CytotherapyPub Date : 2025-05-01DOI: 10.1016/j.jcyt.2025.03.075
S. Azarapetian , E. Hatanaka , E. Gonzalez-Cubero , L. Lydaki , I. Lorenzo Llorente
{"title":"cGMP production of hiPSC-Glial Enriched Progenitors, a cell-based therapy to treat white matter stroke and vascular dementia.","authors":"S. Azarapetian , E. Hatanaka , E. Gonzalez-Cubero , L. Lydaki , I. Lorenzo Llorente","doi":"10.1016/j.jcyt.2025.03.075","DOIUrl":"10.1016/j.jcyt.2025.03.075","url":null,"abstract":"<div><h3>Background and Aims</h3><div>Stroke is the leading cause of adult disability and the second leading cause of dementia. White matter stroke (WMS) constitutes up to 30% of all stroke subtypes, and it is a distinct process from “large artery stroke”. WMS starts as small infarcts in deep penetrating blood vessels in the brain but progresses, accumulates, and expands from preexisting lesions into adjacent white matter to produce hemiparesis with incomplete recovery, gait abnormalities, cognitive decline, and difficulties in executive functioning that present as vascular dementia. Unlike “large artery stroke”, WMS does not damage neuronal cell bodies, but damages axonal tracts and glial cells. Therefore, a cell-based therapy that can replace lost glia and induce structural repair in WMS/VaD is of great promise.</div></div><div><h3>Methodology</h3><div>In this study, we have developed a unique allogenic human induced pluripotent stem cell (hiPSC) derived Glial Enriched Progenitor (GEP) cell therapy product for the treatment of WMS and VaD.</div></div><div><h3>Results</h3><div>We have demonstrated that hiPSC-GEPs transplanted into the brain after WMS/VaD promoted motor and cognitive recovery through 3 mechanisms of action: axonal growth, astrocytic modulation, and oligodendrocyte differentiation and remyelination. To date, we have also qualified the entire manufacturing process for the intended therapeutic candidate, hiPSC-GEPs, through safety, identity, purity, activity, and stability qualification assays. To demonstrate the scale-up manufacturing capabilities of the potential therapeutic product, we have developed a new cGMP manufacturing protocol and produced well above the necessary number of cells for phases 1 and 2 of a future clinical application.</div></div><div><h3>Conclusion</h3><div>This preliminary work could pave the way for a faster route to Pre-IND, FDA approval, and a future clinical application.</div></div>","PeriodicalId":50597,"journal":{"name":"Cytotherapy","volume":"27 5","pages":"Pages S46-S47"},"PeriodicalIF":3.7,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143888010","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CytotherapyPub Date : 2025-05-01DOI: 10.1016/j.jcyt.2025.03.089
C. Annunziata , S. Falci , K. Brown , N. Fereydouni , K. Payte , M. Kele , T. Kallur
{"title":"Biorelevant full-length Biolaminin 521 enhances primary MSCs proliferation and generation of iMSCs with stable expansion and differentiation potential","authors":"C. Annunziata , S. Falci , K. Brown , N. Fereydouni , K. Payte , M. Kele , T. Kallur","doi":"10.1016/j.jcyt.2025.03.089","DOIUrl":"10.1016/j.jcyt.2025.03.089","url":null,"abstract":"<div><h3>Background & Aim</h3><div>Mesenchymal stromal cells (MSCs) are powerful therapeutic tools in regenerative medicine. The functions of MSCs are tightly regulated by the interaction with their microenvironment, where survival and differentiation potential of MSCs is controlled by intrinsic mechano-signalling pathways. However, challenges remain such as source-heterogeneity and use of undefined and xenogenic culture conditions hampering standardization and clinical transition. Protocol adaptation to xenofree and chemically defined cell culture condition, suitable for both cell type and applications, is crucial for expanding their therapeutic potential. Laminins are a family of large extracellular matrix (ECM) proteins found throughout the body, essential for healthy tissue development- and tissue homeostasis, particularly abundant in basement membranes of epithelial- and endothelial tissues. Here, full-length laminins play a critical role in shaping the microenvironment and in mediating cell-ECM interactions via establishment of focal adhesion contacts. Their role is essential for stable cellular survival, identity, and migration. Biolaminin® s are full-length human recombinant laminin proteins, scientifically proven to support human pluripotent stem cells (PSC) and targeted differentiation <em>in vitro,</em> replacing tissue extract products. Biolaminin® 521 is particularly known to support PSCs and adult stem cells niches <em>in vitro</em>.</div></div><div><h3>Methodology</h3><div>Primary adipose derived-MSCs and induced PSC (iPSC)-derived mesenchymal stromal cells (iMSCs) were employed in these studies.</div></div><div><h3>Results</h3><div>Primary MSCs cultivated on Biolaminin® 521 maintain a stable- and fast proliferation compared to plastic (no coat) or other traditional substrates, also in serum-free conditions. Moreover, researchers have demonstrated that full-length Biolaminin 521 supports generation, expansion and differentiation of iMSCs. Biolaminin 521 supports stable and fast proliferation of iMSCs up to 10 passages compared to other substrates such as fragmented laminin and laminin-rich tissue extract products. Full-length laminin significantly increases mineral deposition of iMSCs-derived osteoblast inducing a uniform and efficient osteogenic differentiation over plastic or tissue extracts products.</div></div><div><h3>Conclusion</h3><div>In conclusion, Biolaminin 521 is a biologically relevant, xeno-free and defined substrate, enhancing primary MSCs proliferation and supporting iMSCs culture, from initial generation to downstream differentiation enabling protocol standardization and clinical translation.</div></div>","PeriodicalId":50597,"journal":{"name":"Cytotherapy","volume":"27 5","pages":"Page S53"},"PeriodicalIF":3.7,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143888120","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CytotherapyPub Date : 2025-05-01DOI: 10.1016/j.jcyt.2025.03.065
M. Chambers
{"title":"DEMONSTRATING NEW APPROACHES TO SMALL-BATCH CELL THERAPY MANUFACTURE THROUGH A LABEL-FREE MICROFLUIDIC CELL SORTER","authors":"M. Chambers","doi":"10.1016/j.jcyt.2025.03.065","DOIUrl":"10.1016/j.jcyt.2025.03.065","url":null,"abstract":"<div><h3>Background & Aim</h3><div>Automation of bioprocessing and analysis needs to be at the heart of advances in cell and gene manufacture for the transition to personalised therapies. Small-batch processes still require significant manual control and are limited to off-line quality analysis. Automated and decentralised production technologies, combined with in-line or at-line process analytical technologies, must be the route forward to realise the potential of personalised therapies. Integrating AI and machine learning (ML) into these approaches offers opportunities to reduce costs and access novel capabilities that have not yet been fully explored. Here, we demonstrate that simple and inexpensive devices can be constructed for specific applications, drawing on examples from the development of a microfluidic cell sorter. Driven by an ML algorithm, the device was designed to provide greater freedom to therapy developers without commitment to bulk manufacturing processes.</div></div><div><h3>Methodology</h3><div>The device was built from commercially available components. Fluid is driven by a syringe pump and controlled by a pair of rocker valves. The solution of mixed cells is held in a syringe. The mixed cells pass through a y-channel where they are imaged by an optics system. A ML algorithm identifies the cells and directs the valves to sort them into the correct channel. The total cost of all components is less than $3000.</div></div><div><h3>Results</h3><div>Morphologically dissimilar cells can be identified by optical imaging and separated without relying on extrinsic labelling. Cells do not require pre or post processing to add or remove fluorescent or magnetic labels. Labelling cells prior to sorting adds time and complexity to the process step and labelling physiologically similar cells for purification can be a barrier to the development of autologous cell therapies, where there is a risk of expanding and reintroducing diseased cells. The algorithm is trained to distinguish differences in the size and shape of cells. Once correctly trained, the device is applicable to any morphologically dissimilar mixture of cells delivered into the channel.</div></div><div><h3>Conclusion</h3><div>Increasing automation while reducing process complexity is essential for the development of personalised therapies, particularly autologous cell therapies. Automating small-batch production need not require high investment in manufacturing facilities, while leveraging new technologies and alternative approaches to manufacture can lower production times and costs, providing better outcomes for patients and developers.</div></div>","PeriodicalId":50597,"journal":{"name":"Cytotherapy","volume":"27 5","pages":"Pages S39-S40"},"PeriodicalIF":3.7,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143888154","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CytotherapyPub Date : 2025-05-01DOI: 10.1016/j.jcyt.2025.03.024
A. Lotfy , H. Wei , W. Gou , E. Green , J. Kim , C. Strange , H. Wang
{"title":"Overexpression of PD-L1 in mesenchymal stem/stromal cells and their derived extracellular vesicles enhances immunomodulatory properties and therapeutic efficacy in nonobese diabetic mice","authors":"A. Lotfy , H. Wei , W. Gou , E. Green , J. Kim , C. Strange , H. Wang","doi":"10.1016/j.jcyt.2025.03.024","DOIUrl":"10.1016/j.jcyt.2025.03.024","url":null,"abstract":"<div><h3>Background & Aim</h3><div>Type 1 diabetes (T1D) is an autoimmune disease characterized by the destruction of pancreatic β cells, necessitating lifelong insulin therapy. Programmed death ligand-1 (PD-L1) is critical in maintaining peripheral tolerance and immunological homeostasis. This study investigates the protective effects of mesenchymal stem/stromal cells (MSCs) overexpressing PD-L1 and their derived extracellular vesicles (PEVs) in murine models of T1D.</div></div><div><h3>Methodology</h3><div>PD-L1-engineered MSCs (PD-L1-MSCs) were generated by infecting human bone marrow-derived MSCs with a lentiviral vector encoding the human PD-L1 gene. The immunosuppressive properties and impact on islet cell death of PD-L1 MSCs and PEVs were evaluated <em>in vitro</em> by coculturing them with peripheral blood mononuclear cells (PBMCs) and murine islets. For the <em>in vivo</em> study, 8-week-old female NOD mice were given a single IV infusion of MSCs (n=17), PD-L1-MSCs (n=18), MSC-EVs (n=20), PEVs (n=20), or PBS (control, n=20), respectively. Blood glucose levels were monitored weekly for 25 weeks. In separated groups of mice, the pancreas, pancreatic lymph nodes (PLN), and spleen were collected 3 weeks post-treatment to assess immune cell infiltration, T cell profiling and function via H&E staining and flow cytometry. Statistical differences were analyzed using one-way ANOVA with post-hoc correction.</div></div><div><h3>Results</h3><div><em>In vitro</em>, PD-L1-MSCs and PEVs effectively suppressed T cell proliferation and increased T regulatory cells (Tregs, p<0.01 vs. control), highlighting their immunomodulatory potential. Islet viability was significantly improved when cocultured with PD-L1-MSCs (viability: 75.3±5.3%) or PEVs (91.3±1.9%), vs. controls (59.9±6.0%, p<0.05 vs control in each group). In vivo, PD-L1-MSCs or PEVs significantly reduced blood glucose and delayed T1D onset in NOD mice (CTR vs. PD-L1-MSC, p<0.05; CTR vs. PEV, p<0.01, Logrank test). H&E staining showed a significant decrease in immune cell infiltration in the pancreatic islets. In PLN, PD-L1 MSCs or PEVs decreased CD8<sup>+</sup> T cell number (p< 0.03 vs control), and increased CD8<sup>+</sup> T cell exhaustion and Tregs.</div></div><div><h3>Conclusion</h3><div>These findings suggest that PD-L1 overexpression in MSCs enhances their immunosuppressive and protective effects on pancreatic islets, partly by enhancing Tregs and CD8<sup>+</sup> T cell exhaustion. This underscores the therapeutic potential of PD-L1-MSCs and PEVs in modulating immune responses in T1D and reveals the underlying cellular mechanisms.</div></div>","PeriodicalId":50597,"journal":{"name":"Cytotherapy","volume":"27 5","pages":"Page S19"},"PeriodicalIF":3.7,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143888151","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CytotherapyPub Date : 2025-05-01DOI: 10.1016/j.jcyt.2025.03.046
N. Avni , Y. Geffen , K. Taler , N. Drori Carmi , I. Paz , S. Rubin , N. Kutner , S. Baumgarten-Soueid , I. Solomon , I. Sarel , L. Shaltiel , S. Levenberg
{"title":"ExoPTEN: Allogeneic Exosome Therapy for Spinal Cord Injury with Strong Therapeutic Potential and Clinical Promise","authors":"N. Avni , Y. Geffen , K. Taler , N. Drori Carmi , I. Paz , S. Rubin , N. Kutner , S. Baumgarten-Soueid , I. Solomon , I. Sarel , L. Shaltiel , S. Levenberg","doi":"10.1016/j.jcyt.2025.03.046","DOIUrl":"10.1016/j.jcyt.2025.03.046","url":null,"abstract":"<div><h3>Background & Aim</h3><div>NurExone's ExoPTEN, an exosome-based therapy, represents a novel approach for treating spinal cord injury (SCI). This allogeneic, off-the-shelf therapy utilizes exosomes derived from bone marrow mesenchymal stem cells (BM-MSCs) to deliver a proprietary siRNA targeting PTEN, a key inhibitor in the mTOR pathway. With a scalable and robust manufacturing process, ExoPTEN demonstrates significant therapeutic potential, backed by comprehensive preclinical data and strong regulatory achievements, including orphan drug designation (ODD) and supportive pre-IND feed back.</div></div><div><h3>Methodology</h3><div>Preclinical studies were conducted using two rat models of SCI: complete spinal cord transection and spinal cord compression. ExoPTEN, loaded with siRNA-PTEN, was administered via intranasal and intrathecal routes. Efficacy was evaluated through MRI, BBB motor scoring, von Frey sensory tests, and immunohistochemistry to assess motor, sensory, and structural recovery. The homing capacity of ExoPTEN was evaluated by tracking fluorescently labeled exosomes in rats. Scalability was evaluated by transitioning production from 2D culture systems to 3D bioreactors.</div></div><div><h3>Results</h3><div>ExoPTEN treatment led to significant improvement in motor and sensory functions in SCI rats, as well as structural recovery. Fluorescent labeling confirmed that ExoPTEN successfully homed to the injury site and remained there up to seven days post-injury, establishing a therapeutic window. Process robustness was maintained throughout the scalability from 2D to 3D production, with consistent exosome yields and functionality across different BM donors and siRNA sources. The positive preclinical data, along with orphan drug designation and supportive pre-IND feedback from the FDA, confirm ExoPTEN's potential for clinical translation.</div></div><div><h3>Conclusion</h3><div>ExoPTEN offers a groundbreaking therapeutic option for SCI, backed by robust preclinical data, a demonstrated seven-day therapeutic window, and regulatory support. This scalable, and versatile therapy is designed for clinical use, with the ability to load various therapeutic payloads and provide long-term treatment through repeated administration. These encouraging results demonstrate ExoPTEN's clinical potential and its applicability in additional nerve regeneration conditions.</div></div>","PeriodicalId":50597,"journal":{"name":"Cytotherapy","volume":"27 5","pages":"Pages S30-S31"},"PeriodicalIF":3.7,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143888159","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CytotherapyPub Date : 2025-05-01DOI: 10.1016/j.jcyt.2025.03.006
T. Bronshtein , A. Novak , Y. Shandalov-Levi , L. Kaneti , H. Luxenburg , V. Kivity , D. Ben David , S. Meretzki
{"title":"Standardizing MSC Therapies: A Framework for Addressing Donor Variability and Benchmarking Functional Heterogeneity","authors":"T. Bronshtein , A. Novak , Y. Shandalov-Levi , L. Kaneti , H. Luxenburg , V. Kivity , D. Ben David , S. Meretzki","doi":"10.1016/j.jcyt.2025.03.006","DOIUrl":"10.1016/j.jcyt.2025.03.006","url":null,"abstract":"<div><h3>Background & Aim</h3><div>Inter- and intra-donor variability poses a significant challenge to ensuring consistent quality and performance in cell therapies. This challenge is emphasized in MSC therapies, which rely on the cells' inherent, multifaceted therapeutic capacities. Compared to cells with defined therapeutic targets, MSC efficacy is more sensitive to donor and batch variations, complicating standardization. MSC priming has emerged as a strategy to enhance therapeutic potency and consistency. Previously, we introduced a primed MSC therapy (MesenCure™) that reduced mortality by 68% and the need for invasive ventilation by 57% in respiratory distress patients. Here, we characterize the distinct phenotype of primed MSCs and introduce a framework for assessing functional heterogeneity, enabling the evaluation of cell bank performance against established benchmarks.</div></div><div><h3>Results & Conclusion</h3><div><strong>Results:</strong> RNAseq analysis of primed MSCs from 3 donors revealed 6,909 differentially expressed genes (Fig. 1A, q<0.01) compared to unprimed MSCs. PCA of genes accounting for 99.9% of variability demonstrated distinct clustering of donors by priming status (Fig. 1B). Similar patterns were observed in PCA of cellular and secretome proteomic data from 3 batches derived from a single donor (Figs. 1C, 1D), reaffirming the unique phenotype of primed MSCs.</div><div>Subsequently, donor variability was assessed in 15 donor-derived primed cell batches, examining parameters related to immunomodulatory capacity (IC50 of CD4+ T-cell activation), regenerative potential (FGF7 secretion), and cellular durability (autophagy levels) (Fig. 2A). PCA of these parameters identified a principal component (PC1) linked to enhanced performance, clustering batches into low-, medium-, and high-performing groups (Fig. 2B).</div><div>To evaluate our clinical cell bank performance, we simulated 1,000 normally distributed samples (Fig. 2C) based on observed averages for IC50 (N = 67), FGF7 (N = 16), and autophagy (N = 4), overlaid in Fig. 2A. PC1 values for the simulated samples, calculated using the same transformation that identified the three clusters, were overlaid onto these clusters in Fig. 2B. The distribution of simulated PC1 values demonstrated alignment of clinical cell bank batches with the high-performing cluster (Figs. 2B, 2D).</div></div><div><h3>Conclusion</h3><div>In conclusion, these findings present a robust blueprint for assessing functional heterogeneity among MSC batches, offering a practical framework to benchmark cell bank performance and advance the standardization of MSC-based therapies</div></div>","PeriodicalId":50597,"journal":{"name":"Cytotherapy","volume":"27 5","pages":"Page S9"},"PeriodicalIF":3.7,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143888160","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CytotherapyPub Date : 2025-05-01DOI: 10.1016/j.jcyt.2025.03.037
I. Garitaonandia , C. Vignon , A. Goubaud , A. Cormier , G. Trebuchet , M. de Kalbermatten , P. Henon
{"title":"Results of the Phase I/IIb EXCELLENT trial evaluating the safety and efficacy of transendocardial injection of expanded autologous CD34+ cells in patients with recent myocardial infarction","authors":"I. Garitaonandia , C. Vignon , A. Goubaud , A. Cormier , G. Trebuchet , M. de Kalbermatten , P. Henon","doi":"10.1016/j.jcyt.2025.03.037","DOIUrl":"10.1016/j.jcyt.2025.03.037","url":null,"abstract":"<div><h3>Background & Aim</h3><div>Acute myocardial infarction is followed by irreversible cardiomyocyte necrosis that leads to left ventricular (LV) disfunction and detrimental clinical outcomes. CD34+ cell-based therapy has emerged as an option to repair the myocardium and improve outcomes. After AMI, CD34+ cells are mobilized from the bone marrow to the peripheral blood and the extent of this mobilization is significantly correlated with heart regeneration and functional improvement. In a pilot study, we showed that intramyocardial administration of autologous CD34+ cells leads to long term improvement with average survival of 17 years, compared to the 5-year survival generally observed in 50% of AMI patients. CD34+ cells promote revascularization and cardiomyocyte regeneration through the secretion of paracrine factors. Here, we describe the results of the EXCELLENT study, an international multicenter randomized open label Phase 1/2b clinical trial to evaluate the safety and efficacy of transendocardial injections of autologous expanded CD34+ cells in post-AMI patients (NCT02669810).</div></div><div><h3>Methodology</h3><div>Thirty-three participants with a large AMI and LV ejection fraction <50% were randomized to the treatment plus standard of care arm and 16 participants to the standard of care arm. Participants randomized to intervention were treated with lenograstim for 5 days before blood cell harvest from which autologous CD34+ cells were purified and expanded using an automated GMP compliant platform. The primary endpoint was the incidence of major adverse cardiac events over 6 months. The main secondary endpoints included the improvement of LV end systolic volume index and the viability of the infarcted segments over 6 months.</div></div><div><h3>Results</h3><div>There were no significant differences in baseline values between the patients in the two treatment arms and more than 90% of patients presented with microvascular obstructions. The transendocardial injection procedure was considered feasible and safe and there were no serious adverse events related to the product. There were favorable changes in blinded central reading secondary endpoints. Autologous expanded CD34+ cells improved remodeling as suggested by congruent favorable changes in viability (infarcted zone), left ventricular dimensions, NT-proBNP, and quality of life.</div></div><div><h3>Conclusion</h3><div>The long-term effects on these patients are being evaluated in a 10-year follow-up study. As observed in the pilot study, we believe that this therapy has the potential to improve the long-term clinical outcome of these patients.</div></div>","PeriodicalId":50597,"journal":{"name":"Cytotherapy","volume":"27 5","pages":"Page S26"},"PeriodicalIF":3.7,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143887587","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CytotherapyPub Date : 2025-05-01DOI: 10.1016/j.jcyt.2025.03.067
W. Luo , Y. Soignier , H. Zhu , S. Eguchi , J. Ayello , M. Felices , D.A. Lee , J.S. Miller , M.S. Cairo
{"title":"Circumventing Pediatric Solid Tumor Microenvironment Resistance by Combinatorial CAR NK and Immunomodulating Therapy","authors":"W. Luo , Y. Soignier , H. Zhu , S. Eguchi , J. Ayello , M. Felices , D.A. Lee , J.S. Miller , M.S. Cairo","doi":"10.1016/j.jcyt.2025.03.067","DOIUrl":"10.1016/j.jcyt.2025.03.067","url":null,"abstract":"<div><h3>Background & Aim</h3><div>Patients diagnosed with relapsed/refractory (R/R) Neuroblastoma (NB) and Sarcoma (SARC) have a dismal outcome. Lines of evidence have shown the importance of NK cell immunity against NB and SARC. However, NK cell number and function are low in NB and SARC patients, in large part due to the immunosuppressive tumor microenvironment (TME). Our aim is to improve NK therapy against R/R NB and SARC by strategic combinations that simultaneously enhance tumor specific targeting of NK cells via chimeric antigen receptor (CAR), increase NK migration and tumor infiltration by engineering CAR NK cells to express tumor attracting chemokine receptor CXCR2, improve NK cell ADCC by a GD2 antibody dinutuximab, enhance NK cell persistence and the immune synapse between cancer and NK cells via a tri-specific killer engager Cam1615B7H3, and increase expression of NK activating receptor ligands (MICA/B) on tumor cells by a HDAC inhibitor romidepsin.</div></div><div><h3>Methodology</h3><div>NK cells were expanded using K562-mbIL-21-41BBL feeder cells and IL-2. Expanded NK cells were engineered to co-express ROR1 CAR and CXCR2 using mRNA electroporation. NK cell cytotoxicity was evaluated by luciferase-based cytotoxicity assay. In vitro transwell assays were performed to determine the migratory ability of CAR NK and CAR CXCR2 NK cells. Flow cytometry was utilized to evaluate MICA/B expression on tumor cells treated with or without romidepsin.</div></div><div><h3>Results</h3><div>We found that compared to mock NK cells, ROR1 CAR NK cells had significantly enhanced cytotoxicity against NB (SKNFI) and SARC (A673 and U2OS) cells at various T:E ratios (p<0.05). While the in vitro cytotoxicity of CAR NK and CAR CXCR2 NK cells was very similar (Fig.1A), co-expression of CXCR2 on CAR NK cells significantly enhanced their migration towards the conditioned media of tumor cells (p<0.05 and p<0.01) (Fig.1B). Both dinutuximab and cam1615B7H3 significantly enhanced in vitro cytotoxicity of CAR CXCR2 NK cells against tumor cells (p<0.05 and p<0.01) (Fig.1C and 1D). MICA/B expression on U2OS but not A673 or SKNFI cells was markedly increased (33% vs 72%) after romidepsin treatment (Fig.1E). We observed significantly enhanced cytotoxicity of CAR NK cells when combined with romidepsin compared to CAR NK cells alone against U2OS cells (p<0.01) (Fig.1F).</div></div><div><h3>Conclusion</h3><div>Our results demonstrated the in vitro anti-tumor efficacy of ROR1 CAR CXCR2 NK cells in combination with dinutuximab, cam1615B7H3 and/or romidepsin against malignant NB and SARC. Supported by U54 CA232561 and ALSF reach grant.</div></div>","PeriodicalId":50597,"journal":{"name":"Cytotherapy","volume":"27 5","pages":"Page S41"},"PeriodicalIF":3.7,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143887600","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CytotherapyPub Date : 2025-05-01DOI: 10.1016/j.jcyt.2025.03.061
A. Cantilena , B. Yates , P. Jin , N. Shah , D. Stroncek , A. Dreyzin
{"title":"The Impact of Prior Immunotherapy on Composition of Apheresis Material for CAR T-cell Therapy in Patients with B-Cell Acute Lymphocytic Leukemia (B-ALL)","authors":"A. Cantilena , B. Yates , P. Jin , N. Shah , D. Stroncek , A. Dreyzin","doi":"10.1016/j.jcyt.2025.03.061","DOIUrl":"10.1016/j.jcyt.2025.03.061","url":null,"abstract":"<div><h3>Background & Aim</h3><div>CAR T-cell functionality relies on the composition of the apheresis product used for manufacturing, with higher total nucleated cell (TNC) yield and balanced CD4/CD8 ratios associated with improved disease response. The impact of prior immunotherapy on apheresis composition has not been established. As targeted agents like blinatumomab and inotuzumab are increasingly used for leukemia therapy, an understanding of how these agents affect apheresis composition will be important for treatment planning.</div><div>Our aim was to describe the relationship between blinatumomab and inotuzumab exposure and apheresis composition in patients with relapsed/refractory B-ALL.</div></div><div><h3>Methodology</h3><div>Data from children and young adults with relapsed or refractory B-ALL who underwent apheresis collection for CAR T-cell therapy trials (NCT05442515, NCT03448393, NCT02315612) were analyzed retrospectively. Independent variables included exposure to blinatumomab and/or inotuzumab therapies and timing between immunotherapy and apheresis. Patients who had hematopoietic stem cell transplant following immunotherapy were excluded. Patients who received both blinatumomab and inotuzumab were grouped based on the most proximal therapy (Fig 1). Primary outcomes were TNC yield per patient weight in kilograms, CD3%, and CD4/CD8 ratios as measured by flow cytometry. Outcomes were compared using Mann Whitney U tests.</div></div><div><h3>Results</h3><div>A total of 114 patients with a median age of 18 years (range 4-38), 5 prior lines of therapy (range 2-13), and baseline disease burden of 1% blasts (range 0 to 97%) in bone marrow were evaluated. 29 (25%) patients had blinatumomab exposure at a median of 5.1 months (range 1-58). 22 (19%) had prior inotuzumab at a median of 3.4 months (range 1-30 mo) prior to apheresis.</div><div>Exposure to and timing of blinatumomab were not associated with differences in apheresis composition. In contrast, inotuzumab exposure at any time prior to apheresis was associated with a lower CD4/CD8 ratio (0.77 for those exposed vs 1.11 for those not exposed, p = 0.0047) (Fig 2). The timing of inotuzumab did not impact apheresis composition. TNC yield and CD3% did not differ with blinatumomab or inotuzumab exposure.</div></div><div><h3>Conclusion</h3><div>Inotuzumab exposure may alter CD4 and CD8 proportions in apheresis products, leading to decreased CD4/CD8 ratios, while the same effect was not observed with blinatumomab. More detailed characterization of T-cell phenotypes and fitness after immunotherapy will help guide apheresis timing.</div></div>","PeriodicalId":50597,"journal":{"name":"Cytotherapy","volume":"27 5","pages":"Pages S37-S38"},"PeriodicalIF":3.7,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143886706","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CytotherapyPub Date : 2025-05-01DOI: 10.1016/j.jcyt.2025.03.090
A. Gerth , C.H. Opitz , K.J. Lee
{"title":"From Bench to Bedside – ongoing Clinical Development in Organoids and cell Therapy in Europe","authors":"A. Gerth , C.H. Opitz , K.J. Lee","doi":"10.1016/j.jcyt.2025.03.090","DOIUrl":"10.1016/j.jcyt.2025.03.090","url":null,"abstract":"<div><h3>Background & Aim</h3><div>Two case studies are used to demonstrate the path of clinical development and translation into practice on the European market. Two product developments are presented that are characterised by high patient benefit.</div><div>ATORM-C is an organoid-based therapy for the treatment of chronic inflammatory bowel diseases (IBD). ATORM-C is classified in Europe as a Tissue Engineered Product (TEP) under the ATMP regulations. The active ingredient of ATORM-C is autologous intestinal organoids derived from adult stem cells cultured in vitro. The ability of stem cells to grow into the organ or tissue enables ATORM-C to develop its therapeutic efficacy through tissue regeneration. The therapeutic efficacy of ATORM-C has been investigated in small and medium to large animal models. The first patients were treated in South Korea.</div></div><div><h3>Methodology</h3><div>Current treatment strategies for chronic IBD are aimed at limiting the excessive immune response. However, this strategy is not suitable for reversing the cellular changes that underlie the development of the disease.</div><div>It is planned to establish GMP-compliant production of the organoids in Germany. Clinical testing of the product is planned from 2026. The aim is to obtain marketing authorisation for ATORM-C in Europe.</div></div><div><h3>Results</h3><div>The product MesemCart is an ATMP for the treatment of cartilage damage to the knee joint. The active ingredient is mesenchymal stromal cells derived from human umbilical cord tissue (UC-MSC). MesemCart is intended for allogeneic use. The cell suspension is delivered to the surgeon off the shelf in a defined quality together with a collagen matrix. The cell-laden matrix is applied to the cartilage damage and fixed with fibrin glue or sutures intraoperatively during arthroscopy of the knee joint. Treatment with the UC-MSC aims to modulate inflammation, relieve pain and prevent further damage as well as regenerate hyaline cartilage.</div><div>MesemCart is intended for the following indications: symptomatic grade III/IV cartilage defects according to the International Cartilage Repair Society (ICRS), osteochondral defects with a maximum depth of 3 mm, focal cartilage defects with a defect size of 1 to 6 sq cm. The application for a clinical trial in Europe has been submitted. In the Phase I/II clinical trial, safety as the primary endpoint and efficacy as the secondary endpoint will be tested in 55 patients.</div></div><div><h3>Conclusion</h3><div>MesemCart will be the first allogeneic cell therapy based on UC-MSC for the treatment of cartilage damage to the knee joint in Europe.</div></div>","PeriodicalId":50597,"journal":{"name":"Cytotherapy","volume":"27 5","pages":"Page S54"},"PeriodicalIF":3.7,"publicationDate":"2025-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143888121","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}