17 research outputs found

    Manufacturing of patient specific novel T cell therapies using the Cocoon® Platform automated system

    Get PDF
    Engineered T cell therapies, particularly chimeric antigen receptor T cell (CAR-T) immunotherapies, have proved effective against hematologic cancers. However, CAR-T therapies can potentiate immune responses causing cytokine release syndrome (CRS; “cytokine storm”) leading to adverse events in patients. Additionally, CAR-T has shown sporadic success in solid tumor indications. Novel therapies which activate T cells via the native T cell receptors (TCR) have shown greater tumor antigen recognition providing an alternative therapy which may prove effective against solid tumors. Utilizing novel cell immunotherapy modalities is only part of the solution as challenges remain to scale manufacturing to meet commercial demand. Scaling out commercial patient-specific cell therapy manufacturing for large populations using current methods will be expensive (cleanrooms and FTEs) and complex (logistics). Innovative manufacturing solutions will be required to manufacture patient-specific therapies in a robust and cost-effective manner. The Cocoon® Platform is one such innovation, a functionally-closed, automated, scalable cell therapy manufacturing platform. This abstract highlights a therapeutic T cell process translated from an open, manual process to the Cocoon® Platform. During process translation, the functionally-closed Cocoon® Platform was used to automate cell seeding, activation, transduction, feeding, real-time process monitoring, washing, and final product harvest using the single-use Cocoon cassette. During process development and translation, important process parameters were identified, optimized, and programmed enabling multiple process step automation removing the need for manual intervention. For the process, 200 million CD4+ and CD8+ isolated T cells were inoculated with TransActTM activator. The following day, cells were transduced with HER-2 lentivirus vector at various multiplicities of infection (MOI). Cells were expanded with a predefined feeding strategy in media supplemented with IL-2 until final product harvest. Following harvest, cells were assessed for cell yield, viability, transduction efficiency, and VCN. T cell phenotype and functionality was assessed via flow cytometry. The Cocoon manufacturing processes yielded 2.7 x 109 viable cells on average with viability \u3e85%. The Cocoon processes supported both CD4+ and CD8+ T cell expansion with 68% CD4+ T cells and 31% CD8+ T cells on average. The final product exhibited high T cell purity and viability (i.e. \u3e90% abTCR+ and 89% abTCR+, respectively) with transduction efficiencies varied from ~30% to \u3e65% depending on the process MOI. Vector copy number (VCN) was evaluated after each process and found to be ≤5 copies/transduced T cell. In summary, a gene-modified T cell process was successfully translated to the Cocoon and the harvested final products met all pre-defined acceptable criteria. The Cocoon represents a tool for manufacturing cell therapies in a robust manner, while maintaining comparability, and lowering manufacturing costs via increased automation. Ultimately the Cocoon will enable and accelerate development of cell therapies to address solid tumor indications and meet a critical patient need

    An automated and closed system for patient specific CAR-T cell therapies

    Get PDF
    Autologous cell therapies, particularly chimeric antigen receptor T-cell (CAR-T) immunotherapies, are becoming a promising treatment option for difficult diseases. Immunotherapies for blood cancers have dominated the pipeline, while treatments for solid tumors have started to become more successful. However, as the market continues to grow and more clinical trials begin globally, the challenge of manufacturing autologous cell therapies remains significant. A greater number of patients will lead to an increase in cost, labor, and the complexity of logistics for scaling out the commercial production of patient specific therapies. To enable clinical and commercial success, novel manufacturing platforms, such as closed and automated systems, will be required to produce cost effective and robust therapies. This abstract highlights a successful CAR-T process translation from a manual process to an automated patient scale system. To accomplish a CAR-T process translation, we utilized a platform that automates cell seeding, activation, transduction, real time process monitoring, feeding, washing and concentration, and harvesting. In order to mimic a therapeutic CAR-T cell process, manual research scale processes were optimized, scaled up, and then programmed to run automatically without manual intervention. In these processes, 100 million peripheral blood mononuclear cells (PBMC) were first inoculated with CD3/CD28 activation beads. The following day, cells were transduced with HER-2 lentivirus vector. Cells were then expanded with a defined feeding strategy and IL-2 supplements until harvested when target yields were reached. After harvest, cells were analyzed for cell yield, viability, transduction efficiency, and an array of cell phenotype, potency and functionality via FACS and killing assays. Specifically, CAR-T cells were analyzed for the presence of naïve T cells, T stem cell memory, T central memory, T effector memory, and T effector cells. We show here how we optimized, scaled up, and automated manual processes to reach clinical requirements. Automated runs using the above process with cells transduced by HER-2 virus yielded an average of 2 x 109 cells post harvest with a viability \u3e 90%. Automated runs and associated controls were able to support the expansion of both CD4+ and CD8+ T cells with 73% CD4+ T cells and 20% CD8+ T cells. Harvested cells yielded approximately 80% NGFR+ cells with a higher detection of NGFR in the CD4+ fraction than in the CD8+ fraction for all samples. Both CD4+ and CD8+ subsets demonstrated T cell phenotype such as naïve T cells, T stem cell memory, T central memory, T effector memory, and T effector cells. Both subsets also only expressed between 15-20% of immunosuppressive regulatory T cells. Cell health was evaluated by the levels of exhaustion marker, PD-1, which was 19% in CD4+ T cells and \u3c 1% in CD8+ T cells. Furthermore, there was a negligible amount of senescent T cells and anergic cells and \u3c 10% expression of the apoptotic marker, Caspase-3. Subsequently, cells from multiple automated runs showed the specific killing of NGFR+ tumor line were correlated with high levels of effector cytokines: TNF-alpha (~34%) and IFN-gamma (20-25%) as compared to a manual control. In summary, automated CAR-T process in the Cocoon system yields a healthy populations of T cell subsets. This system is a viable solution to translate labor-intensive CAR-T process into a fully automated system, thus allowing scalability, high yield, reduction of manufacturing cost, and better process control to yield high quality CAR-T cells

    Modulation of the Metabiome by Rifaximin in Patients with Cirrhosis and Minimal Hepatic Encephalopathy

    Get PDF
    Hepatic encephalopathy (HE) represents a dysfunctional gut-liver-brain axis in cirrhosis which can negatively impact outcomes. This altered gut-brain relationship has been treated using gut-selective antibiotics such as rifaximin, that improve cognitive function in HE, especially its subclinical form, minimal HE (MHE). However, the precise mechanism of the action of rifaximin in MHE is unclear. We hypothesized that modulation of gut microbiota and their end-products by rifaximin would affect the gut-brain axis and improve cognitive performance in cirrhosis. Aim To perform a systems biology analysis of the microbiome, metabolome and cognitive change after rifaximin in MHE. Methods Twenty cirrhotics with MHE underwent cognitive testing, endotoxin analysis, urine/serum metabolomics (GC and LC-MS) and fecal microbiome assessment (multi-tagged pyrosequencing) at baseline and 8 weeks post-rifaximin 550 mg BID. Changes in cognition, endotoxin, serum/urine metabolites (and microbiome were analyzed using recommended systems biology techniques. Specifically, correlation networks between microbiota and metabolome were analyzed before and after rifaximin. Results There was a significant improvement in cognition(six of seven tests improved,pVeillonellaceaeand increase inEubacteriaceae was observed. Rifaximin resulted in a significant reduction in network connectivity and clustering on the correlation networks. The networks centered onEnterobacteriaceae, Porphyromonadaceae and Bacteroidaceae indicated a shift from pathogenic to beneficial metabolite linkages and better cognition while those centered on autochthonous taxa remained similar. Conclusions Rifaximin is associated with improved cognitive function and endotoxemia in MHE, which is accompanied by alteration of gut bacterial linkages with metabolites without significant change in microbial abundance. Trial Registration ClinicalTrials.gov NCT0106913

    Preserved hemostatic status in patients with non-alcoholic fatty liver disease

    No full text
    Background & Aims: Non-alcoholic fatty liver disease (NAFLD) is associated with an increased risk of thrombosis. However, it remains unclear if hypercoagulability contributes to this risk. We, therefore, determined an in-depth hemostatic profile in a cohort of well-defined patients with NAFLD. Methods: We drew blood samples from 68 patients with biopsy proven NAFLD (simple steatosis n = 24, NASH n = 22, and NASH cirrhosis n = 22), 30 lean controls, 30 overweight controls (body mass index (BMI) >25 kg/m(2)), and 15 patients with alcoholic (ASH) cirrhosis, and performed in-depth hemostatic profiling. Results: Basal and agonist-induced platelet activation, plasma levels of markers of platelet activation, and plasma levels of the platelet adhesion regulators von Willebrand factor and ADAMTSI3 were comparable between patients with non cirrhotic NAFLD and controls. Agonist-induced platelet activation was decreased in patients with cirrhosis. Thrombomodulin-modified thrombin generation was comparable between all patients and controls, although patients with cirrhosis had a reduced anticoagulant response to thrombomodulin. Thromboelastography test results were comparable between controls and non-cirrhotic NAFLD patients, but revealed moderate hypocoagulability in cirrhosis. Plasma fibrinolytic potential was decreased in overweight controls and non-cirrhotic NAFLD, but accelerated fibrinolysis was observed in ASH cirrhosis. Clot permeability was decreased in overweight controls and patients with NAFLD. Conclusions: The overall hemostatic profile is comparable between patients with non-cirrhotic NAFLD and controls. Additionally, pro-thrombotic features (hypofibrinolysis and a pro thrombotic structure of fibrin clot) in patients with NAFLD are likely driven by obesity. Our study suggests a limited role for hyperactive hemostasis in the increased thrombotic risk in NAFLD. Lay summary: The combined results of this study show that the overall hemostatic status is comparable between healthy individuals and patients with a fatty liver disease. (C) 2016 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved
    corecore