BON3 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
BON3 antibody; At1g08860 antibody; F7G19.25Protein BONZAI 3 antibody
Target Names
BON3
Uniprot No.

Target Background

Function
This antibody targets a protein that acts as a negative regulator of cell death and defense responses. It represses the expression of multiple R genes and may play a role in promoting growth and development. Additionally, it could be involved in membrane trafficking and the fusion of vesicles with the plasma membrane.
Database Links

KEGG: ath:AT1G08860

STRING: 3702.AT1G08860.1

UniGene: At.51165

Protein Families
Copine family
Subcellular Location
Cell membrane; Lipid-anchor.
Tissue Specificity
Expressed at an extremely low level.

Q&A

What is the fundamental mechanism behind bone-targeting antibody technology?

Bone-targeting antibody technology is based on engineering therapeutic antibodies with bone-homing peptide sequences that dramatically enhance their concentration in the bone microenvironment. Inspired by natural biomolecules that target bone tissue, this approach transitions antibody-based therapies from being merely antigen-specific to both antigen and tissue-specific. The technology typically involves modifying antibodies with bisphosphonates or other molecules that have high binding affinity for hydroxyapatite, the main component of hard bone tissue. This modification enables the antibodies to overcome the physical and biological barriers in the bone microenvironment, resulting in improved therapeutic efficacy against bone tumors and metastases .

Why is targeting bone tissue with antibodies particularly challenging?

Delivery of antibodies to bone tissue faces several unique challenges:

  • Low vascularization of bone tissue limits antibody access via blood circulation

  • Physical barriers to penetration due to the hard, mineralized structure of bone

  • Poor tissue accessibility for large macromolecules like antibodies

  • Tendency of therapeutic agents to attach to adjacent healthy tissues rather than bone tumors

  • Limited networks of blood vessels in bone tissue

These challenges collectively contribute to inadequate pharmacokinetics and reduced efficacy of conventional antibody therapies when targeting bone tumors . The acidic microenvironment of bone tumors further complicates targeting, though this property can be leveraged in designing selective delivery systems .

What types of bone-targeting moieties are currently being used in research?

Several bone-targeting moieties have been investigated for enhancing antibody delivery to bone:

Targeting MoietyMechanism of ActionAdvantagesCommon Applications
Bisphosphonates (e.g., alendronate)High binding affinity for hydroxyapatitePreferential binding to acidic sites like bone tumorsAntibody conjugates for bone metastasis treatment
L-Asp6 peptide sequencesBone matrix bindingCan be inserted at multiple sites in antibody structureEngineering antibodies with bone specificity
Bone-homing peptidesSpecific binding to bone tissue componentsMinimally disruptive to native IgG structure and functionEnhanced bone tumor distribution of therapeutic antibodies

Research shows that these targeting moieties can be incorporated at various permissive sites in antibody structures, including light chain, heavy chain, and C-terminus positions, with each configuration offering different degrees of bone-targeting efficacy .

How does the insertion position of bone-homing peptides affect antibody function and bone-targeting efficacy?

The position of bone-homing peptide insertion in the antibody structure significantly impacts both the antibody's native function and its bone-targeting capabilities. Research has identified several permissive insertion sites that maintain antibody integrity while enhancing bone specificity:

  • Light Chain (LC, A153): Insertion at this site preserves antibody structure while adding bone-targeting ability

  • Heavy Chain (CH1, A165): This position allows for bone-homing peptide incorporation with minimal disruption to antigen binding

  • C-terminus (CT, G449): Terminal modification often has the least impact on antibody function

The optimal insertion strategy depends on the specific antibody and target, requiring careful engineering and validation to balance bone-targeting enhancement with preserved therapeutic function.

What are the mechanisms by which bone-targeting antibodies inhibit metastatic progression?

Bone-targeting antibodies inhibit metastatic progression through multiple interconnected mechanisms:

  • Enhanced local concentration: The bone-targeting moieties significantly increase antibody concentration in bone tumor microenvironments, leading to more effective tumor cell killing at the primary bone metastatic site .

  • Prolonged residence time: These modified antibodies remain at the tumor site longer than conventional antibodies, providing extended therapeutic effects .

  • Prevention of secondary metastases: By effectively treating initial bone metastases, these antibodies can prevent secondary metastatic dissemination from bone lesions to other organs .

  • Selective accumulation in acidic environments: Bone-targeting moieties like bisphosphonates preferentially accumulate in acidic sites typical of bone tumors, resulting in higher drug concentration in tumors compared to surrounding healthy tissue .

  • Disruption of the bone metastatic niche: By targeting factors involved in the "fertile soil" environment that bone provides for cancer cells, these antibodies can make bone tissue less hospitable for metastatic growth and further spread .

In experimental models, BonTarg technology has demonstrated significant efficacy not only in reducing initial bone metastasis but also in inhibiting secondary metastasis to organs including the brain, heart, and liver - a critical advantage given the poor prognosis associated with such metastatic progression .

How do bone-targeting antibodies compare with antibody-drug conjugates in treating bone metastases?

Bone-targeting antibodies and antibody-drug conjugates (ADCs) represent two approaches that can be complementary in treating bone metastases:

FeatureBone-Targeting AntibodiesStandard ADCsBone-Targeting ADCs
Targeting mechanismTissue (bone) and antigen specificAntigen specific onlyTissue, antigen, and cytotoxic
Payload requirementMay rely on antibody mechanism of actionRequires cytotoxic payloadCombines bone targeting with cytotoxic payload
Distribution profileEnhanced bone concentrationSystemic with target-cell preferenceEnhanced bone concentration with target-cell specificity
Off-target effectsReduced systemic toxicityCan affect healthy cells expressing targetMinimized through dual targeting
Manufacturing complexityModerate (peptide insertion)High (conjugation chemistry)Very high (both modifications)

Research indicates that the bone-targeting approach can also be applied to prepare bone-targeting antibody-drug conjugates, combining the advantages of both strategies. These hybrid constructs have demonstrated enhanced therapeutic efficacy in experimental models, suggesting that the addition of bone-specific targeting to ADCs provides a powerful strategy to overcome the poor accessibility of antibodies to bone tumors and the consequential resistance to therapy .

What are the recommended protocols for evaluating bone-targeting efficacy of engineered antibodies?

Comprehensive evaluation of bone-targeting efficacy requires a multi-step approach:

  • In vitro characterization:

    • Verify successful antibody modification through SDS-PAGE and electrospray ionization mass spectrometry (ESI-MS)

    • Assess antibody stability and aggregation tendency

    • Confirm preserved antigen binding using surface plasmon resonance or ELISA

    • Evaluate binding affinity to hydroxyapatite or bone mineral components

  • Pharmacokinetic analysis:

    • Compare plasma half-life of modified vs. unmodified antibodies

    • Quantify biodistribution across various tissues with special attention to bone-to-blood and bone-to-organ ratios

    • Track antibody clearance rates and elimination pathways

  • In vivo imaging studies:

    • Use fluorescently labeled antibodies to visualize bone localization

    • Perform time-course imaging to determine optimal dosing intervals

    • Quantify signal intensity at bone tumors vs. healthy bone areas

  • Therapeutic efficacy assessment:

    • Establish appropriate xenograft models of bone metastasis

    • Compare tumor growth inhibition between unmodified and bone-targeting antibodies

    • Evaluate prevention of secondary metastasis from initial bone lesions

    • Analyze survival outcomes and quality of life metrics

A rigorous evaluation should include both breast cancer cell lines that naturally metastasize to bone and direct bone injection models to isolate the effect of the bone-targeting moiety on established bone tumors.

How should researchers modify antibodies with bone-targeting moieties while preserving their therapeutic function?

Successful modification of antibodies with bone-targeting moieties requires careful consideration of multiple factors:

  • Site selection: Choose permissive sites within the antibody structure that tolerate modification without disrupting antigen binding or Fc function. Research has identified specific positions including:

    • Light chain position A153

    • Heavy chain position A165

    • C-terminus position G449
      These sites have been shown to maintain antibody integrity after modification .

  • Conjugation chemistry: Select appropriate bioorthogonal chemistries that enable precise conjugation under mild conditions. pClick technology has proven effective for linking bone-targeting molecules like alendronate to therapeutic antibodies without requiring harsh chemicals, enzymes, or ultraviolet light that could damage the antibody .

  • Optimization of targeting moiety: Consider the following parameters when selecting and optimizing bone-targeting components:

    • Number of targeting moieties per antibody (typically 1-3)

    • Type of targeting moiety (bisphosphonates, L-Asp6 peptides, etc.)

    • Linker length and composition between antibody and targeting moiety

  • Quality control: Implement rigorous testing to ensure:

    • Homogeneity of the modified antibody population

    • Absence of significant aggregation

    • Retention of antigen binding affinity

    • Preservation of effector functions

    • Appropriate bone-targeting capability

The research demonstrates that maintaining a balance between sufficient bone-targeting ability and preserved antibody function is critical, as excessive modification can lead to aggregation and loss of therapeutic efficacy.

What animal models are most appropriate for evaluating bone-targeting antibodies?

Selecting appropriate animal models is crucial for accurately evaluating bone-targeting antibodies:

Model TypeDescriptionAdvantagesLimitationsBest Applications
Xenograft bone metastasisHuman cancer cells injected systemically that metastasize to boneRecapitulates natural metastatic processVariability in metastasis formation; longer experimental timeframeEvaluating prevention of bone metastasis
Direct bone tumor inoculationCancer cells injected directly into bone tissueConsistent tumor location; reduced variabilityBypasses early metastatic stepsAssessing efficacy against established bone tumors
Orthotopic metastasisPrimary tumors grown at original site with spontaneous metastasis to boneBest physiological relevanceTechnically challenging; variable metastasis ratesStudying complete metastatic cascade
Patient-derived xenografts (PDX)Patient tumor samples implanted into immunodeficient micePreserves original tumor heterogeneityRequires immunocompromised hostsTesting against clinically relevant tumors

For bone-targeting antibody research, multiple complementary models should be employed:

  • Use intracardiac injection models for breast cancer cells to study metastasis to bone sites

  • Employ direct bone injection models to evaluate therapeutic efficacy against established bone tumors

  • Implement secondary metastasis models to assess prevention of further tumor spread from bone lesions

  • Consider immunocompetent models when possible to evaluate the contribution of immune responses

The selection should align with specific research questions, such as whether the focus is on preventing initial bone metastasis or treating established bone tumors.

What are the current limitations of bone-targeting antibody approaches and how might they be overcome?

Despite promising advances, bone-targeting antibody technologies face several important limitations:

  • Heterogeneous bone uptake: Current targeting approaches may result in uneven distribution within bone tissue and between different bone sites. This limitation might be addressed through:

    • Development of targeting moieties with affinity for multiple bone components

    • Combined use of complementary bone-targeting strategies

    • Optimized dosing regimens to improve distribution

  • Potential off-target accumulation: Bone-targeting moieties may accumulate in non-target tissues with similar components. Strategies to improve specificity include:

    • Dual-targeting approaches combining bone affinity with tumor cell recognition

    • pH-sensitive linkers that release active compounds predominantly in the acidic tumor microenvironment

    • Design of targeting moieties that recognize tumor-modified bone matrix

  • Limited penetration into solid tumors: Even with bone targeting, large antibodies face challenges penetrating dense tumor tissue. Potential solutions include:

    • Development of smaller antibody fragments with bone-targeting capabilities

    • Combination with agents that modify tumor vasculature or extracellular matrix

    • Utilization of two-step targeting approaches where smaller molecules prepare the site for antibody penetration

  • Antibody-induced immune responses: Modified antibodies may trigger immunogenicity. Approaches to mitigate this include:

    • Careful selection of modification sites to minimize altered epitope exposure

    • Human-derived targeting peptides where possible

    • Engineering strategies to shield modifications from immune recognition

Addressing these limitations will require multidisciplinary approaches combining antibody engineering, bone biology expertise, and advanced delivery technologies.

How might bone-targeting antibody technology be extended to therapeutic applications beyond cancer?

While current research focuses primarily on cancer applications, bone-targeting antibody technology holds promise for various other therapeutic areas:

  • Osteoporosis and metabolic bone diseases:

    • Targeting RANKL with bone-specific antibodies could enhance local efficacy while reducing systemic effects

    • Delivery of anabolic agents specifically to areas of low bone density

    • Targeting of sclerostin with bone-specific antibodies for localized bone formation

  • Orthopedic applications and bone regeneration:

    • Enhancing fracture healing through targeted delivery of growth factors

    • Improving outcomes in spinal fusion procedures

    • Supporting implant integration and reducing aseptic loosening

    • Creating scaffold-antibody combinations for tissue engineering applications

  • Rheumatoid arthritis and inflammatory bone disorders:

    • Targeted delivery of anti-inflammatory antibodies to affected joints

    • Protecting bone from inflammatory damage through localized therapy

    • Combination approaches targeting both inflammation and bone destruction

  • Rare bone disorders:

    • Treatment of localized bone dysplasias

    • Management of Paget's disease with targeted therapies

    • Addressing osteogenesis imperfecta through bone-specific delivery of corrective factors

Future research directions should explore the utility of bone-targeting antibody platforms in these non-oncological applications, potentially using modified versions of technologies like BonTarg that are currently being developed primarily for cancer treatment .

What emerging technologies might enhance the efficacy of bone-targeting antibody approaches?

Several emerging technologies show promise for enhancing bone-targeting antibody approaches:

  • Advanced bioconjugation methods:

    • Site-specific conjugation technologies like pClick that enable precise control over the location and number of bone-targeting moieties

    • Enzymatic approaches for controlled modification of antibodies

    • Biorthogonal chemistries allowing for in vivo assembly of targeting components

  • Multimodal targeting strategies:

    • Antibodies with dual targeting for both bone tissue and specific cell types

    • pH-responsive targeting systems that exploit the acidic microenvironment of bone tumors

    • Combination of bone-targeting with tumor microenvironment-modifying agents

  • Nanoparticle-antibody hybrid systems:

    • Bone-targeting antibody fragments conjugated to nanoparticles for enhanced delivery

    • Liposomal or polymeric nanocarriers modified with bone-targeting antibodies

    • Stimulus-responsive nanoparticles that release their payload upon reaching bone tissue

  • Genetic engineering approaches:

    • Cell-based therapies engineered to express bone-targeting antibodies

    • Gene editing to enhance bone-homing properties of therapeutic cells

    • mRNA delivery systems targeted to bone that enable in situ antibody production

  • Computational and AI-driven design:

    • Machine learning algorithms to predict optimal antibody modification sites

    • Computational modeling of bone-targeting moiety interactions with bone components

    • AI-assisted optimization of dosing regimens for maximal bone tumor exposure

These technologies, particularly when used in combination, have the potential to overcome current limitations and significantly enhance the efficacy of bone-targeting antibody therapies for both oncological and non-oncological applications .

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.