Manufacturing Practice (GMP)-compliant protocol for the production of Treg cells that approaches the whole process with a risk-management methodology, from process design to completion of final product development. High emphasis was given to the description of the quality control (QC) methodologies used for the in-process and release tests (sterility, endotoxin test, mycoplasma, and immunophenotype). Results The GMP-compliant protocol defined in this work allows at least 4.11 × 109 Treg cells to be obtained with an average purity of 95.75 ± 4.38% and can be used in different clinical settings to exploit Treg cell immunomodulatory function. Conclusions These results could be of great use for facilities implementing GMP-compliant cell therapy protocols of these cells for different conditions aimed at restoring the Treg cell number and function, which may slow the progression of certain diseases.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Miryam Mebarki ◽  
Nathan Iglicki ◽  
Céline Marigny ◽  
Camille Abadie ◽  
Claire Nicolet ◽  
...  

Abstract Background Umbilical cord-derived mesenchymal stromal cells (UC-MSCs) revealed their key role in immune regulation, offering promising therapeutic perspectives for immune and inflammatory diseases. We aimed to develop a production process of an UC-MSC-based product and then to characterize UC-MSC properties and immunomodulatory activities in vitro, related to their clinical use and finally, to transfer this technology to a good manufacturing practice (GMP) compliant facility, to manufacture an advanced therapy medicinal product (ATMP). Methods Fifteen human umbilical cords (UCs) were collected to develop the production process. Three batches of UC-MSCs from a single donor were characterized at basal state and after in vitro pro-inflammatory stimulation by interferon-γ (IFNγ) and tumor necrosis factor-α (TNFα). Proliferation, immunophenotype, activation markers’ expression and the inhibition of T cell proliferation were assessed. Finally, this technology was transferred to a GMP-compliant facility to manufacture an UC-MSC-based ATMP, from a single donor, using the explant method followed by the establishment of master and work cell stocks. Results Twelve UCs were processed successfully allowing to isolate UC-MSCs with doubling time and population doubling remaining stable until passage 4. CD90, CD105, CD73, CD44, CD29, CD166 expression was positive; CD14, CD45, CD31, HLA-DR, CD40, CD80 and CD86 expression was negative, while CD146 and HLA-ABC expression was heterogeneous. Cell morphology, proliferation and immunophenotype were not modified by inflammatory treatment. Indoleamine 2,3-dioxygenase (IDO) expression was significantly induced by IFNγ and IFNγ + TNFα versus non-treated cells. Intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) expression was induced significantly after priming. T cell proliferation was significantly decreased in the presence of UC-MSCs in a dose-dependent manner. This inhibitory effect was improved by IFNγ or IFNγ + TNFα, at UC-MSCs:PBMC ratio 1:10 and 1:30, whereas only IFNγ allowed to decrease significantly T cell proliferation at ratio 1:100. The manufacturing process of the UC-MSC-based ATMP was qualified and authorized by the French regulatory agency for clinical use (NCT04333368). Conclusion This work allowed to develop an investigational UC-MSC-based ATMP authorized for clinical use. Our results showed that an inflammatory environment preserves the biological properties of UC-MSCs with an improvement of their immunomodulatory functions.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi97-vi97
Author(s):  
Stefaan Van Gool ◽  
Jennifer Makalowski ◽  
Volker Schirrmacher ◽  
Wilfried Stuecker

Abstract The prognosis of IDH1 wild type MGMT promotor unmethylated (MGMT-p-UM) GBM patients remains poor. Addition of TMZ to radiotherapy shifted the median OS from 11.8 to 12.6 months (Stupp, Lancet Oncol 2019). We retrospectively analysed the value of individualized multimodal immunotherapy (IMI) to improve OS in these patients. Adults with first event of IDH1wt GBM and documented status of MGMT-p-UM, and treated with IMI in the period June 2015 till July 2020, were selected. IMI consisted of 1/ immunogenic cell death (ICD) therapy (NDV injections + modulated electrohyperthermia), 2/ active specific immunotherapy with autologous mature dendritic cells loaded with tumor lysate or ICD therapy-induced serum-derived antigenic extracellular microvesicles and apoptotic bodies (IO-Vac® is an approved advanced therapy medicinal product since 27/05/2015), 3/ modulatory immunotherapy adapted to the patient, and 4/ complementary medicines. Twenty-eight patients (11f, 17m) had a median age of 48y (range 18-69) and a KPI of 90 (50-100). Extent of resection was complete (11), < complete (9) or not documented (8). Seven patients were treated with surgery/radio(chemo)therapy and subsequent IMI (Group-1); 21 patients were treated with radiochemotherapy followed by maintenance TMZ + ICD therapy, followed by DC vaccines (Group-2). Both groups received further maintenance ICD therapy. Age, KPI and extent of resection were not different amongst both groups. PFS was not assessed because of challenges about pseudoprogression. The median OS of group-1 patients was 11m (2y OS: 0%). Surprisingly the median OS of group-2 patients was 18m with 2y OS of 17% (CI95%: +31, -15), which was significantly (Log-rank: p = 0.027) different from group-1. The data suggest that addition of IMI after local therapy on its own has no relevant effect on OS in IDH1 wild type MGMT-p-UM GBM patients, similar to maintenance TMZ. However, the combination of both TMZ + IMI significantly improves median OS.


2021 ◽  
Author(s):  
Miryam MEBARKI ◽  
Nathan Iglicki ◽  
Céline Marigny ◽  
Camille Abadie ◽  
Claire Nicolet ◽  
...  

Abstract Background Umbilical Cord-derived Mesenchymal Stromal Cells (UC-MSCs) revealed their key role in immune regulation, offering promising therapeutic perspectives for immune and inflammatory diseases. We aimed to develop a production process of an UC-MSCs-based product, then to characterize UC-MSCs properties and immunomodulatory activities in vitro, related to their clinical use and finally, to transfer this technology to a good manufacturing practice (GMP) compliant facility, to manufacture an Advanced Therapy Medicinal Product (ATMP). Methods Fifteen human umbilical cords (UCs) were collected to develop the production process. Three batches of UC-MSCs from a single donor were characterized at basal state and after in vitro pro-inflammatory stimulation by interferon-γ (IFNγ) and Tumor Necrosis Factor-α (TNFα). Proliferation, immunophenotype, activation markers expression and the inhibition of T-cells proliferation were assessed. Finally, this technology was transferred to a GMP-compliant facility to manufacture an UC-MSCs-based ATMP, from a single donor, using the explant method followed by the establishment of master and work cell stocks. Results Twelve UCs were processed successfully allowing to isolate UC-MSCs with doubling time and population doubling remaining stable until passage 4. CD90, CD105, CD73, CD44, CD29, CD166 expression was positive; CD14, CD45, CD31, HLA-DR, CD40, CD80 and CD86 negative, while CD146 and HLA-ABC expression was heterogeneous. Cell morphology, proliferation and immunophenotype were not modified by inflammatory treatment. Indoleamine 2,3-dioxygenase (IDO) expression was significantly induced by IFNγ and IFNγ + TNFα versus non-treated cells. Inter Cellular Adhesion Molecule-1 (ICAM-1) and Vascular Cell Adhesion Molecule 1 (VCAM-1) expression was induced significantly after priming. T-cells proliferation was significantly decreased in the presence of UC-MSCs in a dose-dependent manner. This inhibitory effect was improved by IFNγ or IFNγ + TNFα, at UC-MSCs:PBMC ratio 1:10 and 1:30, whereas only IFNγ allowed to decrease significantly T-cells proliferation at ratio 1:100. The manufacturing process of the UC-MSCs-based ATMP was qualified and authorized by the French regulatory agency for clinical use (NCT04333368). Conclusion This work allowed to develop an investigational UC-MSCs-based ATMP authorized for clinical use. Our results showed that an inflammatory environment preserves the biological properties of UC-MSCs with an improvement of their immunomodulatory functions.


Cells ◽  
2021 ◽  
Vol 10 (8) ◽  
pp. 1908
Author(s):  
Anna Labedz-Maslowska ◽  
Agnieszka Szkaradek ◽  
Tomasz Mierzwinski ◽  
Zbigniew Madeja ◽  
Ewa Zuba-Surma

Adipose tissue (AT) represents a commonly used source of mesenchymal stem/stromal cells (MSCs) whose proregenerative potential has been widely investigated in multiple clinical trials worldwide. However, the standardization of the manufacturing process of MSC-based cell therapy medicinal products in compliance with the requirements of the local authorities is obligatory and will allow us to obtain the necessary permits for product administration according to its intended use. Within the research phase (RD), we optimized the protocols used for the processing and ex vivo expansion of AT-derived MSCs (AT-MSCs) for the development of an Advanced Therapy Medicinal Product (ATMP) for use in humans. Critical process parameters (including, e.g., the concentration of enzyme used for AT digestion, cell culture conditions) were identified and examined to ensure the high quality of the final product containing AT-MSCs. We confirmed the identity of isolated AT-MSCs as MSCs and their trilineage differentiation potential according to the International Society for Cellular Therapy (ISCT) recommendations. Based on the conducted experiments, in-process quality control (QC) parameters and acceptance criteria were defined for the manufacturing of hospital exemption ATMP (HE-ATMP). Finally, we conducted a validation of the manufacturing process in a GMP facility. In the current study, we presented a process approach leading to the optimization of processing and the ex vivo expansion of AT-MSCs for the development of ATMP for use in humans.


2021 ◽  
Author(s):  
Cristiana Lavazza ◽  
Silvia Budelli ◽  
Elisa Montelatici ◽  
Mariele Viganò ◽  
Francesca Ulbar ◽  
...  

Abstract BACKGROUND: A growing number of clinical trials have shown that regulatory T (Treg) cells transfer may have a favorable effect on the maintenance of self-tolerance and immune homeostasis in different conditions such as graft-versus-host disease (GvHD), solid organ transplantation, type 1 diabetes, and others. In this context, the availability of a robust manufacturing protocol that is able to produce a sufficient number of functional Treg cells represents a fundamental prerequisite for the success of a cell therapy clinical protocol. However, extended workflow guidelines for nonprofit manufacturers are currently lacking. Despite the fact that different successful manufacturing procedures and cell products with excellent safety profiles have been reported from early clinical trials, the selection and expansion protocols for Treg cells vary a lot. The objective of this study was to validate a Good Manufacturing Practice (GMP)-compliant protocol for the production of Treg cells that approaches the whole process with a risk-management methodology, from process design to completion of final product development. High emphasis was given to the description of the quality control (QC) methodologies used for the in-process and release tests (sterility, endotoxin test, mycoplasma, and immunophenotyping).RESULTS: The GMP-compliant protocol defined in this work allows at least 4.11 × 109 Treg cells to be obtained with an average purity of 95.75 ± 4.38% and can be used in different clinical settings to exploit Treg cell immunomodulatory function.CONCLUSIONS: These results could be of great use for facilities implementing GMP-compliant cell therapy protocols of these cells for different conditions aimed at restoring the Treg cell number and function, which may slow the progression of certain diseases.


Biomedicines ◽  
2021 ◽  
Vol 9 (7) ◽  
pp. 775
Author(s):  
Olimpia Ortiz-Arrabal ◽  
Ramón Carmona ◽  
Óscar-Darío García-García ◽  
Jesús Chato-Astrain ◽  
David Sánchez-Porras ◽  
...  

Because cartilage has limited regenerative capability, a fully efficient advanced therapy medicinal product is needed to treat severe cartilage damage. We evaluated a novel biomaterial obtained by decellularizing sturgeon chondral endoskeleton tissue for use in cartilage tissue engineering. In silico analysis suggested high homology between human and sturgeon collagen proteins, and ultra-performance liquid chromatography confirmed that both types of cartilage consisted mainly of the same amino acids. Decellularized sturgeon cartilage was recellularized with human chondrocytes and four types of human mesenchymal stem cells (MSC) and their suitability for generating a cartilage substitute was assessed ex vivo and in vivo. The results supported the biocompatibility of the novel scaffold, as well as its ability to sustain cell adhesion, proliferation and differentiation. In vivo assays showed that the MSC cells in grafted cartilage disks were biosynthetically active and able to remodel the extracellular matrix of cartilage substitutes, with the production of type II collagen and other relevant components, especially when adipose tissue MSC were used. In addition, these cartilage substitutes triggered a pro-regenerative reaction mediated by CD206-positive M2 macrophages. These preliminary results warrant further research to characterize in greater detail the potential clinical translation of these novel cartilage substitutes.


Cells ◽  
2021 ◽  
Vol 10 (6) ◽  
pp. 1320
Author(s):  
Chantal Lechanteur ◽  
Alexandra Briquet ◽  
Virginie Bettonville ◽  
Etienne Baudoux ◽  
Yves Beguin

Following European regulation 1394/2007, mesenchymal stromal cell (MSCs) have become an advanced therapy medicinal product (ATMP) that must be produced following the good manufacturing practice (GMP) standards. We describe the upgrade of our existing clinical-grade MSC manufacturing process to obtain GMP certification. Staff organization, premises/equipment qualification and monitoring, raw materials management, starting materials, technical manufacturing processes, quality controls, and the release, thawing and infusion were substantially reorganized. Numerous studies have been carried out to validate cultures and demonstrate the short-term stability of fresh or thawed products, as well their stability during long-term storage. Detailed results of media simulation tests, validation runs and early MSC batches are presented. We also report the validation of a new variant of the process aiming to prepare fresh MSCs for the treatment of specific lesions of Crohn’s disease by local injection. In conclusion, we have successfully ensured the adaptation of our clinical-grade MSC production process to the GMP requirements. The GMP manufacturing of MSC products is feasible in the academic setting for a limited number of batches with a significant cost increase, but moving to large-scale production necessary for phase III trials would require the involvement of industrial partners.


Sign in / Sign up

Export Citation Format

Share Document