Severe fever with thrombocytopenia syndrome virus (SFTSV), an emerging tick-borne phlebovirus, poses a growing public health threat, with no approved vaccines or targeted therapies. Its genetic diversity and rapid reassortment have hindered the development of broadly effective interventions. Here, we isolated and characterized broadly neutralizing camelid nanobodies generated by a heterologous immunization strategy. These nanobodies exhibited cross-subtype neutralization and conferred protection against disease in humanized and lethal murine models of SFTSV infection. Structural analysis revealed binding to distinct, nonoverlapping epitopes on the viral glycoprotein complex. A rationally designed cocktail leveraging this epitope diversity achieved complete viral inhibition through synergistic mechanisms. In both murine and immunocompetent ferret models, the cocktail enabled efficient viral clearance and full protection against lethal challenge. These results demonstrate the therapeutic potential of nanobody cocktails for SFTSV infection and establish a generalizable framework for nanobody-based countermeasures against genetically variable emerging viruses, including other members of the Bunyavirales order.
{"title":"A rationally designed cocktail of nanobodies elicited by heterologous vaccination confers protection against SFTSV in preclinical models","authors":"Xilin Wu, Linjing Zhu, Shengjian Liang, Zhen Chang, Yaxin Wang, Qingcui Zou, Zhili Xu, Doudou Zhang, Jiaqian Hu, Shengya Geng, Zhifeng Li, Wenkui Yu, Zhiliang Hu, Ming Chen, Ke Cao, Bei Jia, Hongxia Wei, Rentian Cai, Junyi Long, Xiaoman Yang, Qiaojiang Yang, Zhanjing Yu, Lingen Shi, Binghong Xu, Yangwu Zheng, Shengrui Meng, Jiameng Cai, Guangfeng Zhou, Kerui Zhu, Yunbo Dou, Zhiyong Lou, Minghua Li, Yan Wu, Zhiwei Wu","doi":"10.1126/scitranslmed.ady9025","DOIUrl":"10.1126/scitranslmed.ady9025","url":null,"abstract":"<div >Severe fever with thrombocytopenia syndrome virus (SFTSV), an emerging tick-borne phlebovirus, poses a growing public health threat, with no approved vaccines or targeted therapies. Its genetic diversity and rapid reassortment have hindered the development of broadly effective interventions. Here, we isolated and characterized broadly neutralizing camelid nanobodies generated by a heterologous immunization strategy. These nanobodies exhibited cross-subtype neutralization and conferred protection against disease in humanized and lethal murine models of SFTSV infection. Structural analysis revealed binding to distinct, nonoverlapping epitopes on the viral glycoprotein complex. A rationally designed cocktail leveraging this epitope diversity achieved complete viral inhibition through synergistic mechanisms. In both murine and immunocompetent ferret models, the cocktail enabled efficient viral clearance and full protection against lethal challenge. These results demonstrate the therapeutic potential of nanobody cocktails for SFTSV infection and establish a generalizable framework for nanobody-based countermeasures against genetically variable emerging viruses, including other members of the Bunyavirales order.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 825","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145545438","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Numerous G protein–coupled receptors (GPCRs) expressed in the gastrointestinal tract serve as crucial transducers to regulate a variety of physiological functions upon activation. Takeda G protein–coupled receptor 5 (TGR5), a prominent gastrointestinal GPCR expressed on enteroendocrine L cells, is activated by intestinal bile acids and plays a role in glucose utilization. However, the development of TGR5 agonists has been hindered by the hepatobiliary toxicity associated with long-term supplementation with exogenous agonists. Here, we designed and characterized a biomimetic receptor agonist, which we termed TGR5-targeted carrier-drug conjugate (TGR5-CaDC), that combined the TGR5-activating capabilities of deoxycholic acid, a TGR5 agonist, with the nonabsorbable properties of a carrier. Unlike traditional agonists or carrier-based drug delivery systems, nonabsorbable TGR5-CaDC remained localized in the intestines of mice and pigs, providing high surface concentrations of TGR5 agonists in addition to ensuring strong L cell specificity and TGR5 affinity. TGR5-CaDC treatment also promoted TGR5 cluster aggregation, signal amplification, and increased glucagon-like peptide 1 secretion. Notably, TGR5-CaDC demonstrated sustained glycemic effects with reduced toxicity compared with deoxycholic acid alone or liraglutide in diabetic mice and Bama minipigs. By targeting extracellular binding domains and mimicking native ligand-receptor binding patterns, the design concept underlying this carrier-drug conjugate has the potential for applications in a variety of GPCR-mediated gastrointestinal diseases.
{"title":"Intestinal TGR5-targeted carrier-drug conjugate improves glycemic control in mice and pigs","authors":"Yaqi Zhang, Hui Huang, Yaying Wang, Xiang Li, Peizhou Hou, Miaorong Yu, Zhuan Zhang, Shiyan Guo, Chang Liu, Zilong Zhang, Yan Zhuo, Chunliu Zhu, Pengcheng Zhang, Shisheng Wang, Hu Zhou, Yong Gan","doi":"10.1126/scitranslmed.ado5177","DOIUrl":"10.1126/scitranslmed.ado5177","url":null,"abstract":"<div >Numerous G protein–coupled receptors (GPCRs) expressed in the gastrointestinal tract serve as crucial transducers to regulate a variety of physiological functions upon activation. Takeda G protein–coupled receptor 5 (TGR5), a prominent gastrointestinal GPCR expressed on enteroendocrine L cells, is activated by intestinal bile acids and plays a role in glucose utilization. However, the development of TGR5 agonists has been hindered by the hepatobiliary toxicity associated with long-term supplementation with exogenous agonists. Here, we designed and characterized a biomimetic receptor agonist, which we termed TGR5-targeted carrier-drug conjugate (TGR5-CaDC), that combined the TGR5-activating capabilities of deoxycholic acid, a TGR5 agonist, with the nonabsorbable properties of a carrier. Unlike traditional agonists or carrier-based drug delivery systems, nonabsorbable TGR5-CaDC remained localized in the intestines of mice and pigs, providing high surface concentrations of TGR5 agonists in addition to ensuring strong L cell specificity and TGR5 affinity. TGR5-CaDC treatment also promoted TGR5 cluster aggregation, signal amplification, and increased glucagon-like peptide 1 secretion. Notably, TGR5-CaDC demonstrated sustained glycemic effects with reduced toxicity compared with deoxycholic acid alone or liraglutide in diabetic mice and Bama minipigs. By targeting extracellular binding domains and mimicking native ligand-receptor binding patterns, the design concept underlying this carrier-drug conjugate has the potential for applications in a variety of GPCR-mediated gastrointestinal diseases.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 825","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145545435","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-19DOI: 10.1126/scitranslmed.adu8579
Matthew D. Slein, Iara M. Backes, Lesle M. Jiménez, Natasha S. Kelkar, Callaghan R. Garland, Urjeet S. Khanwalkar, Anton M. Sholukh, Christine Johnston, David A. Leib, Margaret E. Ackerman
Herpes simplex virus (HSV) encodes surface glycoproteins that are host defense evasion molecules. For example, glycoproteins E and I (gE/gI) form a viral Fc receptor (vFcR) for most subclasses and allotypes of human IgG, promoting evasion of humoral immune responses. Although monoclonal antibodies (mAbs) protect mice from neonatal HSV (nHSV) infections, the impact of vFcR activity on mAb-mediated protection is unknown. Using HSV-1 with intact and ablated gE-mediated Fc binding, as well as Fc-engineered mAbs with modified ability to interact with gE/gI, we investigated the role of the vFcR in mAb-mediated protection from nHSV. HSV-specific mAbs modified to lack binding to gE exhibited enhanced neutralization in vitro and superior protection in vivo compared with their native IgG1 forms. Improved protection was dependent on the presence of vFcR activity and was observed for mAbs specific for both glycoprotein D and glycoprotein B, as well as for a nonneutralizing mAb, and for both laboratory-adapted and clinical isolates of HSV-1 and HSV-2. Further, human IgG3 allotypes, including those lacking vFcR binding, also exhibited enhanced antiviral activity in vivo, identifying a unique viral susceptibility to this subclass. In summary, this study demonstrates that rendering mAbs insensitive to the vFcR can improve protection against HSV, offering prospects for antibody-based interventions.
{"title":"Eliminating interactions with the viral Fc receptor improves antibody-mediated protection against neonatal HSV infection in mice","authors":"Matthew D. Slein, Iara M. Backes, Lesle M. Jiménez, Natasha S. Kelkar, Callaghan R. Garland, Urjeet S. Khanwalkar, Anton M. Sholukh, Christine Johnston, David A. Leib, Margaret E. Ackerman","doi":"10.1126/scitranslmed.adu8579","DOIUrl":"10.1126/scitranslmed.adu8579","url":null,"abstract":"<div >Herpes simplex virus (HSV) encodes surface glycoproteins that are host defense evasion molecules. For example, glycoproteins E and I (gE/gI) form a viral Fc receptor (vFcR) for most subclasses and allotypes of human IgG, promoting evasion of humoral immune responses. Although monoclonal antibodies (mAbs) protect mice from neonatal HSV (nHSV) infections, the impact of vFcR activity on mAb-mediated protection is unknown. Using HSV-1 with intact and ablated gE-mediated Fc binding, as well as Fc-engineered mAbs with modified ability to interact with gE/gI, we investigated the role of the vFcR in mAb-mediated protection from nHSV. HSV-specific mAbs modified to lack binding to gE exhibited enhanced neutralization in vitro and superior protection in vivo compared with their native IgG1 forms. Improved protection was dependent on the presence of vFcR activity and was observed for mAbs specific for both glycoprotein D and glycoprotein B, as well as for a nonneutralizing mAb, and for both laboratory-adapted and clinical isolates of HSV-1 and HSV-2. Further, human IgG3 allotypes, including those lacking vFcR binding, also exhibited enhanced antiviral activity in vivo, identifying a unique viral susceptibility to this subclass. In summary, this study demonstrates that rendering mAbs insensitive to the vFcR can improve protection against HSV, offering prospects for antibody-based interventions.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 825","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145545440","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-19DOI: 10.1126/scitranslmed.adr1777
Dingkang Liu, Jing Tian, Lichao Yu, Hong Luo, Haibo Rong, Yue Tong, Xiangdong Gao, Jun Yin
Immunogenic cell death (ICD) is a promising approach for generating antitumor immune responses to treat patients with cancer. However, the stereotactic induction of ICD and spatiotemporal synchronized activation of the tumor-specific immune response pose two critical challenges. Here, we report the basic principles and systematic development of a modular metalloprotein platform designated the protease-activated PSTAGylated in situ tumor vaccine (PPTV). Ferritin was used as a vaccine framework, with its outer surface fused with an optimal mito-disrupt peptide and its inner cavity loaded with manganese ions (Mn2+). Moreover, a protease-activated PSTAGylated prodrug strategy was developed to circumvent the major issues associated with ferritin, such as liver interception and drug leakage. We also demonstrated the activation of prodrugs in tumor lysates from patients. In subcutaneous and orthotopic tumor transplantation mouse models, the PPTV effectively codelivered mito-disrupt peptides and Mn2+ into tumors, leading to a combined effect of ICD and cGAS-STING cyclic GMP–AMP synthase–stimulator of interferon genes (cGAS-STING) pathway activation, thereby achieving a potent antitumor immune response. The combined application of PPTV and anti–programmed death-ligand 1 resulted in the eradication of established tumors. These results underscore the applicability of the PPTV as an innovative in situ vaccine nanoplatform for establishing an antitumor immune response. This modular metalloprotein vaccine has clinical translational potential because of its simple generation and safety profile.
{"title":"A modular metalloprotein in situ vaccine for cancer immunotherapy in mouse models of breast cancer","authors":"Dingkang Liu, Jing Tian, Lichao Yu, Hong Luo, Haibo Rong, Yue Tong, Xiangdong Gao, Jun Yin","doi":"10.1126/scitranslmed.adr1777","DOIUrl":"10.1126/scitranslmed.adr1777","url":null,"abstract":"<div >Immunogenic cell death (ICD) is a promising approach for generating antitumor immune responses to treat patients with cancer. However, the stereotactic induction of ICD and spatiotemporal synchronized activation of the tumor-specific immune response pose two critical challenges. Here, we report the basic principles and systematic development of a modular metalloprotein platform designated the protease-activated PSTAGylated in situ tumor vaccine (PPTV). Ferritin was used as a vaccine framework, with its outer surface fused with an optimal mito-disrupt peptide and its inner cavity loaded with manganese ions (Mn<sup>2+</sup>). Moreover, a protease-activated PSTAGylated prodrug strategy was developed to circumvent the major issues associated with ferritin, such as liver interception and drug leakage. We also demonstrated the activation of prodrugs in tumor lysates from patients. In subcutaneous and orthotopic tumor transplantation mouse models, the PPTV effectively codelivered mito-disrupt peptides and Mn<sup>2+</sup> into tumors, leading to a combined effect of ICD and cGAS-STING cyclic GMP–AMP synthase–stimulator of interferon genes (cGAS-STING) pathway activation, thereby achieving a potent antitumor immune response. The combined application of PPTV and anti–programmed death-ligand 1 resulted in the eradication of established tumors. These results underscore the applicability of the PPTV as an innovative in situ vaccine nanoplatform for establishing an antitumor immune response. This modular metalloprotein vaccine has clinical translational potential because of its simple generation and safety profile.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 825","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145545439","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-12DOI: 10.1126/scitranslmed.adn3993
Xiaoshu Pan, Peixin Huang, Samantha S. Ali, Bryan Renslo, Zachary Greenberg, Nina Erwin, Yanjun Li, Zuo Ding, Tarun E. Hutchinson, Athanasia Warnecke, Natalia E. Fernandez, Hinrich Staecker, Mei He
The clinical translation of gene therapy has been challenging in part because of the limitations of current delivery approaches. Herein, we report an efficient nonviral genome editor delivery approach using extracellular vesicles (EVs) carrying single-guide RNA (sgRNA): CRISPR-Cas9 ribonucleoprotein (RNP) complexes for in vivo gene therapy. By leveraging a high-throughput microfluidic droplet–based electroporation system (μDES), we achieved a 10-fold enhancement in loading efficiency and more than 1000-fold increase in processing throughput for loading RNP complexes into EVs compared with conventional high-voltage pulsed electroporation. μDES generated uniform microdroplets containing EVs and RNPs by applying direct current–controlled low voltage (up to 60 V) to transiently permeabilize membranes and enable efficient cargo encapsulation while maintaining EV integrity at both the protein and morphological levels. In the Myo7aWT/Sh1 mouse model of autosomal dominant progressive hearing loss, which may model MYO7A-associated DFNA11 hearing loss in humans, we demonstrated the effective delivery of RNPs by EVs into cochlear hair cells by cross-sectional and whole-mount confocal imaging. The injection of RNP-EVs via the posterior semicircular canal in 4-week-old Myo7aWT/Sh1 mice resulted in a reduction in Myo7aSh1 messenger RNA expression and evidence of hearing preservation, as measured by auditory brainstem responses, compared with untreated ears and EV only–injected mice. This study highlights the potential of μDES-produced RNP-EVs for gene editing as a treatment for progressive nonsyndromic hearing loss in patients.
{"title":"Extracellular vesicle–mediated gene editing for the treatment of nonsyndromic progressive hearing loss in adult mice","authors":"Xiaoshu Pan, Peixin Huang, Samantha S. Ali, Bryan Renslo, Zachary Greenberg, Nina Erwin, Yanjun Li, Zuo Ding, Tarun E. Hutchinson, Athanasia Warnecke, Natalia E. Fernandez, Hinrich Staecker, Mei He","doi":"10.1126/scitranslmed.adn3993","DOIUrl":"10.1126/scitranslmed.adn3993","url":null,"abstract":"<div >The clinical translation of gene therapy has been challenging in part because of the limitations of current delivery approaches. Herein, we report an efficient nonviral genome editor delivery approach using extracellular vesicles (EVs) carrying single-guide RNA (sgRNA): CRISPR-Cas9 ribonucleoprotein (RNP) complexes for in vivo gene therapy. By leveraging a high-throughput microfluidic droplet–based electroporation system (μDES), we achieved a 10-fold enhancement in loading efficiency and more than 1000-fold increase in processing throughput for loading RNP complexes into EVs compared with conventional high-voltage pulsed electroporation. μDES generated uniform microdroplets containing EVs and RNPs by applying direct current–controlled low voltage (up to 60 V) to transiently permeabilize membranes and enable efficient cargo encapsulation while maintaining EV integrity at both the protein and morphological levels. In the <i>Myo7a<sup>WT/Sh1</sup></i> mouse model of autosomal dominant progressive hearing loss, which may model MYO7A-associated DFNA11 hearing loss in humans, we demonstrated the effective delivery of RNPs by EVs into cochlear hair cells by cross-sectional and whole-mount confocal imaging. The injection of RNP-EVs via the posterior semicircular canal in 4-week-old <i>Myo7a<sup>WT/Sh1</sup></i> mice resulted in a reduction in <i>Myo7a<sup>Sh1</sup></i> messenger RNA expression and evidence of hearing preservation, as measured by auditory brainstem responses, compared with untreated ears and EV only–injected mice. This study highlights the potential of μDES-produced RNP-EVs for gene editing as a treatment for progressive nonsyndromic hearing loss in patients.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 824","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145492714","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-12DOI: 10.1126/scitranslmed.adq6645
Cheng-Chia Wu, Luca Szalontay, Antonios N. Pouliopoulos, Sua Bae, Xander Berg, Hong-Jian Wei, Andrea Webster Carrion, Danae Kokossis, Chankrit Sethi, Jessica Fino, Halina Shatravka, Jennifer Lipina, Robin Ji, Keyu Liu, Omid Yousefian, Matthew Gallitto, Nina Yoh, Zachary Englander, Nicholas McQuillan, Masih Tazhibi, Genesis De Los Santos, Peter Canoll, Zhezhen Jin, James Garvin, Robyn D. Gartrell, Jovana Pavisic, Alexis Maddocks, Angela Lignelli, Neil Feldstein, Elisa E. Konofagou, Stergios Zacharoulis
Focused ultrasound (FUS)–mediated blood-brain barrier (BBB) opening with microbubbles is an emerging technology that enables drug delivery for central nervous system diseases. To date, most clinical trials assessing BBB opening in adults were designed to deliver US with a frequency of one treatment over several weeks. Little is known about the feasibility of shorter intervals of US delivery or whether this can be achieved in a pediatric population using a mobile device. Here, FUS and panobinostat were shown to have additive therapeutic effects in a syngeneic orthotopic model of diffuse midline glioma (DMG). We then conducted a single-arm first-in-pediatric trial to investigate the safety and feasibility of delivering neuronavigation-guided FUS treatment in combination with oral panobinostat in children with relapsed DMGs. We included an intrapatient escalation of FUS delivery to assess the feasibility of opening multiple sites in the brain. We demonstrated successful BBB opening using neuronavigation-guided FUS as frequently as every 2 days. Magnetic resonance imaging with contrast was used to identify the region of BBB opening. Three patients were accrued; 22 FUS procedures were delivered for 1 NOTS (number of tumor sites) treated, and four FUS procedures were delivered for 2 NOTS. All three patients received 1 NOTS, without serious adverse events, and two of the patients received 2 NOTS, all without sedation. For 2 NOTS, prolonged BBB opening and one grade 5 event, unlikely related to FUS, were observed. This study demonstrates feasibility of FUS for BBB opening and drug delivery without sedation in pediatric patients.
{"title":"Blood-brain barrier opening with neuronavigation-guided focused ultrasound in pediatric patients with diffuse midline glioma","authors":"Cheng-Chia Wu, Luca Szalontay, Antonios N. Pouliopoulos, Sua Bae, Xander Berg, Hong-Jian Wei, Andrea Webster Carrion, Danae Kokossis, Chankrit Sethi, Jessica Fino, Halina Shatravka, Jennifer Lipina, Robin Ji, Keyu Liu, Omid Yousefian, Matthew Gallitto, Nina Yoh, Zachary Englander, Nicholas McQuillan, Masih Tazhibi, Genesis De Los Santos, Peter Canoll, Zhezhen Jin, James Garvin, Robyn D. Gartrell, Jovana Pavisic, Alexis Maddocks, Angela Lignelli, Neil Feldstein, Elisa E. Konofagou, Stergios Zacharoulis","doi":"10.1126/scitranslmed.adq6645","DOIUrl":"10.1126/scitranslmed.adq6645","url":null,"abstract":"<div >Focused ultrasound (FUS)–mediated blood-brain barrier (BBB) opening with microbubbles is an emerging technology that enables drug delivery for central nervous system diseases. To date, most clinical trials assessing BBB opening in adults were designed to deliver US with a frequency of one treatment over several weeks. Little is known about the feasibility of shorter intervals of US delivery or whether this can be achieved in a pediatric population using a mobile device. Here, FUS and panobinostat were shown to have additive therapeutic effects in a syngeneic orthotopic model of diffuse midline glioma (DMG). We then conducted a single-arm first-in-pediatric trial to investigate the safety and feasibility of delivering neuronavigation-guided FUS treatment in combination with oral panobinostat in children with relapsed DMGs. We included an intrapatient escalation of FUS delivery to assess the feasibility of opening multiple sites in the brain. We demonstrated successful BBB opening using neuronavigation-guided FUS as frequently as every 2 days. Magnetic resonance imaging with contrast was used to identify the region of BBB opening. Three patients were accrued; 22 FUS procedures were delivered for 1 NOTS (number of tumor sites) treated, and four FUS procedures were delivered for 2 NOTS. All three patients received 1 NOTS, without serious adverse events, and two of the patients received 2 NOTS, all without sedation. For 2 NOTS, prolonged BBB opening and one grade 5 event, unlikely related to FUS, were observed. This study demonstrates feasibility of FUS for BBB opening and drug delivery without sedation in pediatric patients.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 824","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.science.org/doi/reader/10.1126/scitranslmed.adq6645","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145492697","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Therapeutic vaccines against cervical cancer caused by human papillomavirus (HPV) are still an unmet medical need, despite a prophylactic HPV vaccine being available, and the now-licensed systemic vaccines may have a limited effect on the reproductive tract. To specifically inhibit cervical cancer development, the concept of mucosal immunity based on the reproductive-respiratory axis was adopted to develop a nasal HPV therapeutic vaccine. We used a cationic nanogel for the nasal vaccine delivery system and targeted HPV16 E7, an oncoprotein in HPV-driven cervical cancer to demonstrate the feasibility of a nasal therapeutic vaccine. The vaccine was combined with cyclic di–adenosine monophosphate as a cell-mediated immunity-inducing adjuvant. Intranasal immunization with the nanogel vaccine induced E7-specific CD4+ and CD8+ T cells in mouse cervicovaginal tissue. An antitumor effect due to the infiltration of vaccine-induced E7-specific T cells was also observed in an orthotopic tumor model in mice. Furthermore, intranasal immunization of nonhuman primates with the nanogel vaccine using a spray device that is also applicable to humans induced E7-specific T cells in the reproductive tissues. Our findings demonstrated that this nasal therapeutic vaccine effectively controlled cervical cancer and will contribute to preclinical evidence for clinical testing in the near future.
{"title":"Cationic nanogel–based nasal therapeutic HPV vaccine prevents the development of cervical cancer","authors":"Rika Nakahashi-Ouchida, Hiromi Mori, Yoshikazu Yuki, Tomonori Machita, Yuko Katakai, Shingo Umemoto, Yohei Uchida, Tomoyuki Yamanoue, Shin-ichi Sawada, Kazuya Ishige, Takashi Miyazaki, Kohtaro Fujihashi, Kazunari Akiyoshi, Yasuhiro Yasutomi, Kei Kawana, Hiroshi Kiyono","doi":"10.1126/scitranslmed.ado8840","DOIUrl":"10.1126/scitranslmed.ado8840","url":null,"abstract":"<div >Therapeutic vaccines against cervical cancer caused by human papillomavirus (HPV) are still an unmet medical need, despite a prophylactic HPV vaccine being available, and the now-licensed systemic vaccines may have a limited effect on the reproductive tract. To specifically inhibit cervical cancer development, the concept of mucosal immunity based on the reproductive-respiratory axis was adopted to develop a nasal HPV therapeutic vaccine. We used a cationic nanogel for the nasal vaccine delivery system and targeted HPV16 E7, an oncoprotein in HPV-driven cervical cancer to demonstrate the feasibility of a nasal therapeutic vaccine. The vaccine was combined with cyclic di–adenosine monophosphate as a cell-mediated immunity-inducing adjuvant. Intranasal immunization with the nanogel vaccine induced E7-specific CD4<sup>+</sup> and CD8<sup>+</sup> T cells in mouse cervicovaginal tissue. An antitumor effect due to the infiltration of vaccine-induced E7-specific T cells was also observed in an orthotopic tumor model in mice. Furthermore, intranasal immunization of nonhuman primates with the nanogel vaccine using a spray device that is also applicable to humans induced E7-specific T cells in the reproductive tissues. Our findings demonstrated that this nasal therapeutic vaccine effectively controlled cervical cancer and will contribute to preclinical evidence for clinical testing in the near future.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 824","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145492699","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rheumatoid arthritis (RA) is a chronic autoimmune condition characterized by aggressive fibroblast-like synoviocytes (FLSs). Small nucleolar RNAs (snoRNAs), traditionally implicated in ribosome biogenesis, are now recognized as disease regulators. However, their involvement in RA-FLSs remains poorly understood. Here, we identified small nucleolar RNA, C/D box 3 (SNORD3), a specific snoRNA up-regulated by tumor necrosis factor–α and interleukin-17, as a key driver of the aggressive transformation of RA-FLSs in vitro. Using an FLS-specific aptamer-functionalized siRNA delivery system, we demonstrated that silencing Snord3 alleviated arthritic symptoms in collagen-induced arthritis (CIA) mice. Transcriptome analyses revealed that SNORD3 up-regulated endothelial cell–specific molecule 1 (ESM1) by modulating the polycomb repressive complex 2 (PRC2)–mediated trimethylation of histone H3 at lysine-27 (H3K27me3), driving the aggressive transformation of RA-FLSs. Mechanistically, we found that SNORD3 physically interacted with enhancer of zeste homolog 2 (EZH2) and competitively disrupted the association of EZH2 with retinoblastoma binding protein 4 within PRC2, thus diminishing the H3K27me3 mark on the ESM1 gene promoter to relieve the transcriptional repression of ESM1. We screened an ESM1-specific aptamer 04 (ESMA04) by systematic evolution of ligands by exponential enrichment, which neutralized ESM1 and inhibited the aggressive transformation of RA-FLSs in vitro. When administered either alone or in combination with a biologic disease-modifying antirheumatic drug, etanercept, ESMA04 demonstrated therapeutic efficacy in CIA mice. Overall, our findings identified SNORD3-EZH2-ESM1 signaling as a driver of RA-FLS pathogenesis and underscored the promise of aptamer-based therapies for RA treatment.
{"title":"snoRNA Snord3 promotes rheumatoid arthritis by epigenetic regulation of ESM1 in fibroblast-like synoviocytes in mice","authors":"Jie Huang, Xuekun Fu, Runrun Zhang, Zhuqian Wang, Fang Qiu, Xinxin Chen, Junyu Fan, Chunhao Cao, Xu Yang, Jie Li, Yiying Liang, Dongyi He, Aiping Lu, Chao Liang","doi":"10.1126/scitranslmed.adt5340","DOIUrl":"10.1126/scitranslmed.adt5340","url":null,"abstract":"<div >Rheumatoid arthritis (RA) is a chronic autoimmune condition characterized by aggressive fibroblast-like synoviocytes (FLSs). Small nucleolar RNAs (snoRNAs), traditionally implicated in ribosome biogenesis, are now recognized as disease regulators. However, their involvement in RA-FLSs remains poorly understood. Here, we identified <i>small nucleolar RNA, C/D box 3</i> (<i>SNORD3</i>), a specific snoRNA up-regulated by tumor necrosis factor–α and interleukin-17, as a key driver of the aggressive transformation of RA-FLSs in vitro. Using an FLS-specific aptamer-functionalized siRNA delivery system, we demonstrated that silencing <i>Snord3</i> alleviated arthritic symptoms in collagen-induced arthritis (CIA) mice. Transcriptome analyses revealed that <i>SNORD3</i> up-regulated endothelial cell–specific molecule 1 (ESM1) by modulating the polycomb repressive complex 2 (PRC2)–mediated trimethylation of histone H3 at lysine-27 (H3K27me3), driving the aggressive transformation of RA-FLSs. Mechanistically, we found that <i>SNORD3</i> physically interacted with enhancer of zeste homolog 2 (EZH2) and competitively disrupted the association of EZH2 with retinoblastoma binding protein 4 within PRC2, thus diminishing the H3K27me3 mark on the <i>ESM1</i> gene promoter to relieve the transcriptional repression of <i>ESM1</i>. We screened an ESM1-specific aptamer 04 (ESMA04) by systematic evolution of ligands by exponential enrichment, which neutralized ESM1 and inhibited the aggressive transformation of RA-FLSs in vitro. When administered either alone or in combination with a biologic disease-modifying antirheumatic drug, etanercept, ESMA04 demonstrated therapeutic efficacy in CIA mice. Overall, our findings identified <i>SNORD3</i>-EZH2-ESM1 signaling as a driver of RA-FLS pathogenesis and underscored the promise of aptamer-based therapies for RA treatment.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 824","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145492695","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-12DOI: 10.1126/scitranslmed.adz2276
Wataru Akahata, Mai Komori, Amber L. Morey, Andrés A. Quiñones-Molina, James B. Hood, Josiane Fofana, Luis Romero, Elizabeth Peters, Jonathan D. Webber, Tyler Meeks, Paulina A. Przygonska, Isabel Steinberg, Ellison Ober, Tae Kim, Daniel C. Rogan, Kenta Matsuda, Jonathan F. Smith, Suryaram Gummuluru, Mark Connors, Hisashi Akiyama
To improve existing synthetic RNA-based vaccines, we previously developed a self-amplifying RNA (saRNA)–based vaccine expressing a membrane-anchored (TM) receptor binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein (S-RBD-TM) and demonstrated that a low dose of this saRNA vaccine elicits robust immune responses. Moreover, a recent clinical trial with an saRNA vaccine incorporating 5-methylcytidine (5mC) (saRNA-5mC) has demonstrated reduced vaccine reactogenicity while maintaining robust humoral responses. In this study, we investigate the mechanisms by which 5mC incorporation attenuates adverse effects while maintaining immunogenicity. We found that incorporation of 5mC into the saRNA platform led to prolonged and robust expression of antigen and attenuated induction of type I interferon, a key driver of reactogenicity, specifically in plasmacytoid dendritic cells (pDCs). As a result, saRNA-5mC alleviated excessive innate immune responses in vivo without impairing B cell and T cell responses against the SARS-CoV-2 RBD. Mechanistically, we demonstrated that the detection of unmodified saRNA in pDCs was mediated by a host cytosolic RNA sensor, RIG-I, and this sensing was abolished with 5mC incorporation. In contrast, saRNA-5mC induced robust innate activation in professional antigen-presenting cells, such as macrophages, in a RIG-I–independent manner, highlighting distinct host sensing mechanisms for synthetic RNAs. Our study provides support for the potential clinical use of saRNA-5mC vaccine platforms.
{"title":"Incorporation of 5-methylcytidine alleviates RIG-I–mediated innate immune responses to a self-amplifying RNA vaccine","authors":"Wataru Akahata, Mai Komori, Amber L. Morey, Andrés A. Quiñones-Molina, James B. Hood, Josiane Fofana, Luis Romero, Elizabeth Peters, Jonathan D. Webber, Tyler Meeks, Paulina A. Przygonska, Isabel Steinberg, Ellison Ober, Tae Kim, Daniel C. Rogan, Kenta Matsuda, Jonathan F. Smith, Suryaram Gummuluru, Mark Connors, Hisashi Akiyama","doi":"10.1126/scitranslmed.adz2276","DOIUrl":"10.1126/scitranslmed.adz2276","url":null,"abstract":"<div >To improve existing synthetic RNA-based vaccines, we previously developed a self-amplifying RNA (saRNA)–based vaccine expressing a membrane-anchored (TM) receptor binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike (S) protein (S-RBD-TM) and demonstrated that a low dose of this saRNA vaccine elicits robust immune responses. Moreover, a recent clinical trial with an saRNA vaccine incorporating 5-methylcytidine (5mC) (saRNA-5mC) has demonstrated reduced vaccine reactogenicity while maintaining robust humoral responses. In this study, we investigate the mechanisms by which 5mC incorporation attenuates adverse effects while maintaining immunogenicity. We found that incorporation of 5mC into the saRNA platform led to prolonged and robust expression of antigen and attenuated induction of type I interferon, a key driver of reactogenicity, specifically in plasmacytoid dendritic cells (pDCs). As a result, saRNA-5mC alleviated excessive innate immune responses in vivo without impairing B cell and T cell responses against the SARS-CoV-2 RBD. Mechanistically, we demonstrated that the detection of unmodified saRNA in pDCs was mediated by a host cytosolic RNA sensor, RIG-I, and this sensing was abolished with 5mC incorporation. In contrast, saRNA-5mC induced robust innate activation in professional antigen-presenting cells, such as macrophages, in a RIG-I–independent manner, highlighting distinct host sensing mechanisms for synthetic RNAs. Our study provides support for the potential clinical use of saRNA-5mC vaccine platforms.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 824","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145492691","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-12DOI: 10.1126/scitranslmed.ady0210
Shady Younis, Salvinaz I. Moutusy, Sajede Rasouli, Shaghayegh Jahanbani, Mahesh Pandit, Xiaohao Wu, Suman Acharya, Orr Sharpe, Tilini U. Wijeratne, Marlayna L. Harris, Emily Y. Yang, Yashaar Chaichian, Shima Parsafar, Matthew C. Baker, John B. Harley, Eric Meffre, Lawrence Steinman, Ann Marshak-Rothstein, Judith A. James, Olivia M. Martinez, Paul J. Utz, Dana E. Orange, Tobias V. Lanz, William H. Robinson
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease characterized by antinuclear antibodies (ANAs). Epstein-Barr virus (EBV) infection has been epidemiologically associated with SLE, yet its role in pathogenesis remains incompletely defined. Here, we developed an EBV-specific single-cell RNA-sequencing platform and used it to demonstrate that EBV infection reprograms autoreactive antinuclear antigen B cells to drive autoimmunity in SLE. We demonstrated that, in SLE, EBV+ B cells are predominantly CD27+CD21low memory B cells that are present at increased frequencies and express ZEB2, TBX21 (T-bet), and antigen-presenting cell transcriptional pathways. Integrative analysis of chromatin immunoprecipitation sequencing (ChIP-seq), assay for transposase-accessible chromatin sequencing (ATAC-seq), and RNA polymerase II occupancy data revealed EBV nuclear antigen 2 (EBNA2) binding at the transcriptional start sites and regulatory regions of CD27, ZEB2, and TBX21, as well as the antigen-presenting cell genes demonstrated to be up-regulated in SLE EBV+ B cells. We expressed recombinant antibodies from SLE EBV+ B cells and demonstrated that they bind prototypical SLE nuclear autoantigens, whereas those from healthy individuals do not. We further found that SLE EBV+ B cells can serve as antigen-presenting cells to drive activation of T peripheral helper cells with concomitant activation of related EBV− antinuclear double-negative 2 B cells and plasmablasts. Our results provide a mechanistic basis for EBV being a driver of SLE through infecting and reprogramming nuclear antigen-reactive B cells to become activated antigen-presenting cells with the potential to promote systemic disease–driving autoimmune responses.
{"title":"Epstein-Barr virus reprograms autoreactive B cells as antigen-presenting cells in systemic lupus erythematosus","authors":"Shady Younis, Salvinaz I. Moutusy, Sajede Rasouli, Shaghayegh Jahanbani, Mahesh Pandit, Xiaohao Wu, Suman Acharya, Orr Sharpe, Tilini U. Wijeratne, Marlayna L. Harris, Emily Y. Yang, Yashaar Chaichian, Shima Parsafar, Matthew C. Baker, John B. Harley, Eric Meffre, Lawrence Steinman, Ann Marshak-Rothstein, Judith A. James, Olivia M. Martinez, Paul J. Utz, Dana E. Orange, Tobias V. Lanz, William H. Robinson","doi":"10.1126/scitranslmed.ady0210","DOIUrl":"10.1126/scitranslmed.ady0210","url":null,"abstract":"<div >Systemic lupus erythematosus (SLE) is a systemic autoimmune disease characterized by antinuclear antibodies (ANAs). Epstein-Barr virus (EBV) infection has been epidemiologically associated with SLE, yet its role in pathogenesis remains incompletely defined. Here, we developed an EBV-specific single-cell RNA-sequencing platform and used it to demonstrate that EBV infection reprograms autoreactive antinuclear antigen B cells to drive autoimmunity in SLE. We demonstrated that, in SLE, EBV<sup>+</sup> B cells are predominantly CD27<sup>+</sup>CD21<sup>low</sup> memory B cells that are present at increased frequencies and express <i>ZEB2</i>, <i>TBX21</i> (T-bet), and antigen-presenting cell transcriptional pathways. Integrative analysis of chromatin immunoprecipitation sequencing (ChIP-seq), assay for transposase-accessible chromatin sequencing (ATAC-seq), and RNA polymerase II occupancy data revealed EBV nuclear antigen 2 (EBNA2) binding at the transcriptional start sites and regulatory regions of <i>CD27</i>, <i>ZEB2</i>, and <i>TBX21</i>, as well as the antigen-presenting cell genes demonstrated to be up-regulated in SLE EBV<sup>+</sup> B cells. We expressed recombinant antibodies from SLE EBV<sup>+</sup> B cells and demonstrated that they bind prototypical SLE nuclear autoantigens, whereas those from healthy individuals do not. We further found that SLE EBV<sup>+</sup> B cells can serve as antigen-presenting cells to drive activation of T peripheral helper cells with concomitant activation of related EBV<sup>−</sup> antinuclear double-negative 2 B cells and plasmablasts. Our results provide a mechanistic basis for EBV being a driver of SLE through infecting and reprogramming nuclear antigen-reactive B cells to become activated antigen-presenting cells with the potential to promote systemic disease–driving autoimmune responses.</div>","PeriodicalId":21580,"journal":{"name":"Science Translational Medicine","volume":"17 824","pages":""},"PeriodicalIF":14.6,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145492694","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}