DAR4 Antibody

Shipped with Ice Packs
In Stock

Description

Development and Conjugation Technologies

DAR4 ADCs are produced using advanced site-specific conjugation platforms:

TechnologyKey FeaturesExample ApplicationSource
WuXiDAR4™Fab-region conjugation; achieves >97% DAR4 homogeneity; 2–3-week productionNectin-4-targeted ADCs (e.g., 9MW2821)
THIOMAB®Engineered cysteine residues for uniform DAR4; reduced immunogenicityAnti-HER2 ADCs (e.g., trastuzumab emtansine)
ARC-ADCEnzymatic conjugation via CD38 fusion; enables tunable DAR2/DAR4 variantsAnti-CLL-1 ADCs for AML

These methods resolve historical issues with stochastic conjugation, which often produced heterogeneous DAR distributions (0–8) .

Efficacy Enhancements

  • DAR4-ARC-ADC demonstrated 25 pM EC50 against U937 AML cells, compared to 0.9 nM for DAR2 counterparts .

  • In xenograft models, DAR4 ADCs increased median survival by >100% versus controls .

Toxicity Mitigation

  • Homogeneous DAR4 reduces off-target payload release, lowering rates of peripheral neuropathy (common in MMAE-based ADCs) and ocular toxicity (linked to MMAF) .

  • WuXiDAR4™-generated ADCs show 42% fewer aggregate-related adverse events compared to traditional conjugates .

Pharmacokinetic Stability

  • DAR4 maintains plasma half-life (~7 days in humans) closer to native antibodies, unlike high-DAR (≥6) variants cleared 3× faster .

Anti-CLL-1 DAR4-ARC-ADC

ParameterDAR2-ARC-ADCDAR4-ARC-ADC
In vitro EC50 (U937)0.9 ± 0.4 nM25 ± 8 pM
Median survival18 days27 days
Tumor suppression68% at 3 mg/kg94% at 5 mg/kg

Source:

9MW2821 (Nectin-4-targeted DAR4 ADC)

  • Induced complete regression in 60% of PDX bladder cancer models at 3 mg/kg .

  • Hydrophobic interaction chromatography (HIC) confirmed 97.6% DAR4 purity versus 42.8% for legacy conjugates .

Emerging Applications and Challenges

  • Dual-payload ADCs: Combining MMAE and MMAF in DAR4 configurations overcomes tumor heterogeneity in breast cancer .

  • Solid tumors: DAR4 DXd ADCs show MTDs of 6.4 mg/kg, enabling higher tolerated doses than DAR8 variants .

Persistent challenges:

  • Balancing Fc-mediated effector functions with payload release kinetics .

  • Engineering stable linkers for intracellular trafficking in low-antigen-density tumors .

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
DAR4 antibody; CHS3 antibody; At5g17890 antibody; MPI7.5 antibody; MPI7.6Protein DA1-related 4 antibody; Protein CHILLING SENSITIVE 3 antibody
Target Names
DAR4
Uniprot No.

Target Background

Function
DAR4 is a NB-LRR receptor-like protein that plays a crucial role in modulating growth, cell death, and freezing tolerance in a temperature-dependent manner. It is also likely involved in defense responses.
Gene References Into Functions
  1. The chs3-1 mutation, which enhances freezing tolerance, was correlated with growth defects and cell death phenotypes resulting from activated defense responses. PMID: 20444230
Database Links

KEGG: ath:AT5G17890

STRING: 3702.AT5G17890.1

UniGene: At.43401

Q&A

What is a DAR4 antibody and how does it differ from other DAR configurations?

A DAR4 antibody is an antibody-drug conjugate (ADC) that carries an average of four drug molecules per antibody molecule. The Drug-to-Antibody Ratio (DAR) is a critical parameter in ADC development that significantly influences both efficacy and safety profiles.

What methodological approaches are used to measure DAR in antibody-drug conjugates?

Accurate DAR determination requires careful consideration of the chemical properties of both the payload and conjugation chemistry. Multiple complementary analytical methods are typically employed:

MethodApplicabilityStrengthsLimitations
UV-Vis SpectroscopyBroad range of conjugatesSimple, non-destructiveLess precise for complex mixtures
Mass Spectrometry (MS)All conjugate typesProvides detailed DAR distributionRequires specialized equipment
Reverse-Phase HPLCCysteine/site-specific conjugatesRobust, reproducible, quantitativeLess effective for lysine conjugates
Hydrophobic Interaction Chromatography (HIC)Cysteine conjugatesMaintains native protein structureLimited to certain conjugation types
Size-Exclusion Chromatography (SEC)All ADC typesEvaluates aggregationLower resolution for DAR determination

Researchers should implement multiple orthogonal methods to ensure accurate DAR characterization. As noted by experienced ADC researchers, "have multiple methods to measure DAR even if one is the 'workhorse'" . The specific analytical approach should consider both the antibody characteristics and payload properties to optimize accuracy and precision.

How does DAR4 impact pharmacokinetics and in vivo efficacy of antibody-drug conjugates?

The impact of DAR4 on pharmacokinetics and efficacy represents a complex interplay between drug loading, target binding, and tissue distribution. In comparative studies, DAR4 antibodies typically demonstrate enhanced in vitro and in vivo potency compared to lower DAR counterparts.

  • Target expression levels in tumor versus normal tissues

  • Target internalization rates

  • Maximum tolerated dose (MTD) of the specific payload

  • Tumor penetration capabilities

The clearance of higher DAR antibodies may be accelerated due to increased hydrophobicity or altered recognition by clearance receptors, potentially offsetting some efficacy advantages through reduced exposure . Researchers should carefully evaluate the pharmacokinetic profile of DAR4 antibodies in relevant models before proceeding to more advanced studies.

How do site-specific conjugation technologies enable the generation of homogeneous DAR4 antibodies?

Site-specific conjugation represents a significant advance over traditional conjugation methods, allowing precise control over drug attachment sites and resulting in more homogeneous DAR4 ADCs. Several technologies have been developed to achieve site-specific DAR4 antibodies:

ADP-ribosyl cyclase-enabled ADCs (ARC-ADCs) utilize genetic fusion of CD38 catalytic domains to antibody heavy and light chains, enabling enzymatic attachment of payloads at specific sites. This approach has successfully generated homogeneous DAR4 antibodies by fusing CD38 to C-termini of both light and heavy chains, followed by single-step enzymatic conjugation reactions . Importantly, these engineered antibodies maintain their target binding properties, with ELISA analysis confirming that "both DAR2-ARC-IgG and DAR4-ARC-IgG exhibit tight binding to recombinant hCLL-1, comparable to that of native anti-hCLL-1 antibody" .

Glycoengineering approaches offer another pathway to site-specific DAR4 ADCs through enzymatic modification of IgG Fc glycans. The AGLink site-specific conjugation kit described in research literature enables controlled generation of DAR4 antibodies with minimal impact on antigen binding capacity . ELISA analysis verified that "HER2 antigen binding to Trastuzumab-MMAE (DAR2/4) was unaffected after the AGLink conjugation, preserving antibody bioactivity" .

These methodologies overcome the heterogeneity limitations of traditional conjugation approaches, resulting in more consistent drug loading and potentially more predictable clinical performance.

What experimental evidence supports differential tumor penetration between DAR4 and other DAR configurations?

Tumor penetration represents a critical parameter for ADC efficacy, with emerging evidence suggesting important differences between various DAR configurations. Research using tumor spheroid models has demonstrated that lower DAR antibodies may achieve enhanced tumor penetration compared to higher DAR variants. Studies have shown "a two-fold enhancement in tumor penetration for a DAR1 ADC compared to its DAR2 equivalent" , suggesting that penetration may decrease as DAR increases.

This penetration differential appears to result from several factors:

  • Increased hydrophobicity with higher DAR values

  • Altered binding kinetics to the target antigen

  • Potential "binding site barrier" effects where high-affinity ADCs primarily bind to peripheral tumor cells

These observations have significant implications for DAR4 antibodies, suggesting that while they offer enhanced potency on a per-molecule basis, their tumor penetration characteristics may be suboptimal compared to lower DAR variants. Researchers working with DAR4 antibodies should carefully evaluate tumor penetration in three-dimensional models that better recapitulate in vivo architecture.

How does payload chemistry influence the optimal DAR for specific cancer targets?

The selection of optimal DAR is highly dependent on payload properties, with different cytotoxic agents demonstrating distinct relationships between DAR and efficacy/toxicity profiles. Research has established that payload chemistry significantly influences the maximum tolerated dose (MTD) of ADCs:

Payload TypeCommon ToxicitiesTypical MTD for DAR4Considerations for DAR Selection
MMAEPeripheral neuropathy~1.8 mg/kgDAR2 MMAE ADCs tolerated at ~3.6 mg/kg
MMAFOcular toxicityVaries by targetHigher DAR possible with hydrophilic linkers
Deruxtecan (DXd)GI toxicity, ILD~8-12 mg/kg for DAR4DAR8 DXd ADCs tolerated at half the dose of DAR4
PBD dimersHematological toxicityUltra-potent, lower DAR preferredDAR1 shows only 2.4-fold potency reduction vs. DAR2

For highly potent payloads like PBD dimers, lower DAR values may provide sufficient efficacy while minimizing toxicity. Comparative studies have shown that "the difference in potency between the DAR1 and DAR2 constructs was 1.5-fold" for maytansinoid payloads and "a 2.4-fold difference in IC50 value was determined for the DAR1 and DAR2 constructs" with PBD dimers , suggesting that lower DAR values may be preferable for ultra-potent payloads.

What strategies can researchers employ to optimize the therapeutic window of DAR4 antibodies?

Optimizing the therapeutic window of DAR4 antibodies requires balancing efficacy and toxicity through careful modulation of multiple parameters. Several evidence-based approaches have emerged from the research literature:

  • Target saturation optimization: Research indicates that "to achieve tumor saturation and direct cellular drug delivery, which are associated with improved efficacy, it is often better to lower the DAR of an ADC with a sub-saturating dose to increase the total antibody administered to a dose that penetrates farther into the tumor" . This approach maximizes tumor exposure while potentially reducing off-target effects.

  • Linker chemistry selection: The choice of linker can significantly impact the stability and toxicity profile of DAR4 antibodies. Stability studies have demonstrated that properly designed DAR4 ADCs maintain excellent stability in human plasma over extended periods, with "both types of ADCs [DAR2/4] stable in vitro throughout the experiment" spanning 8 days .

  • Target selection considerations: The expression pattern of the target antigen critically influences the optimal DAR. For targets with significant normal tissue expression, lower DAR values may provide a better therapeutic window. Clinical studies of various ADCs targeting different antigens have shown that the maximum tolerated dose varies substantially depending on target expression in normal tissues .

  • Payload-specific DAR optimization: Each payload class has an optimal DAR range based on its potency and toxicity profile. Ultra-potent payloads may achieve sufficient efficacy at lower DAR values, while moderately potent payloads may benefit from higher DAR values to achieve therapeutic concentrations in tumors.

  • Aggregation minimization: Higher DAR antibodies may be more prone to aggregation, potentially affecting both efficacy and immunogenicity. SEC-HPLC analysis of well-designed DAR4 ADCs should show "no observable peaks... in the resulting chromatogram, with less than 5% of antibody aggregation" .

What analytical challenges are specific to DAR4 antibodies during characterization?

DAR4 antibodies present unique analytical challenges that must be addressed through comprehensive characterization strategies. These challenges stem from the complexity added by the four conjugated drug molecules and their potential impact on antibody properties.

The analytical workflow for DAR4 antibodies typically requires multiple orthogonal methods to fully characterize:

  • Accurate DAR determination: The heterogeneity within DAR4 preparations (containing antibodies with 0-8 drug molecules) requires sophisticated analytical methods. Mass spectrometry provides detailed DAR distribution profiles but may face ionization challenges with hydrophobic payloads. Chromatographic methods like HIC can separate and quantify different DAR species but may not fully resolve complex mixtures.

  • Conformational stability assessment: The attachment of four drug molecules can potentially impact the conformational stability of the antibody. Differential scanning calorimetry (DSC) and size-exclusion chromatography (SEC) are essential to evaluate whether DAR4 conjugation alters thermal stability or promotes aggregation.

  • Functional characterization complexities: Drug conjugation at multiple sites may impact binding kinetics or Fc-mediated functions. Binding assays must verify that "antigen binding capacity of both DAR2 and DAR4 ADCs" remains intact after conjugation .

  • Batch-to-batch consistency challenges: Achieving consistent DAR4 preparations requires precise control of conjugation conditions. Even small variations in reaction parameters can shift DAR distribution, necessitating robust analytical methods to ensure batch consistency.

  • Stability-indicating methods development: Appropriate stability-indicating methods must be developed to detect potential degradation products specific to DAR4 antibodies, including partial deconjugation or payload modification during storage.

Researchers should implement comprehensive analytical panels that include multiple complementary techniques to fully characterize DAR4 antibodies and ensure consistent quality attributes across batches.

How do in vitro cytotoxicity assays correlate with in vivo efficacy for DAR4 antibodies?

The translation from in vitro potency to in vivo efficacy for DAR4 antibodies involves complex pharmacological relationships that researchers must carefully evaluate. Published data reveal several important patterns:

In vitro cytotoxicity assays typically demonstrate that DAR4 antibodies exhibit significantly higher potency than lower DAR variants against target-positive cell lines. For example, DAR4-ARC-ADC exhibits an EC50 of approximately 25±8 pM against hCLL-1-positive U937 cells, while DAR2-ARC-ADC shows lower potency with an EC50 of 0.9±0.4 nM . This represents an approximately 36-fold potency advantage for the DAR4 variant in this model system.

Several factors contribute to the complex relationship between in vitro and in vivo outcomes:

  • Differential tumor penetration between DAR variants

  • Variations in pharmacokinetic profiles and exposure

  • Target-mediated drug disposition effects

  • Microenvironmental factors not captured in vitro

Researchers should design in vitro assay panels that incorporate three-dimensional models and physiologically relevant conditions to better predict in vivo performance of DAR4 antibodies.

What considerations are important when designing preclinical studies to evaluate DAR4 antibodies?

Preclinical evaluation of DAR4 antibodies requires carefully designed studies that address their unique pharmacological properties. Key considerations include:

  • Model selection based on target expression: The preclinical models must accurately represent the target expression levels and distribution expected in human malignancies. Models with varying target expression levels should be included to evaluate the relationship between expression and response.

  • Comparative DAR evaluation: Studies should include direct comparisons between DAR4 and other DAR variants (e.g., DAR2, DAR8) to identify the optimal configuration for specific applications. Evidence shows that "DAR4-ARC-ADC exhibits notably enhanced potency in both cellular and animal AML models" , but this must be evaluated in the context of specific targets and payloads.

  • Toxicity assessment in relevant species: For targets with cross-reactivity to non-human primates (NHPs), NHP toxicity studies provide valuable insights into potential on-target, off-tumor effects. Research indicates that "antibodies that are cross-reactive to NHP antigens can be used... to help assess antibody target-mediated toxicity" .

  • Tumor penetration evaluation: Three-dimensional models or imaging studies should be incorporated to assess tumor penetration, as evidence suggests that "a two-fold enhancement in tumor penetration for a DAR1 ADC compared to its DAR2 equivalent" may indicate penetration challenges for higher DAR antibodies.

  • Dosing schedule optimization: The optimal dosing schedule may differ for DAR4 antibodies compared to other ADCs due to their unique pharmacokinetic properties. Multiple dosing regimens should be evaluated to identify schedules that maximize efficacy while minimizing toxicity.

  • Resistance mechanism investigations: Studies should investigate potential resistance mechanisms specific to DAR4 antibodies, including alterations in internalization pathways or efflux transporter upregulation.

By addressing these considerations, researchers can develop robust preclinical packages that accurately predict the clinical performance of DAR4 antibodies and guide early clinical development decisions.

How might the principles of DAR4 antibody design extend to other therapeutic modalities?

The technological principles underlying DAR4 antibody development have broad potential applications beyond traditional cytotoxic ADCs. Several emerging applications demonstrate the versatility of these approaches:

  • Bispecific antibody development: The site-specific conjugation technologies used for DAR4 antibodies can be adapted to generate well-defined bispecific antibodies. As noted in the literature, "the presented DAR1 technology is also perfectly suitable for attachment of small protein formats, e.g., scFv's or cytokines, for the generation of T-cell and NK-cell-engaging bispecific antibodies" .

  • Antibody-oligonucleotide conjugates: The controlled conjugation methods developed for DAR4 antibodies can be applied to the attachment of oligonucleotides for targeted delivery of nucleic acid therapeutics, including siRNA and antisense oligonucleotides.

  • Radioimmunoconjugates: Precise control over conjugation sites and stoichiometry is essential for radioimmunoconjugates to ensure consistent biodistribution and radiation dosimetry. The technologies underlying DAR4 antibodies can be adapted for radioisotope attachment.

  • Immunocytokine development: Site-specific attachment of cytokines to antibodies can generate immunocytokines with improved pharmacological properties compared to traditional chemical conjugation approaches.

  • Antibody-enzyme conjugates for prodrug activation: Technologies used for DAR4 antibody development can be applied to the generation of antibody-enzyme conjugates for targeted prodrug activation strategies.

The fundamental principles of site-specific conjugation with defined stoichiometry established in DAR4 antibody research provide a versatile platform for diverse therapeutic applications beyond traditional cytotoxic ADCs.

What evidence supports the potential advantages of variable DAR in addressing tumor heterogeneity?

Tumor heterogeneity represents a significant challenge in cancer therapy, and emerging evidence suggests that ADCs with variable DAR may offer unique advantages in addressing this challenge. While research specifically comparing DAR4 with other configurations in heterogeneous tumor models remains limited, several principles have emerged:

Different DAR values confer distinct pharmacological properties that may complement each other in addressing tumor heterogeneity:

  • Penetration vs. potency trade-offs: Lower DAR antibodies typically demonstrate enhanced tumor penetration but reduced cell-killing potency. Research showing "a two-fold enhancement in tumor penetration for a DAR1 ADC compared to its DAR2 equivalent" suggests that lower DAR variants may better access poorly vascularized tumor regions.

  • Differential efficacy against varying target expression levels: Higher DAR antibodies may be more effective against tumor cells with lower target expression, where delivering more payload molecules per binding event is advantageous. Conversely, for high-expressing cells, even lower DAR antibodies may deliver sufficient payload to achieve cell killing.

  • Varying pharmacokinetic profiles: The different clearance rates of various DAR configurations may create beneficial exposure profiles across heterogeneous tumors, with lower DAR antibodies potentially providing more sustained exposure due to improved pharmacokinetic properties.

These considerations suggest potential advantages for mixed-DAR approaches or sequential administration strategies that leverage the complementary properties of different DAR configurations to more effectively address tumor heterogeneity.

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.