BBM Antibody

Shipped with Ice Packs
In Stock

Description

Overview of BBM Antibodies

BBM antibodies are a class of antibodies that have diverse applications, ranging from identifying specific protein structures to therapeutic uses in gene therapy and disease monitoring. They include:

  • Monoclonal antibody BBM.1, which recognizes an antigenic determinant of human beta 2-microglobulin .

  • Bispecific antibodies (BsAbs), which have two binding sites that target two different antigens or two different epitopes on the same antigen .

  • BBM-H901, a gene therapy that aims to restore FIX production in hemophilia B patients .

  • Blood-based markers (BBM) used in Alzheimer's disease (AD) research and clinical practice .

BBM.1 Monoclonal Antibody Targeting Beta-2 Microglobulin

Monoclonal antibody BBM.1 identifies an antigenic determinant of human beta 2-microglobulin (β2-M) . Beta-2 microglobulin is a component of the class I major histocompatibility complex (MHC I), which functions in antigen processing and presentation .

Key characteristics of BBM.1:

  • Binds to free and HLA-A, B-associated β2-M with similar affinity, showing that the BBM.1 antigenic determinant does not involve residues of β2-M that interact with HLA-A,B heavy chains .

  • The region around residue 45 of the β2-M polypeptide is exposed to the environment and not involved in binding HLA-A,B heavy chain .

  • Can be used for specific elimination of β2-M from serum .

  • Reacts with many types of human cells, including lymphocytes, thymocytes, monocytes, granulocytes, platelets, endothelial cells, and epithelial cells, but is absent on erythrocytes .

Research Findings:

  • Arginine 45 is a major part of the antigenic determinant of human β2-M recognized by mouse monoclonal antibody BBM.1 .

  • Peptides synthesized from residues 35-50 of the β2-M sequence specifically inhibit the binding of BBM.1 to cell surfaces .

  • BBM.1 does not bind to β2-M of species other than humans, gorillas, and chimpanzees .

BBM-H901 Gene Therapy for Hemophilia B

BBM-H901 is a gene therapy designed to increase factor IX (FIX) activity in individuals with moderate to severe hemophilia B . It delivers a functioning copy of the F9 gene, which is packaged in a modified adeno-associated virus (AAV), to liver cells to restore FIX production .

Clinical Trial Results:

  • In a Phase 1 trial, a single intravenous infusion of BBM-H901 led to increased FIX activity in 10 adult men in China .

  • Within the first week of infusion, mean FIX activity reached 57.1 international units per deciliter of blood (IU/dL), peaking at 64.1 IU/dL at a median of five weeks .

  • The mean increase in FIX activity over the median follow-up period of 58 weeks was 36.9 IU/dL .

Blood-Based Markers (BBM) in Alzheimer's Disease

Blood-based markers (BBM) are used in Alzheimer's disease (AD) research and clinical practice to detect amyloid pathology and monitor disease progression . They are more acceptable, accessible, and scalable than amyloid PET or cerebrospinal fluid (CSF) tests .

Potential uses of BBM in clinical trials:

  • As pre-screening tools to identify individuals with a high probability of not having AD pathology .

  • As an inclusion criterion (diagnosis tool) without confirmatory CSF or PET testing .

  • As pharmacodynamic markers to detect direct target engagement and monitor disease-modifying effects .

Table 2. Potential Roles of Blood-Based Markers (BBM)

Potential RoleWhere?
Risk/ProbabilityTo detect persons at risk for a disease or condition
DiagnosisTo confirm the presence of a disease/condition
PrognosisTo determine risk of worsening and/or progression from MCI to AD
MonitoringTo assess changes in disease severity or the effect of a treatment
PharmacodynamicTo detect changes in response to treatment
PredictiveTo predict a favorable or unfavorable effect of a treatment
SafetyTo detect an adverse event

Minimum acceptable performance of BBM tests for clinical use:

  • For use as a triaging test before subsequent confirmatory tests, a BBM test should have a sensitivity of ≥90% with a specificity of ≥85% in primary care and ≥75–85% in secondary care .

  • For use as a confirmatory test without follow-up tests, a BBM test should have performance equivalent to that of CSF tests — a sensitivity and specificity of ~85% .

Bispecific Antibodies (BsAbs)

Bispecific antibodies (BsAbs) are antibodies with two binding sites directed at two different antigens or two different epitopes on the same antigen . They have broad applications for tumor immunotherapy and the treatment of other diseases .

Mechanisms of action:

  • Act as immune cell connectors, connecting immune cells to tumor cells to enable immune cells to exert their killing effect .

  • Target immune checkpoints and tumor-associated antigens (TAAs) .

  • Target inflammatory factors in the tumor microenvironment to reduce inflammation and cytokine release syndrome (CRS) .

Examples of BsAbs:

  • Catumaxomab and blinatumomab recruit and activate immune cells .

  • AMG 424 targets CD38, which is highly expressed in malignant multiple myeloma cells .

Biological Activity-Based Modeling (BABM) for Antiviral Discovery

Biological activity-based modeling (BABM) is a computational approach that uses compound activity profiles across multiple assays to predict compound activity against new targets . It has been used to identify potential antiviral compounds for SARS-CoV-2 .

Key Points:

  • BABM models were applied to predict 311 compounds with potential anti-SARS-CoV-2 activity .

  • 32% of the predicted compounds had antiviral activity in a cell culture live virus assay .

  • The most potent compounds showed an IC50 in the sub-micromolar range .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
14-16 week lead time (made-to-order)
Synonyms
BBM antibody; AIL2 antibody; At5g17430 antibody; T10B6.90AP2-like ethylene-responsive transcription factor BBM antibody; AtBBM antibody; Protein AINTEGUMENTA-LIKE 2 antibody; Protein BABY BOOM antibody
Target Names
BBM
Uniprot No.

Target Background

Function
This antibody targets a transcription factor that plays a crucial role in promoting cell proliferation, differentiation, and morphogenesis, particularly during embryonic development.
Database Links

KEGG: ath:AT5G17430

STRING: 3702.AT5G17430.1

UniGene: At.50710

Protein Families
AP2/ERF transcription factor family, AP2 subfamily
Subcellular Location
Nucleus.
Tissue Specificity
Mostly expressed in developing seeds. Also expressed in roots, seedlings, and siliques, and, at low levels, in leaves.

Q&A

What is the specificity of BBM.1 antibody and what determines its binding?

BBM.1 is a mouse monoclonal antibody that specifically recognizes human beta-2-microglobulin (β2M), a 12 kDa protein associated with the major histocompatibility complex (MHC) class I heavy chain. The antigenic determinant recognized by BBM.1 has been extensively characterized through structural and biochemical studies. Notably, Arginine 45 is a critical residue in the antigenic determinant recognized by BBM.1, as confirmed through reversible modification studies with cyclohexanedione that resulted in up to 95% loss of BBM.1 inhibitory activity when this residue was modified . Additionally, peptides synthesized from residues 35-50 of the β2M sequence specifically inhibit BBM.1 binding to cell surfaces, with their inhibitory activity being destroyed by trypsin treatment . Recent structural modeling studies have further revealed that Glutamic acid 44 and Arginine 81 also participate in direct interactions with BBM.1, while Aspartic acid 38 contributes by stabilizing Arginine 45 . The species specificity of BBM.1 is notable, as it only binds to β2M from humans, gorillas, and chimpanzees, corresponding to the conservation of the Arginine 45 residue in these species .

What applications has BBM.1 antibody been validated for?

The BBM.1 monoclonal antibody has been validated for multiple research applications as detailed below:

ApplicationValidation StatusNotes
Flow Cytometry (FACS)ValidatedEffective for cell surface staining of β2M
Western Blot (WB)ValidatedDetects ~12 kDa band corresponding to β2M
Immunocytochemistry/Immunofluorescence (ICC/IF)ValidatedFor visualization of β2M distribution
Immunohistochemistry - Frozen sections (IHC-Fr)ValidatedNot validated for paraffin-embedded tissues
Complement-dependent cytotoxicityValidatedCytotoxic in complement-dependent lysis assays

BBM.1 is particularly useful in these applications because it binds to both free β2M and HLA-associated β2M with similar affinity, indicating that the epitope does not involve residues that interact with HLA-A,B heavy chains . The antibody has been used successfully with various human cell types including thymocytes, B cell lines, peripheral blood lymphocytes, fibroblasts, and various cell lines such as Molt 4, HeLa derivatives, and HSB2 .

What is the optimal protocol for using BBM.1 in flow cytometry experiments?

For optimal results in flow cytometry applications using BBM.1 antibody, researchers should follow this methodological approach:

  • Sample preparation: Harvest cells at a concentration of 1×10^6 cells/100μl in cold PBS containing 1% BSA and 0.1% sodium azide (FACS buffer).

  • Blocking: Incubate cells with 5-10% normal serum from the same species as the secondary antibody (if using indirect detection) for 30 minutes at 4°C to reduce non-specific binding.

  • Primary antibody incubation: Add BBM.1 antibody at a starting dilution of 1:100 to 1:500. The optimal dilution should be determined empirically for each application . Incubate for 30-45 minutes at 4°C.

  • Washing: Wash cells twice with 2ml FACS buffer by centrifugation at 400×g for 5 minutes.

  • Secondary antibody (if using unconjugated BBM.1): Add fluorochrome-conjugated anti-mouse IgG2b secondary antibody and incubate for 30 minutes at 4°C in the dark.

  • Final washing: Wash cells twice with FACS buffer.

  • Analysis: Resuspend cells in 300-500μl FACS buffer containing a viability dye if needed, and analyze by flow cytometry.

  • Positive control: Raji cells are recommended as a positive control for BBM.1 staining .

  • Negative control: Include an isotype control (Mouse IgG2b, kappa) at the same concentration as the primary antibody.

This protocol can be adapted based on specific experimental requirements and instrument settings. The antibody solution should be gently mixed before use, and a vial spin is recommended prior to opening .

How should BBM.1 antibody be stored to maintain optimal activity?

Proper storage of BBM.1 antibody is critical for maintaining its functionality and specificity. Based on manufacturer recommendations, the following storage guidelines should be followed:

  • Short-term storage (up to one week): Store undiluted antibody at 2-8°C.

  • Long-term storage: Aliquot and store at -20°C or below. Storage in frost-free freezers is not recommended due to temperature fluctuations that can degrade antibody activity .

  • Avoid repeated freeze/thaw cycles: Multiple freeze/thaw cycles can lead to denaturation and aggregation of antibody molecules, resulting in decreased activity and increased background.

  • Preparation before use: Gently mix the antibody solution before use. Spin the vial prior to opening to recover all liquid content that may be lodged in the cap or walls .

  • Working dilution stability: Diluted antibody working solutions typically maintain activity for several weeks when stored at 4°C, but this should be verified experimentally for specific applications.

The BBM.1 antibody is typically supplied in PBS (pH 7.4) containing 0.05% sodium azide and 0.1 mg/ml BSA as stabilizers . These additives help maintain antibody stability during storage. It is important to note that sodium azide is toxic, and proper precautions should be taken when handling the antibody solution.

What controls should be included when working with BBM.1 antibody?

When designing experiments using BBM.1 antibody, appropriate controls are essential for valid interpretation of results. The following controls should be considered:

  • Positive control: Use Raji cells, which are known to express high levels of β2M and have been validated as a positive control for BBM.1 staining . Thymocytes and Molt 4 cells also show considerable expression of β2M .

  • Negative control: Include cells known to lack human β2M expression, such as mouse cells without human chromosome 15 .

  • Isotype control: Use a mouse IgG2b, kappa isotype control antibody at the same concentration as BBM.1 to assess non-specific binding.

  • Secondary antibody control: If using indirect detection methods, include a control with secondary antibody only to evaluate background staining.

  • Blocking peptide control: Consider using synthetic peptides (residues 35-50 of β2M) for competition assays to confirm specificity .

  • Parallel antibody control: Use W6/32 antibody (anti-HLA-A,B,C glycoprotein) in parallel with BBM.1 to compare the relative amounts of β2M and HLA-A,B,C glycoproteins on different cell types .

  • Antibody titration: Perform a titration series to determine the optimal antibody concentration that provides the best signal-to-noise ratio for your specific application.

  • Cross-reactivity control: As BBM.1 has been shown to recognize β2M only from humans, gorillas, and chimpanzees, include cells from other species as negative controls when relevant .

These controls help ensure experimental validity and assist in distinguishing true positive signals from artifacts or non-specific background.

How does the epitope recognition of BBM.1 differ from other anti-beta-2-microglobulin antibodies?

BBM.1 antibody possesses distinct epitope recognition characteristics compared to other anti-β2M antibodies, making it valuable for specific research applications. The defining features of BBM.1's epitope recognition include:

  • Location of epitope: BBM.1 recognizes a surface-exposed region around residue 45 of β2M that forms part of a loop between the two layers of beta-pleated sheet in the immunoglobulin-like domain structure . This location is significant as it is not involved in the interface with MHC class I heavy chains.

  • Key residues: The critical immunodominant residues for BBM.1 binding have been identified as Arginine 45, Glutamic acid 44, and Arginine 81, with Aspartic acid 38 playing a supporting role by stabilizing Arginine 45 . This specific arrangement of amino acids creates a unique epitope fingerprint.

  • Dual recognition capacity: Unlike some other anti-β2M antibodies that recognize either free β2M or MHC-associated β2M, BBM.1 binds both forms with similar affinity . This is possible because the epitope recognized by BBM.1 does not involve residues that interact with MHC class I heavy chains, allowing antibody access even in the assembled complex.

  • Species specificity: BBM.1 demonstrates high species specificity, binding only to β2M from humans, gorillas, and chimpanzees . This is in contrast to some other anti-β2M antibodies and to the W6/32 antibody (which recognizes HLA-A,B,C glycoproteins), where the antigenic determinant appears to be more phylogenetically conserved across species .

  • Structural modeling insights: Analysis of the BBM.1 Fv-β2M complex model reveals no steric conflict between the antibody and the HLA-I heavy chain, explaining mechanistically why BBM.1 can recognize β2M in both its free and MHC-associated forms . This structural understanding provides a basis for comparison with other anti-β2M antibodies that may have different binding modes.

These distinctive epitope recognition properties make BBM.1 particularly useful for applications requiring detection of total β2M (both free and MHC-associated) and for studies requiring species-specific recognition of human β2M.

What structural determinants influence the binding of BBM.1 to beta-2-microglobulin?

The binding of BBM.1 antibody to beta-2-microglobulin involves specific structural determinants that have been elucidated through various biochemical and structural modeling approaches. These determinants include:

  • Critical immunodominant residues: Comprehensive structural modeling and biochemical studies have identified key amino acid residues in β2M that directly interact with BBM.1. Arginine 45 has been established as a major component of the antigenic determinant, as confirmed by reversible modification studies with cyclohexanedione that resulted in up to 95% loss of BBM.1 inhibitory activity . Additionally, Glutamic acid 44 and Arginine 81 have been identified as direct interaction partners with the antibody .

  • Supporting residues: Aspartic acid 38, while not directly contacting BBM.1, plays a crucial supporting role by stabilizing the conformation of Arginine 45 through intramolecular interactions, thus indirectly contributing to epitope formation .

  • Epitope conformation: The epitope lies within a surface-exposed loop between the two layers of beta-pleated sheet in the immunoglobulin-like domain structure of β2M . This conformational arrangement is critical for antibody recognition and accessibility.

  • Non-interference with MHC binding: Structural modeling studies have demonstrated that there is no steric conflict between BBM.1 and the HLA-I heavy chain when β2M is in complex with MHC . This explains the ability of BBM.1 to bind both free and MHC-associated forms of β2M with similar affinity.

  • Peptide inhibition studies: Synthetic peptides derived from residues 35-50 of the β2M sequence have been shown to specifically inhibit BBM.1 binding to cell surfaces, confirming this region as the epitope . The inhibitory activity of these peptides is destroyed by trypsin treatment, highlighting the importance of the intact peptide structure.

  • Hydrophilicity analysis: Analysis of the β2M sequence using local hydrophilicity indices agrees with the region around residue 45 being a major antigenic determinant , consistent with the general principle that hydrophilic, surface-exposed regions often serve as antibody epitopes.

  • Species specificity determinants: The species specificity of BBM.1 (binding only to human, gorilla, and chimpanzee β2M) corresponds to the conservation of Arginine 45 in these species , confirming this residue as a critical determinant of recognition.

Understanding these structural determinants provides insights into the molecular basis of BBM.1 recognition of β2M and can inform the design of experiments involving this antibody as well as potential engineering efforts to modify its binding properties for specific applications.

How can BBM.1 be used to investigate beta-2-microglobulin-related pathologies?

BBM.1 antibody offers valuable methodological approaches for investigating β2M-related pathologies, particularly in the context of dialysis-related amyloidosis (DRA) and other conditions involving β2M dysregulation:

  • Quantification of free vs. bound β2M: BBM.1 can be used alongside antibodies specific for MHC class I heavy chains (such as W6/32) to quantify the relative amounts of free and MHC-associated β2M in patient samples . This is particularly relevant for DRA, where free β2M can accumulate and form amyloid fibrils. The protocol involves:

    • Parallel staining of samples with BBM.1 (total β2M) and W6/32 (MHC-associated)

    • Flow cytometric analysis to determine relative expression levels

    • Calculation of ratios to estimate free β2M levels

  • Therapeutic β2M elimination: BBM.1 has been shown to have potential for specific elimination of β2M from serum , which could be leveraged for therapeutic interventions in DRA. Researchers can explore this application through:

    • Immobilization of BBM.1 on solid supports for extracorporeal β2M removal

    • Evaluation of removal efficiency using Western blot or ELISA for β2M quantification

    • Assessment of the structural integrity of captured β2M to understand amyloid formation mechanisms

  • Amyloid fibril formation studies: Since β2M has a predominantly beta-pleated sheet structure that can form amyloid fibrils in pathological conditions , BBM.1 can be used to:

    • Monitor conformational changes in β2M during fibril formation using epitope accessibility assays

    • Investigate whether BBM.1 binding affects the kinetics of β2M amyloid formation

    • Develop immunohistochemical procedures for detecting β2M amyloid deposits in tissue samples

  • Analysis of β2M mutations: For investigating hypercatabolic hypoproteinemia associated with β2M mutations , BBM.1 can be employed to:

    • Compare binding efficiency to wild-type and mutant β2M

    • Assess the impact of mutations on β2M-MHC interactions

    • Evaluate altered cellular processing of mutant β2M

  • Tumor-related applications: Since BBM.1 can induce apoptosis in several types of tumor cells , researchers can:

    • Determine β2M expression levels in various tumor types using BBM.1 staining

    • Investigate the mechanism of BBM.1-induced apoptosis in β2M-expressing tumor cells

    • Explore the therapeutic potential of BBM.1 or derived antibodies for targeting β2M-overexpressing malignancies

  • Antibody engineering for therapeutic applications: The detailed understanding of BBM.1's epitope recognition provides a foundation for engineering improved anti-β2M antibodies for therapeutic purposes . This could involve:

    • Structure-guided modifications to enhance binding affinity or specificity

    • Development of humanized or fully human versions of BBM.1 for reduced immunogenicity

    • Creation of BBM.1-based bispecific antibodies for targeted therapies

These methodological approaches demonstrate the versatility of BBM.1 as a research tool for investigating β2M-related pathologies and its potential for translation into therapeutic applications.

What experimental approaches using BBM.1 can help distinguish between cell surface and intracellular beta-2-microglobulin?

Distinguishing between cell surface and intracellular β2M populations is crucial for understanding β2M biology and pathology. BBM.1 antibody can be employed in several sophisticated experimental approaches to achieve this differentiation:

  • Flow cytometry with selective permeabilization:

    • Surface staining: Incubate live cells with BBM.1 at 4°C (prevents internalization) in buffer without detergents

    • Total staining: Fix cells with 2-4% paraformaldehyde followed by permeabilization with 0.1% saponin or 0.1% Triton X-100, then stain with BBM.1

    • Analysis: Compare mean fluorescence intensity between permeabilized and non-permeabilized samples to estimate the intracellular fraction

    • Controls: Include W6/32 antibody (which detects assembled MHC-I) to determine the proportion of β2M that is MHC-associated on the cell surface

  • Confocal microscopy with differential staining:

    • Sequential staining protocol: First label surface β2M with BBM.1 conjugated to one fluorophore (e.g., FITC)

    • Fix and permeabilize cells, then label total β2M with BBM.1 conjugated to a different fluorophore (e.g., Cy5)

    • Image analysis: Colocalization analysis to identify regions of overlap (surface) versus Cy5-only signals (intracellular)

    • Quantification: Calculate the Manders' overlap coefficient to determine the proportion of total β2M present on the cell surface

  • Biochemical fractionation combined with Western blotting:

    • Isolate membrane fractions through ultracentrifugation or commercial membrane protein extraction kits

    • Separate cytosolic and organelle fractions

    • Perform Western blotting with BBM.1 on each fraction

    • Controls: Blot for membrane markers (e.g., Na+/K+ ATPase) and cytosolic markers (e.g., GAPDH) to confirm fraction purity

    • Quantification: Densitometric analysis to determine relative distribution between compartments

  • Surface biotinylation assay:

    • Selectively label surface proteins with membrane-impermeable biotin reagents

    • Isolate biotinylated proteins using streptavidin pull-down

    • Perform Western blotting with BBM.1 on both biotinylated (surface) and non-biotinylated (intracellular) fractions

    • Quantification: Compare the amount of β2M in each fraction to determine cellular distribution

  • Enzyme-linked immunosorbent assay (ELISA) for secreted β2M:

    • Collect cell culture supernatants at various time points

    • Perform sandwich ELISA using BBM.1 as capture or detection antibody

    • Controls: Include measurements of total cellular β2M from lysates

    • Analysis: Calculate the ratio of secreted to cellular β2M to assess trafficking patterns

  • Pulse-chase experiments with immunoprecipitation:

    • Metabolically label cells with 35S-methionine/cysteine

    • Chase with unlabeled media for various time points

    • Perform surface biotinylation at each time point

    • Immunoprecipitate with BBM.1 from whole cell lysates

    • Separate biotinylated (surface) from non-biotinylated (intracellular) fractions

    • Analysis: Track the kinetics of β2M movement from intracellular compartments to the cell surface

These methodological approaches provide complementary information about the distribution and trafficking of β2M between cellular compartments, offering valuable insights into normal β2M biology and disease-related alterations.

How can BBM.1 be leveraged in therapeutic development against malignancies with beta-2-microglobulin dysregulation?

BBM.1 antibody presents several promising avenues for therapeutic development against malignancies characterized by β2M dysregulation. These methodological approaches bridge basic research and translational applications:

  • Direct tumor targeting based on apoptosis induction:
    BBM.1 has demonstrated the ability to induce apoptosis in several types of tumor cells . Researchers can leverage this property by:

    • Screening cancer cell lines for susceptibility to BBM.1-induced apoptosis

    • Determining the molecular mechanism of cell death (e.g., through caspase activation assays, mitochondrial membrane potential measurements)

    • Optimizing antibody concentration and exposure time for maximal tumor cell killing with minimal effect on normal cells

    • Developing in vivo models to test efficacy using humanized mice or patient-derived xenografts

  • Antibody engineering based on BBM.1 binding properties:
    The detailed understanding of BBM.1's epitope recognition provides a foundation for creating enhanced therapeutic antibodies:

    • Develop humanized or fully human versions of BBM.1 to reduce immunogenicity

    • Create BBM.1-based antibody-drug conjugates (ADCs) by coupling the antibody to cytotoxic payloads

    • Design bispecific antibodies incorporating the BBM.1 binding domain alongside domains targeting T-cell activation markers (e.g., CD3)

    • Optimize affinity through targeted mutations in the complementarity-determining regions (CDRs)

  • Combination therapies targeting β2M-MHC interactions:
    Since BBM.1 binds both free and MHC-associated β2M , it can be used to disrupt tumor immune evasion mechanisms:

    • Combine BBM.1 treatment with immune checkpoint inhibitors to enhance anti-tumor immune responses

    • Evaluate effects on natural killer (NK) cell recognition of tumor cells, as β2M-MHC complexes regulate NK cell activity

    • Assess impact on antigen presentation and T-cell recognition following BBM.1 treatment

  • Development of BBM.1-based CAR-T cell therapies:
    The single-chain variable fragment (scFv) derived from BBM.1 could be incorporated into chimeric antigen receptor (CAR) constructs:

    • Clone and optimize BBM.1 scFv for CAR expression

    • Test functionality in primary T cells against β2M-expressing tumor cells

    • Evaluate potential on-target, off-tumor effects using normal tissues expressing β2M

    • Develop safety switches or tumor-specific recognition strategies to improve selectivity

  • Theranostic applications:
    BBM.1 can be utilized for both diagnostic imaging and therapeutic delivery:

    • Radiolabel BBM.1 with imaging isotopes (e.g., 89Zr, 124I) to visualize β2M-expressing tumors through PET imaging

    • Use this information to select patients most likely to benefit from β2M-targeted therapies

    • Transition to therapeutic isotopes (e.g., 177Lu, 90Y) for radioimmunotherapy approaches

    • Develop companion diagnostics based on BBM.1 epitope recognition

  • Structural optimization through computer-aided design:
    The availability of structural models of the BBM.1 Fv-β2M complex enables rational design approaches:

    • Perform in silico affinity maturation through computational modeling of binding interface modifications

    • Design smaller binding molecules (e.g., peptides, aptamers) that mimic BBM.1's epitope recognition

    • Create structure-based screening assays to identify small molecules that bind the same epitope

These methodological approaches demonstrate how BBM.1's unique properties can be leveraged for developing novel therapeutic strategies against malignancies with β2M dysregulation, potentially opening new avenues for precision oncology.

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.