bims-engexo Biomed News
on Engineered exosomes
Issue of 2024‒08‒18
twelve papers selected by
Ravindran Jaganathan, Universiti Kuala Lumpur



  1. Zhongguo Fei Ai Za Zhi. 2024 Jul 20. 27(7): 535-540
      The best treatment for non-small cell lung cancer is early surgical treatment, but most lung cancer is diagnosed at an advanced stage. The main treatment methods are drug and radiotherapy. However, drug resistance or no signifi cant effect of the above treatment methods is inevitable. Therefore, more methods are urgently needed for the treatment of lung cancer. Studies have confirmed that engineered exosomes have good clinical application potential in cardiovascular diseases, tumors, tissue regeneration and repair. This paper summarizes the application of engineered exosomes in the treatment of lung cancer at home and abroad.
.
    Keywords:  Engineered exosomes; Gene therapy; Lung neoplasms; Targeted delivery; Tumor microenvironment
    DOI:  https://doi.org/10.3779/j.issn.1009-3419.2024.101.17
  2. Immunity. 2024 Aug 13. pii: S1074-7613(24)00358-3. [Epub ahead of print]57(8): 1752-1768
      Extracellular vesicles (EVs), such as ectosomes and exosomes, contain DNA, RNA, proteins and are encased in a phospholipid bilayer. EVs provide intralumenal cargo for delivery into the cytoplasm of recipient cells with an impact on the function of immune cells, in part because their biogenesis can also intersect with antigen processing and presentation. Motile EVs from activated immune cells may increase the frequency of immune synapses on recipient cells in a proximity-independent manner for local and long-distance modulation of systemic immunity in inflammation, autoimmunity, organ fibrosis, cancer, and infections. Natural and engineered EVs exhibit the ability to impact innate and adaptive immunity and are entering clinical trials. EVs are likely a component of an optimally functioning immune system, with the potential to serve as immunotherapeutics. Considering the evolving evidence, it is possible that EVs could be the original primordial organic units that preceded the creation of the first cell.
    Keywords:  exosomes; extracellular vesicles; immune response; immunity
    DOI:  https://doi.org/10.1016/j.immuni.2024.07.009
  3. Drug Dev Res. 2024 Aug;85(5): e22244
      Nanovaccines have been designed to overcome the limitations associated with conventional vaccines. Effective delivery methods such as engineered carriers or smart nanoparticles (NPs) are critical requisites for inducing self-tolerance and optimizing vaccine immunogenicity with minimum side effects. NPs can be used as adjuvants, immunogens, or nanocarriers to develop nanovaccines for efficient antigen delivery. Multiloaded nanovaccines carrying multiple tumor antigens along with immunostimulants can effectively increase immunity against tumor cells. They can be biologically engineered to boost interactions with dendritic cells and to allow a gradual and constant antigen release. Modifying NPs surface properties, using high-density lipoprotein-mimicking nanodiscs, and developing nano-based artificial antigen-presenting cells such as dendritic cell-derived-exosomes are amongst the new developed technologies to enhance antigen-presentation and immune reactions against tumor cells. The present review provides an overview on the different perspectives, improvements, and barriers of successful clinical application of current cancer therapeutic and vaccination options. The immunomodulatory effects of different types of nanovaccines and the nanoparticles incorporated into their structure are described. The advantages of using nanovaccines to prevent and treat common illnesses such as AIDS, malaria, cancer and tuberculosis are discussed. Further, potential paths to develop optimal cancer vaccines are described. Given the immunosuppressive characteristics of both cancer cells and the tumor microenvironment, applying immunomodulators and immune checkpoint inhibitors in combination with other conventional anticancer therapies are necessary to boost the effectiveness of the immune response.
    Keywords:  cancer immunotherapy; immune system; nanovaccines
    DOI:  https://doi.org/10.1002/ddr.22244
  4. ACS Biomater Sci Eng. 2024 Aug 16.
      Osteosarcoma (OS) is a rare malignant tumor that affects soft tissue and has high rates of lung metastasis and mortality. The primary treatments for OS include preoperative chemotherapy, surgical resection of the lesion, and postoperative chemotherapy. However, OS chemotherapy presents critical challenges related to treatment toxicity and multiple drug resistance. To address these challenges, nanotechnology has developed nanosystems that release drugs directly to OS cells, reducing the drug's toxicity. Extracellular vesicles (EVs) are nanosized lipid-bilayer bound vesicles that act as cell-derived vehicles and drug delivery systems for several cancers. This study aims to utilize EVs for OS management by co-delivering Hdac1 siRNA and zoledronic acid (zol). The EVs' surface is modified with folic acid (FA) and their targeting ability is compared to that of native EVs. The results showed that the EVs' targeting ability depends on the parent cell source, and FA conjugation further enhanced it. Furthermore, EVs were used as the carrier for co-loading drug (zol) and small RNA (Hdac-1). This approach of using surface engineered EVs as carriers for cargo loading and delivery can be a promising strategy for osteosarcoma management.
    Keywords:  Active targeting; Cargo delivery; Co-delivery; Extracellular Vesicles (EVs); Folate conjugation; Nanocarriers
    DOI:  https://doi.org/10.1021/acsbiomaterials.4c00952
  5. Methods Mol Biol. 2024 ;2843 195-216
      Engineered outer membrane vesicles (OMVs) derived from Gram-negative bacteria are a promising vaccine technology for developing immunity against diverse pathogens. However, antigen display on OMVs can be challenging to control and highly variable due to bottlenecks in protein expression and localization to the bacterial host cell's outer membrane, especially for bulky and complex antigens. Here, we describe methods related to a universal vaccine technology called AvidVax (avidin-based vaccine antigen crosslinking) for rapid and simplified assembly of antigens on the exterior of OMVs during vaccine development. The AvidVax platform involves remodeling the OMV surface with multiple copies of a synthetic antigen-binding protein (SNAP), which is an engineered fusion protein comprised of an outer membrane scaffold protein linked to a biotin-binding protein. The resulting SNAPs enable efficient decoration of OMVs with a molecularly diverse array of biotinylated subunit antigens, including globular and membrane proteins, glycans and glycoconjugates, haptens, lipids, nucleic acids, and short peptides. We detail the key steps in the AvidVax vaccine production pipeline including preparation and isolation of SNAP-OMVs, biotinylation and enrichment of vaccine antigens, and formulation and characterization of antigen-loaded SNAP-OMVs.
    Keywords:  Autotransporter protein; Bacterial extracellular vesicles (BEVs); Biotinylation; Cell surface display; Enhanced monoavidin (eMA); Lpp-OmpA; Outer membrane proteins (OMPs); Outer membrane vesicles (OMVs); Rhizavidin; Subunit antigens; avidin
    DOI:  https://doi.org/10.1007/978-1-0716-4055-5_13
  6. Methods Mol Biol. 2024 ;2843 177-194
      Outer membrane vesicles (OMVs) are small, spherical, nanoscale proteoliposomes released from Gram-negative bacteria that play an important role in cellular defense, pathogenesis, and signaling, among other functions. The functionality of OMVs can be enhanced by engineering developed for biomedical and biochemical applications. Here, we describe methods for directed packaging of enzymes into bacterial OMVs of E. coli using engineered molecular systems, such as localizing proteins to the inner or outer surface of the vesicle. Additionally, we detail some modification strategies for OMVs such as lyophilization and surfactant conjugation that enable the protection of activity of the packaged enzyme when exposed to non-physiological conditions such as elevated temperature, organic solvents, and repeated freeze/thaw that otherwise lead to a substantial loss in the activity of the free enzyme.
    Keywords:  Enzymes; Membrane vesicles (MVs); OMV engineering; Outer membrane vesicles (OMVs); Purification; Synthetic biology
    DOI:  https://doi.org/10.1007/978-1-0716-4055-5_12
  7. Pharmacol Res. 2024 Aug 13. pii: S1043-6618(24)00297-4. [Epub ahead of print] 107352
      A cutting-edge approach in cell-based immunotherapy for combating resistant cancer involves genetically engineered chimeric antigen receptor T (CAR-T) lymphocytes. In recent years, these therapies have demonstrated effectiveness, leading to their commercialization and clinical application against certain types of cancer. However, CAR-T therapy faces limitations, such as the immunosuppressive tumour microenvironment (TME) that can render CAR-T cells ineffective, and the adverse side effects of the therapy, including cytokine release syndrome (CRS). Extracellular vesicles (EVs) are a diverse group of membrane-bound particles released into the extracellular environment by virtually all cell types. They are essential for intercellular communication, transferring cargoes such as proteins, lipids, various types of RNAs, and DNA fragments to target cells, traversing biological barriers both locally and systemically. EVs play roles in numerous physiological processes, with those from both immune and non-immune cells capable of modulating the immune system through activation or suppression. Leveraging this capability of EVs to enhance CAR-T cell therapy could represent a significant advancement in overcoming its current limitations. This review examines the current landscape of CAR-T cell immunotherapy and explores the potential role of EVs in augmenting its therapeutic efficacy.
    Keywords:  (CAR) T-CELL THERAPY; Extracellular vesicles; cancer; immune modulation; tumour microenvironment
    DOI:  https://doi.org/10.1016/j.phrs.2024.107352
  8. Biotechnol Bioeng. 2024 Aug 13.
      Engineered bacteria-based cancer therapy has increasingly been considered to be a promising therapeutic strategy due to the development of synthetic biology. Wherein, engineering bacteria-mediated photodynamic therapy (PDT)-immunotherapy shows greater advantages and potential in treatment efficiency than monotherapy. However, the unsustainable regeneration of photosensitizers (PSs) and weak immune responses limit the therapeutic efficiency. Herein, we developed an engineered bacteria-based delivery system for sequential delivery of PSs and checkpoint inhibitors in cancer PDT-immunotherapy. The biosynthetic pathway of 5-aminolevulinic acid (5-ALA) was introduced into Escherichia coli, yielding a supernatant concentration of 172.19 mg/L after 10 h of growth. And another strain was endowed with the light-controllable releasement of anti-programmed cell death-ligand 1 nanobodies (anti-PD-L1). This system exhibited a collaborative effect, where PDT initiated tumor cell death and the released tumor cell fragments stimulated immunity, followed by the elimination of residual tumor cells. The tumor inhibition rate reached 74.97%, and the portion of activated T cells and inflammatory cytokines were reinforced. The results demonstrated that the engineered bacteria-based collaborative system could sequentially deliver therapeutic substance and checkpoint inhibitors, and achieve good therapeutic therapy. This paper will provide a new perspective for the cancer PDT-immunotherapy.
    Keywords:  checkpoint inhibitor; engineered bacteria; optogenetics; photodynamic‐immunotherapy
    DOI:  https://doi.org/10.1002/bit.28829
  9. J Control Release. 2024 Aug 13. pii: S0168-3659(24)00556-X. [Epub ahead of print]
      mRNA incorporated in lipid nanoparticles (LNPs) became a new class of vaccine modality for induction of immunity against COVID-19 and ushered in a new era in vaccine development. Here, we report a novel, easy-to-execute, and cost effective engineered extracellular vesicles (EVs)-based combined mRNA and protein vaccine platform (EVX-M+P vaccine) and explore its utility in proof-of-concept immunity studies in the settings of cancer and infectious disease. As a first example, we engineered EVs to contain ovalbumin mRNA and protein (EVOvaM+P) to serve as cancer vaccine against ovalbumin-expressing melanoma tumors. EVOvaM+P administration to mice with established melanoma tumors resulted in tumor regression associated with effective humoral and adaptive immune responses. As a second example, we generated engineered EVs, natural nanoparticle carriers shed by all cells, that contain mRNA and protein Spike (S) protein to serve as a combined mRNA and protein vaccine (EVSpikeM+P vaccine) against SARS-CoV-2 infection. EVSpikeM+P vaccine administration in mice and baboons elicited robust production of neutralizing IgG antibodies against RBD (receptor binding domain) of S protein and S protein specific T cell responses. Our proof-of-concept study describes a new platform with an ability for rapid development of combination mRNA and protein vaccines employing EVs for deployment against cancer and other diseases.
    DOI:  https://doi.org/10.1016/j.jconrel.2024.08.017
  10. Hum Gene Ther. 2024 Aug 16.
      The invention of next-generation CRISPR/Cas gene editing tools, like base and prime editing, for correction of gene variants causing disease, has created hope for in vivo use in patients leading to wider clinical translation. To realize this potential, delivery vehicles that can ferry gene editing tool kits safely and effectively into specific cell populations or tissues are in great demand. Here, we describe the development of enveloped retrovirus-derived particles as carriers of 'ready-to-work' ribonucleoprotein complexes consisting of Cas9-derived editor proteins and single guide RNAs. We present arguments for adapting viruses for cell-targeted protein delivery and describe the status after a decade-long development period, which has already shown effective editing in primary cells, including T-cells and hematopoietic stem cells, and in tissues targeted in vivo, including mouse retina, liver, and brain. Emerging evidence has demonstrated that engineered virus-derived nanoparticles can accommodate both base and prime editors, and seem to fertilize a sprouting hope that such particles can be further developed and produced on a large scale for therapeutic applications.
    DOI:  https://doi.org/10.1089/hum.2024.105
  11. Biomater Sci. 2024 Aug 13.
      Nanomedicine has emerged as a promising avenue for advancing cancer treatment, but the challenge of mitigating its in vivo side effects necessitates the development of innovative structures and materials. Recent investigation has unveiled nanogels as particularly compelling candidates, characterized by a porous, three-dimensional network architecture that exhibits exceptional drug loading capacity. Beyond this, nanogels boast a substantial specific surface area and can be tailored with specific chemical functionalities. Consequently, nanogels are frequently engineered as a multi-modal synergistic platform for combating cancer, wherein photothermal therapy stands out due to its capacity to penetrate deep tissues and achieve localized tumor eradication through the application of elevated temperatures. In this review, we delve into the synthesis of diverse varieties of photothermal nanogels capable of controlled drug release triggered by either chemical or physical stimuli. It also summarizes their potential for synergistic integration with photothermal therapy alongside other therapeutic modalities to realize effective tumor ablation. Moreover, we analyze the primary mechanisms underlying the contribution of photothermal nanogels to cancer treatment while underscoring their adeptness in regulating therapeutic temperatures for repairing bone defects resulting from tumor-associated trauma. Envisioned as an auspicious strategy in the realm of cancer therapy, photothermal nanogels hold promise for furnishing controlled drug delivery and precise thermal ablation capabilities.
    DOI:  https://doi.org/10.1039/d4bm00662c
  12. ACS Synth Biol. 2024 Aug 16.
      Through the use of CRISPR-assisted transposition, we have engineered a safe Escherichia coli chassis that integrates an orthogonal translation system (OTS) directly into the chromosome. This approach circumvents the limitations and genetic instability associated with conventional plasmid vectors. Precision in genome modification is crucial for the top-down creation of synthetic cells, especially in the orthogonalization of vital cellular processes, such as metabolism and protein translation. Here, we targeted multiple loci in the E. coli chromosome to integrate the OTS simultaneously, creating a synthetic auxotrophic chassis with an altered genetic code to establish a reliable, robust, and safe synthetic protein producer. Our OTS-integrated chassis enabled the site-specific incorporation of m-oNB-Dopa through in-frame amber stop codon readthrough. This allowed for the expression of advanced underwater bioglues containing Dopa-Lysine motifs, which are crucial for wound healing and tissue regeneration. Additionally, we have enhanced the expression process by incorporating scaffold-stabilizing fluoroprolines into bioglues, utilizing our chassis, which has been modified through metabolic engineering (i.e., by introducing proline auxotrophy). We also engineered a synthetic auxotroph reliant on caged Dopa, creating a genetic barrier (genetic firewall) between the synthetic cells and their surroundings, thereby boosting their stability and safety.
    Keywords:  CRISPR-associated transposases (CASTs); genetic code expansion; genetic firewall and synthetic auxotrophy; genomic integration; orthogonal translation system (OTS)
    DOI:  https://doi.org/10.1021/acssynbio.4c00437