DVR Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
DVR antibody; PCB2 antibody; At5g18660 antibody; T1A4.40Divinyl chlorophyllide a 8-vinyl-reductase antibody; chloroplastic antibody; EC 1.3.1.75 antibody; Protein PALE-GREEN AND CHLOROPHYLL B REDUCED 2 antibody
Target Names
DVR
Uniprot No.

Target Background

Function
This antibody targets the DVR enzyme, which catalyzes the conversion of divinyl chlorophyllide to monovinyl chlorophyllide. This process involves the reduction of the 8-vinyl group of the tetrapyrrole to an ethyl group, utilizing NADPH as the reductant. The enzyme exhibits optimal activity with (3,8-divinyl)-chlorophyllide a (DV-Chlidea). It demonstrates very low activity with (3,8-divinyl)-protochlorophyllide a (DV-Pchlidea) and (3,8-divinyl)-magnesium-protoporphyrin IX monomethyl ester (DV-MPE). Notably, the enzyme shows no activity with (3,8-divinyl)-chlorophyllide b (DV-Chlideb), (3,8-divinyl)-magnesium-protoporphyrin IX (DV-Mg-Proto), or either (3,8-divinyl)-chlorophyll a (DV-Chla) or b (DV-Chlb).
Gene References Into Functions
  1. An Arabidopsis mutant accumulating divinyl chlorophyll led to the identification of a gene displaying sequence similarity with isoflavone reductase genes. This gene, DVR, plays a crucial role in monovinyl chlorophyll synthesis. [DVR] PMID: 15632054
Database Links

KEGG: ath:AT5G18660

STRING: 3702.AT5G18660.1

UniGene: At.21772

Subcellular Location
Plastid, chloroplast.
Tissue Specificity
Highly expressed in leaves, stems and flower buds. Detected in roots.

Q&A

What is the mechanism of action for the antibody component in DVR-d therapy?

The antibody component in DVR-d therapy, daratumumab (Darzalex), is a monoclonal antibody that targets CD38, a surface protein highly expressed on multiple myeloma cells. Its mechanism involves multiple immune-mediated actions including complement-dependent cytotoxicity, antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and direct induction of apoptosis. The synergistic effects in the DVR-d regimen occur because the proteasome inhibitor (Velcade/bortezomib) increases CD38 expression on myeloma cells, making them more susceptible to daratumumab-mediated killing, while the immunomodulatory agent (Revlimid/lenalidomide) enhances immune effector cell function, and dexamethasone provides additional anti-myeloma activity .

What factors influence the selection of DVR-d versus other therapeutic regimens?

Selection of DVR-d versus other regimens depends on several clinical and biological factors. Cytogenetics plays a crucial role, as explained by researchers: "Anybody who has high-risk cytogenetics; that is the t(4;14) deletion, 17p, or TP53, even amplification 1q, we tend to use quadruplets such as Darzalex-Velcade-Revlimid-dexamethasone (DVR-d) versus a triplet" . Additional considerations include beta-2 microglobulin, albumin, and lactate dehydrogenase (LDH) test results that help determine disease staging. Higher Revised International Staging System (RISS) stages 2 and 3 often warrant quadruplet therapy. The trend in clinical practice has moved toward quadruplet therapy as standard of care for newly diagnosed patients regardless of risk status, as clinicians note that "more and more, the myeloma community has moved towards a quadruplet in general" .

How do emerging AI technologies impact antibody development for therapies like DVR-d?

Emerging AI technologies are revolutionizing antibody development through sophisticated computational approaches. Vanderbilt University Medical Center's ARPA-H funded project aims to use artificial intelligence to generate antibody therapies against any antigen target of interest. This represents a paradigm shift from traditional methods that are "limited by inefficiency, high costs and fail rates, logistical hurdles, long turnaround times and limited scalability" . The AI approach involves building massive antibody-antigen atlases and developing algorithms to engineer antigen-specific antibodies.

Recent advances in computational biology demonstrate that "a fine-tuned RFdiffusion network is capable of designing de novo antibody variable heavy chains (VHH's) that bind user-specified epitopes" . This enables atomically accurate design of antibodies against specific targets, potentially facilitating development of next-generation therapeutic antibodies with improved binding properties or novel mechanisms of action that could enhance or replace components in regimens like DVR-d.

What are the current challenges in achieving and maintaining MRD negativity with DVR-d therapy?

Achieving and maintaining minimal residual disease (MRD) negativity with DVR-d therapy presents several complex challenges. Technical limitations include standardizing detection methods and addressing potential spatial heterogeneity in bone marrow sampling. As one expert notes, "Reimbursement for the test is a big challenge, as well. For that to become widespread, there has to be solid data" . This highlights the practical implementation barriers despite MRD being recognized as an important clinical endpoint.

Research challenges include determining the optimal duration of MRD negativity required to predict long-term outcomes and understanding patterns of recurrence. Current clinical trials are investigating whether therapy can be discontinued after achieving sustained MRD negativity, potentially reducing toxicity while maintaining efficacy. Additionally, researchers must address the challenge of clonal evolution, where persistent subclones may develop resistance to therapy components over time, necessitating longitudinal monitoring with sensitive techniques.

How can analysis of large-scale antibody databases inform DVR-d component optimization?

Large-scale antibody sequence databases offer unprecedented opportunities for developing next-generation therapeutic antibodies. Research has compiled "four billion productive human heavy variable region sequences and 385 million unique complementarity-determining region (CDR)-H3s" in the AbNGS database . Analysis reveals that "270,000 (0.07% of 385 million) unique CDR-H3s are highly public in that they occur in at least five of 135 bioprojects" , suggesting convergent evolution toward optimal binding solutions.

For DVR-d optimization, these databases can be mined to identify naturally occurring anti-CD38 antibodies with properties that complement or improve upon daratumumab. Statistical analysis of CDR composition in therapeutic-like antibodies can guide rational design of variants with enhanced effector functions or reduced immunogenicity. Machine learning models trained on sequence-function relationships from these databases can predict binding characteristics and developability profiles, potentially addressing resistance mechanisms observed in clinical settings.

What experimental design considerations are essential when evaluating DVR-d in combination with novel agents?

Rigorous evaluation of DVR-d with novel agents requires systematic experimental design. Initial preclinical studies should test mechanism-based synergy hypotheses using cell line panels and patient-derived samples. Researchers must determine sequence-dependent effects (concurrent vs. sequential administration) and identify pharmacodynamic biomarkers for each combination component.

Clinical trial designs should incorporate:

Design ElementImplementation StrategyRationale
Adaptive designInterim analyses with decision rulesRapid identification of promising combinations
Biomarker stratificationPatient cohorts based on target expressionMatching biology to therapeutic mechanism
Factorial componentsArm omitting single agentsAssess contribution of each component
Model-informed dosingPK/PD simulationsMitigate overlapping toxicities

What are optimal methods for monitoring immunogenicity in patients receiving DVR-d therapy?

Monitoring immunogenicity in patients receiving DVR-d therapy requires comprehensive methodological approaches to detect, characterize, and manage anti-drug antibodies (ADAs). A robust monitoring program should employ multiple complementary assay formats:

  • Bridging ELISA as a primary screening method

  • Electrochemiluminescence (ECL) for improved sensitivity

  • Surface Plasmon Resonance for real-time kinetic analysis

  • Cell-based assays to determine neutralizing capacity

Sampling strategies should include baseline (pre-treatment) collection to identify pre-existing antibodies, early sampling (weeks 2-4) to detect rapid-onset immunogenicity, regular monitoring throughout treatment, and extended post-treatment monitoring. Multiple myeloma presents specific challenges including high levels of circulating M-protein that may interfere with assays and profound immune dysregulation affecting antibody production.

ADA characterization should include isotype determination, epitope mapping, affinity determination, and cross-reactivity testing with similar therapeutic antibodies. This comprehensive approach provides critical data for managing immunogenicity during treatment and informs future antibody engineering efforts.

How can bispecific antibody approaches complement or enhance DVR-d therapy?

Bispecific antibody approaches represent a significant evolution beyond the monoclonal antibody approach used in DVR-d therapy . While daratumumab targets a single epitope (CD38) and relies on natural immune effector mechanisms, bispecific antibodies simultaneously engage two different epitopes, often bringing T cells (via CD3) into proximity with tumor cells, creating artificial synapses independent of T-cell receptor specificity.

This approach offers several methodological advantages:

FeatureMonoclonal Antibody (DVR-d)Bispecific Antibody
Effector mechanismDependent on Fc receptor engagementDirectly recruits T cells without MHC restriction
Resistance vulnerabilityAntigen loss, Fc receptor polymorphismsLess affected by Fc variations
Therapeutic windowGenerally wider, lower cytokine riskPotentially narrower due to T-cell activation
Manufacturing complexityEstablished platformsRequires specialized engineering approaches

In multiple myeloma treatment, bispecific antibodies targeting BCMA or GPRC5D plus CD3 have shown remarkable efficacy in patients previously treated with daratumumab-containing regimens like DVR-d. Current methodological research focuses on optimizing bispecific antibody design through affinity tuning, conditional activation mechanisms, and novel target combinations to maintain efficacy while improving safety profiles.

How might de novo antibody design approaches transform DVR-d components?

For DVR-d therapy, this approach could enable:

  • Design of antibodies targeting novel epitopes on CD38 that remain accessible in daratumumab-resistant myeloma cells

  • Creation of antibodies with optimized effector functions specifically tailored to the bone marrow microenvironment

  • Development of smaller antibody formats with enhanced tissue penetration properties

  • Generation of antibodies with reduced immunogenicity profiles

These computational design methods could potentially reduce development timelines from years to months, allowing rapid iteration and optimization of therapeutic antibodies based on clinical feedback and resistance mechanisms observed in patients.

What are emerging approaches to overcome resistance mechanisms to DVR-d therapy?

Multiple myeloma cells develop resistance to DVR-d therapy through various mechanisms, primarily including CD38 downregulation, complement inhibitory protein upregulation, and immunosuppressive microenvironment modulation. Emerging research approaches to address these challenges include:

  • Epigenetic modification strategies using HDAC inhibitors or DNA methyltransferase inhibitors to prevent or reverse CD38 downregulation

  • Dual-targeting antibodies that simultaneously engage CD38 and secondary myeloma-specific antigens

  • Antibody-drug conjugates combining anti-CD38 targeting with payload delivery

  • Microenvironment modifiers that enhance effector cell function or overcome immunosuppression

  • Intermittent dosing schedules that allow CD38 re-expression

With the development of AI technologies for therapeutic antibody discovery as described in the VUMC ARPA-H project, researchers can potentially "build a massive antibody-antigen atlas, develop AI-based algorithms to engineer antigen-specific antibodies, and apply the AI technology to identify and develop potential therapeutic antibodies" . This approach could rapidly generate next-generation anti-CD38 antibodies or complementary antibodies targeting resistance-associated proteins to enhance DVR-d efficacy in refractory patients.

What computational methods best analyze antibody-antigen binding interfaces in DVR-d components?

Computational analysis of antibody-antigen binding interfaces requires sophisticated methodologies to understand interaction mechanisms and guide optimization. For DVR-d's antibody component (daratumumab), several complementary approaches provide comprehensive insights:

Computational MethodApplicationOutcome Measure
Molecular dynamics simulationsBinding stability analysisResidence time, conformational changes
Free Energy PerturbationMutation effect predictionΔΔG binding energy differences
Hydrogen bond network analysisInterface stability assessmentBond persistence and water bridges
Computational alanine scanningCritical binding residue identificationContribution to binding energy
Machine learning-based interface predictionNovel binding site discoveryProbability scores for epitope regions

Advanced AI approaches mentioned in the VUMC project aim to "develop AI-based algorithms to engineer antigen-specific antibodies" . These methods can integrate structural prediction with binding energy calculations to design optimized variants with improved affinity or novel binding properties to overcome resistance mechanisms observed in DVR-d therapy.

How should researchers interpret contradictory clinical data regarding DVR-d efficacy across different patient populations?

Interpreting contradictory clinical data regarding DVR-d efficacy requires systematic analysis of multiple factors that may contribute to heterogeneous outcomes. Researchers should:

  • Stratify analysis by cytogenetic risk groups, as high-risk features significantly impact outcomes. As noted in clinical research, cytogenetics drive treatment decisions: "Anybody who has high-risk cytogenetics; that is the t(4;14) deletion, 17p, or TP53, even amplification 1q, we tend to use quadruplets such as Darzalex-Velcade-Revlimid-dexamethasone (DVR-d) versus a triplet" .

  • Consider prior lines of therapy and specific previous exposures, particularly to CD38-targeting agents.

  • Evaluate protocol adherence regarding dosing intensity and duration of each component, as modifications may substantially impact efficacy.

  • Assess response criteria standardization across studies, particularly regarding MRD assessment methods and sensitivity thresholds.

  • Analyze pharmacokinetic data to identify suboptimal drug exposure in certain populations.

  • Examine molecular and cellular biomarkers of response, including CD38 expression levels, immunological parameters, and bone marrow microenvironment characteristics.

Meta-analytical approaches with random effects models can accommodate between-study heterogeneity while identification of effect modifiers through interaction testing can reveal population-specific factors influencing outcomes.

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 2025 TheBiotek. All Rights Reserved.