bims-drudre Biomed News
on Targeted drug delivery and programmed release mechanisms
Issue of 2022‒01‒23
fourteen papers selected by
Ceren Kimna
Technical University of Munich


  1. ACS Nano. 2022 Jan 21.
      Biomineralization technology has become a trend for the arrest and prevention of dental caries. In particular, the bioactivity and ability to release large amounts of Ca2+ and PO43- ions make amorphous calcium phosphate (ACP) for hard tissue remineralization are highly desired. However, the instability of ACP limits its clinical application. Under continuous bacterial challenge in the oral cavity, the currently developed ACP-based remineralization system lacks the ability to inhibit bacterial adhesion and biofilm formation. Here, a dual-functional nanocomposite with antibiofilm and remineralization properties was designed by combining zwitterionic poly(carboxybetaine acrylamide) (PCBAA) and ACP. The resulting nanocomposite was stable in solution for at least 3 days without any aggregation. The PCBAA/ACP nanocomposite exerted a significant inhibitory effect on the adhesion and biofilm formation of Streptococcus mutans and exhibited bactericidal activities under acidic conditions resulting from bacteria. Moreover, compared with fluoride, this nanocomposite demonstrated superior effects in promoting the remineralization of demineralized enamel and the occlusion of exposed dentinal tubules in vivo and in vitro. The present work provides a theoretical and experimental basis for the use of the PCBAA/ACP nanocomposite as a potential dual-functional agent for arresting and preventing caries.
    Keywords:  amorphous calcium phosphate; antibiofilm; dentinal tubule occlusion; enamel remineralization; zwitterionic polymer
    DOI:  https://doi.org/10.1021/acsnano.1c10812
  2. Adv Mater. 2022 Jan 19. e2106350
      Chemo-immunotherapy is a combination of "standard-of-care" chemotherapy with immunotherapy and it is considered the most advanced therapeutic modality for various types of cancers. However, many cancer patients still poorly respond to current regimen of chemo-immunotherapy and suggest nanotherapeutics as a boosting agent. Recently, heme oxygenase-1 (HO1) was shown to act as an immunotherapeutic molecule in tumor myeloid cells, in addition to general chemoresistance function in cancer cells suggesting that HO1-targeted therapeutics could become a novel, optimal strategy for boosting chemo-immunotherapy in the clinic. Currently the available HO1-inhibitors demonstrate serious adverse effects in clinical use. Herein, tumor myeloid cell- and cancer cell- dual targeted HO1-inhibiting lipid nanotherapeutic boost (T-iLNTB) is developed using RNAi loaded lipid nanoparticles (LNPs). T-iLNTB-mediated HO1 inhibition sensitizes cancer cells to "standard-of-care" chemotherapeutics by increasing immunogenic cell death, and directly reprograms tumor myeloid cells with distinguished phenotype. Furthermore, tumor myeloid cell reprogramming by T-iLNTB induces CD8+ cytotoxic T cell recruitment, which drives "Cold-to-Hot" transition and correlates with improved responsiveness to immune checkpoint inhibitor in combination therapy. Finally, ex vivo study proves that HO1-inhibition directly affects tumor macrophage differentiation. This study demonstrates the potential of T-iLNTB as a novel therapeutic modality for boosting chemo-immunotherapy. This article is protected by copyright. All rights reserved.
    Keywords:  Cancer-targeted therapy; Chemo-immunotherapy; HO1-targeted nanotherapeutics; Ionizable LNP; Targeted lipid nanoparticle
    DOI:  https://doi.org/10.1002/adma.202106350
  3. Adv Mater. 2022 Jan 19. e2110490
      The limited anticancer drug library and the frequent occurrence of drug resistance have driven monotherapy-based cancer therapy into a difficult situation. Considering the formidable process of new drug discovery, combination therapy using currently available drugs is a potential alternative. Nevertheless, the barrier between in vitro combination screening and precise in vivo delivery remains insurmountable in the current free drug- or nanoparticle-based combination therapy, which substantially hinders the application of combination therapy. Herein, we proposed a novel, precise drug delivery strategy to realize efficient and individualized combination therapy. We engineered nano-medicine using a microfluidics-based mixer by combining rationally designed polymeric prodrugs of three commercial chemotherapeutics and a cascade-responsive block copolymer; the nano-medicine possessed ratiometric drug loading and synchronized drug release. In addition to quantitative drug loading and precisely controlled drug combination, consistent nano-properties of these nanoparticles made their in vivo fate predictable. Consequently, tumor growth and metastasis could be effectively inhibited by precisely prescribed nanoparticles derived from in vitro combination screening. This proof-of-concept study clearly revealed the feasibility of overcoming the current drug library limitations through precise delivery of any predetermined drug combination, facilitating translational research of individualized combination therapy. This article is protected by copyright. All rights reserved.
    Keywords:  cancer chemotherapy; cascade-responsive; multi-drug resistance; ratiometric drug delivery; synchronized drug release
    DOI:  https://doi.org/10.1002/adma.202110490
  4. Adv Mater. 2022 Jan 19. e2108817
      Over-activated T cells and over-produced pro-inflammatory cytokines form a self-amplified signaling loop to continuously exacerbate the dysregulated inflammatory response and propel the progression of autoimmune diseases (AIDs). Herein, immuno-engineered nanodecoys (NDs) based on poly(lactic-co-glycolic acid) nanoparticles (PLGA NPs) coated with PD-L1-expressing macrophage membrane (PRM) were developed to mediate multi-target interruption of the self-promoted inflammatory cascade in AIDs. PRM collected from IFN-γ-treated RAW 264.7 cells possessed elevated surface levels of adhesion molecule receptors and pro-inflammatory cytokine receptors, and thus systemically administered PRM NDs afforded higher accumulation level in the inflamed tissues and stronger scavenging efficiency toward multiple pro-inflammatory cytokines. More importantly, IFN-γ treatment induced remarkable PD-L1 expression on PRM, thereby allowing PRM NDs to bind mPD-1 on CD4+ T cell surfaces or neutralize free sPD-1, which reconstructed the PD-1/PD-L1 inhibitory axis to suppress CD4+ T cell activation and restore immune tolerance. As such, PRM NDs provoked potent and cooperative anti-inflammatory and immune-suppressive efficacies to alleviate autoimmune damages in Zymosan A-induced arthritis mice and dextran sulfate sodium-induced ulcerative colitis mice. This study provides an enlightened example for the immuno-engineering of cell membrane-based NDs, rendering promising implications into the treatment of AIDs via multi-target immune-modulation. This article is protected by copyright. All rights reserved.
    Keywords:  PD-L1/PD-1 inhibitory axis; T cell activation; autoimmune diseases; macrophage membrane-coated nanodecoys (NDs); pro-inflammatory cytokine scavenging
    DOI:  https://doi.org/10.1002/adma.202108817
  5. Mater Horiz. 2022 Jan 17.
      Bacterial infection has become a global concern owing to the significant morbidity and mortality. Although the phagocytosis of bacteria by immune cells acts as the front line to protect human body from invading pathogens, the relatively slow encounter and insufficient capture of bacteria by immune cells often lead to an inefficient clearance of pathogens. Herein, a supramolecular artificial receptor-modified macrophage (SAR-Macrophage) was developed to enhance the recognition and latch of bacteria in the systemic circulation, mediated via strong and multipoint host-guest interactions between the artificial receptors (cucurbit[7]uril) on the macrophage and the guest ligands (adamantane) selectively anchored on Escherichia coli (E. coli). As a result, the SAR-Macrophage could significantly accelerate the recognition of E. coli, catch and internalize more pathogens, which subsequently induced the M1 polarization of macrophages to generate ROS and effectively kill the intracellular bacteria. Therefore, the SAR-Macrophage represents a simple, yet powerful anti-bacterial approach.
    DOI:  https://doi.org/10.1039/d1mh01813b
  6. Sci Adv. 2022 Jan 21. 8(3): eabk2485
      Cas13 nucleases are a class of programmable RNA-targeting CRISPR effector proteins that are capable of silencing target gene expression in mammalian cells. Here, we demonstrate that RfxCas13d, a Cas13 ortholog with favorable characteristics to other family members, can be delivered to the mouse spinal cord and brain to silence neurodegeneration-associated genes. Intrathecally delivering an adeno-associated virus vector encoding an RfxCas13d variant programmed to target superoxide dismutase 1 (SOD1), a protein whose mutation can cause amyotrophic lateral sclerosis, reduced SOD1 mRNA and protein in the spinal cord by >50% and improved outcomes in a mouse model of the disorder. We further show that intrastriatally delivering an RfxCas13d variant programmed to target huntingtin (HTT), a protein whose mutation is causative for Huntington's disease, led to a ~50% reduction in HTT protein in the mouse brain. Our results establish RfxCas13d as a versatile platform for knocking down gene expression in the nervous system.
    DOI:  https://doi.org/10.1126/sciadv.abk2485
  7. Proc Natl Acad Sci U S A. 2022 Jan 25. pii: e2103099119. [Epub ahead of print]119(4):
      The use of biologics in the treatment of numerous diseases has increased steadily over the past decade due to their high specificities, low toxicity, and limited side effects. Despite this success, peptide- and protein-based drugs are limited by short half-lives and immunogenicity. To address these challenges, we use a genomically recoded organism to produce genetically encoded elastin-like polypeptide-protein fusions containing multiple instances of p ara-azidophenylalanine (pAzF). Precise lipidation of these pAzF residues generated a set of sequence-defined synthetic biopolymers with programmable binding affinity to albumin without ablating the activity of model fusion proteins, and with tunable blood serum half-lives spanning 5 to 94% of albumin's half-life in a mouse model. Our findings present a proof of concept for the use of genetically encoded bioorthogonal conjugation sites for multisite lipidation to tune protein stability in mouse serum. This work establishes a programmable approach to extend and tune the half-life of protein or peptide therapeutics and a technical foundation to produce functionalized biopolymers endowed with programmable chemical and biophysical properties with broad applications in medicine, materials science, and biotechnology.
    Keywords:  genome recoding; noncanonical amino acids; protein engineering; serum protein half-life extension; synthetic biology
    DOI:  https://doi.org/10.1073/pnas.2103099119
  8. Mol Cancer. 2022 Jan 15. 21(1): 17
      BACKGROUND: miRNAs are regulatory transcripts established as repressors of mRNA stability and translation that have been functionally implicated in carcinogenesis. miR-10b is one of the key onco-miRs associated with multiple forms of cancer. Malignant gliomas exhibit particularly striking dependence on miR-10b. However, despite the therapeutic potential of miR-10b targeting, this miRNA's poorly investigated and largely unconventional properties hamper the clinical translation.METHODS: We utilized Covalent Ligation of Endogenous Argonaute-bound RNAs and their high-throughput RNA sequencing to identify miR-10b interactome and a combination of biochemical and imaging approaches for target validation. They included Crosslinking and RNA immunoprecipitation with spliceosomal proteins, a combination of miRNA FISH with protein immunofluorescence in glioma cells and patient-derived tumors, native Northern blotting, and the transcriptome-wide analysis of alternative splicing.
    RESULTS: We demonstrate that miR-10b binds to U6 snRNA, a core component of the spliceosomal machinery. We provide evidence of the direct binding between miR-10b and U6, in situ imaging of miR-10b and U6 co-localization in glioma cells and tumors, and biochemical co-isolation of miR-10b with the components of the spliceosome. We further demonstrate that miR-10b modulates U6 N-6-adenosine methylation and pseudouridylation, U6 binding to splicing factors SART3 and PRPF8, and regulates U6 stability, conformation, and levels. These effects on U6 result in global splicing alterations, exemplified by the altered ratio of the isoforms of a small GTPase CDC42, reduced overall CDC42 levels, and downstream CDC42 -mediated effects on cell viability.
    CONCLUSIONS: We identified U6 snRNA, the key RNA component of the spliceosome, as the top miR-10b target in glioblastoma. We, therefore, present an unexpected intersection of the miRNA and splicing machineries and a new nuclear function for a major cancer-associated miRNA.
    Keywords:  CDC42; Glioblastoma; Nucleus; Splicing machinery; U6 snRNA; miR-10b
    DOI:  https://doi.org/10.1186/s12943-022-01494-z
  9. Adv Mater. 2022 Jan 16. e2108818
      Dynamically crosslinked gels are appealing materials for applications that require time-dependent mechanical responses. DNA duplexes are ideal crosslinkers for building such gels because of their excellent sequence addressability and flexible tunability in bond energy. However, the mechanical responses of most DNA gels are complicated and unpredictable despite the high potential of DNA. Here, we demonstrate a DNA gel with a highly homogeneous gel network and well-predictable mechanical behaviors by using a pair of star-polymer-DNA precursors with presimulated DNA sequences showing the two-state transition. The melting curve analysis of the DNA gels reveals the good correspondence between the thermodynamic potentials of the DNA crosslinkers and the presimulated values by DNA calculators. Stress-relaxation tests and dissociation kinetics measurements show that the macroscopic relaxation time of the DNA gels is approximately equal to the lifetime of the DNA crosslinkers over four orders of magnitude from 0.1-2,000 sec. Furthermore, a series of durability tests find the DNA gels are hysteresis-less and self-healable after the applications of repeated temperature and mechanical stimuli. These results demonstrate the great potential of star-polymer-DNA precursors for building gels with predictable and tunable viscoelastic properties, suitable for applications such as stress-response extracellular matrices, injectable solids, and soft robotics. This article is protected by copyright. All rights reserved.
    Keywords:  kinetics; self-assembly; stress relaxation; thermodynamics; viscoelasticity
    DOI:  https://doi.org/10.1002/adma.202108818
  10. J Am Chem Soc. 2022 Jan 21.
      Cellular functions are regulated with high spatial control through the local activation of chemical processes in a complex inhomogeneous matrix. The development of synthetic macroscopic systems with a similar capacity allows fundamental studies aimed at understanding the relationship between local molecular events and the emergence of functional properties at the macroscopic level. Here, we show that a kinetically stable inhomogeneous hydrogel matrix is spontaneously formed upon the local injection of ATP. Locally, ATP templates the self-assembly of amphiphiles into large nanoreactors with a much lower diffusion rate compared to unassembled amphiphiles. The local depletion of unassembled amphiphiles near the injection point installs a concentration gradient along which unassembled amphiphiles diffuse from the surroundings to the center. This allows for a progressive local accumulation of self-assembled nanoreactors in the matrix upon repetitive cycles of ATP injection separated by time intervals during which diffusion of unassembled amphiphiles takes place. Contrary to the homogeneous matrix containing the same components, in the inhomogeneous matrix the local upregulation of a chemical reaction occurs. Depending on the way the same amount of injected ATP is administered to the hydrogel matrix different macroscopic distributions of nanoreactors are obtained, which affect the location in the matrix where the chemical reaction is upregulated.
    DOI:  https://doi.org/10.1021/jacs.1c13504
  11. ACS Appl Mater Interfaces. 2022 Jan 19.
      Although nanotheranostics have displayed striking potential toward precise nanomedicine, their targeting delivery and tumor penetration capacities are still impeded by several biological barriers. Besides, the current antitumor strategies mainly focus on killing tumor cells rather than antiangiogenesis. Enlightened by the fact that the smart transformable self-targeting nanotheranostics can enhance their targeting efficiency, tumor penetration, and cellular uptake, we herein report carrier-free Trojan-horse diameter-reducible metal-organic nanotheranostics by the coordination-driven supramolecular sequential co-assembly of the chemo-drug pemetrexed (PEM), transition-metal ions (FeIII), and antiangiogenesis pseudolaric acid B. Such nanotheranostics with both a high dual-drug payload efficiency and outstanding physiological stability are responsively decomposed into numerous ultra-small-diameter nanotheranostics under stimuli of the moderate acidic tumor microenvironment and then internalized into tumor cells through tumor-receptor-mediated self-targeting, synergistically enhancing tumor penetration and cellular uptake. Besides, such nanotheranostics enable visualization of self-targeting capacity under the macroscopic monitor of computed tomography/magnetic resonance imaging, thereby realizing efficient oncotherapy. Moreover, tumor microvessels are precisely monitored by optical coherence tomography angiography/laser speckle imaging during chemo-antiangiogenic therapy in vivo, visually verifying that such nanotheranostics possess an excellent antiangiogenic effect. Our work will provide a promising strategy for further tumor diagnosis and targeted therapy.
    Keywords:  antiangiogenic therapy; chemotherapy; diameter reduction; macroscopic/microscopic imaging; supramolecular chemistry
    DOI:  https://doi.org/10.1021/acsami.1c22350
  12. ACS Appl Mater Interfaces. 2022 Jan 18.
      Microglia are the major innate immune cells in the brain and are essential for maintaining homeostasis in a neuronal microenvironment. Currently, a genetic tool to modify microglial gene expression in specific brain regions is not available. In this report, we introduce a tailor-designed method that uses lipid and polymer hybridized nanoparticles (LPNPs) for the local delivery of small interfering RNAs (siRNAs), allowing the silencing of specific microglial genes in the hypothalamus. Our physical characterization proved that this LPNP-siRNA was uniform and stable. We demonstrated that, due to their natural phagocytic behavior, microglial cells are the dominant cell type taking up these LPNPs in the hypothalamus of rats. We then tested the silencing efficiency of LPNPs carrying a cluster of differentiation molecule 11b (CD11b) or Toll-like receptor 4 (TLR4) siRNA using different in vivo and in vitro approaches. In cultured microglial cells treated with LPNP-CD11b siRNA or LPNP-TLR4 siRNA, we found a silencing efficiency at protein expression levels of 65 or 77%, respectively. In line with this finding, immunohistochemistry and western blotting results from in vivo experiments showed that LPNP-CD11b siRNA significantly inhibited microglial CD11b protein expression in the hypothalamus. Furthermore, following lipopolysaccharide (LPS) stimulation of cultured microglial cells, gene expression of the TLR4 downstream signaling component myeloid differentiation factor 88 and its associated cytokines was significantly inhibited in LPNP-TLR4 siRNA-treated microglial cells compared with cells treated with LPNP-scrambled siRNA. Finally, after LPNP-TLR4 siRNA injection into the rat hypothalamus, we observed a significant reduction in microglial activation in response to LPS compared with the control rats injected with LPNP-scrambled siRNA. Our results indicate that LPNP-siRNA is a promising tool to manipulate microglial activity locally in the brain and may serve as a prophylactic approach to prevent microglial dysfunction-associated diseases.
    Keywords:  CD11b; TLR4; hypothalamus; microglia; nanoparticles; phagocytosis; siRNA
    DOI:  https://doi.org/10.1021/acsami.1c22434
  13. Trends Cardiovasc Med. 2022 Jan 17. pii: S1050-1738(22)00006-8. [Epub ahead of print]
      Diabetes mellitus is a global public health problem whose cases will continue to rise along with the progressive increase in obesity and the aging of the population. People with diabetes exhibit higher risk of cardiovascular complications, especially myocardial infarction (MI). microRNAs (miRNAs) are evolutionary conserved small non-coding RNAs involved in the regulation of biological processes by interfering in gene expression at the post-transcriptional level. Accumulating studies in the last two decades have uncovered the role of stage-specific miRNAs associated with key pathobiological events observed in the hearts of people with diabetes and MI, including cardiomyocyte death, angiogenesis, inflammatory response, myocardial remodeling, and myocardial lipotoxicity. A better understanding of the importance of these miRNAs and their targets may provide novel opportunities for RNA-based therapeutic interventions to address the increased risk of MI in diabetes.
    Keywords:  diabetes; microRNA; myocardial infarction
    DOI:  https://doi.org/10.1016/j.tcm.2022.01.004
  14. Sci Adv. 2022 Jan 21. 8(3): eabl8096
      Although atherosclerosis preferentially develops at arterial curvatures and bifurcations where disturbed flow (DF) activates endothelium, therapies targeting flow-dependent mechanosensing pathways in the vasculature are unavailable. Here, we provided experimental evidence demonstrating a previously unidentified causal role of DF-induced endothelial TXNDC5 (thioredoxin domain containing 5) in atherosclerosis. TXNDC5 was increased in human and mouse atherosclerotic lesions and induced in endothelium subjected to DF. Endothelium-specific Txndc5 deletion markedly reduced atherosclerosis in ApoE-/- mice. Mechanistically, DF-induced TXNDC5 increases proteasome-mediated degradation of heat shock factor 1, leading to reduced heat shock protein 90 and accelerated eNOS (endothelial nitric oxide synthase) protein degradation. Moreover, nanoparticles formulated to deliver Txndc5-targeting CRISPR-Cas9 plasmids driven by an endothelium-specific promoter (CDH5) significantly increase eNOS protein and reduce atherosclerosis in ApoE-/- mice. These results delineate a new molecular paradigm that DF-induced endothelial TXNDC5 promotes atherosclerosis and establish a proof of concept of targeting endothelial mechanosensitive pathways in vivo against atherosclerosis.
    DOI:  https://doi.org/10.1126/sciadv.abl8096