HTNV

Hantavirus Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Epitope Mapping

HTNV encodes glycoproteins (Gn and Gc) critical for viral entry and fusion. Structural studies of related hantaviruses (e.g., PUUV Gc) reveal a class II fusion glycoprotein architecture with three domains (I–III) forming a pre-fusion conformation . Key epitopes recognized by neutralizing antibodies, such as those targeting the Gc glycoprotein junction between domains I and II, have been characterized through crystallography and cryo-EM .

Table 1: Key Molecular Features of HTNV Glycoproteins

FeatureDescriptionSource
Gc glycoproteinClass II fusion architecture with domains I–III; pre-fusion conformation
Neutralizing epitopeJunction of Gc domains I and II (e.g., PUUV Gc epitope)
MHC-II epitopes289 unique epitopes predicted across HTNV Gn/Gc; 42 amino acid variants

Antiviral Therapies: Small-Molecule Inhibitors and Repurposed Drugs

Multiple small-molecule inhibitors and repurposed drugs have demonstrated in vitro and in vivo efficacy against HTNV:

Table 2: Antiviral Compounds Targeting HTNV

CompoundMechanismEC50 (μM)CC50 (μM)SIIn Vivo EfficacySource
8G1AKT/mTOR pathway inhibition1.5399.8865.28Reduces viral titer in A549 cells
RibavirinRNA polymerase inhibition2.65N/AN/AIC90 = 10.49 μM
FavipiravirRNA polymerase inhibition3.89N/AN/AIC90 = 9.79 μM
Cepharanthine (CEP)Entry inhibition0.99N/A5094% reduction in HTNV pseudovirus
N6 (Coumarin)Unknown7.0677.0810.9Reduces HTNV-S RNA expression

Key Findings:

  • 8G1 inhibits AKT phosphorylation, reducing mTOR and eIF4E signaling, critical for viral translation .

  • Ribavirin and favipiravir show comparable in vitro potency, with potential for combination therapy to mitigate resistance .

  • CEP demonstrates broad-spectrum antiviral activity, including against SARS-CoV and HIV-1, with in vivo efficacy in murine models .

Host Response and Pathogenesis

HTNV infection triggers complex immune responses, including apoptosis and innate signaling:

Table 3: Immune Mechanisms in HTNV Infection

MechanismObservationSource
TRAIL-mediated apoptosisHTNV upregulates TRAIL, inducing caspase-8-dependent apoptosis in in vivo models
MAIT cell activationHTNV reduces CD8+ MAIT cells, correlating with disease severity
Type I IFN responseHTNV infection activates IRF3/IRF7, enhancing IFN-β production

Key Insights:

  • TRAIL treatment reduces viral load and mortality in HTNV-infected mice, suggesting therapeutic potential .

  • MAIT cell depletion exacerbates HTNV pathogenesis, highlighting their role in antiviral immunity .

Genetic Diversity and Evolution

Phylogenetic analysis of HTNV isolates reveals significant geographic and genetic variation:

Table 4: Genetic Diversity of HTNV

ParameterDetailSource
Phylogenetic groups11 distinct groups with geographic clustering
Recombination events9 identified, particularly in Guizhou strains
Amino acid variants42 significant sites, 19 in immune epitopes

Evolutionary Hotspots:

  • Guizhou Province: High recombination rates and divergent lineages, suggesting a potential origin hub .

  • Zhejiang Province: Proposed as an ancestral origin site based on Bayesian analysis .

Vaccine Development

Phase 1 trials of DNA vaccines targeting HTNV and Puumala virus (PUUV) glycoproteins (GnGc) have shown promise:

Table 5: Neutralizing Antibody Responses to HTNV DNA Vaccines

CohortPsVNA50 Titer (GMT)PRNT50 Titer (GMT)Cross-ReactivitySource
HTNV alone10,000+1,000+43% vs. PUUV
PUUV alone1,000<20None
HTNV/PUUV combo500<20Limited

Notable Outcomes:

  • HTNV-specific vaccines elicit higher neutralizing titers than PUUV vaccines .

  • Cross-reactivity: ~43% of HTNV-vaccinated subjects showed neutralizing antibodies against PUUV .

Immunological Epitope Discovery

Bioinformatic and experimental approaches have identified MHC-II-restricted epitopes in HTNV glycoproteins:

Table 6: Predicted and Experimental HTNV Epitopes

Epitope RegionPredicted MHC-II AllelesExperimental ValidationSource
Gn 289–303H2-Ad, DRB1Mouse models
Gn 445–459H2-A, DRB1Mouse models
Gc 781–795H2-E, DRB5Mouse models

Conservation Analysis:

  • Gn 289–303: High conservation across strains, suggesting a stable target for vaccine design .

Product Specs

Introduction
Hantaviruses, belonging to the Bunyaviridae family, are transmitted through aerosolized rodent excreta or bites. Infection, known as hemorrhagic fever with renal syndrome (HFRS), carries high mortality. Different Hantavirus strains cause HFRS, Hantavirus pulmonary syndrome (HPS), or Hantavirus cardiopulmonary syndrome (HCPS). These viruses increase vascular permeability, decrease blood pressure, and damage the endothelium, primarily affecting the kidneys, lungs, spleen, and gallbladder. HFRS is prevalent in China, the Korean Peninsula, Russia, and parts of Europe. HPS and HCPS are more common in Argentina, Chile, Brazil, the United States, Canada, and Panama. Andes virus, a Hantavirus strain in South America, causes HCPS and can spread between humans. Fatality rates reach 25-35% in Argentina and 37% in Chile.
Description
The Hantavirus nucleocapsid (N) fusion protein, expressed in E. coli, includes a 6xHis tag. Conserved across Hantavirus strains, it's used in tests detecting Hantavirus-specific IgM and IgG antibodies.
Physical Appearance
A clear, sterile-filtered solution.
Formulation

HTNV is supplied in a solution of 1xPBS at pH 7.4 with 0.05% sodium azide.

Stability

Store at -20°C upon receipt. Avoid repeated freeze-thaw cycles.

Purity

HTNV protein purity exceeds 95%, determined by 12% PAGE (Coomassie staining).

Synonyms
HTNV, Hantaan Virus, Hantanvirus.
Source
E.Coli
Amino Acid Sequence

GSMATMEELQREINAHEGQLVIARQKVRDAEKQYEKDPDELNKRTLTDRE GVAVSIQAKIDELKRQLADRIATGKNLGKEQDPTGVEPGDHLKERSMLSY GNVLDLNHLDIDEPTGQTADWLSIVVYVD

Purification Method
Purified by proprietary chromatographic technique.

Q&A

What is HTNV and what is its significance in infectious disease research?

HTNV (Hantaan virus) is a zoonotic pathogen belonging to the Bunyavirales family that is primarily transmitted by rodents. It is clinically significant as a causative agent of hemorrhagic fever with renal syndrome (HFRS) in humans, particularly prevalent in East Asia. From a research perspective, HTNV serves as an important model for understanding zoonotic virus transmission, pathogenesis of hemorrhagic diseases, and host immune responses to emerging infectious diseases. Unlike other hantaviruses such as Sin Nombre virus (SNV) and Andes virus (ANDV) that cause hantavirus cardiopulmonary syndrome (HCPS) in the Americas, HTNV primarily targets the kidneys, making it a distinct model for organ-specific viral pathogenesis studies .

How does HTNV differ from other hantaviruses in terms of geographic distribution and clinical manifestations?

HTNV is predominantly found in East Asia, whereas other hantaviruses have different geographic distributions: Dobrava virus and Puumala virus (PUUV) are prevalent in Europe, Seoul virus has worldwide distribution, and Sin Nombre virus (SNV) and Andes virus (ANDV) are found in North and South America, respectively. The clinical manifestations also differ significantly. HTNV causes hemorrhagic fever with renal syndrome (HFRS) with the highest mortality rate among Old World Hantaviruses (OWHs), characterized by vascular leakage, thrombocytopenia, and acute kidney injury. In contrast, New World hantaviruses like SNV and ANDV cause hantavirus cardiopulmonary syndrome (HCPS), where pulmonary edema and respiratory failure are predominant. Another important distinction is that ANDV has been documented to transmit from person to person through respiratory routes and saliva, while HTNV transmission remains primarily zoonotic .

What animal models are currently available for HTNV research, and what are their limitations?

  • Pathogenesis limitations: Mice do not fully replicate the human disease course of HFRS

  • Immune response differences: Murine immune responses may not perfectly mirror human responses to HTNV infection

  • Challenge model limitations: Establishing consistent viral challenge parameters can be difficult

For advanced studies, suckling mouse brain models have been used for vaccine development, as seen with the licensed HTNV vaccines in South Korea and China. For future research, non-human primate models are suggested for improved clinical relevance, particularly for vaccine efficacy testing before clinical development. Researchers should be cognizant that the choice of animal model significantly impacts the translational value of their findings and should clearly acknowledge these limitations in their study design and interpretations .

How does HTNV infection of CD8+ T cells contribute to disease pathogenesis and severity?

HTNV's ability to infect CD8+ T cells represents a crucial pathogenic mechanism that directly influences disease outcomes. Research with a clinical cohort of 280 HFRS patients has established that circulating HTNV-infected CD8+ T cell quantities directly correlate with disease severity. The pathogenic mechanisms involve several processes:

  • Altered immune function: HTNV infection compromises CD8+ T cell functionality, potentially impairing viral clearance

  • Enhanced cytoplasmic multivesicular bodies: Electron microscopy studies reveal significant increases in cytoplasmic multivesicular bodies within infected CD8+ T cells, which facilitates virus production

  • Immune dysregulation: Infected CD8+ T cells may contribute to cytokine dysregulation and immunopathology

This finding has significant implications for disease monitoring and therapeutic development, suggesting that HTNV-infected CD8+ T cell quantification could serve as a biomarker for disease progression. Methodologically, researchers investigating this pathway should employ flow cytometry with viral antigen staining, electron microscopy for ultrastructural analysis, and correlational statistical methods to establish relationships between cell infection rates and clinical outcomes .

What is the current understanding of the humoral versus cellular immune responses in controlling HTNV infection?

The immune response to HTNV infection involves both humoral and cellular components, though recent research suggests differential contributions:

Humoral immunity components:

  • Neutralizing antibodies (NAbs) against HTNV glycoproteins are critical for protection

  • Single-dose rVSV-HTNV-GP vaccination elicits robust NAb responses (mean titers 72.0-104.0) that exceed those from traditional inactivated vaccines (mean titer 28.0)

  • Cross-reactive NAbs against related viruses like SEOV can be generated through targeting conserved epitopes

Cellular immunity components:

  • Both Th1 (IFN-γ, TNFα) and Th2 (IL-4) cytokine responses are detectable following vaccination

  • Additional cytokines including IL-2 and IL-10 are part of the cellular response

  • T cell responses to rVSV-HTNV-GP show no clear advantage over inactivated vaccines

Passive transfer experiments demonstrate that humoral immunity plays the dominant protective role against HTNV infection. When designing studies to investigate immune responses to HTNV, researchers should include comprehensive assessment of both neutralizing antibody titers (using focus-reduction neutralization tests) and T cell responses (through intracellular cytokine staining and ELISpot assays) .

How do viral ultrastructures and life cycle events in HTNV-infected cells correlate with virion production and pathogenesis?

The ultrastructure and life cycle of HTNV in infected cells reveal critical insights into viral pathogenesis. Electron microscopy studies of HTNV-infected CD8+ T cells have identified several key structural and morphological changes:

Ultrastructural FeatureObservation in HTNV-infected CellsCorrelation with Viral Production
Multivesicular bodies (MVBs)Significant increase in cytoplasmStrongly positive correlation with virion assembly
Virus-containing vesiclesPresent in cytoplasmDirect evidence of active viral replication
Cell membrane alterationsDetectable modificationsFacilitates viral budding and release

The cycle progresses through attachment via integrins, endocytosis, RNA replication in cytoplasm, protein synthesis, assembly associated with MVBs, and eventual release. Researchers investigating HTNV cellular pathogenesis should utilize transmission electron microscopy with immunogold labeling for viral proteins, confocal microscopy with time-lapse imaging, and correlative light and electron microscopy (CLEM) to link ultrastructural observations with functional outcomes. These methodological approaches allow for the tracking of virion morphogenesis and can help identify potential targets for antiviral intervention .

What are the experimental approaches for developing and evaluating HTNV vaccines?

Current experimental approaches for HTNV vaccine development and evaluation follow systematic workflows that integrate multiple methodologies:

  • Vaccine platform selection and construction:

    • Recombinant viral vectors (e.g., VSV expressing HTNV glycoproteins)

    • Inactivated whole virus preparations

    • Subunit vaccines based on immunogenic viral components

  • In vitro characterization:

    • Antigen expression confirmation via immunofluorescence and Western blotting

    • Cross-reactivity analysis with convalescent sera (e.g., neutralization patterns from HFRS patients)

    • Antigenic similarity assessment between vaccine candidates and authentic HTNV

  • In vivo immunogenicity evaluation:

    • Dose-ranging studies (e.g., 2×10⁴, 2×10⁵, and 2×10⁶ PFUs)

    • Single-dose versus multi-dose regimens

    • Antibody response quantification (both binding and neutralizing antibodies)

    • T cell response analysis via cytokine production measurement

  • Challenge studies:

    • Viral challenge with authentic HTNV in appropriate animal models

    • Assessment of sterile immunity versus disease modification

    • Correlates of protection identification

This methodological framework enables systematic comparison between different vaccine candidates. For instance, the rVSV-HTNV-GP vaccine demonstrated superior immunogenicity compared to traditional inactivated vaccines, inducing higher neutralizing antibody titers (mean 77.4-80 versus 37.2) following a single dose versus three sequential injections of inactivated vaccine .

How can cross-reactivity against different hantavirus species be evaluated in HTNV vaccine research?

Evaluating cross-reactivity against different hantavirus species is essential for developing broadly protective vaccines. A comprehensive methodological approach includes:

  • Neutralization assays against multiple virus species:

    • Focus reduction neutralization tests (FRNT) against both HTNV and related hantaviruses

    • Serum neutralization titration against multiple virus strains

  • Epitope mapping for cross-reactive antibodies:

    • Peptide array analysis to identify conserved epitopes

    • Competition binding assays to determine antibody recognition patterns

  • Comparative immunogenicity assessment:

    • Side-by-side evaluation of immune responses against multiple viral species

    • Quantification of cross-reactivity indices (ratio of heterologous to homologous neutralization)

Recent research demonstrated that rVSV-HTNV-GP vaccination induced neutralizing antibodies against both HTNV (mean titers 77.4-80) and the related SEOV (mean titers 29.6-44.6), showing substantially higher cross-reactivity than inactivated vaccines (SEOV mean titer 13.6). This cross-reactivity assessment methodology allows researchers to predict broader protection potential and optimize vaccine formulations to target conserved epitopes across Old World Hantaviruses (OWHs) .

What immune correlates of protection should be measured in HTNV vaccine trials?

Comprehensive evaluation of HTNV vaccine efficacy requires measurement of multiple immune correlates of protection:

In evaluating vaccine candidates, researchers should prioritize neutralizing antibody responses as the primary endpoint, as passive transfer studies demonstrate their crucial role in protection. Secondary endpoints should include cellular immune responses, though these appear less critical for direct protection against HTNV. The optimal approach integrates multiple immune parameters to develop a comprehensive protection profile, as demonstrated in the rVSV-HTNV-GP vaccine evaluation where both humoral and cellular responses were comprehensively characterized .

How can HTNV-infected CD8+ T cells be detected and quantified for disease monitoring?

Detection and quantification of HTNV-infected CD8+ T cells requires a multi-parameter methodological approach:

  • Flow cytometry-based quantification:

    • Surface staining for CD8+ T cell markers

    • Intracellular staining for viral antigens (e.g., nucleocapsid protein or glycoproteins)

    • Viability discrimination to exclude non-specific staining

    • Fluorescence minus one (FMO) controls for accurate gating

  • Molecular confirmation:

    • RNA extraction from sorted CD8+ T cells

    • RT-PCR targeting viral segments (S, M, or L segments)

    • Digital droplet PCR for absolute quantification

    • RNA-seq to assess transcriptional changes in infected cells

  • Visualization techniques:

    • Immunofluorescence microscopy of isolated cells

    • Confocal microscopy for co-localization of viral components

    • Electron microscopy to detect viral particles within cells

The clinical significance of these measurements has been established in a cohort of 280 HFRS patients, where the quantity of circulating HTNV-infected CD8+ T cells directly correlated with disease severity. This finding suggests that implementing these methods could provide a valuable biomarker for monitoring disease progression and therapeutic response in clinical settings .

What experimental techniques are available for studying HTNV virion structure and assembly?

Advanced experimental techniques for studying HTNV virion structure and assembly include:

  • High-resolution imaging methods:

    • Cryo-electron microscopy for near-atomic resolution of virion structure

    • Transmission electron microscopy with negative staining for basic morphology

    • Scanning electron microscopy for surface features

    • Atomic force microscopy for topographical analysis

    • Immunogold labeling for localization of specific viral proteins

  • Biochemical characterization approaches:

    • Virion purification through density gradient centrifugation

    • Mass spectrometry for comprehensive protein composition analysis

    • Western blotting for specific viral protein detection

    • Protease protection assays to determine protein topology

  • Dynamic assembly visualization:

    • Live-cell imaging with fluorescently tagged viral components

    • Fluorescence recovery after photobleaching (FRAP) to study component mobility

    • Fluorescence resonance energy transfer (FRET) for protein-protein interactions

    • Time-lapse microscopy to follow assembly process

Studies of HTNV-infected CD8+ T cells have revealed important insights about virion assembly, particularly the association between cytoplasmic multivesicular bodies and virus production. These structures appear to serve as platforms for viral assembly before release. Researchers should combine multiple techniques for comprehensive analysis, as each method provides complementary information about the structural and functional aspects of HTNV virion formation .

How should researchers design experiments to study HTNV antigenicity and antibody neutralization mechanisms?

Experimental design for studying HTNV antigenicity and antibody neutralization mechanisms should follow a systematic approach:

  • Antigenic characterization:

    • Differential staining of infected cells with antibodies against viral proteins

    • Comparative immunofluorescence between authentic HTNV and vaccine constructs

    • Epitope mapping using peptide arrays or alanine scanning mutagenesis

    • Competition binding assays to identify immunodominant regions

  • Neutralization assay optimization:

    • Focus-reduction neutralization test (FRNT) protocol standardization

    • Determination of optimal virus input and cell substrate

    • Inclusion of appropriate controls (negative sera, non-neutralizing antibodies)

    • Quantitative readouts through automated image analysis

  • Mechanistic studies of neutralization:

    • Pre- versus post-attachment neutralization assays

    • Temperature-shift experiments to distinguish attachment from fusion inhibition

    • Single-particle tracking to visualize neutralization effects

    • Structural biology approaches (e.g., cryo-EM of antibody-virion complexes)

These methods have been successfully applied to demonstrate the antigenic similarity between rVSV-HTNV-GP and authentic HTNV, showing similar dose-dependent neutralization patterns from convalescent plasma. When designing such experiments, researchers should ensure appropriate controls, standardized protocols, and sufficient replication to generate robust and reproducible results that accurately characterize HTNV antigenicity and antibody neutralization mechanisms .

What are the most promising approaches for developing single-dose HTNV vaccines with enhanced efficacy?

Several innovative approaches show promise for developing single-dose HTNV vaccines with enhanced efficacy:

  • Viral vector platforms:

    • Recombinant vesicular stomatitis virus (rVSV) expressing HTNV glycoproteins has demonstrated superior immunogenicity compared to traditional inactivated vaccines

    • Alternative vectors (adenovirus, modified vaccinia Ankara) merit exploration for potentially different immune response profiles

    • The combination of viral vectors in prime-boost strategies may enhance both breadth and durability

  • Antigen engineering strategies:

    • Inclusion of both glycoproteins and nucleocapsid protein in a single construct

    • Structure-guided design of stabilized pre-fusion glycoprotein conformations

    • Identification and incorporation of conserved neutralizing epitopes for broader protection

  • Adjuvant and delivery innovations:

    • Novel adjuvant formulations to enhance immunogenicity

    • Controlled-release technologies for extended antigen presentation

    • Mucosal delivery systems to induce local immunity at potential sites of viral entry

The rVSV-HTNV-GP platform has demonstrated significant advantage over traditional approaches, inducing robust neutralizing antibody responses (mean titers 77.4-80) after a single dose compared to three doses of inactivated vaccine (mean titer 37.2). Future research should focus on optimizing these promising platforms through comparative immunogenicity studies, identifying correlates of protection, and conducting challenge studies in appropriate animal models before advancing to clinical trials .

How can systems biology approaches enhance our understanding of HTNV pathogenesis?

Systems biology approaches offer powerful frameworks for comprehensively understanding HTNV pathogenesis:

  • Multi-omics integration methodologies:

    • Transcriptomics to characterize host gene expression changes during infection

    • Proteomics to identify altered protein expression and post-translational modifications

    • Metabolomics to detect metabolic pathway dysregulation

    • Integration of these datasets to construct network models of host-pathogen interactions

  • Single-cell analysis technologies:

    • Single-cell RNA sequencing to identify cell-specific responses to infection

    • Mass cytometry (CyTOF) for high-dimensional immune phenotyping

    • Spatial transcriptomics to map infection and host response in tissues

  • Computational modeling approaches:

    • Predictive modeling of disease progression based on molecular signatures

    • Network analysis to identify central nodes in host response pathways

    • Machine learning for biomarker identification and outcome prediction

These approaches can address key questions in HTNV research, including why CD8+ T cells are specifically targeted by the virus, how infection leads to hemorrhagic manifestations and kidney damage, and what molecular determinants drive disease severity. Researchers employing systems biology should design longitudinal studies with appropriate sampling timepoints, include diverse patient populations, and establish robust computational pipelines for data integration and interpretation .

What are the challenges and methodological considerations for studying HTNV in the context of co-infections or pre-existing immunity to related viruses?

Studying HTNV in the context of co-infections or pre-existing immunity presents significant challenges that require specific methodological considerations:

  • Co-infection experimental design:

    • Sequential versus simultaneous infection protocols

    • Controlling for viral interference effects

    • Quantitative methods for distinguishing responses to each pathogen

    • Appropriate animal models that support multiple infections

  • Pre-existing immunity assessment:

    • Baseline serological screening for cross-reactive antibodies

    • Depletion studies to determine the impact of pre-existing antibodies

    • T cell epitope mapping to identify cross-reactive cellular responses

    • Longitudinal studies to track how pre-existing immunity shapes outcomes

  • Analysis and interpretation challenges:

    • Accounting for immunological background variation in study populations

    • Developing statistical approaches for multi-pathogen data

    • Distinguishing protective versus enhancing effects of heterologous immunity

    • Establishing appropriate control groups and reference standards

These considerations are particularly relevant given the geographical overlap of HTNV with other hantaviruses and the observation that rVSV-HTNV-GP vaccination induces cross-reactive neutralizing antibodies against related viruses like SEOV. Researchers should specifically design studies that can distinguish between homologous and heterologous immune responses, potentially through the use of chimeric viruses, epitope-specific assays, or adoptive transfer experiments in appropriate animal models .

Product Science Overview

Introduction

Hantaviruses are a genus within the family Hantaviridae, known for causing serious illnesses in humans, such as Hantavirus Pulmonary Syndrome (HPS) and Hemorrhagic Fever with Renal Syndrome (HFRS) . These viruses are primarily transmitted to humans through contact with rodent excreta, including urine, droppings, or saliva .

Structure and Genome

Hantaviruses are enveloped viruses with a tripartite negative-stranded RNA genome, consisting of three segments: the small (S), medium (M), and large (L) segments . The S segment encodes the nucleocapsid protein, the M segment encodes the glycoproteins (Gn and Gc), and the L segment encodes the RNA-dependent RNA polymerase .

Hantavirus Recombinant Proteins

Recombinant proteins are proteins that are genetically engineered in the laboratory by inserting the gene encoding the protein into a host organism, such as bacteria or yeast, which then produces the protein. In the context of Hantaviruses, recombinant proteins are used for various purposes, including vaccine development, diagnostic assays, and therapeutic research.

Applications of Hantavirus Recombinant Proteins
  1. Vaccine Development: Recombinant Hantavirus proteins are being explored as potential vaccine candidates. These proteins can elicit an immune response in the host, providing protection against Hantavirus infections. For example, the glycoproteins Gn and Gc are key targets for vaccine development due to their role in virus entry and immune recognition .

  2. Diagnostic Assays: Recombinant Hantavirus proteins are used in diagnostic assays to detect Hantavirus infections. These assays can identify antibodies against Hantavirus in patient samples, aiding in the diagnosis of HPS and HFRS .

  3. Therapeutic Research: Recombinant proteins are also used in therapeutic research to develop treatments for Hantavirus infections. For instance, neutralizing antibodies generated from Hantavirus convalescent patients have shown efficacy against Hantavirus infections . Additionally, RNA interference (RNAi) and small interfering RNA (siRNA) therapies are being investigated to target specific gene segments of the Hantavirus .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.