HIV-1 Polyvalent

HIV-1 Polyvalent Recombinant
Shipped with Ice Packs
In Stock

Description

Polyvalent HIV1 antigens contain HIV1 gp41 (long sequence) and new discovered HIV1 protein P30 from HIV1 genome.

Product Specs

Description
Polyvalent HIV1 antigens contain HIV1 gp41 (long sequence) and new discovered HIV1 protein P30 from HIV1 genome.
Physical Appearance
Sterile Filtered clear solution.
Formulation
HIV-1 Polyvalent solution contains 25mM K2CO3 & PBS.
Stability
Store at 4°C if entire vial will be used within 2-4 weeks. Store, frozen at -20°C for longer periods of time. For long term storage it is recommended to add a carrier protein (0.1% HSA or BSA). Avoid multiple freeze-thaw cycles.
Purity
Protein is >95% pure as determined by 10% PAGE (coomassie staining).
Applications
Immunoassay.
Source

Escherichia Coli.

Q&A

What is a polyvalent HIV-1 vaccine and why is this approach necessary?

A polyvalent HIV-1 vaccine contains multiple HIV-1 antigens from different viral strains or clades, designed to address the remarkable genetic diversity of HIV-1. This approach is necessary because the diversity of HIV-1 subtypes and high frequency of viral mutations has virtually eliminated the possibility of developing an effective vaccine based on a single natural HIV-1 envelope (Env) antigen .

The scientific rationale for polyvalent approaches includes:

  • Evidence that combinations of neutralizing monoclonal antibodies from different sources provide broader neutralizing activities against heterologous isolates

  • The existence of functional constraints in HIV-1 proteins that limit structural diversity despite sequence variation

  • Observations that HIV superinfection, while possible, is relatively rare, suggesting some cross-protection mechanisms may exist

For nearly three decades, HIV vaccine development has focused on either inducing T cell immune responses or antibody responses, rarely addressing both components simultaneously. The polyvalent approach aims to overcome this limitation by generating broader immune responses targeting multiple viral epitopes .

How do polyvalent HIV-1 vaccines differ from traditional monovalent approaches?

Polyvalent HIV-1 vaccines differ from monovalent approaches in several fundamental ways:

  • Antigen composition: While monovalent vaccines contain a single HIV-1 antigen (typically from one viral strain), polyvalent formulations include multiple antigens representing different HIV-1 clades or variants.

  • Immune response breadth: Polyvalent formulations have demonstrated the ability to generate antibody responses with greater breadth against heterologous HIV-1 isolates. Studies show that the breadth of antibody responses can be significantly improved when multiple gp120 antigens are delivered in a polyvalent formulation .

  • Neutralization capacity: Polyvalent approaches can elicit neutralizing antibodies against primary HIV-1 isolates that are typically resistant to neutralization, including viruses from clades not represented in the vaccine formulation .

  • T-cell response diversity: Polyvalent vaccines generate T-cell responses against a wider array of epitopes, potentially reducing the likelihood of viral escape and providing recognition of diverse viral sequences.

  • Manufacturing complexity: Polyvalent vaccines are more complex to develop, requiring consistent production of multiple antigens with preserved conformational epitopes.

What are the key immunological challenges in HIV-1 vaccine development?

HIV-1 vaccine development faces several critical immunological challenges that must be addressed for successful protection:

  • Viral diversity: HIV-1's remarkable genetic diversity presents a fundamental challenge, with multiple clades (subtypes) circulating globally. After the STEP trial failure, it became evident that T cell-based vaccines would likely not be protective if responses were limited to a few dominant epitopes .

  • Rapid mutation: HIV-1's high mutation rate allows it to quickly escape immune recognition, requiring vaccines to target conserved viral regions or generate extremely broad responses.

  • Establishment of viral reservoirs: HIV-1 rapidly establishes latent reservoirs that persist even during antiretroviral therapy, making sterilizing immunity difficult to achieve.

  • Immune evasion mechanisms: HIV-1 employs numerous strategies to evade immune detection, including glycan shielding of key epitopes, conformational masking, and targeted destruction of CD4+ T cells.

  • Correlates of protection: The precise immune responses required for protection remain incompletely understood, with broadly neutralizing antibodies difficult to elicit through conventional vaccination.

  • Mucosal immunity: As sexual transmission is the primary route of HIV-1 infection, vaccines must generate effective mucosal immune responses, which are challenging to induce through parenteral immunization.

Following VaxGen's failed Phase III trials, a key criticism was the limited antigen breadth in the vaccine formulations. Polyvalent approaches directly address this limitation by including multiple viral antigens to expand immunological coverage .

What strategies exist for selecting HIV-1 envelope proteins for polyvalent formulations?

Several sophisticated strategies have been developed for optimal selection of HIV-1 envelope proteins in polyvalent vaccine formulations:

  • Clade representation: Selecting Env proteins that represent major circulating HIV-1 clades (A, B, C, D, E, etc.) to provide broad coverage of global viral diversity. This approach was used in clinical studies combining DNA vaccines expressing env genes from multiple HIV-1 clades with protein boosts .

  • Antigenicity-based selection: Choosing Env variants based on their antigenicity profiles:

    • Binding patterns with broadly neutralizing antibodies

    • Sensitivity to neutralization by polyclonal sera from HIV-1 infected individuals

    • Complementary neutralization sensitivity patterns

  • Evolutionary distance-based selection: Selecting Env proteins that maximize the evolutionary distance between included variants to cover as much of the viral phylogenetic tree as possible.

  • Natural escape variant inclusion: Incorporating Env proteins from documented cases of viral escape to account for common immune evasion strategies. Studies have evaluated the use of natural escape mutant viruses as part of polyvalent formulations .

  • Neutralization fingerprinting: Selecting variants with distinct neutralization sensitivity patterns to induce complementary antibody responses.

These strategies are not mutually exclusive, and effective polyvalent formulations often employ multiple selection criteria to optimize both the breadth and potency of immune responses.

How do DNA-based vaccination approaches facilitate polyvalent HIV-1 vaccine development?

DNA-based vaccination approaches have significantly expanded the technical flexibility of testing polyvalent HIV vaccines through several key advantages:

  • Simplified formulation testing: DNA vaccines with multiple env gene inserts can be tested directly, avoiding the more complicated process of producing recombinant Env proteins .

  • Antigen co-delivery: Several Env-expressing DNA vaccines can be mixed and delivered in a single formulation without concerns about potential interactions among different recombinant Env protein antigens .

  • Flexible prime-boost strategies: DNA vaccines can be effectively combined with protein boost components to enhance the immunogenicity of polyvalent formulations, as demonstrated in clinical studies .

  • In vivo expression advantages: DNA vaccines lead to in vivo synthesis of antigens, potentially resulting in proper protein folding and post-translational modifications.

  • Balanced immune responses: DNA vaccines can induce both humoral and cellular immune responses, critical for comprehensive immunity against HIV-1.

A Phase I clinical trial evaluated a multi-gene, polyvalent formulation consisting of DNA vaccines expressing env genes from multiple clades of HIV-1 and a gag gene from a single clade, followed by a gp120 protein boost. This approach demonstrated robust cross-subtype HIV-1-specific T cell responses and high-titer serum antibody responses .

What methodologies are used to evaluate cross-clade protection in polyvalent HIV-1 vaccines?

Evaluating cross-clade protection for polyvalent HIV-1 vaccines requires sophisticated methodologies that assess immune responses against diverse viral strains:

  • Neutralization assays with global virus panels:

    • Testing vaccine-induced sera against standardized panels of HIV-1 pseudoviruses representing global diversity

    • Categorizing viruses by neutralization sensitivity (Tier 1, 2, and 3)

    • Calculating breadth (percentage of strains neutralized) and potency (geometric mean titer)

  • T-cell epitope mapping across clades:

    • Using peptide pools representing different HIV-1 clades to stimulate vaccine-induced T cells

    • Measuring cross-reactive T-cell responses via ELISpot or intracellular cytokine staining

    • Identifying conserved epitopes recognized across multiple HIV-1 subtypes

  • Challenge studies in animal models:

    • SHIV (simian-human immunodeficiency virus) challenge in non-human primates with heterologous Env

    • Evaluation of protection against viruses containing Env from clades not in the vaccine

    • Assessment of post-challenge viral load and CD4+ T-cell preservation

  • Sieve analysis of breakthrough infections:

    • Comparing viral sequences from vaccinated and unvaccinated individuals who become infected

    • Identifying genetic signatures associated with vaccine escape

    • Determining if the vaccine exerted selective pressure on certain viral epitopes

Studies have demonstrated that polyvalent approaches can elicit immune responses against HIV-1 isolates from clades not represented in the vaccine formulation, highlighting the potential for cross-clade protection .

How are heterologous prime-boost strategies optimized for polyvalent HIV-1 vaccines?

Optimization of heterologous prime-boost strategies for polyvalent HIV-1 vaccines involves several critical considerations:

  • Platform selection and sequencing:

    • DNA priming followed by protein boosting has shown success in generating both cellular and humoral responses

    • Viral vector priming (e.g., adenovirus or poxvirus) followed by protein boosting can enhance immunogenicity

    • The specific order of different platforms significantly impacts immune response quality

  • Timing optimization:

    • Interval between prime and boost doses affects response magnitude and quality

    • Longer intervals (3-6 months) often generate more durable responses than shorter intervals

    • Multiple boost doses may be required for optimal response maturation

  • Antigen modifications between prime and boost:

    • Using identical antigens maintains focus on specific epitopes

    • Sequential exposure to slightly varied antigens can guide antibody maturation

    • Presenting different forms of Env (e.g., gp120 vs. gp140) in prime versus boost may target different epitopes

  • Adjuvant selection:

    • Different adjuvants for prime versus boost can activate complementary immune pathways

    • Tailoring adjuvants to specific delivery platforms enhances response quality

    • Novel adjuvant combinations may preferentially enhance either cellular or humoral immunity

Studies have demonstrated that DNA vaccines expressing env genes from multiple HIV-1 clades, when combined with a gp120 protein boost, can elicit neutralizing antibodies against both homologous strains and heterologous HIV-1 isolates, including those from clades not present in the vaccine formulation .

The RV144 Thai trial, which showed modest protection (31% efficacy), employed a heterologous prime-boost strategy combining a recombinant canarypox vector vaccine with a bivalent recombinant gp120 subunit vaccine, suggesting the potential of this approach when combined with polyvalent antigen formulations .

What clinical trial data exists for polyvalent HIV-1 vaccines?

Several polyvalent HIV-1 vaccine candidates have advanced to clinical trials, with varying degrees of success:

  • Multi-gene, polyvalent DNA plus protein boost approach:
    A Phase I clinical trial evaluated a formulation consisting of:

    • DNA vaccines expressing env genes from multiple HIV-1 clades

    • A gag gene from a single HIV-1 clade

    • A gp120 protein boost homologous to the DNA vaccine components

    Results demonstrated:

    • Robust cross-subtype HIV-1-specific T cell responses

    • High-titer serum antibody responses

    • Safety and tolerability in human subjects

  • RV144 Thai Trial:
    This trial employed a prime-boost strategy with:

    • Recombinant canarypox vector vaccine vCP1521 (prime)

    • Bivalent recombinant gp120 subunit vaccine AIDSVAX B/E (boost)

    The trial showed:

    • 31% decrease in HIV-1 infection in vaccinated individuals compared to placebo

    • Protection identified in the early phase of HIV-1 infection

    • Evidence that antibody responses may have played an important role in protection

  • HVTN 111 Trial:
    This study evaluated:

    • DNA prime (multiple env genes and gag)

    • Protein boost with different adjuvants

    • Safety and immunogenicity in healthy adults

These clinical trials have demonstrated the feasibility and immunogenicity of polyvalent HIV-1 vaccine approaches. The modest efficacy signal in the RV144 trial, while not sufficient for vaccine approval, provided proof-of-concept that protection against HIV-1 acquisition is possible and supported further development of polyvalent vaccination strategies.

How does the breadth of neutralizing antibody responses compare between monovalent and polyvalent HIV-1 vaccines?

Comparative studies between monovalent and polyvalent HIV-1 vaccines reveal significant differences in neutralizing antibody response breadth:

  • Cross-clade neutralization capacity:

    • Polyvalent formulations consistently demonstrate the ability to induce antibodies that neutralize a wider range of HIV-1 isolates

    • Studies show that gp120 antigens delivered in a polyvalent formulation significantly improve the breadth of antibody responses

    • Polyvalent approaches can elicit neutralizing antibodies against primary HIV-1 isolates that are typically resistant to neutralization

  • Heterologous strain coverage:

    • Polyvalent vaccines can generate antibodies that neutralize HIV-1 isolates from clades not included in the vaccine formulation

    • For example, studies have demonstrated neutralization of clade A isolates by antibodies induced by vaccines that did not include clade A antigens

  • Neutralization potency versus breadth:

    • While breadth is typically improved with polyvalent formulations, the potency of neutralization against any individual strain may be less compared to a monovalent vaccine optimized for that specific strain

    • This represents an important trade-off in polyvalent vaccine design

  • Conserved epitope targeting:

    • Polyvalent formulations may better focus the immune response on conserved epitopes shared across diverse HIV-1 strains

    • This focusing effect likely contributes to the improved breadth of neutralization observed with polyvalent approaches

These findings suggest that polyvalent HIV-1 vaccines offer significant advantages for generating broadly neutralizing antibody responses, a critical requirement for effective protection against the diverse HIV-1 strains circulating globally.

What insights have been gained from the study of T-cell responses to polyvalent HIV-1 vaccines?

Studies of T-cell responses to polyvalent HIV-1 vaccines have yielded several important insights:

  • Cross-reactive T-cell generation:

    • Polyvalent formulations induce T cells capable of recognizing epitopes from multiple HIV-1 clades

    • This cross-reactivity results from recognition of conserved epitopes and related variant epitopes

    • Broader T-cell recognition potentially reduces the likelihood of viral escape

  • Balanced CD4+ and CD8+ responses:

    • Polyvalent HIV-1 vaccines can generate both helper CD4+ T-cell responses and cytotoxic CD8+ T-cell responses

    • This balance is crucial since CD4+ T cells provide essential help for antibody development while CD8+ T cells directly target infected cells

  • Epitope hierarchy effects:

    • Inclusion of multiple HIV-1 antigens can alter immunodominance patterns observed with single antigens

    • This can lead to recognition of subdominant epitopes that might be more conserved across viral variants

    • Broader epitope targeting may contribute to improved vaccine efficacy

  • Polyfunctional T-cell induction:

    • Polyvalent vaccines have demonstrated the ability to generate T cells with multiple effector functions

    • These polyfunctional T cells (producing multiple cytokines/chemokines) are associated with better antiviral efficacy

Studies have shown that polyvalent formulations containing env genes from multiple clades of HIV-1 and a gag gene from a single clade can elicit robust cross-subtype HIV-1-specific T-cell responses, demonstrating the potential of polyvalent approaches to generate broadly reactive cellular immunity .

What manufacturing and quality control considerations are unique to polyvalent HIV-1 vaccines?

Polyvalent HIV-1 vaccines present unique manufacturing and quality control challenges that require specialized approaches:

  • Antigen production complexity:

    • Ensuring consistent production of multiple antigens with preserved conformational epitopes

    • Managing increased process development requirements for diverse components

    • Developing scalable manufacturing protocols for different HIV-1 Env proteins

    • Maintaining stability of complex formulations containing multiple antigens

  • Quality control requirements:

    • Identity testing for each component in the polyvalent formulation

    • Potency assays that evaluate each antigen independently and in combination

    • Stability monitoring of all components under various storage conditions

    • Batch-to-batch consistency testing for complex formulations

  • Formulation considerations:

    • Optimizing antigen ratios to prevent immunodominance of specific components

    • Ensuring compatibility between multiple antigens in a single formulation

    • Selecting appropriate adjuvants that enhance responses to all components

    • Preventing negative interference between components

  • Regulatory challenges:

    • Developing appropriate reference standards for complex formulations

    • Establishing acceptance criteria for each component and the complete formulation

    • Designing lot release tests that adequately characterize polyvalent products

    • Meeting regulatory expectations for demonstration of consistency

These considerations highlight the technical complexity of developing polyvalent HIV-1 vaccines and underscore the need for specialized expertise and infrastructure in vaccine manufacturing and quality control.

How might next-generation technologies advance polyvalent HIV-1 vaccine development?

Several emerging technologies show significant promise for advancing polyvalent HIV-1 vaccine development:

  • Structure-based vaccine design:

    • Using high-resolution structures of HIV-1 Env to design improved immunogens

    • Computational design of antigens that focus immune responses on conserved epitopes

    • Structure-guided stabilization of Env trimers in native-like conformations

    • Design of chimeric antigens that present multiple conserved epitopes from diverse strains

  • mRNA vaccine platforms:

    • Rapid production of multiple antigens for polyvalent formulations

    • Ability to co-express different HIV-1 antigens from a single construct

    • Potential for self-amplifying mRNA to enhance antigen expression

    • Simplified manufacturing processes for complex polyvalent formulations

  • Nanoparticle delivery systems:

    • Multivalent display of HIV-1 antigens on nanoparticle surfaces

    • Co-delivery of antigens and immune stimulatory molecules

    • Targeted delivery to specific immune cell populations

    • Enhanced germinal center reactions for improved antibody development

  • Germline-targeting approaches:

    • Sequential immunization strategies to guide B-cell maturation

    • Design of immunogens that activate precursors of broadly neutralizing antibody lineages

    • Combination of germline-targeting antigens with polyvalent boosting

    • Personalized vaccination based on B-cell receptor repertoire analysis

  • Systems biology integration:

    • Comprehensive profiling of vaccine responses using multi-omics approaches

    • Computational modeling to predict optimal antigen combinations

    • Machine learning to identify correlates of protection

    • Network analysis to understand interactions between different immune components

These technologies may overcome current limitations in polyvalent HIV-1 vaccine development and lead to more effective vaccination strategies for this challenging pathogen.

What lessons from other polyvalent vaccine successes might apply to HIV-1 vaccine development?

Successful polyvalent vaccines for other pathogens offer valuable lessons that may inform HIV-1 vaccine development:

  • Influenza vaccines:

    • Annual reformulation based on surveillance data demonstrates the feasibility of updating polyvalent formulations

    • Quadrivalent vaccines containing multiple influenza strains show that balanced immune responses are possible

    • High-dose formulations for elderly populations suggest antigen dose optimization may be crucial for vulnerable groups

    • Universal influenza vaccine research provides insights on targeting conserved epitopes across diverse strains

  • Human papillomavirus (HPV) vaccines:

    • Progression from quadrivalent to nonavalent formulations demonstrates the feasibility of increasing valency

    • Long-term protection suggests durable immunity is possible with polyvalent formulations

    • Cross-protection against non-vaccine HPV types provides evidence for epitope sharing benefits

    • Success in preventing HPV-associated cancers demonstrates the value of focusing on disease endpoints

  • Pneumococcal conjugate vaccines:

    • Expansion from 7 to 13 to 15 serotypes shows manufacturing scalability for increasingly complex formulations

    • Herd immunity effects highlight population-level benefits even with partial serotype coverage

    • Serotype replacement phenomena underscore the importance of strain selection and surveillance

    • Carrier protein conjugation strategies suggest potential advantages of presenting HIV-1 antigens in specific molecular contexts

  • Translation to HIV-1 vaccine development:

    • Rational selection of strains based on global epidemiology

    • Optimization of manufacturing processes for consistent production of multiple antigens

    • Implementation of robust surveillance systems to monitor for viral escape

    • Development of adjuvant formulations that enhance responses to all components

The successes of polyvalent vaccine approaches against other antigenically variable pathogens encourage implementation of similar strategies for the design of HIV-1 vaccines .

How might polyvalent approaches be integrated with other HIV prevention strategies?

Integrating polyvalent HIV-1 vaccines with other prevention strategies offers promising opportunities for comprehensive HIV control:

  • Combination with pre-exposure prophylaxis (PrEP):

    • Polyvalent vaccines could provide durable background protection while PrEP offers high efficacy during periods of exposure

    • Vaccines might reduce the need for strict PrEP adherence

    • Combined approaches could address both sexual and mother-to-child transmission routes

    • Sequential or simultaneous implementation strategies could be evaluated for maximum impact

  • Integration with broadly neutralizing antibody (bNAb) delivery:

    • Passive immunization with bNAbs could complement active immunization with polyvalent vaccines

    • Long-acting bNAb formulations might provide immediate protection while vaccine-induced responses develop

    • Polyvalent vaccines could boost and broaden responses initially primed by bNAb treatment

    • Combined approaches could target different viral epitopes for more comprehensive coverage

  • Synergy with treatment as prevention:

    • Polyvalent vaccines could reduce transmission in populations where antiretroviral therapy (ART) coverage is incomplete

    • Vaccination might help control viral rebound in individuals with suboptimal ART adherence

    • Combined approaches could accelerate progress toward HIV elimination in high-prevalence regions

    • Mathematical modeling could identify optimal implementation strategies for specific contexts

  • Implementation science considerations:

    • Developing integrated delivery platforms for multiple prevention modalities

    • Training healthcare workers to provide comprehensive prevention packages

    • Addressing potential risk compensation following vaccination

    • Designing monitoring and evaluation systems for combination prevention programs

Integrating polyvalent vaccines with existing prevention tools could provide synergistic effects and potentially overcome the limitations of individual approaches.

Product Science Overview

Introduction

Human Immunodeficiency Virus type 1 (HIV-1) is a rapidly evolving virus that poses significant challenges for vaccine development due to its high genetic diversity. The virus is classified into four groups: M (main), O (outlier), N (non-M/non-O), and P (pending). Group M is further subdivided into nine distinct subtypes and numerous additional circulating recombinant forms (CRFs). This diversity necessitates the development of polyvalent vaccines that can elicit broad immune responses.

HIV-1 Polyvalent Recombinant Vaccines

Polyvalent recombinant vaccines are designed to target multiple antigens or strains of a virus to provide broader protection. In the context of HIV-1, these vaccines aim to induce immune responses against various subtypes and CRFs of the virus. The development of such vaccines involves the use of recombinant DNA technology to create vaccine candidates that express multiple HIV-1 antigens.

Mechanism of Action

The primary goal of HIV-1 polyvalent recombinant vaccines is to elicit broadly neutralizing antibodies (bNAbs) and T-cell responses. These vaccines typically include multiple HIV-1 antigens, such as Gag, Pol, and Env proteins, which are crucial for viral replication and entry into host cells. By presenting these antigens to the immune system, the vaccines aim to stimulate the production of bNAbs that can neutralize diverse HIV-1 strains.

Challenges and Strategies

One of the major challenges in developing HIV-1 polyvalent recombinant vaccines is the extreme genetic diversity of the virus. The high mutation rate and recombination events contribute to the rapid evolution of HIV-1, making it difficult to design a vaccine that can provide long-lasting protection. To address this, researchers are exploring various strategies, including:

  1. Sequential Vaccination: Administering a series of vaccines that target different HIV-1 antigens to facilitate B-cell maturation and the production of bNAbs.
  2. Prime-Boost Regimens: Combining different types of vaccines, such as DNA vaccines and viral vector vaccines, to enhance immune responses.
  3. Adjuvants: Using adjuvants, such as IL-12 plasmid DNA, to boost the immune response to the vaccine antigens.
Recent Developments

Recent studies have shown promising results in the development of HIV-1 polyvalent recombinant vaccines. For example, a phase 1 clinical trial tested the safety and tolerability of an HIV-1 multiantigen DNA vaccine co-administered with IL-12 plasmid DNA via electroporation. The vaccine regimen was found to be safe and generally well-tolerated, with some participants experiencing mild to moderate reactogenicity .

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.