Virological Characterization of a New Isolated Strain of Andes Virus Involved in the Recent Person-to-Person Transmission Outbreak Reported in Argentina ======================================================================================================================================================== * Rocío M. Coelho * Sebastian Kehl * Natalia Periolo * Emiliano Biondo * Daniel O. Alonso * Celeste Perez * Darío Fernández Do Porto * Gustavo Palacios * Alexis Edelstein * Carla Bellomo * Valeria P. Martinez ## Summary On November 2018, a person-to-person transmission outbreak of Andes virus (ANDV) began in the small town of Epuyen, Argentina. The strain involved demonstrated a high capacity for sustained transmission among the human population requiring the implementation of quarantine measures, rigorous contact tracing, isolation of close contacts, and active clinical monitoring to prevent further spread. In this study, we report the isolation of this strain, which we name the ARG-Epuyen after just three passages in cell culture. Complete sequencing revealed only a single amino acid change post-isolation, revealing that it is a non-adapted, wild-type ANDV strain, and its isolation probably represents a critical step toward the development of medical countermeasures against this emerging pathogen. The pathogenicity and transmissibility potential of ARG-Epuyen were evaluated in hamsters, the only animal model for Hantavirus Pulmonary Syndrome. Additionally, this strain was compared with ARG, an ANDV strain previously isolated from the same geographical area in the Argentinian Patagonia, from a rodent specimen. Our findings revealed high infectiousness and efficient hamster-to-hamster transmission through direct contact experiments, although ARG-Epuyen appeared to be less pathogenic than ARG. Keywords * Hantavirus * Andes virus * Hantavirus pulmonary syndrome * Infection diseases * culture isolation * Animal model * Hamster * person-to-person transmission ## 1. Introduction Hantaviruses (*Bunyavirales: Hantaviridae: Mammantavirinae*) are enveloped, single stranded, negative sense RNA viruses with three-segmented genome. The genomic segments consist of a small segment (S), a medium segment (M), and a large segment (L), which encode the nucleocapsid (N) protein, a nonstructural protein (NSs) in some species, surface glycoproteins (Gn and Gc), and an RNA-dependent RNA polymerase (RdRp), respectively (*1*). Hantaviruses are distributed worldwide and are hosted by various vertebrate animal species. Pathogenic hantaviruses are primarily associated with rodents as natural reservoirs and are classified under the genus *Orthohantavirus*. These viruses establish seemingly asymptomatic and chronic infections in several rodent species. The risks of viral spillover have increased due to new farming practices, climate change, the expansion of rural human settlements, and disruptions to the zoonotic interface. Additionally, rural tourism has led to travel-related cases (*2–4*). Several species of orthohantaviruses are responsible for Hantavirus Pulmonary Syndrome (HPS) in the Americas and Hemorrhagic Fever with Renal Syndrome (HFRS) in Asia and Europe. HPS, first described in 1993 in the US (*5*), is caused by at least 24 distinct viruses (*6*). Humans generally become infected through the inhalation of aerosolized rodent excreta. In Argentina, most HPS cases are caused by seven viruses closely related to Andes virus (ANDV), species *Orthohantavirus andesense*. ANDV was the first hantavirus characterised in Argentina (*7*) and it was associated with the long-tailed pygmy rice rat *Oligoryzomys longicaudatus* in the Patagonian Andean region. The signs and symptoms of the disease can manifest after a long period of up to 40 days after infection (*8,9*). ANDV-HPS is associated with high case-lethality rates ranging from 21–50% (*10,11*). Before 1996, the route of orthohantavirus transmission was considered strictly zoonotic, resulting in “Dead-End” human infections (*7*). However, in 1996, an ANDV-caused HPS outbreak began in the small city of El Bolsón and then expanded to distant cities, such as Bariloche (121 km) and Buenos Aires (1700 km), involving 16 epidemiologically linked cases. This outbreak became a focal point for orthohantavirus research because molecular and epidemiological evidence suggested person-to-person (PTP) transmission (*12,13*). A larger PTP transmission outbreak that began in 2018 and involved 34 cases, could be curtailed by the implementation of strict quarantine measures. In this outbreak, several individuals were identified as superspreaders, predicting the high transmission potential of this strain (*10*). To date, no vaccines or therapeutic treatments are available for any HPS agent, therefore, there is an urgent need for vaccine candidates and/or therapeutics. The paucity of vaccine developments relayed for many years on the lack of animal models, due to their zoonotic origin, and on the difficulty to isolate New World Hantaviruses in cell culture. Only three strains of ANDV had been propagated in cell culture (CH-9717869; CH-7913 and ARG). CHI-7913 was obtained from a serum sample of a Chilean patient before the onset of symptoms (14). CH-9717869 and ARG were isolated from long tailed pygmy rice rats captured in Chile and Argentina respectively (15,16). It was first described that CH-9717869, and later ARG, caused a lethal and very similar disease to HPS in Syrian Golden Hamsters (17,18) and, since then, the model ANDV/hamster became a unique resource for the study of HPS. Most recently, the human CHI-7913 strain resulted in an asymptomatic infection in the same animal model suggesting that the lethal ANDV model of HPS is strain specific, reinforcing the necessity to obtain and characterise new human strains of ANDV to understand their pathogenesis in humans (*19*). In this work we report the isolation in cell culture of the strain responsible for the largest ANDV PTP-transmission outbreak ever reported, ARG-Epuyen. In addition, we performed a preliminary virological characterization of this strain proving its ability to infect hamsters causing high levels of viremia and to spread efficiently between them through direct contact experiments. ## 2. Materials and methods ### 2.1. Virus isolation on cell culture Biological samples involved in this study were obtained during the Ministry of Health activities for the epidemiological investigation of the PTP outbreak form November 2018 to March 2019. The specimens were originally obtained from living individuals in a form that neither the recipient nor the provider can link the biospecimens directly to identifiable private information of the original source. Unidentified remnant biological specimens were utilized for virus isolation. The samples with higher viral load and absence of antibodies were selected. Complete genome sequencing revealed 100% nucleotide identity with cases involved in the PTP transmission outbreak (*8*). The selected serum samples were inoculated onto Vero E6 cell monolayer (CRL-1586; ATCC, Manassas, VA, USA) and incubated for one hour at 37 °C in a humidified atmosphere containing 5% CO2. After the incubation, fresh complete medium (MEM, 10% FBS, antibiotics, and antimycotics) was added to each flask, and incubated at 37 °C under the same conditions. After 15 days, the cells were trypsinized, washed, and seeded onto a new monolayer, and incubated under identical conditions (first blind passage). For each flask, one-third of the cells were stored at -80 °C, another third was tested by indirect immunofluorescence assay (IFA), and the final third was seeded onto a new monolayer. The medium was centrifuged, aliquoted, and stored at -80 °C. Three blind passages were performed in total. Each passage was monitored by real-time RT-PCR for ANDV RNA in the culture media and by IFA for detecting viral antigen in the cells. Mock-infected Vero E6 cells, treated under identical conditions, were used as a negative control. ### 2.2. Immunofluorescence Assay Infected and mock-infected cells were resuspended in 1X phosphate-buffered saline (PBS) and washed three times by centrifugation at 400 x g for five minutes at 4 °C. The pellets were then resuspended in 1 ml PBS, and several 10 µl drops of each suspension were spotted onto a slide. The slides were left inside a laminar flow cabinet until completely dried. The slides were then immersed in ice-cold acetone for 10 minutes at -20 °C, removed from the acetone, allowed to dry, and stored at -20 °C until further processing. The slides were blocked with 3% horse serum in PBS-Triton for 15 minutes at room temperature and then washed with PBS. The cells were treated with a 1:500 dilution of rabbit polyclonal serum against ANDV in PBS-Triton-BSA and incubated for one hour at 37 °C in a humid chamber. After washing, the slides were incubated with FITC-conjugated anti-rabbit IgG (Kirkegaard & Perry) in PBS-Triton-BSA under the same conditions. Finally, the cells were incubated with 1 µg/ml DAPI in PBS for 15 minutes at room temperature and then washed. The slides were allowed to drain, mounted, and analyzed under a fluorescent microscope. ### 2.3. Virus infection in Vero E6 and A549 cell lines Vero E6 and A549 (human epithelial lung cell, ATCC CCL-185) cells were maintained in Dulbecco’s Modified Eagle’s Medium (DMEM high glucose; Sigma) supplemented with 5% fetal bovine serum (FBS, Gibco), 10 mM HEPES buffer, and 2 mM L-glutamine. The cells were infected with ARG and ARG-Epuyen at the indicated multiplicity of infection (MOI) for 60 minutes at 37 °C. After incubation, the monolayers were washed three times with PBS, and the cells were maintained in complete DMEM (Sigma). Supernatants were collected at various days post-infection, centrifuged to remove cells, and stored at -80°C until use. Viral RNA was isolated using the Qiagen QIAamp® Viral RNA kit according to the manufacturer’s protocol. Viral growth kinetics were determined by quantifying ANDV RNA at different days post-infection using real-time RT-PCR as described below. ### 2.4. Evaluation of ARG-Epuyen virulence in the hamster model To evaluate whether ANDV could be transmitted between hamsters in a manner similar to human transmission, Syrian Golden Hamsters were infected with either the ARG-Epuyen or ARG strains. Seventeen 9-week-old hamsters were distributed across seven ventilated cages. One hamster per cage was infected via intramuscular injection (rear thigh) with 100 µl (1 × 104 FFU) of ARG-Epuyen passage No. 4 (two males and four females) or ARG passage No. 19 (one male). The infected hamsters were placed in direct contact with one or two non-infected individuals in the same ventilated cages on the day they were inoculated (day 0). As a control group, four hamsters were inoculated with cell culture supernatant from non-infected Vero E6 cells. All individuals were monitored daily for 30 days for signs of disease, including fatigue, inappetence, lethargy (reluctance to move), and/or dyspnea. Animals with fatal outcomes were necropsied on the day of death to collect lung samples, which were stored at -80 °C until processing. On day 30, all survivors were anesthetized by inhalation of isoflurane, terminally bled by cardiac puncture, and necropsied to obtain lung tissue. ### 2.5. Enzyme Linked Immunosorbent Assay The detection of IgG antibodies against the viral nucleoprotein (NP) in hamsters was performed by ELISA, as previously described, to confirm infection (*18*). Briefly, serial dilutions of hamster sera were incubated in polystyrene plates coated with recombinant ANDV-NP and a nonspecific recombinant protein in a humid chamber at 37 °C for 1 hour. After washing, peroxidase-labelled goat anti-hamster IgG (H+L) (Kirkegaard & Perry) was added and incubated under the same conditions. TMB solution was used as a substrate, and optical density (OD) was measured at 450 nm. ΔOD values were calculated by subtracting the OD measured for each sample incubated with the specific (ANDV-NP) and nonspecific recombinant proteins. Samples were considered IgG positive if ΔOD values were greater than 0.4. The IgG titre was calculated as the inverse of the highest dilution that yielded a positive result. ### 2.6. Viral RNA detection and genomic sequencing Total RNA was extracted from lung samples and from the culture medium of infected cell passages using Trizol (Invitrogen) according to the manufacturer’s protocol. RT-qPCR was performed as previously described (20). Briefly, each RNA sample was amplified in duplicate using the One Step RT-qPCR qScript kit (Quanta Biosciences) following the manufacturer’s instructions. Each reaction mixture contained 12.5 μl of 2X Master Mix, 2 μl of an oligonucleotide mix designed to amplify the ANDV S-segment (1 μM each), 4.75 μl of nuclease-free water, 0.5 μl of the ANDV probe (5’FAM-BHQ-3’), 0.25 μl of qScript One Step RT, and 5 μl of template RNA. Reverse transcription was performed at 50 °C for 15 minutes, followed by denaturation at 95 °C for 5 minutes, and 40 cycles of amplification at 95 °C for 15 seconds and 60 °C for 60 seconds in a Real-Time PCR Detection System CFX-96 (Bio-Rad, CA). To obtain complete genomic sequences, an amplicon-based method was selected using a one-step RT-PCR strategy to enrich vRNA, followed by library preparation as previously described (21). The library was sequenced on a MiSeq sequencing platform (Illumina, San Diego, CA) using 2 x 151-bp paired-end sequencing. Bioinformatic analysis was performed as described previously (*8*). ### 2.7. Biosafety precautions and Ethics statements The entire procedure of viral isolation, subsequent propagation, and sample analysis before viral inactivation was conducted at the biosafety level 3 facility at the Unidad Operativa Centro de Contención Biológica (UOCCB-ANLIS). All procedures involving human samples for viral isolation used in this study were approved by the Bioethics Committee, Area Programatica Comodoro Rivadavia, Chubut province, to ensure compliance with ethical standards. All procedures involving animal handling were carried out within an animal biosafety level 3 facility at UOCCB-ANLIS and hamsters were housed in ventilated cage racks. Research was conducted in compliance with institutional guidelines and the national law no.: 14,346, that regulates experiments involving animals, and adheres to principles stated in the Guide for the Care and Use of Laboratory Animals, National Research Council, 2011 (22). An institutional committee for animal use and care approved the protocols used. ## 3. Results ### 3.1. A new strain of Andes virus was isolated in cell culture from a human serum sample A human serum sample with a high viral load (6.3 × 107 copies/ml) and no detectable specific antibodies against ANDV, was selected to infect a Vero E6 cell monolayer. After three blind passages, viral RNA was detectable in the supernatant of the infected cell culture (2.5 × 108 copies/ml). Infection was confirmed in the cells by IFA, showing the presence of viral antigen in approximately 50% of the cells in passage No. 3 and 80% in passage No. 4 (p4) (Figure 1). A large viral stock was prepared from the supernatant of passage No. 3, as described above, for use in subsequent experiments. The infectious titre of the viral stock obtained was 2.1 × 106 FFU/ml. This new strain was designated as ARG-Epuyen. ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/10/08/2024.10.08.24312738/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2024/10/08/2024.10.08.24312738/F1) Figure 1. Visualization of ARG-Epuyen by Indirect immunofluorescence assay. The micrograph shows Vero E6 cells infected with the ANDV ARG-Epuyen strain (p4, 7 days post-infection), micrograph with DAPI, FITC and merge (A, B, C). Vero E6 cell control (mock infected cells), equal condition (D, E, F). Cells were stained with rabbit polyclonal serum against the nucleoprotein of ANDV, followed by an FITC-conjugated antibody against rabbit IgG. DAPI was used for DNA staining (4’,6-diamidino-2-phenylindole). The images were captured at 400X magnification. ### 3.2. ARG-Epuyen and ARG showed differential kinetics of replication and infectivity in cell cultures The growth kinetics of the ARG and ARG-Epuyen strains were first assessed in Vero E6 cells using an MOI of 0.002 (Figure 2). Quantification of viral RNA in the culture supernatants of cells infected with each strain showed that ARG replicates faster than ARG-Epuyen. Both strains produced high levels of infectious viral particles, with the maximum level observed for ARG, which reached up to 2.1 × 107 FFU/ml, whereas ARG-Epuyen did not exceed 2.1 × 106 FFU/ml (Figure 2B). The growth kinetics were then compared using the epithelial cell line A549 with a higher MOI. The results were consistent, showing that ARG replicates faster than ARG-Epuyen in both Vero E6 and A549 cells (Figure 3). ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/10/08/2024.10.08.24312738/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2024/10/08/2024.10.08.24312738/F2) Figure 2. Comparative kinetics curves of Andes/ARG and ARG-Epuyen strains in Vero E6 (A) and comparative levels of infective viral particles of Andes/ARG and ARG-Epuyen strains in Vero E6 (B). ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/10/08/2024.10.08.24312738/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2024/10/08/2024.10.08.24312738/F3) Figure 3. Comparative kinetics curves of ANDV strains in Vero E6 and A549 cells. ### 3.3. Both strains of Andes virus are highly transmissible in Hamsters Seven Syrian Golden hamsters of different sexes (3 males: MI; 4 females: FI) were inoculated intramuscularly with high doses of ANDV (approximately 1 × 104 FFU) and placed in separate cages with one or two non-inoculated cage-mates (receptors: MR and FR). During the 30 days post-infection (p.i.) period, neither the hamsters inoculated with ARG-Epuyen (n=6) nor the receptors exposed to them (n=9) showed any visible signs of illness. However, infection was confirmed in all of them through seroconversion and/or genomic RNA detection in lung tissues (Table 1). In contrast, the single hamster inoculated with ARG (MI-1) exhibited inappetence and lethargy starting from day five p.i., and dyspnea was evident from day 13 to day 16 p.i. After 11 days of illness, MI-1 gradually improved and fully recovered by the end of the experiment. However, the two hamsters exposed to MI-1, MR-1 and MR-2, became ill 16 and 22 days after the onset of symptoms in MI-1, respectively, and both rapidly died. After 30 days p.i., all the survivors were sampled and sacrificed. The six index hamsters showed a strong humoral response (IgG titers >25,600), while four out of 11 of their cage-mates (receptors) exhibited low to moderate IgG titers. Nevertheless, all of them (n=11) were infected, as evidenced by the detection of viral RNA in blood (up to 1.5 × 107 RNA copies/ml) and/or in lung tissues (up to 1.6 × 108 copies/100 ng RNA). View this table: [Table 1.](http://medrxiv.org/content/early/2024/10/08/2024.10.08.24312738/T1) Table 1. Evaluation of virulence of Andes virus strains in the golden hamster model. ### 3.4. ARG-Epuyen grew in cell culture without need of adaptation To analyze potential changes that could lead to viral adaptation for growth in cell culture and replication in hamsters, the complete genomes of the isolated virus (p4) and from the lung of an infected hamster (MI-3) were sequenced and compared with the previously published sequence from the human sample (GenBank [MN258238](http://medrxiv.org/lookup/external-ref?link\_type=GEN&access\_num=MN258238&atom=%2Fmedrxiv%2Fearly%2F2024%2F10%2F08%2F2024.10.08.24312738.atom), [MN258204](http://medrxiv.org/lookup/external-ref?link\_type=GEN&access_num=MN258204&atom=%2Fmedrxiv%2Fearly%2F2024%2F10%2F08%2F2024.10.08.24312738.atom), [MN258171](http://medrxiv.org/lookup/external-ref?link_type=GEN&access_num=MN258171&atom=%2Fmedrxiv%2Fearly%2F2024%2F10%2F08%2F2024.10.08.24312738.atom)). The isolated strain ARG-Epuyen p4 showed only one nucleotide change in the non-coding region of the S segment (position 1522) and one non-synonymous change, K2116E, in the L segment (A6381G). No changes were observed between the isolated strain (p4) and the virus recovered from the infected hamster. To investigate the molecular basis for the observed differences in pathogenicity between ARG-Epuyen (p4) and ARG (p19) in the hamster model, we compared the complete sequences of both strains. We identified 25 non-synonymous substitutions: three in the S segment (one in the N ORF and two in the NSs ORF), 10 in the M segment (six in Gn and four in Gc), and 12 in the L segment (Table 2). View this table: [Table 2.](http://medrxiv.org/content/early/2024/10/08/2024.10.08.24312738/T2) Table 2. Evaluation of amino acid changes of Andes virus strains. ## 4. Discussion The limited progress in vaccine development and other medical countermeasures (MCMs) against hantaviruses has been partly due to restricted access to wild-type viral strains and the lack of appropriate animal models of the disease. Additionally, no reverse genetic system for hantaviruses has been reported to date. Generally, in vivo and in vitro studies of hantavirus infections have been conducted using cell-cultured strains derived from moderate to high passage numbers. The success of MCM development depends on selecting strains that are actively circulating and have proven pathogenicity in humans. Following the COVID-19 pandemic, the National Institute of Allergy and Infectious Diseases proposed a prototype pathogen approach to develop a generalizable MCM strategy. This approach can be applied to other viruses within the same viral family, enabling the rapid development of MCMs and shortening the timeline between pathogen outbreak and regulatory authorization if a virus with similar properties emerges. ANDV has been mentioned as a prototype pathogen of the Hantaviridae family and should be considered for vaccine development and pre-clinical and clinical testing (23,24). For these purposes, the availability of well-characterized ANDV strains is critical. In this work, we described the isolation of the strain responsible for the largest ANDV PTP transmission outbreak, which occurred in the small town of Epuyen and began on 2 November 2018. This strain, ARG-Epuyen, exhibited a high capacity for PTP transmission, necessitating the implementation of quarantine measures to curtail further spread (*8*). The median reproductive number (the mean number of secondary cases caused by an infected person) was 2.12 before control measures were implemented and subsequently dropped to below 1.0 by late January. Early intervention allowed for the collection of samples leading to the isolation of this new ANDV strain from an asymptomatic case. An early passage of this strain was sequenced, revealing only one amino acid difference from the virus recovered from the patient, indicating that it can be considered a wild-type strain. Like the ARG strain, this strain was able to grow in a new host without needing adaptation (25). Two of the three previously isolated ANDV strains in cell culture caused lethal disease in hamsters (17–19,25). Although this animal model has become crucial for studying the pathogenesis of HPS, the spreading capacity and horizontal transmission of ANDV have only been evaluated in one previous study with the CH-9717869 strain (26). In this study, we assessed the pathogenicity and spreading capacity of the strain responsible for sustained PTP transmission in this animal model and compared it with the highly lethal ARG strain in direct exposure experiments. All hamsters—both those directly inoculated and those exposed to infected hamsters—became infected. Our data indicated that ARG-Epuyen was less virulent than ARG, as none of the six index hamsters inoculated with ARG-Epuyen developed severe disease during the 30-day period, despite showing high viral titres in blood and lungs. However, as five hamsters exposed to the index hamsters were sacrificed during the incubation period—when they had no IgG titres and rising viral RNA loads in blood and lungs—their final outcomes could not be assessed. We demonstrated that both ARG and ARG-Epuyen were highly transmissible in direct exposure experiments. Although the mechanism of transmission remains to be confirmed through indirect exposure experiments, we hypothesize that the route was likely respiratory or digestive, due to the absence of wounds in the animals. Further studies are required to assess the pathogenic characteristics of this strain in the animal model. Our preliminary data revealed differences in lethality between the two Argentinean strains. A previous comparative study of two Chilean strains found similar results. The strain obtained from a human serum sample (CHI-7913) caused an asymptomatic infection in hamsters, while the strain obtained from a rodent (CHI-9717869) was highly lethal (*19*). The molecular basis for the observed differences in pathogenesis between these strains was evaluated through in silico studies (19,25). However, the specific differences observed need further investigation through directed mutagenesis experiments. The N protein plays no role, as both strains share 100% identity. In vitro studies suggested that ANDV virulence could be determined by its ability to alter cellular signalling pathways by restricting the early induction of beta interferon (IFNB) and IFN-stimulated genes (ISGs) (*26–28*). Additionally, compared with other pathogenic hantaviruses, ANDV is unique in that three viral proteins—N, NSs, and Gn/Gc—can block the same signalling pathway at different levels. However, the few differences found in the Gn/Gc between the strains seem to be located outside conserved motifs or domains involved in the regulation of the IFNB response. Further characterization of the NSs and RdRp proteins is needed to evaluate the effects of the observed changes. As a result of comparing the complete sequences of both ANDV strains, several amino acid changes were predicted. None of the substitutions represented drastic changes that could affect key motifs already described for the structural conformation and/or function of the N, Gn/Gc, and RdRp proteins. Exceptions included the changes P97S and T641I in the M-segment; the P residue is in the Gn ectodomain and may impact the flexibility of a loop structure, affecting recognition by some immune response components and/or interaction with cell receptors. The T641I change, located in an alpha-helix structure near the WAASA motif, may result in the loss of an oxygen molecule capable of forming a hydrogen bond with the adjacent residue C642. In conclusion, we obtained a low-passaged isolate of a prototype pathogen: ANDV. The new strain, ARG-Epuyen, is a well-characterized isolate due to its known outcome in a recent HPS outbreak and its high PTP transmission potential. Furthermore, the isolation was obtained directly from a clinical sample with a low number of passages (p=4) and only one amino acid change. Therefore, it can be considered an ANDV wild-type strain, representing a critical step toward developing MCMs against this emerging pathogen. ## Data Availability All data produced in the present work are contained in the manuscript. ## Biographical Sketch Miss Coelho and Mr. Kehl are part of the staff of the Hantavirus National Reference Laboratory. Coelho’s primary research interests include diagnoses of infectious diseases and is coordinating the serology section of the laboratory. Mr. Kehl is a junior researcher interested in infectious diseases and emergent viral zoonosis. ## Acknowledgments We thank Silvia Girard and Lara Martín for laboratory support. Financial support: this work was supported by the Instituto Nacional de Enfermedades Infecciosas, Administración Nacional de Laboratorios e Institutos de Salud “Dr. C. Malbrán”. All authors: No reported conflicts of interest. * Received October 8, 2024. * Revision received October 8, 2024. * Accepted October 8, 2024. * © 2024, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## REFERENCES 1. 1.Plyusnin A, Vapalahti O, Vaheri A. Hantaviruses: genome structure, expression and evolution. J Gen Virol. noviembre de 1996;77 ( Pt 11):2677–87. 2. 2.Castillo C, Nicklas C, Mardones J, Ossa G. Andes Hantavirus as possible cause of disease in travellers to South America. Travel Med Infect Dis. enero de 2007;5(1):30–4. 3. 3.Kuenzli AB, Marschall J, Schefold JC, Schafer M, Engler OB, Ackermann-Gäumann R, et al. Hantavirus Cardiopulmonary Syndrome Due to Imported Andes Hantavirus Infection in Switzerland: A Multidisciplinary Challenge, Two Cases and a Literature Review. Clin Infect Dis Off Publ Infect Dis Soc Am. 13 de noviembre de 2018;67(11):1788–95. 4. 4.Kofman A, Eggers P, Kjemtrup A, Hall R, Brown SM, Morales-Betoulle M, et al. Notes from the Field: Contact Tracing Investigation after First Case of Andes Virus in the United States - Delaware, February 2018. MMWR Morb Mortal Wkly Rep. 19 de octubre de 2018;67(41):1162–3. 5. 5.Nichol ST, Spiropoulou CF, Morzunov S, Rollin PE, Ksiazek TG, Feldmann H, et al. Genetic identification of a hantavirus associated with an outbreak of acute respiratory illness. Science. 5 de noviembre de 1993;262(5135):914–7. 6. 6.Abudurexiti A, Adkins S, Alioto D, Alkhovsky SV, Avšič-Županc T, Ballinger MJ, et al. Taxonomy of the order Bunyavirales: update 2019. Arch Virol. julio de 2019;164(7):1949–65. 7. 7.López N, Padula P, Rossi C, Lázaro ME, Franze-Fernández MT. Genetic identification of a new hantavirus causing severe pulmonary syndrome in Argentina. Virology. 1 de junio de 1996;220(1):223–6. 8. 8.Martínez VP, Di Paola N, Alonso DO, Pérez-Sautu U, Bellomo CM, Iglesias AA, et al. «Super-Spreaders» and Person-to-Person Transmission of Andes Virus in Argentina. N Engl J Med. 3 de diciembre de 2020;383(23):2230–41. 9. 9.Iglesias AA, Períolo N, Bellomo CM, Lewis LC, Olivera CP, Anselmo CR, et al. Delayed viral clearance despite high number of activated T cells during the acute phase in Argentinean patients with hantavirus pulmonary syndrome. EBioMedicine. 2 de enero de 2022;75:103765. 10. 10.Martinez VP, Bellomo CM, Cacace ML, Suarez P, Bogni L, Padula PJ. Hantavirus pulmonary syndrome in Argentina, 1995-2008. Emerg Infect Dis. diciembre de 2010;16(12):1853–60. 11. 11.Tortosa F, Carrasco G, Gallardo D, Prandi D, Parodi V, Santamaría G, et al. [Prognostic factors for cardio-pulmonary syndrome and death by hantavirus Andes Sur: cohort study in San Carlos de Bariloche and health influence area]. Medicina (Mex). 2022;82(3):351–60. 12. 12.Wells RM, Sosa Estani S, Yadon ZE, Enria D, Padula P, Pini N, et al. An unusual hantavirus outbreak in southern Argentina: person-to-person transmission? Hantavirus Pulmonary Syndrome Study Group for Patagonia. Emerg Infect Dis. junio de 1997;3(2):171–4. 13. 13.Padula PJ, Edelstein A, Miguel SD, López NM, Rossi CM, Rabinovich RD. Hantavirus pulmonary syndrome outbreak in Argentina: molecular evidence for person-to-person transmission of Andes virus. Virology. 15 de febrero de 1998;241(2):323–30. 14. 14.Galeno H, Mora J, Villagra E, Fernandez J, Hernandez J, Mertz GJ, et al. First human isolate of Hantavirus (Andes virus) in the Americas. Emerg Infect Dis. julio de 2002;8(7):657–61. 15. 15.Padula PJ, Sanchez AJ, Edelstein A, Nichol ST. Complete nucleotide sequence of the M RNA segment of Andes virus and analysis of the variability of the termini of the virus S, M and L RNA segments. J Gen Virol. septiembre de 2002;83(Pt 9):2117–22. 16. 16.Meissner JD, Rowe JE, Borucki MK, St Jeor SC. Complete nucleotide sequence of a Chilean hantavirus. Virus Res. octubre de 2002;89(1):131–43. 17. 17.Hooper JW, Larsen T, Custer DM, Schmaljohn CS. A lethal disease model for hantavirus pulmonary syndrome. Virology. 10 de octubre de 2001;289(1):6–14. 18. 18.Martinez VP, Padula PJ. Induction of protective immunity in a Syrian hamster model against a cytopathogenic strain of Andes virus. J Med Virol. enero de 2012;84(1):87–95. 19. 19.Warner BM, Sloan A, Deschambault Y, Dowhanik S, Tierney K, Audet J, et al. Differential pathogenesis between Andes virus strains CHI-7913 and Chile-9717869in Syrian Hamsters. J Virol. 24 de febrero de 2021;JVI.00108-21. 20. 20.Bellomo CM, Pires-Marczeski FC, Padula PJ. Viral load of patients with hantavirus pulmonary syndrome in Argentina. J Med Virol. noviembre de 2015;87(11):1823–30. 21. 21.Alonso DO, Kehl S, Coelho R, Periolo N, Poklepovich T, Perez-Sautu U, et al. Orthohantavirus Diversity in Central-East Argentina: Insights from Complete Genomic Sequencing on Phylogenetics, Geographic patterns and Transmission scenarios. bioRxiv. 1 de enero de 2024;2024.03.25.586579. 22. 22.National Research Council (US) Committee for the Update of the Guide for theCare and Use of Laboratory Animals. Guide for the Care and Use of Laboratory Animals [Internet]. 8th ed. Washington (DC): National Academies Press (US); 2011 [citado 20 de agosto de 2024]. (The National Academies Collection: Reports funded by National Institutes of Health). Disponible en: [http://www.ncbi.nlm.nih.gov/books/NBK54050/](http://www.ncbi.nlm.nih.gov/books/NBK54050/) 23. 23.Cassetti MC, Pierson TC, Patterson LJ, Bok K, DeRocco AJ, Deschamps AM, et al. Prototype Pathogen Approach for Vaccine and Monoclonal Antibody Development: A Critical Component of the NIAID Plan for Pandemic Preparedness. J Infect Dis. 15 de junio de 2023;227(12):1433–41. 24. 24.Morabito KM, Cassetti MC, DeRocco AJ, Deschamps AM, Pierson TC. Viral Prototypes for Pandemic Preparedness: The Road Ahead. J Infect Dis. 18 de octubre de 2023;228(Suppl 6):S460–4. 25. 25.Bellomo CM, Alonso DO, Pérez-Sautu U, Prieto K, Kehl S, Coelho RM, et al. Andes Virus Genome Mutations That Are Likely Associated with Animal Model Attenuation and Human Person-to-Person Transmission. mSphere. 22 de junio de 2023;8(3):e0001823. 26. 26.Riesle-Sbarbaro SA, Kirchoff N, Hansen-Kant K, Stern A, Kurth A, Prescott JB. Human-to-Human Transmission of Andes Virus Modeled in Syrian Hamsters. Emerg Infect Dis. octubre de 2023;29(10):2159–63. 27. 27.Matthys VS, Cimica V, Dalrymple NA, Glennon NB, Bianco C, Mackow ER. Hantavirus GnT elements mediate TRAF3 binding and inhibit RIG-I/TBK1-directed beta interferon transcription by blocking IRF3 phosphorylation. J Virol. febrero de 2014;88(4):2246–59. 28. 28.Cimica V, Dalrymple NA, Roth E, Nasonov A, Mackow ER. An innate immunity-regulating virulence determinant is uniquely encoded by the Andes virus nucleocapsid protein. mBio. 18 de febrero de 2014;5(1):e01088–13. 29. 29.Geimonen E, LaMonica R, Springer K, Farooqui Y, Gavrilovskaya IN, Mackow ER. Hantavirus pulmonary syndrome-associated hantaviruses contain conserved and functional ITAM signaling elements. J Virol. enero de 2003;77(2):1638–43.