以智能工程开拓生物医学应用。

IF 10.7 2区 材料科学 Q1 CHEMISTRY, PHYSICAL
Zhi-bei Qu, Tao Sun, Lu Zhou
{"title":"以智能工程开拓生物医学应用。","authors":"Zhi-bei Qu,&nbsp;Tao Sun,&nbsp;Lu Zhou","doi":"10.1002/smtd.202501013","DOIUrl":null,"url":null,"abstract":"<p>Intelligent engineered biomedicine fundamentally integrates dynamic environmental responsiveness with sophisticated data-driven architectures incorporating biochips, microfluidics, and robotic automation, collectively enabling next-generation personalized therapeutics. Contemporary advancements prioritize adaptive biomaterial systems that continuously detect, process, and react to complex biological signals through integrated technological frameworks spanning artificial intelligence (AI)-optimized chemical synthesis, molecular computation, and precision fluidic control. These interconnected platforms drive end-to-end biopharmaceutical innovation – from smart manufacturing and targeted delivery to closed-loop feedback mechanisms – positioning them as core enablers of precision medicine through enhanced therapeutic accuracy, minimized adverse effects, and patient-specific treatment paradigms. Implementation strategies bifurcate into top-down modifications of conventional biomedical platforms with programmable functionality and bottom-up engineering of intrinsically intelligent biomaterials through molecular-level design principles. To highlight the latest advancements in these fields, we have put together a Special Section titled “Intelligent Biomedical Engineering.” This collection includes 10 research articles, 1 perspective and 13 reviews from prominent scientists with their valuable insights.</p><p>Drug delivery emerges as a transformative cornerstone in intelligent biomedical engineering, where responsive nanomaterials synergize with AI-driven pharmacokinetic modeling and biofeedback-enabled nanodevices to enable dynamic, physiology-responsive therapeutics. These advanced systems utilize stimuli-activated carriers that intelligently modulate drug release in real-time based on biomarker fluctuations, achieving unprecedented spatiotemporal precision through engineered ligand-receptor targeting and biosensor-integrated automatic control. Tao Sun et al. develop a reactive oxygen species (ROS)-responsive heterodimer prodrug (VTO) combining oaldehyde dehydrogenase 2roxylin A (OA) and all-trans retinol (VA) linked via a thioketal bond to synergistically treat liver fibrosis (smtd.202402247). Encapsulated in biomimetic high-density lipoprotein, the co-delivery platform named PL-VTO nanoparticle leveraged natural liver targeting via scarvenger receptor type 1 binding. Upon reaching fibrotic liver, PL-VTO released OA and VA in response to elevated ROS levels, effectively scavenging ROS, deactivating hepatic stellate cells, and reducing collagen deposition. In vitro and in vivo studies demonstrated enhanced ROS clearance, targeted delivery, and fibrosis reversal with minimal toxicity, offering a promising multi-drug nanotherapeutic approach for liver fibrosis treatment. Hendrick W. de Haan and his coworkers investigate comb-shaped polymers for protein molecular sieving coatings (smtd.202500140). Simulations reveal three interaction regimes: no-, weak-, and strong-interactions between polymer chains, governed by steric hindrance, microenvironmental effects, and altered protein dynamics. Systematic parameter analysis identifies strategies to tune coating permeability and size selectivity, providing a universal framework for designing molecular sieving systems with controlled thresholds.</p><p>Hydrogels, as promising drug delivery platforms especially in the field of wound healing, were integrated into intelligent biomedicine by various groups. Yanli Zhao et al. introduce a photodynamic hydrogel activating aldehyde dehydrogenase 2 (ALDH2) to treat diabetic wounds by synergizing immune-vascular repair (smtd.202500391). ALDH2 deficiency in diabetic wounds exacerbates inflammation via neutrophil extracellular traps (NETs) and impairs angiogenesis. The hydrogel integrates ALDA-1 (ALDH2 activator) and chlorin e6, reducing NETs, polarizing macrophages to anti-inflammatory M2, and enhancing vascularization. In vivo tests show accelerated healing through dual modulation of inflammation and angiogenesis, offering a novel therapeutic strategy for chronic diabetic wounds. Chang Du and her coworkers develop an intelligent hydrogel platform (OGM) using gelatin modified with phenylboronic acid, oxidized dextran, and myricetin to accelerate diabetic wound healing (smtd.202401127). The hydrogel exhibits pH/ROS/glucose-responsive release of myricetin, effectively scavenging ROS, alleviating oxidative stress, and promoting angiogenesis via nuclear factor erythroid 2-related factor 2 pathway activation. In vitro and in vivo tests demonstrated enhanced cell proliferation, migration, and vascularization. This triple-responsive platform offers a promising strategy for diabetic wound care. Li Kong and his collaborators present a hydrogel-based immunochemotherapy system (smtd.202401425), to prevent post-surgical recurrence and metastasis in triple-negative breast cancer. The system combines doxorubicin-loaded nanoparticles (NPD) attached to platelets and interleukin-12 (IL-12) within a dopamine-modified alginate hydrogel. Upon activation in the tumor microenvironment, platelets release NPD-loaded microparticles that target residual/metastatic tumor cells, inducing immunogenic cell death. This strategy offers a promising localized therapy to enhance chemotherapeutic efficacy and immune activation post-surgery.</p><p>Exosomes are emerging as promising natural nanocarriers in drug delivery due to their innate biocompatibility, target-homing capabilities, and membrane stability. Current research focuses on engineering exosomal surfaces for enhanced tissue specificity, optimizing cargo loading techniques, and harnessing endogenous trafficking mechanisms. Seungpyo Hong et al. review researches in exosomes as theranostic tools in gastrointestinal cancers, highlighting their dual role as diagnostic biomarkers (miRNAs, lncRNAs, circRNAs, proteins) and engineered therapeutic carriers (smtd.202402058). It discusses advancements in exosome-based strategies for early detection, overcoming drug resistance, metastasis suppression, and immune modulation, addressing clinical challenges and future directions. Jiyong Liu et al. highlighted exosome-based vaccines as promising platforms for infectious diseases and cancer, leveraging their immunomodulatory properties, antigen presentation, and drug delivery capabilities (smtd.202402222). It discusses their biogenesis, components, preparation, and clinical applications, emphasizing personalized vaccine design while addressing challenges in standardization and clinical translation of exosome-based vaccines.</p><p>Advancements in other aspects of drug delivery systems including microneedles and oral drug delivery were highlighted by the contributors. Shutao Guo et al. discuss responsive microneedles for ocular drug delivery, addressing traditional limitations like low bioavailability (smtd.202402048). It categorizes responsive microneedles (pH, ROS, enzyme, glucose-triggered), highlighting their mechanisms and applications in treating diseases like glaucoma, diabetic retinopathy, and infections, emphasizing precision and reduced invasiveness. Wei Tao et al. summarize challenges in oral biologics delivery, including gastric acid, enzymatic degradation, and poor intestinal permeability (smtd.202401355). It highlights strategies like nanoparticle systems, ionic liquids, microneedles, and colon-targeted approaches to enhance bioavailability. Advances in delivery technologies may enable effective oral administration of biologics for chronic diseases.</p><p>Nanomedicine is revolutionizing intelligent biomedical engineering through the development of stimuli-responsive nanoplatforms that synergistically combine molecular targeting, biosensing capabilities, and controlled therapeutic delivery. Current research focuses on engineering smart materials with environmental sensitivity and precision targeting via ligand-receptor modifications, while integrating AI-driven nanoparticle design and adaptive biosensing systems. Notable advances include tumor-specific delivery platforms and blood-brain barrier penetrating systems, though challenges persist in predicting complex in vivo behaviors and establishing scalable manufacturing protocols. Emerging frontiers encompass DNA nanostructures for programmable drug carriers, organ-on-chip validation platforms, and computational pharmacokinetic models that simulate nanoparticle-body interactions. The field continues to evolve toward multifunctional theranostic systems capable of simultaneous diagnosis, real-time monitoring, and adaptive treatment modulation. Jinjin Shi et al. develop neutrophil-targeting biomimetic nanoparticles (MPB NPs) to treat atherosclerosis by inhibiting neutrophil extracellular trap (NET) formation (smtd.202402019). MPB NPs, composed of Prussian blue nanoparticles coated with bacterial membranes, specifically hitchhike on neutrophils via pathogen-associated molecular pattern recognition. By scavenging intracellularROS, MPB NPs suppress NET release induced by inflammatory stimuli. In atherosclerotic mouse models, MPB NPs remarkably reduced plaque area, stabilized plaques, and halted disease progression. This targeted approach demonstrates significant therapeutic potential for atherosclerosis and offers a platform for NET-related inflammatory diseases. Lin Mei et al. report a novel nanoparticle for treating idiopathic pulmonary fibrosis (IPF) by co-loading luteolin (Lut, an anti-fibrotic flavonoid) and hyaluronidase (HAase) into PEG-crosslinked nanoparticles (smtd.202400980). The Lut@HAase demonstrated enhanced lung accumulation via inhalation and deep tissue penetration through HAase-mediated hyaluronic acid degradation. In vitro, Lut@HAase suppressed TGF-β1-induced fibroblast activation (reducing α-SMA/FN expression), inflammatory cytokines (IL-6, IL-1β, TNF-α), and ROS in MRC5 cells. In bleomycin-induced IPF mice, inhaled Lut@HAase improved lung function (reduced resistance, increased compliance), decreased hydroxyproline content, attenuated fibrosis markers, and enhanced survival rates compared to free Lut or HAase, showcasing synergistic anti-fibrotic and anti-inflammatory effects. Si Li et al. develop chiral hydrogel microspheres (HMSs) incorporating L/D-Co₃O₄ nanoparticles to enhance lipase catalytic performance (smtd.202400918). By embedding lipase within L-Co₃O₄-HMSs, catalytic activity significantly increased compared to free lipase and was doubled than D-Co₃O₄-HMSs, attributed to the chiral microenvironment. The L-Co₃O₄-HMSs also demonstrated superior inhibition of 3T3-L1 fibroblast differentiation into adipocytes, highlighting their efficacy in lipid metabolism regulation. This work underscores the role of chirality in optimizing enzyme activity and offers a novel strategy for designing biomimetic catalytic systems with enhanced performance in industrial and biomedical applications. Jiao Yan review explorations in intelligent nanomaterials for osteoarthritis management, addressing its complex pathogenesis and current treatment limitations (smtd.202402263). It highlights nanomaterial innovations enabling targeted drug delivery, anti-inflammatory effects, and cartilage regeneration through engineered properties, emphasizing their clinical potential for personalized and effective osteoarthritis therapies. Hua Kuang et al. provide a review in nanomaterials (metal/metal oxide nanoparticles, liposomes, polymers) as vaccine adjuvants (smtd.202402059), highlighting their mechanisms: depot effect, NLRP3 inflammasome activation, receptor targeting, and cGAS-STING pathway activation. It discusses current clinical applications and challenges in optimizing safety, efficacy, and scalability for future use. Huan Meng et al. write a review discussing how nanomaterials induce pyroptosis, a lytic cell death releasing inflammatory cytokines, for therapeutic applications (smtd.202401290). Nanomaterials’ tunable properties (size, shape, charge) enable precise modulation of pyroptosis via inflammasome activation, enhancing targeted cancer and infection treatments while minimizing toxicity. Strategies include mRNA-based nanomedicine and controlled delivery systems to optimize efficacy. Another review paper in nanomedicine comes from Kui Luo et al. This paper reviews advancements in nanomedicine for enhancing non-small cell lung cancer (NSCLC) immunotherapy (smtd.202401783). It highlights strategies like immune checkpoint inhibitors, antibody-drug conjugates, cell engagers, adoptive cell therapy, and cancer vaccines. Nanomedicine improves drug targeting, bioavailability, and reduces toxicity while overcoming immune resistance. Innovations include nanoparticle-based delivery systems for PD-1/PD-L1 inhibitors, combination therapies, and TME modulation. Clinical trials show promising synergy with existing treatments, offering safer, more precise NSCLC therapies. Future directions focus on multifunctional nanoplatforms to optimize immunotherapy efficacy and patient outcomes. Let's see the last review paper in nanomedicine by Quanyin Hu. It reviews nanomedicine strategies for treating abdominal aortic aneurysms (AAA), addressing challenges like insufficient drug targeting and dynamic aortic environments (smtd.202402268). It highlights the role of inflammation, ECM degradation, and intraluminal thrombus in AAA progression, discussing nanoparticle-based therapies leveraging pathophysiological cues for localized drug delivery and improved clinical outcomes.</p><p>Biosensors are advancing intelligent biomedical engineering through innovations in wearable/implantable devices integrating nanomaterials, AI algorithms, and electro-bio interfaces. Current research emphasizes continuous biomarker monitoring (glucose, cytokines), multi-analyte detection, and on-site therapeutic systems. Nanoscale aptasensors and clustered regularly interspaced short palindromic repeats (CRISPR)-based detectors achieve unprecedented sensitivity. Challenges persist in long-term biocompatibility, signal drift correction, and interference minimization in complex biofluids. Emerging trends include self-powered biosensors using biopotential energy and synthetic biology-engineered living sensors for dynamic pathophysiological response tracking. Fengqin Li et al. report a DNA nanoflower (DNF)-powered CRISPR/Cas12a biosensing platform (DNF-CRISPR) for ultrasensitive protein detection (smtd.202402130). By integrating DNFs for upstream signal amplification and CRISPR/Cas12a for downstream trans-cleavage, the platform achieves a detection limit of sub-mg/L level, a broad dynamic range, and rapid results within 2 h. Validated using neutrophil gelatinase-associated lipocalin (NGAL) in kidney injury patients’ blood and urine, the platform demonstrated high accuracy, outperforming conventional ELISA. This approach enables precise, high-sensitivity protein marker detection, expanding CRISPR-based diagnostics to non-nucleic acid targets in clinical settings. Jiang Li et al. summarize recent advances in DNA-templated protein patterning, leveraging DNA nanotechnology for precise, programmable protein arrangements (smtd.202402249). It discusses covalent (e.g., crosslinkers, click chemistry) and non-covalent (e.g., biotin-streptavidin, aptamers) conjugation strategies. Applications span 1D to 3D nanostructures, dynamic patterns, and biomedicine (biosensing, nanovaccines, targeted therapies). DNA's addressability enables sub-10 nm precision, biocompatibility, and dynamic control. Challenges include conjugation efficiency, stability, and scalability. Future directions focus on integrating dynamic DNA systems for smart diagnostics/therapeutics and expanding functional complexity. Haisheng He et al. report a research work in PN-C18, a near-infrared-II (NIR-II) polarity-sensitive fluorescent probe with aggregation-caused quenching (ACQ) properties, to track in vivo lipolysis of lipid nanocarriers (smtd.202402249). PN-C18 overcomes interference from free probes and mixed micelles, enabling precise monitoring of lipid nanoparticle translocation and degradation. In vitro and in vivo experiments demonstrate its superior correlation with lipolysis progression and deep-tissue imaging capability. This approach advances understanding of lipid-based drug delivery systems’ in vivo fate, offering insights for optimizing oral drug formulations. Chiral biosensing is a research “hot-spot” in intelligent biosensors because chirality provided additional physical dimensions to biological systems. Ji-Young Kim et al. write a perspective highlighting advances in chirality quantification methods for optimizing nanophotonic biosensors (smtd.202500112). Chiral nanomaterials enhance sensitivity for detecting biomolecules via amplified chiroptical signals, but require deeper understanding of structural-optical chirality relationships. The perspective discusses near-field studies to probe chiral nano-bio interfaces and superchiral field effects. Key challenges include standardizing quantification metrics, improving nano-bio interactions, and unifying sensor signal analysis. Future directions focus on precise material design and standardized protocols to advance enantioselective biosensing for disease diagnostics and personalized medicine.</p><p>Finally, we are glad to see achievements in cutting-edge investigations of intelligent biomedicines in combinations of large-scale scientific facilities, smart soft robots, and deep-learning methodologies, largely expanding the scope of this special issue. Ying Zhu and her group review recent advances in synchrotron-based X-ray molecular probes for intelligent biomedicine (smtd.202401890). It highlights advanced imaging techniques and probes like antibody-coupled nanoparticles, genetically encoded tags, and targeted nanomaterials. Applications span cellular imaging, drug analysis, and disease diagnosis/therapy, with future trends focusing on AI-enhanced precision and personalized medicine. Zhengzhi Mu and his interdisciplinary teams gather research work in bioinspired soft robots (BSR) integrating biological motion mechanisms and rigid-flexible coupling systems for biomedical applications (smtd.202402264). Inspired by bending (e.g., Venus flytrap), twisting (e.g., octopus arms), and stretching (e.g., chameleon tongue) organisms, BSR leverage artificial muscle fibers, hydrogels, shape-memory alloys, and soft-rigid composites to achieve multi-degree-of-freedom deformations. Key design strategies mimic biological structures like soft tissues, rigid skeletons, and their synergistic interactions. Applications include in vivo tumor therapy, targeted drug delivery, and in vitro rehabilitative devices (e.g., bionic prostheses). Challenges remain in optimizing motion precision, environmental adaptability, and biocompatibility. Future directions emphasize advanced material integration and biomimetic control systems to enhance clinical translation and functionality in complex biomedical environments. Gang Bao et al. focus on predictions of off-target (OTS) risks of CRISPR/Cas editing (smtd.202500122). Deep learning methods like CRISPR-Net, R-CRISPR, and CRISPR-SGRU show promising OTS prediction capabilities when trained with validated off-target data. In this review, they evaluated six public datasets revealing integrating high-quality experimental OTS data enhances model robustness, particularly with imbalanced datasets. While no universal leader emerged, these models demonstrate improved precision and safety potential for clinical genome editing applications.</p><p>This special section of <i>Small Methods</i> proudly presents global breakthroughs in intelligent biomedicines, dedicated to fostering interdisciplinary collaboration between researchers and practitioners. We extend profound appreciation to all contributors that their research and review work exemplifies frontier innovations in this dynamic field. Gratitude is owed to the <i>Small Methods</i> editorial team, especially Dr. Xi Wen, for supplying us the chance to publish this special section.</p><p>The authors declare no conflict of interest.</p>","PeriodicalId":229,"journal":{"name":"Small Methods","volume":"9 7","pages":""},"PeriodicalIF":10.7000,"publicationDate":"2025-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/smtd.202501013","citationCount":"0","resultStr":"{\"title\":\"Pioneering Biomedical Applications with Intelligent Engineering\",\"authors\":\"Zhi-bei Qu,&nbsp;Tao Sun,&nbsp;Lu Zhou\",\"doi\":\"10.1002/smtd.202501013\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Intelligent engineered biomedicine fundamentally integrates dynamic environmental responsiveness with sophisticated data-driven architectures incorporating biochips, microfluidics, and robotic automation, collectively enabling next-generation personalized therapeutics. Contemporary advancements prioritize adaptive biomaterial systems that continuously detect, process, and react to complex biological signals through integrated technological frameworks spanning artificial intelligence (AI)-optimized chemical synthesis, molecular computation, and precision fluidic control. These interconnected platforms drive end-to-end biopharmaceutical innovation – from smart manufacturing and targeted delivery to closed-loop feedback mechanisms – positioning them as core enablers of precision medicine through enhanced therapeutic accuracy, minimized adverse effects, and patient-specific treatment paradigms. Implementation strategies bifurcate into top-down modifications of conventional biomedical platforms with programmable functionality and bottom-up engineering of intrinsically intelligent biomaterials through molecular-level design principles. To highlight the latest advancements in these fields, we have put together a Special Section titled “Intelligent Biomedical Engineering.” This collection includes 10 research articles, 1 perspective and 13 reviews from prominent scientists with their valuable insights.</p><p>Drug delivery emerges as a transformative cornerstone in intelligent biomedical engineering, where responsive nanomaterials synergize with AI-driven pharmacokinetic modeling and biofeedback-enabled nanodevices to enable dynamic, physiology-responsive therapeutics. These advanced systems utilize stimuli-activated carriers that intelligently modulate drug release in real-time based on biomarker fluctuations, achieving unprecedented spatiotemporal precision through engineered ligand-receptor targeting and biosensor-integrated automatic control. Tao Sun et al. develop a reactive oxygen species (ROS)-responsive heterodimer prodrug (VTO) combining oaldehyde dehydrogenase 2roxylin A (OA) and all-trans retinol (VA) linked via a thioketal bond to synergistically treat liver fibrosis (smtd.202402247). Encapsulated in biomimetic high-density lipoprotein, the co-delivery platform named PL-VTO nanoparticle leveraged natural liver targeting via scarvenger receptor type 1 binding. Upon reaching fibrotic liver, PL-VTO released OA and VA in response to elevated ROS levels, effectively scavenging ROS, deactivating hepatic stellate cells, and reducing collagen deposition. In vitro and in vivo studies demonstrated enhanced ROS clearance, targeted delivery, and fibrosis reversal with minimal toxicity, offering a promising multi-drug nanotherapeutic approach for liver fibrosis treatment. Hendrick W. de Haan and his coworkers investigate comb-shaped polymers for protein molecular sieving coatings (smtd.202500140). Simulations reveal three interaction regimes: no-, weak-, and strong-interactions between polymer chains, governed by steric hindrance, microenvironmental effects, and altered protein dynamics. Systematic parameter analysis identifies strategies to tune coating permeability and size selectivity, providing a universal framework for designing molecular sieving systems with controlled thresholds.</p><p>Hydrogels, as promising drug delivery platforms especially in the field of wound healing, were integrated into intelligent biomedicine by various groups. Yanli Zhao et al. introduce a photodynamic hydrogel activating aldehyde dehydrogenase 2 (ALDH2) to treat diabetic wounds by synergizing immune-vascular repair (smtd.202500391). ALDH2 deficiency in diabetic wounds exacerbates inflammation via neutrophil extracellular traps (NETs) and impairs angiogenesis. The hydrogel integrates ALDA-1 (ALDH2 activator) and chlorin e6, reducing NETs, polarizing macrophages to anti-inflammatory M2, and enhancing vascularization. In vivo tests show accelerated healing through dual modulation of inflammation and angiogenesis, offering a novel therapeutic strategy for chronic diabetic wounds. Chang Du and her coworkers develop an intelligent hydrogel platform (OGM) using gelatin modified with phenylboronic acid, oxidized dextran, and myricetin to accelerate diabetic wound healing (smtd.202401127). The hydrogel exhibits pH/ROS/glucose-responsive release of myricetin, effectively scavenging ROS, alleviating oxidative stress, and promoting angiogenesis via nuclear factor erythroid 2-related factor 2 pathway activation. In vitro and in vivo tests demonstrated enhanced cell proliferation, migration, and vascularization. This triple-responsive platform offers a promising strategy for diabetic wound care. Li Kong and his collaborators present a hydrogel-based immunochemotherapy system (smtd.202401425), to prevent post-surgical recurrence and metastasis in triple-negative breast cancer. The system combines doxorubicin-loaded nanoparticles (NPD) attached to platelets and interleukin-12 (IL-12) within a dopamine-modified alginate hydrogel. Upon activation in the tumor microenvironment, platelets release NPD-loaded microparticles that target residual/metastatic tumor cells, inducing immunogenic cell death. This strategy offers a promising localized therapy to enhance chemotherapeutic efficacy and immune activation post-surgery.</p><p>Exosomes are emerging as promising natural nanocarriers in drug delivery due to their innate biocompatibility, target-homing capabilities, and membrane stability. Current research focuses on engineering exosomal surfaces for enhanced tissue specificity, optimizing cargo loading techniques, and harnessing endogenous trafficking mechanisms. Seungpyo Hong et al. review researches in exosomes as theranostic tools in gastrointestinal cancers, highlighting their dual role as diagnostic biomarkers (miRNAs, lncRNAs, circRNAs, proteins) and engineered therapeutic carriers (smtd.202402058). It discusses advancements in exosome-based strategies for early detection, overcoming drug resistance, metastasis suppression, and immune modulation, addressing clinical challenges and future directions. Jiyong Liu et al. highlighted exosome-based vaccines as promising platforms for infectious diseases and cancer, leveraging their immunomodulatory properties, antigen presentation, and drug delivery capabilities (smtd.202402222). It discusses their biogenesis, components, preparation, and clinical applications, emphasizing personalized vaccine design while addressing challenges in standardization and clinical translation of exosome-based vaccines.</p><p>Advancements in other aspects of drug delivery systems including microneedles and oral drug delivery were highlighted by the contributors. Shutao Guo et al. discuss responsive microneedles for ocular drug delivery, addressing traditional limitations like low bioavailability (smtd.202402048). It categorizes responsive microneedles (pH, ROS, enzyme, glucose-triggered), highlighting their mechanisms and applications in treating diseases like glaucoma, diabetic retinopathy, and infections, emphasizing precision and reduced invasiveness. Wei Tao et al. summarize challenges in oral biologics delivery, including gastric acid, enzymatic degradation, and poor intestinal permeability (smtd.202401355). It highlights strategies like nanoparticle systems, ionic liquids, microneedles, and colon-targeted approaches to enhance bioavailability. Advances in delivery technologies may enable effective oral administration of biologics for chronic diseases.</p><p>Nanomedicine is revolutionizing intelligent biomedical engineering through the development of stimuli-responsive nanoplatforms that synergistically combine molecular targeting, biosensing capabilities, and controlled therapeutic delivery. Current research focuses on engineering smart materials with environmental sensitivity and precision targeting via ligand-receptor modifications, while integrating AI-driven nanoparticle design and adaptive biosensing systems. Notable advances include tumor-specific delivery platforms and blood-brain barrier penetrating systems, though challenges persist in predicting complex in vivo behaviors and establishing scalable manufacturing protocols. Emerging frontiers encompass DNA nanostructures for programmable drug carriers, organ-on-chip validation platforms, and computational pharmacokinetic models that simulate nanoparticle-body interactions. The field continues to evolve toward multifunctional theranostic systems capable of simultaneous diagnosis, real-time monitoring, and adaptive treatment modulation. Jinjin Shi et al. develop neutrophil-targeting biomimetic nanoparticles (MPB NPs) to treat atherosclerosis by inhibiting neutrophil extracellular trap (NET) formation (smtd.202402019). MPB NPs, composed of Prussian blue nanoparticles coated with bacterial membranes, specifically hitchhike on neutrophils via pathogen-associated molecular pattern recognition. By scavenging intracellularROS, MPB NPs suppress NET release induced by inflammatory stimuli. In atherosclerotic mouse models, MPB NPs remarkably reduced plaque area, stabilized plaques, and halted disease progression. This targeted approach demonstrates significant therapeutic potential for atherosclerosis and offers a platform for NET-related inflammatory diseases. Lin Mei et al. report a novel nanoparticle for treating idiopathic pulmonary fibrosis (IPF) by co-loading luteolin (Lut, an anti-fibrotic flavonoid) and hyaluronidase (HAase) into PEG-crosslinked nanoparticles (smtd.202400980). The Lut@HAase demonstrated enhanced lung accumulation via inhalation and deep tissue penetration through HAase-mediated hyaluronic acid degradation. In vitro, Lut@HAase suppressed TGF-β1-induced fibroblast activation (reducing α-SMA/FN expression), inflammatory cytokines (IL-6, IL-1β, TNF-α), and ROS in MRC5 cells. In bleomycin-induced IPF mice, inhaled Lut@HAase improved lung function (reduced resistance, increased compliance), decreased hydroxyproline content, attenuated fibrosis markers, and enhanced survival rates compared to free Lut or HAase, showcasing synergistic anti-fibrotic and anti-inflammatory effects. Si Li et al. develop chiral hydrogel microspheres (HMSs) incorporating L/D-Co₃O₄ nanoparticles to enhance lipase catalytic performance (smtd.202400918). By embedding lipase within L-Co₃O₄-HMSs, catalytic activity significantly increased compared to free lipase and was doubled than D-Co₃O₄-HMSs, attributed to the chiral microenvironment. The L-Co₃O₄-HMSs also demonstrated superior inhibition of 3T3-L1 fibroblast differentiation into adipocytes, highlighting their efficacy in lipid metabolism regulation. This work underscores the role of chirality in optimizing enzyme activity and offers a novel strategy for designing biomimetic catalytic systems with enhanced performance in industrial and biomedical applications. Jiao Yan review explorations in intelligent nanomaterials for osteoarthritis management, addressing its complex pathogenesis and current treatment limitations (smtd.202402263). It highlights nanomaterial innovations enabling targeted drug delivery, anti-inflammatory effects, and cartilage regeneration through engineered properties, emphasizing their clinical potential for personalized and effective osteoarthritis therapies. Hua Kuang et al. provide a review in nanomaterials (metal/metal oxide nanoparticles, liposomes, polymers) as vaccine adjuvants (smtd.202402059), highlighting their mechanisms: depot effect, NLRP3 inflammasome activation, receptor targeting, and cGAS-STING pathway activation. It discusses current clinical applications and challenges in optimizing safety, efficacy, and scalability for future use. Huan Meng et al. write a review discussing how nanomaterials induce pyroptosis, a lytic cell death releasing inflammatory cytokines, for therapeutic applications (smtd.202401290). Nanomaterials’ tunable properties (size, shape, charge) enable precise modulation of pyroptosis via inflammasome activation, enhancing targeted cancer and infection treatments while minimizing toxicity. Strategies include mRNA-based nanomedicine and controlled delivery systems to optimize efficacy. Another review paper in nanomedicine comes from Kui Luo et al. This paper reviews advancements in nanomedicine for enhancing non-small cell lung cancer (NSCLC) immunotherapy (smtd.202401783). It highlights strategies like immune checkpoint inhibitors, antibody-drug conjugates, cell engagers, adoptive cell therapy, and cancer vaccines. Nanomedicine improves drug targeting, bioavailability, and reduces toxicity while overcoming immune resistance. Innovations include nanoparticle-based delivery systems for PD-1/PD-L1 inhibitors, combination therapies, and TME modulation. Clinical trials show promising synergy with existing treatments, offering safer, more precise NSCLC therapies. Future directions focus on multifunctional nanoplatforms to optimize immunotherapy efficacy and patient outcomes. Let's see the last review paper in nanomedicine by Quanyin Hu. It reviews nanomedicine strategies for treating abdominal aortic aneurysms (AAA), addressing challenges like insufficient drug targeting and dynamic aortic environments (smtd.202402268). It highlights the role of inflammation, ECM degradation, and intraluminal thrombus in AAA progression, discussing nanoparticle-based therapies leveraging pathophysiological cues for localized drug delivery and improved clinical outcomes.</p><p>Biosensors are advancing intelligent biomedical engineering through innovations in wearable/implantable devices integrating nanomaterials, AI algorithms, and electro-bio interfaces. Current research emphasizes continuous biomarker monitoring (glucose, cytokines), multi-analyte detection, and on-site therapeutic systems. Nanoscale aptasensors and clustered regularly interspaced short palindromic repeats (CRISPR)-based detectors achieve unprecedented sensitivity. Challenges persist in long-term biocompatibility, signal drift correction, and interference minimization in complex biofluids. Emerging trends include self-powered biosensors using biopotential energy and synthetic biology-engineered living sensors for dynamic pathophysiological response tracking. Fengqin Li et al. report a DNA nanoflower (DNF)-powered CRISPR/Cas12a biosensing platform (DNF-CRISPR) for ultrasensitive protein detection (smtd.202402130). By integrating DNFs for upstream signal amplification and CRISPR/Cas12a for downstream trans-cleavage, the platform achieves a detection limit of sub-mg/L level, a broad dynamic range, and rapid results within 2 h. Validated using neutrophil gelatinase-associated lipocalin (NGAL) in kidney injury patients’ blood and urine, the platform demonstrated high accuracy, outperforming conventional ELISA. This approach enables precise, high-sensitivity protein marker detection, expanding CRISPR-based diagnostics to non-nucleic acid targets in clinical settings. Jiang Li et al. summarize recent advances in DNA-templated protein patterning, leveraging DNA nanotechnology for precise, programmable protein arrangements (smtd.202402249). It discusses covalent (e.g., crosslinkers, click chemistry) and non-covalent (e.g., biotin-streptavidin, aptamers) conjugation strategies. Applications span 1D to 3D nanostructures, dynamic patterns, and biomedicine (biosensing, nanovaccines, targeted therapies). DNA's addressability enables sub-10 nm precision, biocompatibility, and dynamic control. Challenges include conjugation efficiency, stability, and scalability. Future directions focus on integrating dynamic DNA systems for smart diagnostics/therapeutics and expanding functional complexity. Haisheng He et al. report a research work in PN-C18, a near-infrared-II (NIR-II) polarity-sensitive fluorescent probe with aggregation-caused quenching (ACQ) properties, to track in vivo lipolysis of lipid nanocarriers (smtd.202402249). PN-C18 overcomes interference from free probes and mixed micelles, enabling precise monitoring of lipid nanoparticle translocation and degradation. In vitro and in vivo experiments demonstrate its superior correlation with lipolysis progression and deep-tissue imaging capability. This approach advances understanding of lipid-based drug delivery systems’ in vivo fate, offering insights for optimizing oral drug formulations. Chiral biosensing is a research “hot-spot” in intelligent biosensors because chirality provided additional physical dimensions to biological systems. Ji-Young Kim et al. write a perspective highlighting advances in chirality quantification methods for optimizing nanophotonic biosensors (smtd.202500112). Chiral nanomaterials enhance sensitivity for detecting biomolecules via amplified chiroptical signals, but require deeper understanding of structural-optical chirality relationships. The perspective discusses near-field studies to probe chiral nano-bio interfaces and superchiral field effects. Key challenges include standardizing quantification metrics, improving nano-bio interactions, and unifying sensor signal analysis. Future directions focus on precise material design and standardized protocols to advance enantioselective biosensing for disease diagnostics and personalized medicine.</p><p>Finally, we are glad to see achievements in cutting-edge investigations of intelligent biomedicines in combinations of large-scale scientific facilities, smart soft robots, and deep-learning methodologies, largely expanding the scope of this special issue. Ying Zhu and her group review recent advances in synchrotron-based X-ray molecular probes for intelligent biomedicine (smtd.202401890). It highlights advanced imaging techniques and probes like antibody-coupled nanoparticles, genetically encoded tags, and targeted nanomaterials. Applications span cellular imaging, drug analysis, and disease diagnosis/therapy, with future trends focusing on AI-enhanced precision and personalized medicine. Zhengzhi Mu and his interdisciplinary teams gather research work in bioinspired soft robots (BSR) integrating biological motion mechanisms and rigid-flexible coupling systems for biomedical applications (smtd.202402264). Inspired by bending (e.g., Venus flytrap), twisting (e.g., octopus arms), and stretching (e.g., chameleon tongue) organisms, BSR leverage artificial muscle fibers, hydrogels, shape-memory alloys, and soft-rigid composites to achieve multi-degree-of-freedom deformations. Key design strategies mimic biological structures like soft tissues, rigid skeletons, and their synergistic interactions. Applications include in vivo tumor therapy, targeted drug delivery, and in vitro rehabilitative devices (e.g., bionic prostheses). Challenges remain in optimizing motion precision, environmental adaptability, and biocompatibility. Future directions emphasize advanced material integration and biomimetic control systems to enhance clinical translation and functionality in complex biomedical environments. Gang Bao et al. focus on predictions of off-target (OTS) risks of CRISPR/Cas editing (smtd.202500122). Deep learning methods like CRISPR-Net, R-CRISPR, and CRISPR-SGRU show promising OTS prediction capabilities when trained with validated off-target data. In this review, they evaluated six public datasets revealing integrating high-quality experimental OTS data enhances model robustness, particularly with imbalanced datasets. While no universal leader emerged, these models demonstrate improved precision and safety potential for clinical genome editing applications.</p><p>This special section of <i>Small Methods</i> proudly presents global breakthroughs in intelligent biomedicines, dedicated to fostering interdisciplinary collaboration between researchers and practitioners. We extend profound appreciation to all contributors that their research and review work exemplifies frontier innovations in this dynamic field. Gratitude is owed to the <i>Small Methods</i> editorial team, especially Dr. Xi Wen, for supplying us the chance to publish this special section.</p><p>The authors declare no conflict of interest.</p>\",\"PeriodicalId\":229,\"journal\":{\"name\":\"Small Methods\",\"volume\":\"9 7\",\"pages\":\"\"},\"PeriodicalIF\":10.7000,\"publicationDate\":\"2025-06-17\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/smtd.202501013\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Small Methods\",\"FirstCategoryId\":\"88\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/smtd.202501013\",\"RegionNum\":2,\"RegionCategory\":\"材料科学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"CHEMISTRY, PHYSICAL\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Small Methods","FirstCategoryId":"88","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/smtd.202501013","RegionNum":2,"RegionCategory":"材料科学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CHEMISTRY, PHYSICAL","Score":null,"Total":0}
引用次数: 0

摘要

智能工程生物医学从根本上集成了动态环境响应和复杂的数据驱动架构,结合了生物芯片、微流体和机器人自动化,共同实现了下一代个性化治疗。当代的进步优先考虑自适应生物材料系统,通过跨越人工智能(AI)优化的化学合成、分子计算和精密流体控制的集成技术框架,持续检测、处理和反应复杂的生物信号。这些相互关联的平台推动了端到端的生物制药创新——从智能制造和靶向递送到闭环反馈机制——通过提高治疗准确性、最小化不良反应和针对患者的治疗范例,将它们定位为精准医疗的核心推动者。实现策略分为自上而下对具有可编程功能的传统生物医学平台进行修改和自下而上通过分子水平设计原理对本质智能生物材料进行工程设计。为了突出这些领域的最新进展,我们专门制作了一个名为“智能生物医学工程”的专题。本书收录了10篇研究论文,1个观点和13篇评论,这些评论都是来自著名科学家的宝贵见解。药物递送成为智能生物医学工程的变革基石,反应性纳米材料与人工智能驱动的药代动力学建模和生物反馈支持的纳米设备协同作用,实现动态的生理反应治疗。这些先进的系统利用刺激激活的载体,根据生物标志物的波动实时智能调节药物释放,通过工程配体受体靶向和生物传感器集成自动控制,实现前所未有的时空精度。Tao Sun等开发了一种由乙醛脱氢酶2roxylin a (OA)和全反式视黄醇(VA)通过硫酮键连接的活性氧(ROS)响应异二聚体前药(VTO),协同治疗肝纤维化(smtd.202402247)。该共递送平台名为PL-VTO纳米颗粒,包裹在仿生高密度脂蛋白中,通过1型清除剂受体结合利用天然肝脏靶向。PL-VTO到达纤维化肝脏后,释放OA和VA以响应ROS水平升高,有效清除ROS,使肝星状细胞失活,减少胶原沉积。体外和体内研究表明,在毒性最小的情况下,增强ROS清除,靶向递送和纤维化逆转,为肝纤维化治疗提供了一种有前途的多药物纳米治疗方法。Hendrick W. de Haan和他的同事研究梳状聚合物用于蛋白质分子筛选涂层(smtd.202500140)。模拟揭示了三种相互作用机制:由位阻、微环境效应和改变的蛋白质动力学控制的聚合物链之间的无、弱和强相互作用。系统参数分析确定了调整涂层渗透率和尺寸选择性的策略,为设计具有控制阈值的分子筛系统提供了通用框架。水凝胶作为一种非常有前途的药物传递平台,特别是在伤口愈合领域,被各种研究小组整合到智能生物医学中。赵艳丽等人介绍了一种激活醛脱氢酶2 (ALDH2)的光动力水凝胶,通过免疫血管修复协同治疗糖尿病伤口(smtd.202500391)。糖尿病伤口中ALDH2缺乏通过中性粒细胞胞外陷阱(NETs)加剧炎症并损害血管生成。该水凝胶整合了ALDA-1 (ALDH2激活剂)和氯离子e6,减少NETs,使巨噬细胞极化为抗炎M2,并增强血管化。体内试验表明,通过炎症和血管生成的双重调节,加速愈合,为慢性糖尿病伤口提供了一种新的治疗策略。Chang Du和她的同事开发了一种智能水凝胶平台(OGM),该平台使用苯基硼酸、氧化葡聚糖和杨梅素修饰明胶来加速糖尿病伤口愈合(smt .202401127)。水凝胶表现出pH/ROS/葡萄糖反应性释放杨梅素,有效清除ROS,减轻氧化应激,并通过核因子红细胞2相关因子2通路激活促进血管生成。体外和体内试验显示增强细胞增殖、迁移和血管化。这种三重反应平台为糖尿病伤口护理提供了一种有前途的策略。李孔等提出了一种基于水凝胶的免疫化疗系统(smtd.202401425),用于预防三阴性乳腺癌术后复发和转移。 该系统将装载阿霉素的纳米颗粒(NPD)与血小板和白细胞介素-12 (IL-12)结合在多巴胺修饰的海藻酸盐水凝胶中。在肿瘤微环境中激活后,血小板释放装载npd的微粒,靶向残余/转移性肿瘤细胞,诱导免疫原性细胞死亡。这种策略提供了一种有希望的局部治疗,以提高化疗疗效和术后免疫激活。外泌体由于其固有的生物相容性、靶向能力和膜稳定性而成为药物输送中有前途的天然纳米载体。目前的研究主要集中在工程外泌体表面,以增强组织特异性,优化货物装载技术,并利用内源性运输机制。Seungpyo Hong等人回顾了外泌体作为胃肠道癌症治疗工具的研究,强调了它们作为诊断生物标志物(mirna, lncrna, circrna,蛋白质)和工程治疗载体的双重作用(smtd.202402058)。它讨论了基于外泌体的早期检测策略的进展,克服耐药性,转移抑制和免疫调节,解决临床挑战和未来方向。Jiyong Liu等人强调,基于外泌体的疫苗利用其免疫调节特性、抗原呈递和药物递送能力,是治疗传染病和癌症的有希望的平台(smtd.202402222)。它讨论了它们的生物发生、成分、制备和临床应用,强调个性化疫苗设计,同时解决基于外泌体的疫苗的标准化和临床翻译的挑战。与会者强调了包括微针和口服给药在内的其他给药系统方面的进展。郭树涛等人讨论了用于眼部给药的反应性微针,解决了生物利用度低等传统限制(smtd.202402048)。它对反应性微针(pH、ROS、酶、葡萄糖触发)进行了分类,强调了它们在治疗青光眼、糖尿病视网膜病变和感染等疾病中的机制和应用,强调了精确性和降低了侵入性。魏涛等人总结了口服生物制剂递送面临的挑战,包括胃酸、酶降解和肠通透性差(smtd.202401355)。它强调了诸如纳米粒子系统、离子液体、微针和结肠靶向方法等提高生物利用度的策略。给药技术的进步可能使慢性疾病的生物制剂口服给药变得有效。纳米医学通过开发刺激反应纳米平台,将分子靶向、生物传感能力和控制治疗递送协同结合起来,正在彻底改变智能生物医学工程。目前的研究重点是通过配体受体修饰,结合人工智能驱动的纳米颗粒设计和自适应生物传感系统,设计具有环境敏感性和精确靶向性的智能材料。值得注意的进展包括肿瘤特异性递送平台和血脑屏障穿透系统,尽管在预测复杂的体内行为和建立可扩展的制造方案方面仍然存在挑战。新兴领域包括用于可编程药物载体的DNA纳米结构,芯片上的器官验证平台,以及模拟纳米颗粒-体相互作用的计算药代动力学模型。该领域继续向多功能治疗系统发展,能够同时诊断、实时监测和自适应治疗调节。Shi Jinjin等人开发了靶向中性粒细胞的仿生纳米颗粒(MPB NPs),通过抑制中性粒细胞胞外陷阱(NET)的形成来治疗动脉粥样硬化(smtd.202402019)。MPB NPs由包裹有细菌膜的普鲁士蓝纳米颗粒组成,通过病原体相关的分子模式识别特异性地搭便车到中性粒细胞上。通过清除细胞内ros, MPB NPs抑制炎症刺激诱导的NET释放。在动脉粥样硬化小鼠模型中,MPB NPs显著减少斑块面积,稳定斑块,并阻止疾病进展。这种靶向方法显示了动脉粥样硬化的显著治疗潜力,并为net相关炎症性疾病提供了一个平台。林梅等人报道了一种新型纳米颗粒,通过将木犀草素(Lut,一种抗纤维化类黄酮)和透明质酸酶(HAase)共负载到peg交联纳米颗粒中,用于治疗特发性肺纤维化(IPF) (smtd.202400980)。Lut@HAase显示通过吸入和haase介导的透明质酸降解深层组织渗透增强肺部积聚。在体外,Lut@HAase抑制TGF-β1诱导的MRC5细胞成纤维细胞活化(降低α-SMA/FN表达)、炎症因子(IL-6、IL-1β、TNF-α)和ROS。 在博莱霉素诱导的IPF小鼠中,与游离Lut或HAase相比,吸入Lut@HAase可改善肺功能(降低抵抗力,增加依从性),降低羟脯氨酸含量,减轻纤维化标志物,提高生存率,显示协同抗纤维化和抗炎作用。Si Li等人开发了含有L/D-Co₃O₄纳米颗粒的手性水凝胶微球(hms),以提高脂肪酶的催化性能(smtd.202400918)。通过将脂肪酶包埋在L-Co₃O₄- hms中,催化活性比游离脂肪酶明显提高,比D-Co₃O₄- hms的催化活性提高了一倍,这是由于手性微环境的原因。L-Co₃O₄- hms还显示出对3T3-L1成纤维细胞向脂肪细胞分化的良好抑制作用,突出了它们在脂质代谢调节中的功效。这项工作强调了手性在优化酶活性中的作用,并为设计具有增强工业和生物医学应用性能的仿生催化系统提供了一种新的策略。焦岩综述了智能纳米材料在骨关节炎治疗中的研究进展,指出了其复杂的发病机制和目前的治疗局限性(smtd.202402263)。它强调了纳米材料的创新,通过工程特性实现靶向药物传递、抗炎作用和软骨再生,强调了它们在个性化和有效骨关节炎治疗方面的临床潜力。Hua Kuang等人综述了纳米材料(金属/金属氧化物纳米颗粒、脂质体、聚合物)作为疫苗佐剂的研究进展(smtd.202402059),强调了它们的机制:储存效应、NLRP3炎性小体激活、受体靶向和cGAS-STING途径激活。它讨论了当前的临床应用和优化安全性、有效性和可扩展性的挑战,以供未来使用。Huan孟等人写了一篇综述,讨论纳米材料如何诱导热凋亡,一种释放炎症细胞因子的溶解性细胞死亡,用于治疗应用(smtd.202401290)。纳米材料的可调特性(大小、形状、电荷)能够通过炎性小体激活精确调节焦亡,增强靶向癌症和感染治疗,同时最小化毒性。策略包括基于mrna的纳米药物和控制递送系统以优化疗效。另一篇关于纳米医学的综述论文来自于Luo等人。本文综述了纳米药物增强非小细胞肺癌(NSCLC)免疫治疗的研究进展。它强调了免疫检查点抑制剂、抗体-药物偶联物、细胞接合物、过继细胞治疗和癌症疫苗等策略。纳米医学提高了药物的靶向性、生物利用度,并在克服免疫耐药性的同时降低了毒性。创新包括基于纳米颗粒的PD-1/PD-L1抑制剂递送系统、联合疗法和TME调制。临床试验显示有希望与现有治疗协同,提供更安全,更精确的非小细胞肺癌治疗。未来的发展方向是多功能纳米平台,以优化免疫治疗效果和患者预后。让我们来看看胡全音在纳米医学领域的最后一篇综述。它回顾了治疗腹主动脉瘤(AAA)的纳米药物策略,解决了诸如药物靶向性不足和动态主动脉环境等挑战。它强调了炎症、ECM降解和腔内血栓在AAA进展中的作用,讨论了基于纳米颗粒的治疗方法,利用病理生理线索来局部给药和改善临床结果。生物传感器通过集成纳米材料、人工智能算法和电-生物接口的可穿戴/植入式设备的创新,正在推进智能生物医学工程。目前的研究强调持续的生物标志物监测(葡萄糖、细胞因子)、多分析物检测和现场治疗系统。纳米级适配体传感器和聚类规则间隔短回文重复序列(CRISPR)探测器实现了前所未有的灵敏度。在复杂的生物流体中,长期生物相容性、信号漂移校正和干扰最小化仍然存在挑战。新兴趋势包括使用生物势能的自供电生物传感器和用于动态病理生理反应跟踪的合成生物工程活传感器。Fengqin Li等人报道了一种DNA纳米花(DNF)驱动的用于超灵敏蛋白检测的CRISPR/Cas12a生物传感平台(DNF-CRISPR) (smt .202402130)。通过整合上游信号扩增的dnf和下游反式切割的CRISPR/Cas12a,该平台在2小时内实现了亚mg/L水平的检测限、宽动态范围和快速结果。利用肾损伤患者血液和尿液中的中性粒细胞明胶酶相关脂钙蛋白(NGAL)进行验证,该平台具有较高的准确性,优于传统的ELISA。 这种方法可以实现精确、高灵敏度的蛋白质标记检测,将基于crispr的诊断扩展到临床环境中的非核酸靶标。Jiang Li等人总结了DNA模板化蛋白质图谱的最新进展,利用DNA纳米技术进行精确的、可编程的蛋白质排列(smtd.202402249)。它讨论了共价(例如,交联剂,点击化学)和非共价(例如,生物素-链亲和素,适体)偶联策略。应用范围从一维到三维纳米结构、动态模式和生物医学(生物传感、纳米疫苗、靶向治疗)。DNA的可寻址性可实现低于10纳米的精度,生物相容性和动态控制。挑战包括共轭效率、稳定性和可扩展性。未来的方向集中在集成动态DNA系统,用于智能诊断/治疗和扩展功能复杂性。何海生等人报道了一种具有聚集引起猝灭(ACQ)特性的近红外(NIR-II)极性敏感荧光探针PN-C18的研究工作,用于跟踪脂质纳米载体的体内脂质分解(smt .202402249)。PN-C18克服了自由探针和混合胶束的干扰,能够精确监测脂质纳米颗粒的易位和降解。体外和体内实验表明,它与脂肪分解进展和深层组织成像能力具有良好的相关性。这种方法促进了对基于脂质的药物传递系统在体内命运的理解,为优化口服药物配方提供了见解。由于手性为生物系统提供了额外的物理维度,因此手性生物传感是智能生物传感器的研究热点。Ji-Young Kim等人撰写了一篇观点文章,强调了优化纳米光子生物传感器的手性量化方法的进展(smtd.202500112)。手性纳米材料通过放大手性信号来提高检测生物分子的灵敏度,但需要对结构-光学手性关系有更深入的了解。该展望讨论了近场研究,以探索手性纳米生物界面和超手性场效应。关键挑战包括标准化量化指标,改进纳米生物相互作用,统一传感器信号分析。未来的方向是精确的材料设计和标准化方案,以推进疾病诊断和个性化医疗的对映选择性生物传感。最后,我们高兴地看到,结合大规模科学设施、智能软机器人和深度学习方法,在智能生物医学的前沿研究方面取得了成就,这在很大程度上扩大了本期特刊的范围。朱颖等综述了基于同步加速器的智能生物医学x射线分子探针的研究进展(smt .202401890)它突出了先进的成像技术和探针,如抗体偶联纳米粒子、基因编码标签和靶向纳米材料。应用范围包括细胞成像、药物分析和疾病诊断/治疗,未来的趋势将集中在人工智能增强的精准和个性化医疗上。穆正志和他的跨学科团队集中研究生物灵感软机器人(BSR),集成生物运动机制和生物医学应用的刚柔耦合系统(smtd.202402264)。受弯曲(如捕蝇草)、扭曲(如章鱼手臂)和拉伸(如变色龙舌头)生物的启发,BSR利用人造肌肉纤维、水凝胶、形状记忆合金和软刚性复合材料来实现多自由度变形。关键的设计策略模仿生物结构,如软组织、刚性骨骼和它们的协同作用。应用包括体内肿瘤治疗、靶向药物输送和体外康复装置(如仿生假体)。在优化运动精度、环境适应性和生物相容性方面仍然存在挑战。未来的方向是强调先进的材料集成和仿生控制系统,以增强复杂生物医学环境中的临床翻译和功能。Gang Bao等人专注于预测CRISPR/Cas编辑的脱靶(OTS)风险(smtd.202500122)。CRISPR-Net、R-CRISPR和CRISPR-SGRU等深度学习方法在经过验证的脱靶数据训练后,显示出很好的OTS预测能力。在这篇综述中,他们评估了六个公共数据集,揭示了整合高质量的实验OTS数据可以增强模型的鲁棒性,特别是对于不平衡数据集。虽然没有出现普遍的领导者,但这些模型显示了临床基因组编辑应用的精度和安全性潜力。这个小方法的特别部分自豪地展示了智能生物医学的全球突破,致力于促进研究人员和实践者之间的跨学科合作。 我们对所有贡献者表示深切的感谢,他们的研究和审查工作是这一充满活力的领域的前沿创新的典范。感谢《小方法》的编辑团队,特别是奚文博士,为我们提供了发表这一专题的机会。作者声明无利益冲突。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Pioneering Biomedical Applications with Intelligent Engineering

Intelligent engineered biomedicine fundamentally integrates dynamic environmental responsiveness with sophisticated data-driven architectures incorporating biochips, microfluidics, and robotic automation, collectively enabling next-generation personalized therapeutics. Contemporary advancements prioritize adaptive biomaterial systems that continuously detect, process, and react to complex biological signals through integrated technological frameworks spanning artificial intelligence (AI)-optimized chemical synthesis, molecular computation, and precision fluidic control. These interconnected platforms drive end-to-end biopharmaceutical innovation – from smart manufacturing and targeted delivery to closed-loop feedback mechanisms – positioning them as core enablers of precision medicine through enhanced therapeutic accuracy, minimized adverse effects, and patient-specific treatment paradigms. Implementation strategies bifurcate into top-down modifications of conventional biomedical platforms with programmable functionality and bottom-up engineering of intrinsically intelligent biomaterials through molecular-level design principles. To highlight the latest advancements in these fields, we have put together a Special Section titled “Intelligent Biomedical Engineering.” This collection includes 10 research articles, 1 perspective and 13 reviews from prominent scientists with their valuable insights.

Drug delivery emerges as a transformative cornerstone in intelligent biomedical engineering, where responsive nanomaterials synergize with AI-driven pharmacokinetic modeling and biofeedback-enabled nanodevices to enable dynamic, physiology-responsive therapeutics. These advanced systems utilize stimuli-activated carriers that intelligently modulate drug release in real-time based on biomarker fluctuations, achieving unprecedented spatiotemporal precision through engineered ligand-receptor targeting and biosensor-integrated automatic control. Tao Sun et al. develop a reactive oxygen species (ROS)-responsive heterodimer prodrug (VTO) combining oaldehyde dehydrogenase 2roxylin A (OA) and all-trans retinol (VA) linked via a thioketal bond to synergistically treat liver fibrosis (smtd.202402247). Encapsulated in biomimetic high-density lipoprotein, the co-delivery platform named PL-VTO nanoparticle leveraged natural liver targeting via scarvenger receptor type 1 binding. Upon reaching fibrotic liver, PL-VTO released OA and VA in response to elevated ROS levels, effectively scavenging ROS, deactivating hepatic stellate cells, and reducing collagen deposition. In vitro and in vivo studies demonstrated enhanced ROS clearance, targeted delivery, and fibrosis reversal with minimal toxicity, offering a promising multi-drug nanotherapeutic approach for liver fibrosis treatment. Hendrick W. de Haan and his coworkers investigate comb-shaped polymers for protein molecular sieving coatings (smtd.202500140). Simulations reveal three interaction regimes: no-, weak-, and strong-interactions between polymer chains, governed by steric hindrance, microenvironmental effects, and altered protein dynamics. Systematic parameter analysis identifies strategies to tune coating permeability and size selectivity, providing a universal framework for designing molecular sieving systems with controlled thresholds.

Hydrogels, as promising drug delivery platforms especially in the field of wound healing, were integrated into intelligent biomedicine by various groups. Yanli Zhao et al. introduce a photodynamic hydrogel activating aldehyde dehydrogenase 2 (ALDH2) to treat diabetic wounds by synergizing immune-vascular repair (smtd.202500391). ALDH2 deficiency in diabetic wounds exacerbates inflammation via neutrophil extracellular traps (NETs) and impairs angiogenesis. The hydrogel integrates ALDA-1 (ALDH2 activator) and chlorin e6, reducing NETs, polarizing macrophages to anti-inflammatory M2, and enhancing vascularization. In vivo tests show accelerated healing through dual modulation of inflammation and angiogenesis, offering a novel therapeutic strategy for chronic diabetic wounds. Chang Du and her coworkers develop an intelligent hydrogel platform (OGM) using gelatin modified with phenylboronic acid, oxidized dextran, and myricetin to accelerate diabetic wound healing (smtd.202401127). The hydrogel exhibits pH/ROS/glucose-responsive release of myricetin, effectively scavenging ROS, alleviating oxidative stress, and promoting angiogenesis via nuclear factor erythroid 2-related factor 2 pathway activation. In vitro and in vivo tests demonstrated enhanced cell proliferation, migration, and vascularization. This triple-responsive platform offers a promising strategy for diabetic wound care. Li Kong and his collaborators present a hydrogel-based immunochemotherapy system (smtd.202401425), to prevent post-surgical recurrence and metastasis in triple-negative breast cancer. The system combines doxorubicin-loaded nanoparticles (NPD) attached to platelets and interleukin-12 (IL-12) within a dopamine-modified alginate hydrogel. Upon activation in the tumor microenvironment, platelets release NPD-loaded microparticles that target residual/metastatic tumor cells, inducing immunogenic cell death. This strategy offers a promising localized therapy to enhance chemotherapeutic efficacy and immune activation post-surgery.

Exosomes are emerging as promising natural nanocarriers in drug delivery due to their innate biocompatibility, target-homing capabilities, and membrane stability. Current research focuses on engineering exosomal surfaces for enhanced tissue specificity, optimizing cargo loading techniques, and harnessing endogenous trafficking mechanisms. Seungpyo Hong et al. review researches in exosomes as theranostic tools in gastrointestinal cancers, highlighting their dual role as diagnostic biomarkers (miRNAs, lncRNAs, circRNAs, proteins) and engineered therapeutic carriers (smtd.202402058). It discusses advancements in exosome-based strategies for early detection, overcoming drug resistance, metastasis suppression, and immune modulation, addressing clinical challenges and future directions. Jiyong Liu et al. highlighted exosome-based vaccines as promising platforms for infectious diseases and cancer, leveraging their immunomodulatory properties, antigen presentation, and drug delivery capabilities (smtd.202402222). It discusses their biogenesis, components, preparation, and clinical applications, emphasizing personalized vaccine design while addressing challenges in standardization and clinical translation of exosome-based vaccines.

Advancements in other aspects of drug delivery systems including microneedles and oral drug delivery were highlighted by the contributors. Shutao Guo et al. discuss responsive microneedles for ocular drug delivery, addressing traditional limitations like low bioavailability (smtd.202402048). It categorizes responsive microneedles (pH, ROS, enzyme, glucose-triggered), highlighting their mechanisms and applications in treating diseases like glaucoma, diabetic retinopathy, and infections, emphasizing precision and reduced invasiveness. Wei Tao et al. summarize challenges in oral biologics delivery, including gastric acid, enzymatic degradation, and poor intestinal permeability (smtd.202401355). It highlights strategies like nanoparticle systems, ionic liquids, microneedles, and colon-targeted approaches to enhance bioavailability. Advances in delivery technologies may enable effective oral administration of biologics for chronic diseases.

Nanomedicine is revolutionizing intelligent biomedical engineering through the development of stimuli-responsive nanoplatforms that synergistically combine molecular targeting, biosensing capabilities, and controlled therapeutic delivery. Current research focuses on engineering smart materials with environmental sensitivity and precision targeting via ligand-receptor modifications, while integrating AI-driven nanoparticle design and adaptive biosensing systems. Notable advances include tumor-specific delivery platforms and blood-brain barrier penetrating systems, though challenges persist in predicting complex in vivo behaviors and establishing scalable manufacturing protocols. Emerging frontiers encompass DNA nanostructures for programmable drug carriers, organ-on-chip validation platforms, and computational pharmacokinetic models that simulate nanoparticle-body interactions. The field continues to evolve toward multifunctional theranostic systems capable of simultaneous diagnosis, real-time monitoring, and adaptive treatment modulation. Jinjin Shi et al. develop neutrophil-targeting biomimetic nanoparticles (MPB NPs) to treat atherosclerosis by inhibiting neutrophil extracellular trap (NET) formation (smtd.202402019). MPB NPs, composed of Prussian blue nanoparticles coated with bacterial membranes, specifically hitchhike on neutrophils via pathogen-associated molecular pattern recognition. By scavenging intracellularROS, MPB NPs suppress NET release induced by inflammatory stimuli. In atherosclerotic mouse models, MPB NPs remarkably reduced plaque area, stabilized plaques, and halted disease progression. This targeted approach demonstrates significant therapeutic potential for atherosclerosis and offers a platform for NET-related inflammatory diseases. Lin Mei et al. report a novel nanoparticle for treating idiopathic pulmonary fibrosis (IPF) by co-loading luteolin (Lut, an anti-fibrotic flavonoid) and hyaluronidase (HAase) into PEG-crosslinked nanoparticles (smtd.202400980). The Lut@HAase demonstrated enhanced lung accumulation via inhalation and deep tissue penetration through HAase-mediated hyaluronic acid degradation. In vitro, Lut@HAase suppressed TGF-β1-induced fibroblast activation (reducing α-SMA/FN expression), inflammatory cytokines (IL-6, IL-1β, TNF-α), and ROS in MRC5 cells. In bleomycin-induced IPF mice, inhaled Lut@HAase improved lung function (reduced resistance, increased compliance), decreased hydroxyproline content, attenuated fibrosis markers, and enhanced survival rates compared to free Lut or HAase, showcasing synergistic anti-fibrotic and anti-inflammatory effects. Si Li et al. develop chiral hydrogel microspheres (HMSs) incorporating L/D-Co₃O₄ nanoparticles to enhance lipase catalytic performance (smtd.202400918). By embedding lipase within L-Co₃O₄-HMSs, catalytic activity significantly increased compared to free lipase and was doubled than D-Co₃O₄-HMSs, attributed to the chiral microenvironment. The L-Co₃O₄-HMSs also demonstrated superior inhibition of 3T3-L1 fibroblast differentiation into adipocytes, highlighting their efficacy in lipid metabolism regulation. This work underscores the role of chirality in optimizing enzyme activity and offers a novel strategy for designing biomimetic catalytic systems with enhanced performance in industrial and biomedical applications. Jiao Yan review explorations in intelligent nanomaterials for osteoarthritis management, addressing its complex pathogenesis and current treatment limitations (smtd.202402263). It highlights nanomaterial innovations enabling targeted drug delivery, anti-inflammatory effects, and cartilage regeneration through engineered properties, emphasizing their clinical potential for personalized and effective osteoarthritis therapies. Hua Kuang et al. provide a review in nanomaterials (metal/metal oxide nanoparticles, liposomes, polymers) as vaccine adjuvants (smtd.202402059), highlighting their mechanisms: depot effect, NLRP3 inflammasome activation, receptor targeting, and cGAS-STING pathway activation. It discusses current clinical applications and challenges in optimizing safety, efficacy, and scalability for future use. Huan Meng et al. write a review discussing how nanomaterials induce pyroptosis, a lytic cell death releasing inflammatory cytokines, for therapeutic applications (smtd.202401290). Nanomaterials’ tunable properties (size, shape, charge) enable precise modulation of pyroptosis via inflammasome activation, enhancing targeted cancer and infection treatments while minimizing toxicity. Strategies include mRNA-based nanomedicine and controlled delivery systems to optimize efficacy. Another review paper in nanomedicine comes from Kui Luo et al. This paper reviews advancements in nanomedicine for enhancing non-small cell lung cancer (NSCLC) immunotherapy (smtd.202401783). It highlights strategies like immune checkpoint inhibitors, antibody-drug conjugates, cell engagers, adoptive cell therapy, and cancer vaccines. Nanomedicine improves drug targeting, bioavailability, and reduces toxicity while overcoming immune resistance. Innovations include nanoparticle-based delivery systems for PD-1/PD-L1 inhibitors, combination therapies, and TME modulation. Clinical trials show promising synergy with existing treatments, offering safer, more precise NSCLC therapies. Future directions focus on multifunctional nanoplatforms to optimize immunotherapy efficacy and patient outcomes. Let's see the last review paper in nanomedicine by Quanyin Hu. It reviews nanomedicine strategies for treating abdominal aortic aneurysms (AAA), addressing challenges like insufficient drug targeting and dynamic aortic environments (smtd.202402268). It highlights the role of inflammation, ECM degradation, and intraluminal thrombus in AAA progression, discussing nanoparticle-based therapies leveraging pathophysiological cues for localized drug delivery and improved clinical outcomes.

Biosensors are advancing intelligent biomedical engineering through innovations in wearable/implantable devices integrating nanomaterials, AI algorithms, and electro-bio interfaces. Current research emphasizes continuous biomarker monitoring (glucose, cytokines), multi-analyte detection, and on-site therapeutic systems. Nanoscale aptasensors and clustered regularly interspaced short palindromic repeats (CRISPR)-based detectors achieve unprecedented sensitivity. Challenges persist in long-term biocompatibility, signal drift correction, and interference minimization in complex biofluids. Emerging trends include self-powered biosensors using biopotential energy and synthetic biology-engineered living sensors for dynamic pathophysiological response tracking. Fengqin Li et al. report a DNA nanoflower (DNF)-powered CRISPR/Cas12a biosensing platform (DNF-CRISPR) for ultrasensitive protein detection (smtd.202402130). By integrating DNFs for upstream signal amplification and CRISPR/Cas12a for downstream trans-cleavage, the platform achieves a detection limit of sub-mg/L level, a broad dynamic range, and rapid results within 2 h. Validated using neutrophil gelatinase-associated lipocalin (NGAL) in kidney injury patients’ blood and urine, the platform demonstrated high accuracy, outperforming conventional ELISA. This approach enables precise, high-sensitivity protein marker detection, expanding CRISPR-based diagnostics to non-nucleic acid targets in clinical settings. Jiang Li et al. summarize recent advances in DNA-templated protein patterning, leveraging DNA nanotechnology for precise, programmable protein arrangements (smtd.202402249). It discusses covalent (e.g., crosslinkers, click chemistry) and non-covalent (e.g., biotin-streptavidin, aptamers) conjugation strategies. Applications span 1D to 3D nanostructures, dynamic patterns, and biomedicine (biosensing, nanovaccines, targeted therapies). DNA's addressability enables sub-10 nm precision, biocompatibility, and dynamic control. Challenges include conjugation efficiency, stability, and scalability. Future directions focus on integrating dynamic DNA systems for smart diagnostics/therapeutics and expanding functional complexity. Haisheng He et al. report a research work in PN-C18, a near-infrared-II (NIR-II) polarity-sensitive fluorescent probe with aggregation-caused quenching (ACQ) properties, to track in vivo lipolysis of lipid nanocarriers (smtd.202402249). PN-C18 overcomes interference from free probes and mixed micelles, enabling precise monitoring of lipid nanoparticle translocation and degradation. In vitro and in vivo experiments demonstrate its superior correlation with lipolysis progression and deep-tissue imaging capability. This approach advances understanding of lipid-based drug delivery systems’ in vivo fate, offering insights for optimizing oral drug formulations. Chiral biosensing is a research “hot-spot” in intelligent biosensors because chirality provided additional physical dimensions to biological systems. Ji-Young Kim et al. write a perspective highlighting advances in chirality quantification methods for optimizing nanophotonic biosensors (smtd.202500112). Chiral nanomaterials enhance sensitivity for detecting biomolecules via amplified chiroptical signals, but require deeper understanding of structural-optical chirality relationships. The perspective discusses near-field studies to probe chiral nano-bio interfaces and superchiral field effects. Key challenges include standardizing quantification metrics, improving nano-bio interactions, and unifying sensor signal analysis. Future directions focus on precise material design and standardized protocols to advance enantioselective biosensing for disease diagnostics and personalized medicine.

Finally, we are glad to see achievements in cutting-edge investigations of intelligent biomedicines in combinations of large-scale scientific facilities, smart soft robots, and deep-learning methodologies, largely expanding the scope of this special issue. Ying Zhu and her group review recent advances in synchrotron-based X-ray molecular probes for intelligent biomedicine (smtd.202401890). It highlights advanced imaging techniques and probes like antibody-coupled nanoparticles, genetically encoded tags, and targeted nanomaterials. Applications span cellular imaging, drug analysis, and disease diagnosis/therapy, with future trends focusing on AI-enhanced precision and personalized medicine. Zhengzhi Mu and his interdisciplinary teams gather research work in bioinspired soft robots (BSR) integrating biological motion mechanisms and rigid-flexible coupling systems for biomedical applications (smtd.202402264). Inspired by bending (e.g., Venus flytrap), twisting (e.g., octopus arms), and stretching (e.g., chameleon tongue) organisms, BSR leverage artificial muscle fibers, hydrogels, shape-memory alloys, and soft-rigid composites to achieve multi-degree-of-freedom deformations. Key design strategies mimic biological structures like soft tissues, rigid skeletons, and their synergistic interactions. Applications include in vivo tumor therapy, targeted drug delivery, and in vitro rehabilitative devices (e.g., bionic prostheses). Challenges remain in optimizing motion precision, environmental adaptability, and biocompatibility. Future directions emphasize advanced material integration and biomimetic control systems to enhance clinical translation and functionality in complex biomedical environments. Gang Bao et al. focus on predictions of off-target (OTS) risks of CRISPR/Cas editing (smtd.202500122). Deep learning methods like CRISPR-Net, R-CRISPR, and CRISPR-SGRU show promising OTS prediction capabilities when trained with validated off-target data. In this review, they evaluated six public datasets revealing integrating high-quality experimental OTS data enhances model robustness, particularly with imbalanced datasets. While no universal leader emerged, these models demonstrate improved precision and safety potential for clinical genome editing applications.

This special section of Small Methods proudly presents global breakthroughs in intelligent biomedicines, dedicated to fostering interdisciplinary collaboration between researchers and practitioners. We extend profound appreciation to all contributors that their research and review work exemplifies frontier innovations in this dynamic field. Gratitude is owed to the Small Methods editorial team, especially Dr. Xi Wen, for supplying us the chance to publish this special section.

The authors declare no conflict of interest.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Small Methods
Small Methods Materials Science-General Materials Science
CiteScore
17.40
自引率
1.60%
发文量
347
期刊介绍: Small Methods is a multidisciplinary journal that publishes groundbreaking research on methods relevant to nano- and microscale research. It welcomes contributions from the fields of materials science, biomedical science, chemistry, and physics, showcasing the latest advancements in experimental techniques. With a notable 2022 Impact Factor of 12.4 (Journal Citation Reports, Clarivate Analytics, 2023), Small Methods is recognized for its significant impact on the scientific community. The online ISSN for Small Methods is 2366-9608.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信