scholarly journals Transition from static culture to stirred tank bioreactor for the allogeneic production of therapeutic discogenic cell spheres

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Daniel Rodriguez-Granrose ◽  
Jeff Zurawski ◽  
Will Heaton ◽  
Terry Tandeski ◽  
Galina Dulatov ◽  
...  

Abstract Background Culturing cells as cell spheres results in a tissue-like environment that drives unique cell phenotypes, making it useful for generating cell populations intended for therapeutic use. Unfortunately, common methods that utilize static suspension culture have limited scalability, making commercialization of such cell therapies challenging. Our team is developing an allogeneic cell therapy for the treatment of lumbar disc degeneration comprised of discogenic cells, which are progenitor cells expanded from human nucleus pulposus cells that are grown in a sphere configuration. Methods We evaluate sphere production in Erlenmeyer, horizontal axis wheel, stirred tank bioreactor, and rocking bag format. We then explore the use of ramped agitation profiles and computational fluid dynamics to overcome obstacles related to cell settling and the undesired impact of mechanical forces on cell characteristics. Finally, we grow discogenic cells in stirred tank reactors (STRs) and test outcomes in vitro (potency via aggrecan production and identity) and in vivo (rabbit model of disc degeneration). Results Computation fluid dynamics were used to model hydrodynamic conditions in STR systems and develop statistically significant correlations to cell attributes including potency (measured by aggrecan production), cell doublings, cell settling, and sphere size. Subsequent model-based optimization and testing resulted in growth of cells with comparable attributes to the original static process, as measured using both in vitro and in vivo models. Maximum shear rate (1/s) was maintained between scales to demonstrate feasibility in a 50 L STR (200-fold scale-up). Conclusions Transition of discogenic cell production from static culture to a stirred-tank bioreactor enables cell sphere production in a scalable format. This work shows significant progress towards establishing a large-scale bioprocess methodology for this novel cell therapy that can be used for other, similar cell therapies.

2021 ◽  
Vol 9 (2) ◽  
pp. e001608
Author(s):  
Debottam Sinha ◽  
Sriganesh Srihari ◽  
Kirrliee Beckett ◽  
Laetitia Le Texier ◽  
Matthew Solomon ◽  
...  

BackgroundEpstein-Barr virus (EBV), an oncogenic human gammaherpesvirus, is associated with a wide range of human malignancies of epithelial and B-cell origin. Recent studies have demonstrated promising safety and clinical efficacy of allogeneic ‘off-the-shelf’ virus-specific T-cell therapies for post-transplant viral complications.MethodsTaking a clue from these studies, we developed a highly efficient EBV-specific T-cell expansion process using a replication-deficient AdE1-LMPpoly vector that specifically targets EBV-encoded nuclear antigen 1 (EBNA1) and latent membrane proteins 1 and 2 (LMP1 and LMP2), expressed in latency II malignancies.ResultsThese allogeneic EBV-specific T cells efficiently recognized human leukocyte antigen (HLA)-matched EBNA1-expressing and/or LMP1 and LMP2-expressing malignant cells and demonstrated therapeutic potential in a number of in vivo models, including EBV lymphomas that emerged spontaneously in humanized mice following EBV infection. Interestingly, we were able to override resistance to T-cell therapy in vivo using a ‘restriction-switching’ approach, through sequential infusion of two different allogeneic T-cell therapies restricted through different HLA alleles. Furthermore, we have shown that inhibition of the programmed cell death protein-1/programmed death-ligand 1 axis in combination with EBV-specific T-cell therapy significantly improved overall survival of tumor-bearing mice when compared with monotherapy.ConclusionThese findings suggest that restriction switching by sequential infusion of allogeneic T-cell therapies that target EBV through distinct HLA alleles may improve clinical response.


2020 ◽  
Vol 20 (1) ◽  
pp. 138-149 ◽  
Author(s):  
Lara Ionescu Silverman ◽  
Galina Dulatova ◽  
Terry Tandeski ◽  
Isaac E. Erickson ◽  
Beverly Lundell ◽  
...  

2020 ◽  
Vol 9 (1) ◽  
pp. 261 ◽  
Author(s):  
Tereza Filipi ◽  
Zuzana Hermanova ◽  
Jana Tureckova ◽  
Ondrej Vanatko ◽  
Miroslava Anderova

Amyotrophic lateral sclerosis (ALS) is a fatal neurological disease, which is characterized by the degeneration of motor neurons in the motor cortex and the spinal cord and subsequently by muscle atrophy. To date, numerous gene mutations have been linked to both sporadic and familial ALS, but the effort of many experimental groups to develop a suitable therapy has not, as of yet, proven successful. The original focus was on the degenerating motor neurons, when researchers tried to understand the pathological mechanisms that cause their slow death. However, it was soon discovered that ALS is a complicated and diverse pathology, where not only neurons, but also other cell types, play a crucial role via the so-called non-cell autonomous effect, which strongly deteriorates neuronal conditions. Subsequently, variable glia-based in vitro and in vivo models of ALS were established and used for brand-new experimental and clinical approaches. Such a shift towards glia soon bore its fruit in the form of several clinical studies, which more or less successfully tried to ward the unfavourable prognosis of ALS progression off. In this review, we aimed to summarize current knowledge regarding the involvement of each glial cell type in the progression of ALS, currently available treatments, and to provide an overview of diverse clinical trials covering pharmacological approaches, gene, and cell therapies.


2021 ◽  
Author(s):  
Sevil Kestane

This overview was evaluated by the development of diabetic retinopathy (DR) and the stem cell therapy approach. DR is a microvascular complication of diabetes mellitus, characterized by damage to the retinal blood vessels leading to progressive loss of vision. However, the pathophysiological mechanisms are complicated and not completely understood yet. The current treatment strategies have included medical, laser, intravitreal, and surgical approaches. It is known that the use of mesenchymal stem cells (MSC), which has a great potential, is promising for the treatment of many degenerative disorders, including the eye. In retinal degenerative diseases, MSCs were ameliorated retinal neurons and retinal pigmented epithelial cells in both in vitro and in vivo studies. Stem cell therapies show promise in neurodegenerative diseases. However, it is very important to know which type of stem cell will be used in which situations, the amount of stem cells to be applied, the method of application, and its physiological/neurophysiological effects. Therefore, it is of great importance to evaluate this subject physiologically. After stem cell application, its safety and efficacy should be followed for a long time. In the near future, widespread application of regenerative stem cell therapy may be a standard treatment in DR.


Antioxidants ◽  
2021 ◽  
Vol 10 (6) ◽  
pp. 832
Author(s):  
Katherine E. Burns ◽  
Robert F. Uhrig ◽  
Maggie E. Jewett ◽  
Madison F. Bourbon ◽  
Kristen A. Krupa

Silver nanoparticles (AgNPs) are being employed in numerous consumer goods and applications; however, they are renowned for inducing negative cellular consequences including toxicity, oxidative stress, and an inflammatory response. Nanotoxicological outcomes are dependent on numerous factors, including physicochemical, biological, and environmental influences. Currently, NP safety evaluations are carried out in both cell-based in vitro and animal in vivo models, with poor correlation between these mechanisms. These discrepancies highlight the need for enhanced exposure environments, which retain the advantages of in vitro models but incorporate critical in vivo influences, such as fluid dynamics. This study characterized the effects of dynamic flow on AgNP behavior, cellular interactions, and oxidative stress within both adherent alveolar (A549) and suspension monocyte (U937) models. This study determined that the presence of physiologically relevant flow resulted in substantial modifications to AgNP cellular interactions and subsequent oxidative stress, as assessed via reactive oxygen species (ROS), glutathione levels, p53, NFκB, and secretion of pro-inflammatory cytokines. Within the adherent model, dynamic flow reduced AgNP deposition and oxidative stress markers by roughly 20%. However, due to increased frequency of contact, the suspension U937 cells were associated with higher NP interactions and intracellular stress under fluid flow exposure conditions. For example, the increased AgNP association resulted in a 50% increase in intracellular ROS and p53 levels. This work highlights the potential of modified in vitro systems to improve analysis of AgNP dosimetry and safety evaluations, including oxidative stress assessments.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3762-3762
Author(s):  
Challice L. Bonifant ◽  
David Torres ◽  
Mireya Paulina Velasquez ◽  
Kota Iwahori ◽  
Caroline Arber ◽  
...  

Abstract Background: The outcome for patients with high risk acute myeloid leukemia (AML) remains poor. Thus new targeted therapies are needed and immunotherapies have the potential to fulfill this need. Adoptive transfer of tumor-specific T cells is one promising approach; however infused T cells do not redirect the large reservoir of resident T cells to tumors. To overcome this limitation we have recently developed a new approach to render T cells specific for tumor cells, which relies on genetically modifying T cells with a secretable, bispecific T cell engager (ENG-T cells). Secretion of bispecific protein should activate infused cells as well as bystander T cells against tumor. Consistent and prolonged synthesis of engagers by T cells should be superior to the intermittent, direct infusion of the recombinant bispecific antibody, not only because these recombinant proteins have short half-lives but also because they do not accumulate at tumor sites. The goal of this project was to generate T cells secreting IL3Rα (CD123) and CD3 bispecific T cell engagers (CD123-ENG T cells) and to evaluate their effector function in vitro and in vivo. Methods: CD123-ENG T cells were generated by transducing T cells with a retroviral vector encoding a CD123-specific T cell engager consisting of an scFv recognizing CD123 linked to an scFv recognizing CD3. The retroviral vector was also fashioned to include an mOrange gene downstream of an IRES element. The effector function of CD123-ENG T cells was evaluated in vitro and in a xenograft model. Results: Transduction of CD3/CD28-activated T cells resulted in mOrange expression in transduced T cells (median transduction efficiency 78%, range 49-92%) The presence of CD123-ENG molecules on the cell surface of both transduced and non-transduced T cells was demonstrated by FACS analysis using an F(ab) antibody that recognizes the CD123 scFv. Coculture of CD123+ AML cells (MV-4-11, MOLM-1, KG1a) and K562 cells genetically modified to express CD123 (K562-CD123) with engager T cells resulted in robust T-cell activation as judged by IFNγ and IL2 secretion. In contrast CD123-negative cells (K562) did not activate T cells. Likewise, control engager T cells (targeting an irrelevant antigen) were not activated when cultured with CD123+ cells. Antigen-dependent recognition was confirmed with cytotoxicity assays, in which engager T cells specifically killed CD123+ AML cells at an effector:target ratios ranging from 40:1-5:1 (p<0.05) Since CD123 is expressed at low levels on normal hematopoietic progenitor cells (HPCs), we evaluated the ability of CD123-ENG T cells to recognize normal HPCs in colony formation assays. Only at high CD123-ENG to HPC ratios did we observe a decline in colony formation, indicating that CD123+ AML cells can be targeted while preserving normal HPCs. In vivo anti-tumor activity was assessed using a modified KG1a AML cell line expressing firefly luciferase (KG1a.ffluc) to allow for serial bioluminescence imaging. NSG (NOD-SCID, IL2γR deficient) mice were sublethally irradiated 24 hours prior to leukemia infusion and were then treated with two intravenous doses of CD123-ENG T cells or control T cells on days 7 and 14. CD123-ENG T cells had potent anti-leukemia activity resulting in a significant survival advantage of treated animals (p=0.002; n=5 CD123-ENG, n=5 Control-ENG, n=10 control animals). Conclusions: We have generated CD123-ENG T cells with the potential to direct bystander T cells to CD123+ AML in a tumor antigen-specific manner. These CD123-ENG T cells have potent anti-AML activity in vivo, presenting a promising addition to currently available AML therapies. Disclosures Bonifant: Celgene, Bluebird bio: Baylor College of Medicine has a Research Collaboration with Celgene and Bluebirdbio to develop gene-modified T-cell Therapies. MPV, KI, XT, and SG have patent applications in the field of T-cell and gene-modified T-cell Therapy for cancer Other. Torres:Celgene, Bluebird bio: Baylor College of Medicine has a Research Collaboration with Celgene and Bluebirdbio to develop gene-modified T-cell Therapies. MPV, KI, XT, and SG have patent applications in the field of T-cell and gene-modified T-cell Therapy for cancer Other. Velasquez:Celgene, Bluebird bio: Baylor College of Medicine has a Research Collaboration with Celgene and Bluebirdbio to develop gene-modified T-cell Therapies. MPV, KI, XT, and SG have patent applications in the field of T-cell and gene-modified T-cell Therapy for cancer Other. Iwahori:Celgene, Bluebird bio: Baylor College of Medicine has a Research Collaboration with Celgene and Bluebirdbio to develop gene-modified T-cell Therapies. MPV, KI, XT, and SG have patent applications in the field of T-cell and gene-modified T-cell Therapy for cancer Other. Arber:Celgene, Bluebird bio: Baylor College of Medicine has a Research Collaboration with Celgene and Bluebirdbio to develop gene-modified T-cell Therapies. MPV, KI, XT, and SG have patent applications in the field of T-cell and gene-modified T-cell Therapy for cancer Other. Song:Celgene, Bluebird bio: Baylor College of Medicine has a Research Collaboration with Celgene and Bluebirdbio to develop gene-modified T-cell Therapies. MPV, KI, XT, and SG have patent applications in the field of T-cell and gene-modified T-cell Therapy for cancer Other. Gottschalk:Celgene, Bluebird bio: Baylor College of Medicine has a Research Collaboration with Celgene and Bluebirdbio to develop gene-modified T-cell Therapies. MPV, KI, XT, and SG have patent applications in the field of T-cell and gene-modified T-cell Therapy for cancer Other.


2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Kyong-Su Park ◽  
Elga Bandeira ◽  
Ganesh V. Shelke ◽  
Cecilia Lässer ◽  
Jan Lötvall

Abstract After the initial investigations into applications of mesenchymal stem cells (MSCs) for cell therapy, there was increased interest in their secreted soluble factors. Following studies of MSCs and their secreted factors, extracellular vesicles (EVs) released from MSCs have emerged as a new mode of intercellular crosstalk. MSC-derived EVs have been identified as essential signaling mediators under both physiological and pathological conditions, and they appear to be responsible for many of the therapeutic effects of MSCs. In several in vitro and in vivo models, EVs have been observed to have supportive functions in modulating the immune system, mainly mediated by EV-associated proteins and nucleic acids. Moreover, stimulation of MSCs with biophysical or biochemical cues, including EVs from other cells, has been shown to influence the contents and biological activities of subsequent MSC-derived EVs. This review provides on overview of the contents of MSC-derived EVs in terms of their supportive effects, and it provides different perspectives on the manipulation of MSCs to improve the secretion of EVs and subsequent EV-mediated activities. In this review, we discuss the possibilities for manipulating MSCs for EV-based cell therapy and for using EVs to affect the expression of elements of interest in MSCs. In this way, we provide a clear perspective on the state of the art of EVs in cell therapy focusing on MSCs, and we raise pertinent questions and suggestions for knowledge gaps to be filled.


Blood ◽  
2020 ◽  
Author(s):  
Fraser Soares ◽  
Branson Chen ◽  
Jong Bok Lee ◽  
Musaddeque Ahmed ◽  
Dalam Ly ◽  
...  

Acute myeloid leukemia (AML) remains a devastating disease in need of new therapies to improve patient survival. Targeted, adoptive T cell therapies have achieved impressive clinical outcomes in some B-cell leukemias and lymphomas but not in AML. Double negative T cells (DNTs) effectively kill blast cells from the majority of AML patients and are now being tested in clinical trials. However, AML blasts obtained from ~30% of patients show resistance to DNT cell-mediated cytotoxicity; the markers or mechanisms underlying this resistance have not been elucidated. Here, we used a targeted CRISPR/Cas9 screen to identify genes that confer susceptibility of AML cells to DNT cell therapy. Inactivation of the SAGA deubiquitinating complex components sensitized AML cells to DNT-mediated cytotoxicity. In contrast, CD64 inactivation resulted in resistance to DNT-mediated cytotoxicity. Importantly, the level of CD64 expression strongly correlated with the sensitivity of AML cells to DNT cell treatment. Furthermore, the ectopic expression of CD64 overcame AML resistance to DNTs both in vitro and in vivo. Altogether, our data demonstrate the utility of CRISPR/Cas9 screens to uncover mechanisms underlying the sensitivity to DNT cell therapy and suggest CD64 as a predictive marker for response in AML patients.


2020 ◽  
pp. 53-68
Author(s):  
Sergey Vadimovich Yargin ◽  

Stem cells and cell therapy are one of the most widely discussed topics in the medical and biological literature. It is supposed to differentiate exogenous SC into various cell lineages and to replace senescent, dysfunctional, and damaged cells. However, in vitro differentiation with the expression of certain markers does not prove replacement of functioning cells in vivo. The application of cell therapies in cardiovascular, hepatic, neurodegenerative diseases, osteoarthritis, and diabetes mellitus is discussed in this article. Some publications exaggerate the successes of cell therapies without giving due consideration to potential adverse effects. In recent years, there has been a global increase in the number of clinics offering stem cell treatment with unproven efficiency. In conclusion, therapeutic methods with unproven efficacy and potential adverse effects should be applied within the framework of high-quality scientific research programs that are free of conflicts of interest.Key words: stem cells; cell therapy; cardiovascular diseases; osteoarthritis; liver cirrhosis; diabetes mellitus; central nervous system.


Sign in / Sign up

Export Citation Format

Share Document