1
|
Huang Q, Chan KY, Wu J, Botticello-Romero NR, Zheng Q, Lou S, Keyes C, Svanbergsson A, Johnston J, Mills A, Lin CY, Brauer PP, Clouse G, Pacouret S, Harvey JW, Beddow T, Hurley JK, Tobey IG, Powell M, Chen AT, Barry AJ, Eid FE, Chan YA, Deverman BE. An AAV capsid reprogrammed to bind human transferrin receptor mediates brain-wide gene delivery. Science 2024; 384:1220-1227. [PMID: 38753766 DOI: 10.1126/science.adm8386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/08/2024] [Indexed: 05/18/2024]
Abstract
Developing vehicles that efficiently deliver genes throughout the human central nervous system (CNS) will broaden the range of treatable genetic diseases. We engineered an adeno-associated virus (AAV) capsid, BI-hTFR1, that binds human transferrin receptor (TfR1), a protein expressed on the blood-brain barrier. BI-hTFR1 was actively transported across human brain endothelial cells and, relative to AAV9, provided 40 to 50 times greater reporter expression in the CNS of human TFRC knockin mice. The enhanced tropism was CNS-specific and absent in wild-type mice. When used to deliver GBA1, mutations of which cause Gaucher disease and are linked to Parkinson's disease, BI-hTFR1 substantially increased brain and cerebrospinal fluid glucocerebrosidase activity compared with AAV9. These findings establish BI-hTFR1 as a potential vector for human CNS gene therapy.
Collapse
Affiliation(s)
- Qin Huang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Ken Y Chan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Jason Wu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Nuria R Botticello-Romero
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Qingxia Zheng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Shan Lou
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Casey Keyes
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Alexander Svanbergsson
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Jencilin Johnston
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Allan Mills
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Chin-Yen Lin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Pamela P Brauer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Gabrielle Clouse
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Simon Pacouret
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - John W Harvey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Thomas Beddow
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Jenna K Hurley
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Isabelle G Tobey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Megan Powell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Albert T Chen
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Andrew J Barry
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Fatma-Elzahraa Eid
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
- Department of Systems and Computer Engineering, Faculty of Engineering, Al-Azhar University, Cairo 11651, Egypt
| | - Yujia A Chan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Benjamin E Deverman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| |
Collapse
|
2
|
Shen Y, Zhou R, Bi L, Huang G, Yang M, Li Z, Yao J, Xian J, Qiu Y, Ye P, Liu Y, Hou Y, Jin H, Wang Y. Synthesis and Evaluation of [ 64Cu]Cu-NOTA-HFn for PET Imaging of Transferrin Receptor 1 Expression in Nasopharyngeal Carcinoma. ACS OMEGA 2024; 9:17423-17431. [PMID: 38645324 PMCID: PMC11024937 DOI: 10.1021/acsomega.4c00187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/29/2024] [Accepted: 03/20/2024] [Indexed: 04/23/2024]
Abstract
As recurrent and metastatic nasopharyngeal carcinoma (NPC) is the most common cause of death among patients with NPC, there is an urgent clinical need for the development of precision diagnosis to guide personalized treatment. Recent emerging evidence substantiates the increased expression of transferrin receptor 1 (also known as cluster of differentiation 71, CD71) within tumor tissues and the inherent targeting capability of natural heavy-chain ferritin (HFn) toward CD71. This study aimed to synthesize and assess a radiotracer ([64Cu]Cu-NOTA-HFn) designed to target CD71 for positron emission tomography (PET) imaging in an NPC tumor-bearing mouse model. The entire radiolabeling process of [64Cu]Cu-NOTA-HFn was completed within 15 min with high yield (>98.5%) and high molar activity (72.96 ± 21.33 GBq/μmol). The in vitro solubility and stability experiments indicated that [64Cu]Cu-NOTA-HFn had a high water solubility (log P = -2.42 ± 0.52, n = 6) and good stability in phosphate-buffered saline (PBS) for up to 48 h. The cell saturation binding assay indicated that [64Cu]Cu-NOTA-HFn had a nanomolar affinity (Kd = 10.9 ± 6.1 nM) for CD71-overexpressing C666-1 cells. To test the target engagement in vivo, prolonged-time PET imaging was performed at 1, 6, 12, 24, and 36 h postinjection (p.i.) of [64Cu]Cu-NOTA-HFn to C666-1 NPC tumor-bearing mice. The C666-1 tumors could be visualized by [64Cu]Cu-NOTA-HFn and blocked by nonradiolabeled HFn. PET imaging quantitative analysis demonstrated that the uptake of [64Cu]Cu-NOTA-HFn in C666-1 tumors peaked at 6 h p.i. and the best radioactive tumor-to-muscle ratio was 10.53 ± 3.11 (n = 3). Ex vivo biodistribution assay at 6 h p.i. showed that the tumor uptakes were 1.43 ± 0.23%ID/g in the nonblock group and 0.92 ± 0.2%ID/g in the block group (n = 3, p < 0.05). Immunohistochemistry and immunofluorescence staining confirmed positive expression of CD71 and the uptake of HFn in C666-1 tumor tissues. In conclusion, our experiments demonstrated that [64Cu]Cu-NOTA-HFn possesses a very high target engagement for CD71-positive NPC tumors and provided a fundamental basis for further clinical translation.
Collapse
Affiliation(s)
- Yanfang Shen
- Department
of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong-Hong
Kong-Macao University Joint Laboratory of Interventional Medicine,
The Fifth Affiliated Hospital, Sun Yat-sen
University, Zhuhai 519000, China
| | - Renwei Zhou
- Department
of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong-Hong
Kong-Macao University Joint Laboratory of Interventional Medicine,
The Fifth Affiliated Hospital, Sun Yat-sen
University, Zhuhai 519000, China
| | - Lei Bi
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong-Hong
Kong-Macao University Joint Laboratory of Interventional Medicine,
The Fifth Affiliated Hospital, Sun Yat-sen
University, Zhuhai 519000, China
| | - Guolong Huang
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong-Hong
Kong-Macao University Joint Laboratory of Interventional Medicine,
The Fifth Affiliated Hospital, Sun Yat-sen
University, Zhuhai 519000, China
| | - Min Yang
- Department
of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong-Hong
Kong-Macao University Joint Laboratory of Interventional Medicine,
The Fifth Affiliated Hospital, Sun Yat-sen
University, Zhuhai 519000, China
| | - Zhijun Li
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong-Hong
Kong-Macao University Joint Laboratory of Interventional Medicine,
The Fifth Affiliated Hospital, Sun Yat-sen
University, Zhuhai 519000, China
| | - Jijin Yao
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong-Hong
Kong-Macao University Joint Laboratory of Interventional Medicine,
The Fifth Affiliated Hospital, Sun Yat-sen
University, Zhuhai 519000, China
| | - Jianzhong Xian
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong-Hong
Kong-Macao University Joint Laboratory of Interventional Medicine,
The Fifth Affiliated Hospital, Sun Yat-sen
University, Zhuhai 519000, China
| | - Yifan Qiu
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong-Hong
Kong-Macao University Joint Laboratory of Interventional Medicine,
The Fifth Affiliated Hospital, Sun Yat-sen
University, Zhuhai 519000, China
| | - Peizhen Ye
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong-Hong
Kong-Macao University Joint Laboratory of Interventional Medicine,
The Fifth Affiliated Hospital, Sun Yat-sen
University, Zhuhai 519000, China
| | - Yongshan Liu
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong-Hong
Kong-Macao University Joint Laboratory of Interventional Medicine,
The Fifth Affiliated Hospital, Sun Yat-sen
University, Zhuhai 519000, China
| | - Yuyi Hou
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong-Hong
Kong-Macao University Joint Laboratory of Interventional Medicine,
The Fifth Affiliated Hospital, Sun Yat-sen
University, Zhuhai 519000, China
| | - Hongjun Jin
- Guangdong
Provincial Engineering Research Center of Molecular Imaging, The Fifth
Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
- Guangdong-Hong
Kong-Macao University Joint Laboratory of Interventional Medicine,
The Fifth Affiliated Hospital, Sun Yat-sen
University, Zhuhai 519000, China
| | - Ying Wang
- Department
of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China
| |
Collapse
|
3
|
Schmitt J, Poole E, Groves I, Owen DJ, Graham SC, Sinclair J, Kelly BT. Repurposing an endogenous degradation domain for antibody-mediated disposal of cell-surface proteins. EMBO Rep 2024; 25:951-970. [PMID: 38287192 PMCID: PMC10933360 DOI: 10.1038/s44319-024-00063-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 01/31/2024] Open
Abstract
The exquisite specificity of antibodies can be harnessed to effect targeted degradation of membrane proteins. Here, we demonstrate targeted protein removal utilising a protein degradation domain derived from the endogenous human protein Proprotein Convertase Subtilisin/Kexin type 9 (PCSK9). Recombinant antibodies genetically fused to this domain drive the degradation of membrane proteins that undergo constitutive internalisation and recycling, including the transferrin receptor and the human cytomegalovirus latency-associated protein US28. We term this approach PACTAC (PCSK9-Antibody Clearance-Targeting Chimeras).
Collapse
Affiliation(s)
- Janika Schmitt
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Hills Road, CB2 0SP, Cambridge, UK
- Faculty of Medicine, Charité Berlin, 10117, Berlin, Germany
- Faculty of Medicine, University of Heidelberg, 69210, Heidelberg, Germany
| | - Emma Poole
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Hills Road, CB2 0SP, Cambridge, UK
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Ian Groves
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Hills Road, CB2 0SP, Cambridge, UK
- Infection Biology, Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - David J Owen
- Cambridge Institute for Medical Research, Keith Peters Building, Hills Road, Cambridge, CB2 0XY, UK.
| | - Stephen C Graham
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK.
| | - John Sinclair
- Department of Medicine, Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Hills Road, CB2 0SP, Cambridge, UK.
| | - Bernard T Kelly
- Cambridge Institute for Medical Research, Keith Peters Building, Hills Road, Cambridge, CB2 0XY, UK.
| |
Collapse
|
4
|
Huang Q, Chan KY, Lou S, Keyes C, Wu J, Botticello-Romero NR, Zheng Q, Johnston J, Mills A, Brauer PP, Clouse G, Pacouret S, Harvey JW, Beddow T, Hurley JK, Tobey IG, Powell M, Chen AT, Barry AJ, Eid FE, Chan YA, Deverman BE. An AAV capsid reprogrammed to bind human Transferrin Receptor mediates brain-wide gene delivery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572615. [PMID: 38187643 PMCID: PMC10769326 DOI: 10.1101/2023.12.20.572615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Developing vehicles that efficiently deliver genes throughout the human central nervous system (CNS) will broaden the range of treatable genetic diseases. We engineered an AAV capsid, BI-hTFR1, that binds human Transferrin Receptor (TfR1), a protein expressed on the blood-brain barrier (BBB). BI-hTFR1 was actively transported across a human brain endothelial cell layer and, relative to AAV9, provided 40-50 times greater reporter expression in the CNS of human TFRC knock-in mice. The enhanced tropism was CNS-specific and absent in wild type mice. When used to deliver GBA1, mutations of which cause Gaucher disease and are linked to Parkinson's disease, BI-hTFR1 substantially increased brain and cerebrospinal fluid glucocerebrosidase activity compared to AAV9. These findings establish BI-hTFR1 as a promising vector for human CNS gene therapy.
Collapse
Affiliation(s)
- Qin Huang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Ken Y. Chan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Shan Lou
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Casey Keyes
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Jason Wu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | | | - Qingxia Zheng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Jencilin Johnston
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Allan Mills
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Pamela P. Brauer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Gabrielle Clouse
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Simon Pacouret
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - John W. Harvey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Thomas Beddow
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Jenna K. Hurley
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Isabelle G. Tobey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Megan Powell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Albert T. Chen
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Andrew J. Barry
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Fatma-Elzahraa Eid
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
- Department of Systems and Computer Engineering, Faculty of Engineering, Al-Azhar University; Cairo, Egypt
| | - Yujia A. Chan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| | - Benjamin E. Deverman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard; Cambridge, USA
| |
Collapse
|
5
|
Westover JB, Naik S, Bailey KW, Wandersee L, Gantla VR, Hickerson BT, McCormack K, Henkel G, Gowen BB. Severe mammarenaviral disease in guinea pigs effectively treated by an orally bioavailable fusion inhibitor, alone or in combination with favipiravir. Antiviral Res 2022; 208:105444. [PMID: 36243175 PMCID: PMC10187609 DOI: 10.1016/j.antiviral.2022.105444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022]
Abstract
Infections by pathogenic New World mammarenaviruses (NWM)s, including Junín virus (JUNV), can result in a severe life-threatening viral hemorrhagic fever syndrome. In the absence of FDA-licensed vaccines or antivirals, these viruses are considered high priority pathogens. The mammarenavirus envelope glycoprotein complex (GPC) mediates pH-dependent fusion between viral and cellular membranes, which is essential to viral entry and may be vulnerable to small-molecule inhibitors that disrupt this process. ARN-75039 is a potent fusion inhibitor of a broad spectrum of pseudotyped and native mammarenaviruses in cell culture and Tacaribe virus infection in mice. In the present study, we evaluated ARN-75039 against pathogenic JUNV in the rigorous guinea pig infection model. The compound was well-tolerated and had favorable pharmacokinetics supporting once-per-day oral dosing in guinea pigs. Importantly, significant protection against JUNV challenge was observed even when ARN-75039 was withheld until 6 days after the viral challenge when clinical signs of disease are starting to develop. We also show that ARN-75039 combination treatment with favipiravir, a viral polymerase inhibitor, results in synergistic activity in vitro and improves survival outcomes in JUNV-challenged guinea pigs. Our findings support the continued development of ARN-75039 as an attractive therapeutic candidate for treating mammarenaviral hemorrhagic fevers, including those associated with NWM infection.
Collapse
Affiliation(s)
- Jonna B Westover
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA
| | | | - Kevin W Bailey
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Luci Wandersee
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA
| | | | - Brady T Hickerson
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA
| | | | | | - Brian B Gowen
- Institute for Antiviral Research, Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA.
| |
Collapse
|
6
|
Gallo GL, López N, Loureiro ME. The Virus–Host Interplay in Junín Mammarenavirus Infection. Viruses 2022; 14:v14061134. [PMID: 35746604 PMCID: PMC9228484 DOI: 10.3390/v14061134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 02/06/2023] Open
Abstract
Junín virus (JUNV) belongs to the Arenaviridae family and is the causative agent of Argentine hemorrhagic fever (AHF), a severe human disease endemic to agricultural areas in Argentina. At this moment, there are no effective antiviral therapeutics to battle pathogenic arenaviruses. Cumulative reports from recent years have widely provided information on cellular factors playing key roles during JUNV infection. In this review, we summarize research on host molecular determinants that intervene in the different stages of the viral life cycle: viral entry, replication, assembly and budding. Alongside, we describe JUNV tight interplay with the innate immune system. We also review the development of different reverse genetics systems and their use as tools to study JUNV biology and its close teamwork with the host. Elucidating relevant interactions of the virus with the host cell machinery is highly necessary to better understand the mechanistic basis beyond virus multiplication, disease pathogenesis and viral subversion of the immune response. Altogether, this knowledge becomes essential for identifying potential targets for the rational design of novel antiviral treatments to combat JUNV as well as other pathogenic arenaviruses.
Collapse
|
7
|
Hickerson BT, Daniels-Wells TR, Payes C, Clark LE, Candelaria PV, Bailey KW, Sefing EJ, Zink S, Ziegenbein J, Abraham J, Helguera G, Penichet ML, Gowen BB. Host receptor-targeted therapeutic approach to counter pathogenic New World mammarenavirus infections. Nat Commun 2022; 13:558. [PMID: 35091550 PMCID: PMC8799657 DOI: 10.1038/s41467-021-27949-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 12/22/2021] [Indexed: 12/25/2022] Open
Abstract
Five New World mammarenaviruses (NWMs) cause life-threatening hemorrhagic fever (HF). Cellular entry by these viruses is mediated by human transferrin receptor 1 (hTfR1). Here, we demonstrate that an antibody (ch128.1/IgG1) which binds the apical domain of hTfR1, potently inhibits infection of attenuated and pathogenic NWMs in vitro. Computational docking of the antibody Fab crystal structure onto the known structure of hTfR1 shows an overlapping receptor-binding region shared by the Fab and the viral envelope glycoprotein GP1 subunit that binds hTfR1, and we demonstrate competitive inhibition of NWM GP1 binding by ch128.1/IgG1 as the principal mechanism of action. Importantly, ch128.1/IgG1 protects hTfR1-expressing transgenic mice against lethal NWM challenge. Additionally, the antibody is well-tolerated and only partially reduces ferritin uptake. Our findings provide the basis for the development of a novel, host receptor-targeted antibody therapeutic broadly applicable to the treatment of HF of NWM etiology.
Collapse
MESH Headings
- A549 Cells
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacology
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Arenaviridae/drug effects
- Arenaviridae/metabolism
- Arenaviridae/physiology
- Chlorocebus aethiops
- Hemorrhagic Fever, American/metabolism
- Hemorrhagic Fever, American/prevention & control
- Hemorrhagic Fever, American/virology
- Host-Pathogen Interactions/drug effects
- Humans
- Junin virus/drug effects
- Junin virus/physiology
- Mice, Inbred C57BL
- Mice, Transgenic
- Molecular Docking Simulation
- Protein Binding/drug effects
- Receptors, Transferrin/antagonists & inhibitors
- Receptors, Transferrin/immunology
- Receptors, Transferrin/metabolism
- Vero Cells
- Viral Envelope Proteins/metabolism
- Mice
Collapse
Affiliation(s)
- Brady T Hickerson
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA
- Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Tracy R Daniels-Wells
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Cristian Payes
- Instituto de Biología y Medicina Experimental (IBYME CONICET), Buenos Aires, Argentina
| | - Lars E Clark
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Pierre V Candelaria
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kevin W Bailey
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Eric J Sefing
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Samantha Zink
- Department of Chemistry and Biochemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - James Ziegenbein
- Department of Chemistry and Biochemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Jonathan Abraham
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Gustavo Helguera
- Instituto de Biología y Medicina Experimental (IBYME CONICET), Buenos Aires, Argentina.
- Department of Chemistry and Biochemistry, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.
| | - Manuel L Penichet
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- UCLA Molecular Biology Institute, Los Angeles, CA, USA.
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA.
- UCLA AIDS Institute, Los Angeles, CA, USA.
| | - Brian B Gowen
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA.
| |
Collapse
|
8
|
Roman-Sosa G, Leske A, Ficht X, Dau TH, Holzerland J, Hoenen T, Beer M, Kammerer R, Schirmbeck R, Rey FA, Cordo SM, Groseth A. Immunization with GP1 but Not Core-like Particles Displaying Isolated Receptor-Binding Epitopes Elicits Virus-Neutralizing Antibodies against Junín Virus. Vaccines (Basel) 2022; 10:vaccines10020173. [PMID: 35214632 PMCID: PMC8874384 DOI: 10.3390/vaccines10020173] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/01/2023] Open
Abstract
New World arenaviruses are rodent-transmitted viruses and include a number of pathogens that are responsible for causing severe human disease. This includes Junín virus (JUNV), which is the causative agent of Argentine hemorrhagic fever. The wild nature and mobility of the rodent reservoir host makes it difficult to control the disease, and currently passive immunization with high-titer neutralizing antibody-containing plasma from convalescent patients is the only specific therapy. However, dwindling supplies of naturally available convalescent plasma, and challenges in developing similar resources for other closely related viruses, have made the development of alternative antibody-based therapeutic approaches of critical importance. In this study, we sought to induce a neutralizing antibody response in rabbits against the receptor-binding subunit of the viral glycoprotein, GP1, and the specific peptide sequences in GP1 involved in cellular receptor contacts. While these specific receptor-interacting peptides did not efficiently induce the production of neutralizing antibodies when delivered as a particulate antigen (as part of hepatitis B virus core-like particles), we showed that recombinant JUNV GP1 purified from transfected mammalian cells induced virus-neutralizing antibodies at high titers in rabbits. Further, neutralization was observed across a range of unrelated JUNV strains, a feature that is critical for effectiveness in the field. These results underscore the potential of GP1 alone to induce a potent neutralizing antibody response and highlight the importance of epitope presentation. In addition, effective virus neutralization by rabbit antibodies supports the potential applicability of this species for the future development of immunotherapeutics (e.g., based on humanized monoclonal antibodies). Such information can be applied in the design of vaccines and immunogens for both prevention and specific therapies against this and likely also other closely related pathogenic New World arenaviruses.
Collapse
Affiliation(s)
- Gleyder Roman-Sosa
- Department of Internal Medicine I, Ulm University Hospital, 89081 Ulm, Germany; (X.F.); (R.S.)
- Correspondence: (G.R.-S.); (A.G.)
| | - Anne Leske
- Laboratory for Arenavirus Biology, Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany; (A.L.); (J.H.)
| | - Xenia Ficht
- Department of Internal Medicine I, Ulm University Hospital, 89081 Ulm, Germany; (X.F.); (R.S.)
| | - Tung Huy Dau
- Laboratory for Immunogenetics, Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany; (T.H.D.); (R.K.)
| | - Julia Holzerland
- Laboratory for Arenavirus Biology, Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany; (A.L.); (J.H.)
| | - Thomas Hoenen
- Laboratory for Integrative Cell and Infection Biology, Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany;
| | - Martin Beer
- National and OIE Reference Laboratory for BHV-1, Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany;
| | - Robert Kammerer
- Laboratory for Immunogenetics, Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany; (T.H.D.); (R.K.)
| | - Reinhold Schirmbeck
- Department of Internal Medicine I, Ulm University Hospital, 89081 Ulm, Germany; (X.F.); (R.S.)
| | - Felix A. Rey
- Structural Virology Unit, CNRS UMR3569, Institut Pasteur, Université de Paris, 75015 Paris, France;
| | - Sandra M. Cordo
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), University of Buenos Aires, Ciudad Universitaria, Pabellón II, Piso 4, Buenos Aires 1428, Argentina;
| | - Allison Groseth
- Laboratory for Arenavirus Biology, Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany; (A.L.); (J.H.)
- Correspondence: (G.R.-S.); (A.G.)
| |
Collapse
|
9
|
Lee M, Koma T, Iwasaki M, Urata S. [South American Hemorrhagic Fever viruses and the cutting edge of the vaccine and antiviral development]. Uirusu 2022; 72:7-18. [PMID: 37899233 DOI: 10.2222/jsv.72.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
South American Hemorrhagic Fever is caused by the Arenavirus, which belong to the Family Arenaviridae, genus mammarenavirus, infection at South America. South American Hemorrhagic Fever includes 1. Argentinian Hemorrhagic fever caused by Junin virus, 2. Brazilian hemorrhagic fever caused by Sabia virus, 3. Venezuelan Hemorrhagic fever caused by Guanarito virus, 4. Bolivian Hemorrhagic fever caused by Machupo virus, and 5. Unassigned hemorrhagic fever caused by Chapare virus. These viruses are classified in New World (NW) Arenavirus, which is different from Old World Arenavirus (ex. Lassa virus), based on phylogeny, serology, and geographic differences. In this review, the current knowledge of the biology and the development of the vaccines and antivirals of NW Arenaviruses which cause South American Hemorrhagic Fever will be described.
Collapse
Affiliation(s)
- Meion Lee
- National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University
- Department of Cell Regulation, Graduate School of Biomedical Sciences, Nagasaki University
| | - Takaaki Koma
- Department of Microbiology, Graduate School of Medicine, Tokushima University
| | - Masaharu Iwasaki
- Laboratory of Emerging Viral Diseases, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University
| | - Shuzo Urata
- National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University
| |
Collapse
|