Parainfluenza Type-3

Parainfluenza Virus Type-3
Shipped with Ice Packs
In Stock

Description

MA 104 cells inoculated with Parainfluenza-3 virus, strain III v2932.

Product Specs

Introduction
Parainfluenza virus (PIV) encompasses four distinct serotypes and two subtypes (4a and 4b) of single-stranded RNA viruses within the paramyxovirus family. These negative-sense viruses feature fusion and hemagglutinin-neuraminidase glycoprotein "spikes" on their surface. Virion size and shape are variable, with an average diameter ranging from 150 to 300 nm.
Description
This product contains MA 104 cells infected with the Parainfluenza-3 virus, specifically strain III v2932.
Inactivation
This product has been treated with thimerosal and beta-propiolactone to ensure inactivation. Standard laboratory practices for safe handling of microbiological and viral agents are essential when working with this product.
Physical Appearance
This solution appears opaque and should not be centrifuged. Thorough mixing is required before use.
Formulation
The Parainfluenza type-3 solution is formulated in STE buffer containing 0.09% sodium azide (NaN3) and 0.005% thimerosal. Avoid centrifugation and ensure thorough mixing before use.
Stability
While Parainfluenza type-3 remains stable for up to 4 weeks at 4°C, storage below -18°C is recommended. Minimize freeze-thaw cycles.
Purity
SDS-PAGE analysis confirms a purity exceeding 90.0%.
Immunological Activity
ELISA, HIT.

Q&A

What are the key characteristics of HPIV-3 that distinguish it from other respiratory viruses?

HPIV-3 is a single-stranded, negative-sense RNA paramyxovirus that primarily causes lower respiratory tract infections. Unlike other respiratory viruses, HPIV-3 is notable for its ability to cause reinfection despite prior exposure, unlike measles or mumps viruses which typically confer lifelong immunity. Additionally, HPIV-3 is considered the most virulent of the four HPIV serotypes, particularly in causing bronchiolitis and pneumonia in young children .

The virus has a unique incubation period of 2-7 days with symptoms typically resolving within 7-10 days. HPIV-3 infections show seasonal patterns, with lower respiratory tract infections more common during spring and summer, potentially continuing into fall .

What are current gold standard methods for detecting and quantifying HPIV-3 in laboratory settings?

The current gold standard methods for HPIV-3 detection and quantification in research settings include:

  • Direct fluorescent antibody (DFA) staining: Used for initial diagnosis from respiratory secretions, providing rapid results but with moderate sensitivity .

  • Real-time RT-PCR: Considered the most sensitive method for viral quantification in nasopharyngeal aspirates or bronchoalveolar lavage samples. This technique can detect viral loads as high as >1.0×10^6 RNA copies/mL, making it valuable for monitoring infection progression and treatment response .

  • Hemagglutinin-neuraminidase gene sequencing: Particularly the variable regions (nt 1–569 and nt 762–1239) for phylogenetic analysis and strain identification, essential for tracking outbreaks and transmission patterns .

These methods should be selected based on research objectives, with RT-PCR preferred for quantitative studies and genetic sequencing for epidemiological investigations.

How does HPIV-3 cellular tropism influence experimental design in respiratory infection models?

HPIV-3 demonstrates specific cellular tropism that significantly impacts experimental design. Recent research using multiplex fluorescence RNAscope and immunohistochemistry followed by confocal microscopy has demonstrated that HPIV-3 primarily targets ciliated cells and club cells of the bronchiolar epithelium .

This tropism has important implications for experimental design:

  • Cell culture selection: Researchers should prioritize human lung epithelial (A549) or epithelium-like (HT1080) cell lines that reflect the virus's natural targets .

  • Animal model considerations: When designing in vivo experiments, researchers should select models that accurately recapitulate human respiratory epithelial architecture. AG129 mice (double IFNα/β and IFNγ receptor knockout) have been validated as supporting viral replication in both upper and lower airways following intranasal inoculation .

  • Infection protocol optimization: For consistent experimental outcomes, intranasal inoculation protocols should be standardized to ensure virus delivery to target cells in the bronchiolar epithelium. This is particularly important when evaluating therapeutic interventions .

Understanding this tropism is essential for correctly interpreting experimental results and developing relevant therapeutic approaches.

How does HPIV-3 modulate host immune responses, and what are the implications for immunopathology?

HPIV-3 employs sophisticated mechanisms to modulate host immune responses, with significant implications for immunopathology. Research has revealed several key pathways:

  • ICAM-1 Upregulation: HPIV-3 strongly induces ICAM-1 (CD54) expression in infected cells, primarily through a direct viral antigen-mediated mechanism rather than through cytokine induction. This process occurs independently of JAK/STAT signaling pathways, suggesting a unique interaction between viral components and host cellular machinery .

  • MHC Regulation: HPIV-3 infection significantly upregulates both MHC class I and II molecules on lung epithelial and epithelium-like cells. While MHC class I upregulation appears to be mediated by released factors (likely type I interferons), MHC class II induction occurs through direct viral mechanisms .

  • Cytokine-Independent Pathways: Unlike many respiratory viruses that trigger immunopathology through cytokine storms, HPIV-3 appears to induce inflammation directly through viral antigen interactions with host cells. Culture supernatants from infected cells contain minimal levels of pro-inflammatory cytokines like IFN-γ, TGF-β, and TNF-α .

These findings suggest that HPIV-3 immunopathology may result from direct virus-host cell interactions rather than dysregulated cytokine production. This has significant implications for therapeutic strategies, suggesting that approaches targeting viral replication directly may be more effective than broad immunomodulatory interventions.

What animal models best represent HPIV-3 infection dynamics, and what are their limitations?

Several animal models have been developed for HPIV-3 research, each with specific advantages and limitations:

Table 1: Comparison of Animal Models for HPIV-3 Research

Animal ModelAdvantagesLimitationsBest Research Applications
AG129 Mice- Support viral replication in upper and lower airways
- Develop bronchopneumonia and pneumocyte hyperplasia
- Allow assessment of antiviral compounds
- Lack functional IFNα/β and IFNγ receptors
- May not fully represent immunocompetent host responses
- No virus transmission between animals
- Antiviral efficacy testing
- Viral pathogenesis studies
- Initial therapeutic screening
Cotton Rats- Support lower respiratory tract viral replication
- Previously used to predict efficacy of antibody immunoprophylaxis
- More permissive than standard mice
- Limited reagents available
- Not as well characterized as mouse models
- May not fully represent human disease
- Antibody neutralization studies
- Prophylactic intervention testing
- Correlates of protection studies
Standard Mice- Widely available
- Well-characterized immune system
- Numerous genetic modifications available
- Do not support efficient HPIV-3 replication
- Require model adaptation
- Not recommended for HPIV-3 research

Cotton rats have been validated for antibody testing, including determining effective dosing of neutralizing antibodies. This model was used to assess the neutralizing antibody PI3-E12, demonstrating protection at doses of 0.625–5 mg/kg when administered intramuscularly prior to infection .

Researchers should select their model based on specific research questions, recognizing that no single model fully recapitulates human HPIV-3 infection.

What methodological approaches are most effective for analyzing HPIV-3 genetic diversity in nosocomial outbreak investigations?

Nosocomial HPIV-3 outbreaks represent significant challenges in healthcare settings, particularly for immunocompromised patients. Effective genetic analysis approaches include:

  • Targeted sequencing of hemagglutinin-neuraminidase (HN) gene variable regions: Sequencing specific variable regions (nt 1–569 and nt 762–1239) of the HN gene provides sufficient discriminatory power to identify strain relatedness. This approach successfully identified multiple distinct clusters during a multicluster outbreak, revealing that 16/32 strains were identical in one major cluster, with three additional smaller clusters .

  • Phylogenetic analysis: Construction of phylogenetic trees using sequence data from the HN gene enables visualization of strain relationships and transmission patterns. This methodology is essential for distinguishing between a single introduction with subsequent spread versus multiple independent introductions .

  • Integration with epidemiological data: Genetic analysis should be combined with detailed patient location tracking, contact tracing, and temporal analysis to establish likely transmission routes. In the documented outbreak, phylogenetic analysis coupled with epidemiological investigation revealed distinct transmission chains within a pediatric oncohematology unit .

For comprehensive outbreak investigations, researchers should:

  • Collect samples from all suspected cases

  • Process samples consistently using standardized protocols

  • Sequence multiple viral genes when possible

  • Construct phylogenetic trees using appropriate evolutionary models

  • Correlate genetic findings with spatial and temporal epidemiological data

This methodological approach enables evidence-based infection control interventions by distinguishing between environmental persistence, healthcare worker transmission, and multiple community introductions.

What are the current experimental approaches to developing neutralizing antibodies against HPIV-3, and how is their efficacy evaluated?

Current experimental approaches to developing neutralizing antibodies against HPIV-3 follow a multi-step process:

  • Isolation of B cells from convalescent donors: Researchers isolate memory B cells from individuals who have recovered from HPIV-3 infection, using flow cytometry to identify and sort antigen-specific B cells .

  • Single-cell PCR amplification: Individual B cells are processed for PCR amplification of heavy and light chain variable regions, which are then cloned into expression vectors .

  • Recombinant antibody expression: Cloned antibody genes are expressed in mammalian cell lines to produce recombinant monoclonal antibodies for testing .

  • Epitope mapping: Advanced techniques including cryo-electron microscopy and hydrogen-deuterium exchange mass spectrometry are employed to precisely define antibody binding sites on viral proteins .

Efficacy evaluation follows a structured progression:

Table 2: Hierarchical Evaluation Pipeline for Anti-HPIV-3 Neutralizing Antibodies

Evaluation StageMethodologyKey MetricsExample from Research
In vitro neutralizationPlaque reduction neutralization testIC50/IC90 valuesPI3-E12 demonstrated potent neutralization in cell culture
Binding characterizationSurface plasmon resonance/Bio-layer interferometryBinding affinity (KD), on/off ratesMost potent antibodies targeted the F protein apex (site Ø)
Conformational specificityELISA using pre- and post-fusion F proteinsSelective binding to prefusion conformationsPI3-E12 bound specifically to prefusion F, unlike antibody PIA174
Prophylactic efficacyCotton rat model, antibody administration prior to challengeReduction in viral titers in lung tissuePI3-E12 at 0.625–5 mg/kg provided protection against 10^5 pfu challenge
Therapeutic efficacyImmunocompromised animal models, antibody administration post-infectionViral clearance, symptom reductionPI3-E12 demonstrated suppression of viral replication in immunocompromised animals

The most promising candidate identified in recent research, PI3-E12, targets the site Ø on the fusion (F) protein and demonstrated both prophylactic and therapeutic efficacy in animal models . This methodological pipeline represents the current gold standard for evaluating potential therapeutic antibodies against HPIV-3.

How do research methodologies differ when studying HPIV-3 in immunocompetent versus immunocompromised populations?

Research methodologies for studying HPIV-3 require significant adaptations when transitioning between immunocompetent and immunocompromised populations:

  • Study Design Considerations:

    • Immunocompetent studies typically focus on acute infection dynamics, viral clearance, and development of protective immunity.

    • Immunocompromised studies must address prolonged viral shedding, risk of nosocomial transmission, and potential for viral evolution during persistent infection .

  • Sampling Protocols:

    • Immunocompetent protocols may prioritize upper respiratory sampling (nasopharyngeal aspirates) at limited timepoints.

    • Immunocompromised protocols should include both upper and lower respiratory sampling (including bronchoalveolar lavage when indicated) with longitudinal collection to track viral persistence and evolution .

  • Viral Load Quantification:

    • Both populations require sensitive RT-PCR quantification, but immunocompromised studies should employ lower detection thresholds and monitor for longer periods (weeks to months rather than days) .

  • Immunological Assessments:

    • Immunocompetent assessments focus on typical immune response markers including neutralizing antibody titers and T-cell responses.

    • Immunocompromised assessments must be tailored to the specific immune defect (e.g., post-HSCT patients require evaluation of reconstituting immune compartments) .

  • Therapeutic Evaluation:

    • Immunocompetent therapeutic studies primarily assess reduction in symptom duration and viral load.

    • Immunocompromised therapeutic studies must evaluate capacity to clear persistent infection and prevent progression to lower respiratory disease .

Research findings from immunocompromised populations highlight the profound impact of immune status on HPIV-3 infection outcomes. In one documented outbreak among pediatric oncology patients, of 32 HPIV-3-positive patients (19 HSCT recipients, 8 with hematologic malignancies, and 5 immunocompetent children), 16 developed lower respiratory tract infections with extremely high viral loads (>1.0×10^6 RNA copies/mL). One immunocompromised patient died from respiratory failure with high viral load in bronchoalveolar lavage .

These differences necessitate specialized research approaches when studying HPIV-3 in vulnerable immunocompromised populations.

What experimental compounds have demonstrated efficacy against HPIV-3 in preclinical models?

Recent research has identified several promising experimental compounds with demonstrated efficacy against HPIV-3 in preclinical models:

  • Neutralizing antibodies targeting the F protein:

    • PI3-E12: This antibody targets the apex region (site Ø) of the HPIV-3 fusion protein and has demonstrated both prophylactic and therapeutic efficacy in cotton rat models. When administered prophylactically at doses of 0.625–5 mg/kg, it provided protection against challenge with 10^5 plaque-forming units of HPIV-3. Importantly, PI3-E12 also demonstrated therapeutic potential in immunocompromised animal models .

  • Nucleoside analog inhibitors:

    • GS-441524: This compound, the parent nucleoside of remdesivir, has shown significant antiviral activity against HPIV-3 in AG129 mouse models. Oral treatment with GS-441524 reduced infectious virus titers in the lung and preserved normal lung histology. Intranasal administration also demonstrated antiviral effects, suggesting potential for both systemic and localized delivery methods .

  • Other neutralizing antibodies with potential:

    • Additional antibodies targeting non-overlapping epitopes of the HPIV-3 F protein have been isolated from human B cells, with varying degrees of neutralizing potency. While not all have been tested in animal models, their identification provides a pipeline of potential therapeutic candidates for further development .

What methodological challenges exist in designing clinical trials for HPIV-3 therapeutics?

Designing clinical trials for HPIV-3 therapeutics presents numerous methodological challenges that researchers must address:

  • Patient Population Heterogeneity:

    • HPIV-3 affects diverse populations from healthy children to severely immunocompromised patients, each with different disease manifestations and outcomes.

    • Trials must define inclusion criteria carefully, potentially stratifying by age, immune status, and presence of comorbidities .

  • Endpoint Selection Complexities:

    • Defining appropriate endpoints is challenging due to the variable natural history of infection.

    • Potential endpoints include viral load reduction, symptom duration, progression to lower respiratory tract infection, need for supplemental oxygen, or hospital length of stay.

    • For immunocompromised populations, viral clearance and prevention of prolonged shedding may be more relevant than symptom improvement .

  • Diagnostic Standardization:

    • Inconsistent diagnostic methodologies between clinical sites can complicate trial execution.

    • Standardized PCR protocols with established cycle threshold cutoffs are essential for accurate patient selection and outcome assessment .

  • Seasonal Variability and Enrollment Challenges:

    • HPIV-3 demonstrates seasonal patterns with varying intensity year to year.

    • Trials must account for seasonal variation in background immunity and viral transmission dynamics.

    • Rapid identification and enrollment systems are needed to capture patients early in their disease course .

  • Control Group Considerations:

    • For severe infections in immunocompromised patients, placebo-controlled trials may raise ethical concerns.

    • Adaptive trial designs or comparison to standardized historical controls may be necessary alternatives .

Researchers designing HPIV-3 therapeutic trials should consider these methodological challenges early in protocol development, potentially employing adaptive trial designs, multicenter coordination, and year-round recruitment strategies to overcome these obstacles.

How do viral mechanisms of immune evasion impact therapeutic development strategies?

HPIV-3 employs several immune evasion mechanisms that significantly impact therapeutic development strategies:

  • Modulation of Cell Surface Immune Molecules:

    • HPIV-3 upregulates ICAM-1 (CD54) in infected cells through a direct viral antigen-mediated mechanism rather than through cytokine induction .

    • The virus also induces MHC class I and II expression on lung epithelial cells, potentially altering normal antigen presentation pathways .

    • Therapeutic implication: Treatments targeting inflammatory pathways may have limited efficacy since viral-induced inflammation occurs through direct cellular mechanisms rather than through classic cytokine cascades.

  • JAK/STAT Signaling Independence:

    • HPIV-3's ability to induce ICAM-1 independently of JAK/STAT signaling indicates the virus has evolved mechanisms to bypass normal immune activation pathways .

    • Therapeutic implication: JAK inhibitors and other immunomodulatory drugs targeting this pathway may have limited utility against HPIV-3-induced pathology.

  • Reinfection Capacity:

    • Unlike measles or mumps viruses, HPIV-3 can cause reinfection despite prior exposure, suggesting sophisticated evasion of long-term protective immunity .

    • Therapeutic implication: Antibody-based therapies must target highly conserved epitopes to prevent escape mutants, and combination approaches may be necessary for effective treatment.

  • Viral Persistence in Immunocompromised Hosts:

    • HPIV-3 can establish persistent infection in immunocompromised individuals, potentially evolving within a single host over time .

    • Therapeutic implication: Antiviral strategies must achieve complete viral clearance rather than simply suppressing viral replication, and resistance monitoring becomes critical during extended treatment courses.

Understanding these immune evasion mechanisms has driven researchers toward two primary therapeutic approaches:

  • Direct-acting antivirals targeting viral replication machinery, such as GS-441524, which circumvent the virus's immune evasion tactics by preventing viral replication directly .

  • Broadly neutralizing antibodies targeting conserved epitopes on the F protein, such as PI3-E12, which can bind and neutralize the virus regardless of its immune evasion capabilities .

These approaches represent the most promising avenues for therapeutic development against this sophisticated viral pathogen.

What gene editing approaches show promise for studying HPIV-3 pathogenesis?

Modern gene editing technologies offer powerful approaches for elucidating HPIV-3 pathogenesis mechanisms:

  • CRISPR/Cas9 Applications:

    • Viral genome modification: CRISPR/Cas9 systems can be used to create recombinant HPIV-3 strains with specific mutations or reporter genes, enabling detailed studies of viral protein functions in replication and pathogenesis.

    • Host factor screening: Genome-wide CRISPR screens in susceptible cell lines can identify essential host factors required for HPIV-3 entry, replication, and immune evasion.

  • Reverse Genetics Systems:

    • Construction of full-length HPIV-3 cDNA clones allows systematic mutation of viral genes to determine their roles in pathogenesis, particularly focusing on the proteins involved in immune modulation identified in previous studies, such as those affecting ICAM-1 and MHC expression .

  • Organoid Models with Gene Editing:

    • Human airway epithelial organoids can be genetically modified using CRISPR to create models with specific genetic backgrounds, particularly useful for studying HPIV-3 interactions with ciliated cells and club cells of the bronchiolar epithelium .

    • These systems would allow detailed investigation of the virus's cellular tropism and pathogenesis in a physiologically relevant context.

  • Transcriptome-Wide Association Studies:

    • Combining RNA-seq approaches with gene editing validation can identify host response pathways that influence HPIV-3 pathogenesis, providing new targets for therapeutic intervention.

These gene editing approaches would address significant knowledge gaps, particularly in understanding the molecular mechanisms behind HPIV-3's ability to induce ICAM-1 independently of cytokine signaling and the virus's capacity to modulate MHC expression in infected cells .

How might next-generation sequencing enhance our understanding of HPIV-3 transmission dynamics?

Next-generation sequencing (NGS) technologies offer transformative approaches for understanding HPIV-3 transmission dynamics:

  • Whole Genome Deep Sequencing:

    • High-throughput sequencing of complete HPIV-3 genomes enables tracking of minor variants and quasispecies within individual patients and across transmission chains.

    • This approach can reveal within-host viral evolution and adaptation, particularly in immunocompromised patients with prolonged infections .

  • Metagenomic Surveillance:

    • NGS of unbiased respiratory samples can detect HPIV-3 alongside co-infecting pathogens, providing ecological context for transmission dynamics.

    • This approach may reveal previously unrecognized interactions between HPIV-3 and other respiratory viruses or bacteria that influence transmission efficiency.

  • Transmission Chain Reconstruction:

    • Building upon previous work using targeted sequencing of the hemagglutinin-neuraminidase gene, whole-genome phylogenetic analysis can provide higher-resolution transmission mapping in outbreak settings .

    • This enhanced resolution allows discrimination between closely related strains and can identify superspreader events or environmental reservoirs.

  • Environmental Sampling and Sequencing:

    • NGS of environmental samples from healthcare settings can identify potential fomite transmission routes and evaluate the effectiveness of infection control measures.

    • This approach could be particularly valuable in understanding nosocomial transmission in healthcare facilities with vulnerable populations .

  • Integration with Contact Tracing Data:

    • Combining genomic data with spatial-temporal contact patterns enables mathematical modeling of HPIV-3 transmission dynamics and effective reproduction numbers under various conditions.

Implementation of these NGS approaches would significantly enhance our understanding of HPIV-3 epidemiology, potentially leading to more effective infection control strategies, particularly in healthcare settings where nosocomial outbreaks pose significant risks to vulnerable populations .

What are the most promising approaches for developing a protective vaccine against HPIV-3?

Despite decades of research, no licensed vaccine exists for HPIV-3. Based on current understanding, several promising approaches warrant further investigation:

  • Live-Attenuated Vaccines:

    • Development of temperature-sensitive mutants that replicate efficiently in the cooler upper respiratory tract but poorly in the warmer lower respiratory tract.

    • Engineering of recombinant viruses with mutations in key virulence or replication genes to create safe, immunogenic candidates.

    • Challenge: Balancing sufficient attenuation with adequate immunogenicity, particularly for vulnerable populations.

  • Subunit and VLP-Based Approaches:

    • Focusing on the F and HN proteins as primary immunogens, particularly targeting the prefusion F conformation that is recognized by the most potent neutralizing antibodies like PI3-E12 .

    • Development of virus-like particles (VLPs) displaying these key antigens in their native conformations.

    • Advantage: Potentially safer profile for immunocompromised populations compared to live-attenuated approaches.

  • mRNA Vaccine Platforms:

    • Adaptation of now-validated mRNA vaccine technology to express HPIV-3 F and/or HN proteins.

    • Potential for rapid development, manufacturing scalability, and precise antigen design.

    • Could incorporate structure-based design to present F protein in its prefusion conformation, similar to successful RSV vaccine approaches.

  • Vectored Vaccine Approaches:

    • Use of viral vectors (adenovirus, VSV, etc.) to express HPIV-3 immunogens.

    • Potential for inducing both humoral and cellular immunity.

    • Could be particularly valuable for immunocompromised populations requiring robust protection.

  • Combined Evaluation Strategies:

    • Implementation of standardized animal models (including AG129 mice and cotton rats) with consistent challenge protocols .

    • Development of immune correlates of protection to accelerate clinical evaluation.

    • Comparison of vaccine-induced antibodies to naturally occurring neutralizing antibodies like PI3-E12 .

The most promising candidates will likely need to induce neutralizing antibodies targeting conserved epitopes on the F protein, particularly site Ø at the apex, which has been identified as the target of potent neutralizing antibodies in recent research .

Product Science Overview

Introduction

Human Parainfluenza Virus Type-3 (HPIV-3) is one of the four types of human parainfluenza viruses (HPIVs) that belong to the Paramyxoviridae family. These viruses are enveloped, negative-sense, single-stranded RNA viruses. HPIV-3 is a significant cause of respiratory infections, particularly in infants, young children, the elderly, and immunocompromised individuals .

Classification

HPIV-3 is classified under the genus Respirovirus within the Paramyxoviridae family. The HPIVs are divided into four types: HPIV-1, HPIV-2, HPIV-3, and HPIV-4, with HPIV-4 further subdivided into HPIV-4A and HPIV-4B .

Epidemiology

HPIV-3 is known for its annual epidemic cycles, typically peaking in late spring or summer . It is the most prevalent type among the HPIVs, causing the majority of infections and outbreaks . The virus spreads through respiratory secretions via close contact with infected individuals or contaminated surfaces .

Clinical Features

HPIV-3 primarily affects the respiratory tract, causing a range of illnesses from mild upper respiratory infections to severe lower respiratory tract diseases such as bronchiolitis, bronchitis, and pneumonia . Symptoms usually develop between 2 to 7 days after exposure and can include fever, runny nose, cough, and sore throat . In severe cases, especially in immunocompromised patients, HPIV-3 can lead to significant morbidity and mortality .

Biological Properties and Mode of Action

HPIV-3 initiates infection using two surface glycoproteins: the haemagglutinin-neuraminidase (HN) and fusion (F) proteins . The HN protein facilitates the attachment of the virus to the host cell, while the F protein mediates the fusion of the viral envelope with the host cell membrane, allowing the viral RNA to enter the host cell and begin replication .

Treatment and Prevention

Currently, there are no specific antiviral treatments or licensed vaccines available for HPIV-3 . Management of HPIV-3 infections primarily involves supportive care to alleviate symptoms. Preventive measures include good hygiene practices such as regular handwashing and avoiding close contact with infected individuals .

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.