Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.365
Filtrar
1.
Sci Rep ; 14(1): 14141, 2024 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-38898154

RESUMO

Secretion levels required of industrial Chinese hamster ovary (CHO) cell lines can challenge endoplasmic reticulum (ER) homeostasis, and ER stress caused by accumulation of misfolded proteins can be a bottleneck in biomanufacturing. The unfolded protein response (UPR) is initiated to restore homeostasis in response to ER stress, and optimization of the UPR can improve CHO cell production of therapeutic proteins. We compared the fed-batch growth, production characteristics, and transcriptomic response of an immunoglobulin G1 (IgG1) producer to its parental, non-producing host cell line. We conducted differential gene expression analysis using high throughput RNA sequencing (RNASeq) and quantitative polymerase chain reaction (qPCR) to study the ER stress response of each cell line during fed-batch culture. The UPR was activated in the IgG1 producer compared to the host cell line and our analysis of differential expression profiles indicated transient upregulation of ATF6α target mRNAs in the IgG1 producer, suggesting two upstream regulators of the ATF6 arm of the UPR, ATF6ß and WFS1, are rational engineering targets. Although both ATF6ß and WFS1 have been reported to negatively regulate ATF6α, this study shows knockdown of either target elicits different effects in an IgG1-producing CHO cell line. Stable knockdown of ATF6ß decreased cell growth without decreasing titer; however, knockdown of WFS1 decreased titer without affecting growth. Relative expression measured by qPCR indicated no direct relationship between ATF6ß and WFS1 expression, but upregulation of WFS1 in one pool was correlated with decreased growth and upregulation of ER chaperone mRNAs. While knockdown of WFS1 had negative impacts on UPR activation and product mRNA expression, knockdown of ATF6ß improved the UPR specifically later in fed-batch leading to increased overall productivity.


Assuntos
Fator 6 Ativador da Transcrição , Cricetulus , Imunoglobulina G , Resposta a Proteínas não Dobradas , Animais , Células CHO , Fator 6 Ativador da Transcrição/metabolismo , Fator 6 Ativador da Transcrição/genética , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Resposta a Proteínas não Dobradas/genética , Estresse do Retículo Endoplasmático/genética , Técnicas de Silenciamento de Genes , Engenharia Celular/métodos , Técnicas de Cultura Celular por Lotes/métodos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética
2.
Acta Biochim Pol ; 71: 12185, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38721308

RESUMO

Human chemokine receptor 8 (CCR8) is a promising drug target for immunotherapy of cancer and autoimmune diseases. Monoclonal antibody-based CCR8 targeted treatment shows significant inhibition in tumor growth. The inhibition of CCR8 results in the improvement of antitumor immunity and patient survival rates by regulating tumor-resident regulatory T cells. Recently monoclonal antibody drug development targeting CCR8 has become a research hotspot, which also promotes the advancement of antibody evaluation methods. Therefore, we constructed a novel engineered customized cell line HEK293-cAMP-biosensor-CCR8 combined with CCR8 and a cAMP-biosensor reporter. It can be used for the detection of anti-CCR8 antibody functions like specificity and biological activity, in addition to the detection of antibody-dependent cell-mediated cytotoxicity and antibody-dependent-cellular-phagocytosis. We obtained a new CCR8 mAb 22H9 and successfully verified its biological activities with HEK293-cAMP-biosensor-CCR8. Our reporter cell line has high sensitivity and specificity, and also offers a rapid kinetic detection platform for evaluating anti-CCR8 antibody functions.


Assuntos
Anticorpos Monoclonais , Técnicas Biossensoriais , AMP Cíclico , Receptores CCR8 , Humanos , Células HEK293 , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Receptores CCR8/imunologia , Receptores CCR8/metabolismo , AMP Cíclico/metabolismo , Técnicas Biossensoriais/métodos , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Engenharia Celular/métodos
3.
Sci Transl Med ; 16(749): eadg9814, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38809963

RESUMO

T cell-based cancer immunotherapy has typically relied on membrane-bound cytotoxicity enhancers such as chimeric antigen receptors expressed in autologous αß T cells. These approaches are limited by tonic signaling of synthetic constructs and costs associated with manufacturing. γδ T cells are an emerging alternative for cellular therapy, having innate antitumor activity, potent antibody-dependent cellular cytotoxicity, and minimal alloreactivity. We present an immunotherapeutic platform technology built around the innate properties of the Vγ9Vδ2 T cell, harnessing specific characteristics of this cell type and offering an allocompatible cellular therapy that recruits bystander immunity. We engineered γδ T cells to secrete synthetic tumor-targeting opsonins in the form of an scFv-Fc fusion protein and a mitogenic IL-15Rα-IL-15 fusion protein (stIL15). Using GD2 as a model antigen, we show that GD2-specific opsonin-secreting Vγ9Vδ2 T cells (stIL15-OPS-γδ T cells) have enhanced cytotoxicity and promote bystander activity of other lymphoid and myeloid cells. Secretion of stIL-15 abrogated the need for exogenous cytokine supplementation and further mediated activation of bystander natural killer cells. Compared with unmodified γδ T cells, stIL15-OPS-γδ T cells exhibited superior in vivo control of subcutaneous tumors and persistence in the blood. Moreover, stIL15-OPS-γδ T cells were efficacious against patient-derived osteosarcomas in animal models and in vitro, where efficacy could be boosted with the addition of zoledronic acid. Together, the data identify stIL15-OPS-γδ T cells as a candidate allogeneic cell therapy platform combining direct cytolysis with bystander activation to promote tumor control.


Assuntos
Osteossarcoma , Receptores de Antígenos de Linfócitos T gama-delta , Animais , Osteossarcoma/terapia , Osteossarcoma/imunologia , Osteossarcoma/patologia , Humanos , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Camundongos , Linfócitos T/imunologia , Ácido Zoledrônico/farmacologia , Efeito Espectador , Interleucina-15 , Engenharia Celular
4.
Adv Protein Chem Struct Biol ; 140: 91-156, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38762281

RESUMO

This book chapter highlights a comprehensive exploration of the transformative innovations in the field of cancer immunotherapy. CAR (Chimeric Antigen Receptor) T-cell therapy represents a groundbreaking approach to treat cancer by reprogramming a patient immune cells to recognize and destroy cancer cells. This chapter underscores the critical role of synthetic biology in enhancing the safety and effectiveness of CAR T-cell therapies. It begins by emphasizing the growing importance of personalized medicine in cancer treatment, emphasizing the shift from one-size-fits-all approaches to patient-specific solutions. Synthetic biology, a multidisciplinary field, has been instrumental in customizing CAR T-cell therapies, allowing for fine-tuned precision and minimizing unwanted side effects. The chapter highlights recent advances in gene editing, synthetic gene circuits, and molecular engineering, showcasing how these technologies are optimizing CAR T-cell function. In summary, this book chapter sheds light on the remarkable progress made in the development of CAR T-cell therapies using synthetic biology, providing hope for cancer patients and hinting at a future where highly personalized and effective cancer treatments are the norm.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Biologia Sintética , Humanos , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/genética , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/genética , Imunoterapia Adotiva/métodos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Edição de Genes , Engenharia Celular
5.
Acc Chem Res ; 57(12): 1722-1735, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38819691

RESUMO

ConspectusIn human cells, intracellular access and therapeutic cargo transport, including gene-editing tools (e.g., CRISPR-Cas9 and transposons), nucleic acids (e.g., DNA, mRNA, and siRNA), peptides, and proteins (e.g., enzymes and antibodies), are tightly constrained to ensure healthy cell function and behavior. This principle is exemplified in the delivery mechanisms of chimeric antigen receptor (CAR)-T cells for ex-vivo immunotherapy. In particular, the clinical success of CAR-T cells has established a new standard of care by curing previously incurable blood cancers. The approach involves the delivery, typically via the use of electroporation (EP) and lentivirus, of therapeutic CAR genes into a patient's own T cells, which are then engineered to express CARs that target and combat their blood cancer. But the key difficulty lies in genetically manipulating these cells without causing irreversible damage or loss of function─all the while minimizing complexities of manufacturing, safety concerns, and costs, and ensuring the efficacy of the final CAR-T cell product.Nanoinjection─the process of intracellular delivery using nanoneedles (NNs)─is an emerging physical delivery route that efficiently negotiates the plasma membrane of many cell types, including primary human T cells. It occurs with minimal perturbation, invasiveness, and toxicity, with high efficiency and throughput at high spatial and temporal resolutions. Nanoinjection promises greatly improved delivery of a broad range of therapeutic cargos with little or no damage to those cargos. A nanoinjection platform allows these cargos to function in the intracellular space as desired. The adaptability of nanoinjection platforms is now bringing major advantages in immunomodulation, mechanotransduction, sampling of cell states (nanobiopsy), controlled intracellular interrogation, and the primary focus of this account─intracellular delivery and its applications in ex vivo cell engineering.Mechanical nanoinjection typically exerts direct mechanical force on the cell membrane, offering a straightforward route to improve membrane perturbation by the NNs and subsequent transport of genetic cargo into targeted cell type (adherent or suspension cells). By contrast, electroactive nanoinjection is controlled by coupling NNs with an electric field─a new route for activating electroporation (EP) at the nanoscale─allowing a dramatic reduction of the applied voltage to a cell and so minimizing post-EP damage to cells and cargo, and overcoming many of the limitations of conventional bulk EP. Nanoinjection transcends mere technique; it is an approach to cell engineering ex vivo, offering the potential to endow cells with new, powerful features such as generating chimeric antigen receptor (CAR)-T cells for future CAR-T cell technologies.We first discuss the manufacturing of NN devices (Section 2), then delve into nanoinjection-mediated cell engineering (Section 3), nanoinjection mechanisms and interfacing methodologies (Section 4), and emerging applications in using nanoinjection to create functional CAR-T cells (Section 5).


Assuntos
Engenharia Celular , Humanos , Engenharia Celular/métodos , Receptores de Antígenos Quiméricos/metabolismo , Nanotecnologia/métodos , Linfócitos T/citologia , Linfócitos T/metabolismo , Eletroporação/métodos , Injeções
6.
Front Immunol ; 15: 1360237, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38576617

RESUMO

Comprising only 1-10% of the circulating T cell population, γδT cells play a pivotal role in cancer immunotherapy due to their unique amalgamation of innate and adaptive immune features. These cells can secrete cytokines, including interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α), and can directly eliminate tumor cells through mechanisms like Fas/FasL and antibody-dependent cell-mediated cytotoxicity (ADCC). Unlike conventional αßT cells, γδT cells can target a wide variety of cancer cells independently of major histocompatibility complex (MHC) presentation and function as antigen-presenting cells (APCs). Their ability of recognizing antigens in a non-MHC restricted manner makes them an ideal candidate for allogeneic immunotherapy. Additionally, γδT cells exhibit specific tissue tropism, and rapid responsiveness upon reaching cellular targets, indicating a high level of cellular precision and adaptability. Despite these capabilities, the therapeutic potential of γδT cells has been hindered by some limitations, including their restricted abundance, unsatisfactory expansion, limited persistence, and complex biology and plasticity. To address these issues, gene-engineering strategies like the use of chimeric antigen receptor (CAR) T therapy, T cell receptor (TCR) gene transfer, and the combination with γδT cell engagers are being explored. This review will outline the progress in various engineering strategies, discuss their implications and challenges that lie ahead, and the future directions for engineered γδT cells in both monotherapy and combination immunotherapy.


Assuntos
Neoplasias , Receptores de Antígenos de Linfócitos T gama-delta , Receptores de Antígenos de Linfócitos T gama-delta/genética , Linfócitos T , Imunoterapia , Imunoterapia Adotiva , Engenharia Celular , Neoplasias/terapia
7.
Trends Pharmacol Sci ; 45(5): 406-418, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38614815

RESUMO

T cells modified to express intelligently designed chimeric antigen receptors (CARs) are exceptionally powerful therapeutic agents for relapsed and refractory blood cancers and have the potential to revolutionize therapy for many other diseases. To circumvent the complexity and cost associated with broad-scale implementation of ex vivo manufactured adoptive cell therapy products, alternative strategies to generate CAR T cells in vivo by direct infusion of nanoparticle-formulated nucleic acids or engineered viral vectors under development have received a great deal of attention in the past few years. Here, we outline the ex vivo manufacturing process as a motivating framework for direct in vivo strategies and discuss emerging data from preclinical models to highlight the potency of the in vivo approach, the applicability for new disease indications, and the remaining challenges associated with clinical readiness, including delivery specificity, long term efficacy, and safety.


Assuntos
Imunoterapia Adotiva , Receptores de Antígenos Quiméricos , Linfócitos T , Humanos , Animais , Linfócitos T/imunologia , Receptores de Antígenos Quiméricos/imunologia , Imunoterapia Adotiva/métodos , Engenharia Celular/métodos , Receptores de Antígenos de Linfócitos T/imunologia , Neoplasias/terapia , Neoplasias/imunologia
8.
Curr Opin Biotechnol ; 87: 103131, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38599012

RESUMO

Glial cells are important in maintaining homeostasis for neurons in the central nervous system (CNS). During CNS disease or after injury, glia react to altered microenvironments and often acquire altered functions that contribute to disease pathology. A major focus for research is utilizing stem cell (SC)-derived glia as a potential renewable source for cell replacement to restore function, including neuronal support, and as a model for disease states to identify therapeutic targets. In this review, we focus on SC differentiation protocols for deriving three types of glial cells, astrocytes, oligodendrocytes, and microglia. These SC-derived glia can be used to identify critical cues that contribute to CNS disease progression and aid in investigation of therapeutic targets.


Assuntos
Doenças do Sistema Nervoso Central , Neuroglia , Humanos , Neuroglia/metabolismo , Doenças do Sistema Nervoso Central/terapia , Doenças do Sistema Nervoso Central/metabolismo , Animais , Diferenciação Celular , Células-Tronco/citologia , Engenharia Celular/métodos
9.
Nat Biomed Eng ; 8(4): 337-338, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38654129
10.
Methods Mol Biol ; 2774: 31-41, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38441756

RESUMO

Protein interactions play a crucial role in a variety of biological processes. Therefore, regulation of these interactions has received considerable attention in terms of synthetic biology tool development. Of those, a toolbox of small peptides known as coiled coils (CCs) represents a unique effective tool for mediating protein-protein interactions because their binding specificity and affinity can be designed and controlled. CC peptides have been used as a building module for designing synthetic regulatory circuits in mammalian cells, construction of fast response to a signal, amplification of the response, and localization and regulation of function of diverse proteins. In this chapter, we describe a designed set of CCs used for mammalian cell engineering and provide a protocol for the construction of CC-mediated logic circuits in mammalian cells. Ultimately, these tools could be used for diverse biotechnological and therapeutic applications.


Assuntos
Biotecnologia , Engenharia Celular , Animais , Domínios Proteicos , Biologia Sintética , Peptídeos , Mamíferos
11.
ACS Appl Mater Interfaces ; 16(13): 15893-15906, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38512725

RESUMO

Polymer-mediated cell surface engineering can be a powerful tool to modify the cell's biological behavior, but a simple ligation strategy must be identified. This manuscript assessed the use of transglutamination as a versatile and adaptable approach for cell surface engineering in various cellular models relevant to biomedical applications. This enzymatic approach was evaluated for its feasibility and potential for conjugating polymers to diverse cell surfaces and its biological effects. Transglutaminase-mediated ligation was successfully performed at temperatures ranging from 4 to 37 °C in as quickly as 30 min, while maintaining biocompatibility and preserving cell viability. This approach was successfully applied to nine different cell surfaces (including adherent cells and suspension cells) by optimizing the enzyme source (guinea pig liver vs microbial), buffer compositions, and incubation conditions. Finally, polymer-mediated cell surface engineering using transglutaminase exhibited immunocamouflage abilities for endothelial cells, T cells, and red blood cells by preventing the recognition of cell surface proteins by antibodies. Employing transglutaminase in polymer-mediated cell surface engineering is a promising approach to maximize its application in cell therapy and other biomedical applications.


Assuntos
Polímeros , Transglutaminases , Animais , Cobaias , Polímeros/metabolismo , Transglutaminases/metabolismo , Células Endoteliais/metabolismo , Membrana Celular/metabolismo , Engenharia Celular
12.
J Mater Chem B ; 12(14): 3356-3375, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38505950

RESUMO

Clinical advances in genetically modified immune cell therapies, such as chimeric antigen receptor T cell therapies, have raised hope for cancer treatment. The majority of these biotechnologies are based on viral methods for ex vivo genetic modification of the immune cells, while the non-viral methods are still in the developmental phase. Nanocarriers have been emerging as materials of choice for gene delivery to immune cells. This is due to their versatile physicochemical properties such as large surface area and size that can be optimized to overcome several practical barriers to successful gene delivery. The in vivo nanocarrier-based gene delivery can revolutionize cell-based cancer immunotherapies by replacing the current expensive autologous cell manufacturing with an off-the-shelf biomaterial-based platform. The aim of this research is to review current advances and strategies to overcome the challenges in nanoparticle-based gene delivery and their impact on the efficiency, safety, and specificity of the process. The main focus is on polymeric and lipid-based nanocarriers, and their recent preclinical applications for cancer immunotherapy.


Assuntos
Terapia Genética , Imunoterapia Adotiva , Imunoterapia Adotiva/métodos , Técnicas de Transferência de Genes , Imunoterapia/métodos , Engenharia Celular
13.
Biomater Sci ; 12(9): 2244-2258, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38482903

RESUMO

As a vital component of blood, platelets play crucial roles in hemostasis and maintaining vascular integrity, and actively participate in inflammation and immune regulation. The unique biological properties of natural platelets have enabled their utilization as drug delivery vehicles. The advancement and integration of various techniques, including biological, chemical, and physicochemical methods, have enabled the preparation of engineered platelets. Platelets can serve as drug delivery platforms combined with immunotherapy and chemokine therapy to enhance their therapeutic impact. This review focuses on the recent advancements in the application of unactivated platelets for drug delivery. The construction strategies of engineered platelets are comprehensively summarized, encompassing internal loading, surface modification, and genetic engineering techniques. Engineered platelets hold vast potential for treating cardiovascular diseases, cancers, and infectious diseases. Furthermore, the challenges and potential considerations in creating engineered platelets with natural activity are discussed.


Assuntos
Plaquetas , Sistemas de Liberação de Medicamentos , Humanos , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Plaquetas/química , Animais , Engenharia Celular
14.
Metab Eng ; 82: 89-99, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38325641

RESUMO

Precise control over mammalian cell growth dynamics poses a major challenge in biopharmaceutical manufacturing. Here, we present a multi-level cell engineering strategy for the tunable regulation of growth phases in mammalian cells. Initially, we engineered mammalian death phase by employing CRISPR/Cas9 to knockout pro-apoptotic proteins Bax and Bak, resulting in a substantial attenuation of apoptosis by improving cell viability and extending culture lifespan. The second phase introduced a growth acceleration system, akin to a "gas pedal", based on an abscidic acid inducible system regulating cMYC gene expression, enabling rapid cell density increase and cell cycle control. The third phase focused on a stationary phase inducing system, comparable to a "brake pedal". A tetracycline inducible genetic circuit based on BLIMP1 gene led to cell growth cessation and arrested cell cycle upon activation. Finally, we developed a dual controllable system, combining the "gas and brake pedals", enabling for dynamic and precise orchestration of mammalian cell growth dynamics. This work exemplifies the application of synthetic biology tools and combinatorial cell engineering, offering a sophisticated framework for manipulating mammalian cell growth and providing a unique paradigm for reprogramming cell behaviour for enhancing biopharmaceutical manufacturing and further biomedical applications.


Assuntos
Produtos Biológicos , Redes Reguladoras de Genes , Divisão Celular , Sistemas CRISPR-Cas , Engenharia Genética/métodos , Engenharia Celular
16.
J Biosci Bioeng ; 137(4): 245-253, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38336581

RESUMO

In the practical scale of cyanobacterial cultivation, the golden algae Poterioochromonas malhamensis is a well-known predator that causes devastating damage to the culture, referred to as pond crash. The establishment and maintenance of monoculture conditions are ideal for large-scale cultures. However, this is a difficult challenge because microbial contamination is unavoidable in practical-scale culture facilities. In the present study, we unexpectedly observed the pond crash phenomenon during the pilot-scale cultivation of Synechococcus elongatus PCC 7942 using a 100-L photobioreactor. This was due to the contamination with P. malhamensis, which probably originated from residual fouling. Interestingly, we found that S.elongatus PCC 7942 can alter its morphological structure when subjected to continuous grazing pressure from predators, resulting in cells that were more than 100 times longer than those of the wild-type strain. These hyper-elongated S.elongatus PCC 7942 cells had mutations in the genes encoding FtsZ or Ftn2 which are involved in bacterial cell division. Importantly, the elongated phenotype remained stable during cultivation, enabling S.elongatus PCC 7942 to thrive and resist grazing. The cultivation of the elongated S.elongatus PCC 7942 mutant strain in a 100-L pilot-scale photobioreactor under non-sterile conditions resulted in increased cyanobacterial biomass without encountering pond crash. This study demonstrates an efficient strategy for cyanobacterial cell culture in practical-scale bioreactors without the need for extensive decontamination or sterilization of the growth medium and culture facility, which can contribute to economically viable cultivation and bioprocessing of microalgae.


Assuntos
Synechococcus , Synechococcus/genética , Engenharia Celular , Mutação
17.
Lab Chip ; 24(6): 1782-1793, 2024 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-38358122

RESUMO

Non-invasive droplet manipulation with no physical damage to the sample is important for the practical value of manipulation tools in multidisciplinary applications from biochemical analysis and diagnostics to cell engineering. It is a challenge to achieve this for most existing photothermal, electric stimuli, and magnetic field-based technologies. Herein, we present a droplet handling toolbox, the ferrofluid transporter, for non-invasive droplet manipulation in an oil environment. It involves the transport of droplets with high robustness and efficiency owing to low interfacial friction. This capability caters to various scenarios including droplets with varying components and solid cargo. Moreover, we fabricated a droplet array by transporter positioning and achieved droplet gating and sorting for complex manipulation in the droplet array. Benefiting from the ease of scale-up and high biocompatibility, the transporter-based droplet array can serve as a digital microfluidic platform for on-chip droplet-based bioanalysis, cell spheroid culture, and downstream drug screening tests.


Assuntos
Coloides , Microfluídica , Engenharia Celular , Técnicas de Cultura de Células
18.
Adv Drug Deliv Rev ; 208: 115215, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38401848

RESUMO

Adoptive T cell transfer has shown great success in treating blood cancers, resulting in a growing number of FDA-approved therapies using chimeric antigen receptor (CAR)-engineered T cells. However, the effectiveness of this treatment for solid tumors is still not satisfactory, emphasizing the need for improved T cell engineering strategies and combination approaches. Currently, CAR T cells are mainly manufactured using gammaretroviral and lentiviral vectors due to their high transduction efficiency. However, there are concerns about their safety, the high cost of producing them in compliance with current Good Manufacturing Practices (cGMP), regulatory obstacles, and limited cargo capacity, which limit the broader use of engineered T cell therapies. To overcome these limitations, researchers have explored non-viral approaches, such as membrane permeabilization and carrier-mediated methods, as more versatile and sustainable alternatives for next-generation T cell engineering. Non-viral delivery methods can be designed to transport a wide range of molecules, including RNA, which allows for more controlled and safe modulation of T cell phenotype and function. In this review, we provide an overview of non-viral RNA delivery in adoptive T cell therapy. We first define the different types of RNA therapeutics, highlighting recent advancements in manufacturing for their therapeutic use. We then discuss the challenges associated with achieving effective RNA delivery in T cells. Next, we provide an overview of current and emerging technologies for delivering RNA into T cells. Finally, we discuss ongoing preclinical and clinical studies involving RNA-modified T cells.


Assuntos
Neoplasias , Linfócitos T , Humanos , RNA/metabolismo , Imunoterapia Adotiva/métodos , Neoplasias/patologia , Engenharia Celular/métodos
19.
ACS Appl Bio Mater ; 7(3): 1801-1809, 2024 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-38416780

RESUMO

Bacterial nanocellulose (BNC) is an attractive green-synthesized biomaterial for biomedical applications and various other applications. However, effective engineering of BNC production has been limited by our poor knowledge of the related metabolic processes. In contrast to the traditional perception that genome critically determines biosynthesis behaviors, here we discover that the glucose metabolism could also drastically affect the BNC synthesis in Gluconacetobacter hansenii. The transcriptomic profiles of two model BNC-producing strains, G. hansenii ATCC 53582 and ATCC 23769, which have highly similar genomes but drastically different BNC yields, were compared. The results show that their BNC synthesis capacities were highly related to metabolic activities such as ATP synthesis, ion transport protein assembly, and carbohydrate metabolic processes, confirming an important role of metabolism-related transcriptomes in governing the BNC yield. Our findings provide insights into the microbial biosynthesis behaviors from a transcriptome perspective, potentially guiding cellular engineering for biomaterial synthesis.


Assuntos
Perfilação da Expressão Gênica , Transcriptoma , Transcriptoma/genética , Materiais Biocompatíveis , Engenharia Celular , Transporte de Íons
20.
Adv Mater ; 36(16): e2308155, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38295870

RESUMO

Following the success of the dendritic cell (DC) vaccine, the cell-based tumor vaccine shows its promise as a vaccination strategy. Except for DC cells, targeting other immune cells, especially myeloid cells, is expected to address currently unmet clinical needs (e.g., tumor types, safety issues such as cytokine storms, and therapeutic benefits). Here, it is shown that an in situ injected macroporous myeloid cell adoptive scaffold (MAS) not only actively delivers antigens (Ags) that are triggered by scaffold-infiltrating cell surface thiol groups but also releases granulocyte-macrophage colony-stimulating factor and other adjuvant combos. Consequently, this promotes cell differentiation, activation, and migration from the produced monocyte and DC vaccines (MASVax) to stimulate antitumor T-cell immunity. Neoantigen-based MASVax combined with immune checkpoint blockade induces rejection of established tumors and long-term immune protection. The combined depletion of immunosuppressive myeloid cells further enhances the efficacy of MASVax, indicating the potential of myeloid cell-based therapies for immune enhancement and normalization treatment of cancer.


Assuntos
Vacinas Anticâncer , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Vacinação , Engenharia Celular , Células Mieloides , Células Dendríticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...