The term "DI-B4" appears in clinical trial data as a monoclonal antibody targeting CD19, a protein expressed on B cells. Key findings from a Phase I/II trial (NCT identifier not provided) include:
| Parameter | DI-B4 Trial Results |
|---|---|
| Target | CD19-positive B-cell malignancies |
| Study Population | Relapsed/refractory low-grade B-cell lymphoma and CLL |
| Dosing | Weekly infusions for 4 weeks |
| Response Rate (lymphoma) | 3/21 partial response; 12/21 stable disease |
| Safety Profile | Dose-dependent toxicity observed |
This trial highlighted modest efficacy but underscored CD19 as a validated therapeutic target in B-cell disorders .
CD19-targeted therapies are a cornerstone of B-cell malignancy treatment. Notable agents include:
Mechanism: Humanized anti-CD19 monoclonal antibody.
Indications: FDA-approved for neuromyelitis optica spectrum disorder (NMOSD) .
Key Data:
Structure: Anti-CD19 antibody conjugated to pyrrolobenzodiazepine (PBD) dimer.
Feature: Non-depleting anti-CD19 antibody inhibiting B-cell activation.
Preclinical Impact: Suppressed B-cell proliferation (IC₅₀ < 1 nM) .
Unrelated to immunology, Di19-3 refers to a Drought-induced zinc-finger transcription factor in Arabidopsis thaliana:
This underscores the importance of distinguishing between immunotherapeutics and plant biology terminology.
While unrelated to "DI19-4," advances in antibody engineering for dengue virus (e.g., J9 bNAb) and aquaporin-4 autoantibodies highlight methodological parallels:
Dengue bNAbs: Target domain I/III epitopes (IC₅₀ = 6–39 ng/mL) .
AQP4-IgG: Pathogenicity linked to Fc-driven complement activation .
If "DI19-4" refers to an experimental or preclinical compound, the following steps are recommended:
Nomenclature Clarification: Cross-reference identifiers (e.g., INN, CAS).
Patent Databases: Search for undisclosed developmental candidates.
Structural Analysis: Compare with CD19, CD20, or aquaporin-4-targeted agents.
KEGG: ath:AT3G06760
UniGene: At.40468
DI-B4 is an anti-CD19 monoclonal antibody designed to target CD19-positive B-cell malignancies. CD19 is a B-cell surface protein present on both normal and malignant B-cells, including those found in non-Hodgkin Lymphoma (NHL), chronic lymphocytic leukemia (CLL), and acute lymphoblastic leukemia (ALL). The antibody recognizes specific epitopes on CD19 and attaches to the cell surface to initiate immune-mediated cytotoxicity .
DI-B4 functions primarily by binding to CD19 on B-cells and recruiting host immune cells toward the tumor site, ultimately leading to cancer cell death. This targeting strategy allows for selective attack against CD19-expressing malignant cells while potentially minimizing off-target effects .
CD19 has emerged as a significant target for hematological malignancies, with several antibodies in development. DI-B4 is being investigated in a Phase I clinical trial specifically for advanced CD19-positive indolent B-cell malignancies . Unlike some other anti-CD19 approaches, DI-B4 is initially being studied as a standalone antibody rather than as part of a conjugated therapeutic.
The following table compares key properties of various CD19-targeting strategies:
| Therapeutic Approach | Format | Mechanism of Action | Current Development Stage |
|---|---|---|---|
| DI-B4 | Monoclonal Antibody | Immune cell recruitment | Phase I clinical trial |
| CD19 ADCs | Antibody-Drug Conjugate | Targeted cytotoxic delivery | Various stages |
| CD19 CAR-T | Cellular Therapy | Engineered T-cell attack | FDA approved for some indications |
| CD19 BiTEs | Bispecific Antibody | T-cell engagement | Various stages |
The Phase I trial for DI-B4 follows a dose escalation design to determine the maximum tolerated dose (MTD) up to 1000mg. The study consists of two phases:
Dose escalation phase: Recruiting approximately 15-20 patients to determine MTD
Dose expansion cohort: Recruiting up to an additional 20 patients at the identified optimal dose
The trial targets patients with relapsed or refractory CD19-positive indolent B-cell lymphoma or chronic lymphocytic leukemia. DI-B4 is administered intravenously on a weekly schedule for four weeks .
Primary objectives include safety assessment and determination of the maximum tolerated dose, while secondary objectives likely include preliminary efficacy evaluation and pharmacokinetic characterization .
Based on the clinical trial information, researchers should consider the following patient populations for DI-B4 investigation:
Patients with relapsed or refractory disease
Confirmed CD19-positive status through immunohistochemistry or flow cytometry
Indolent B-cell lymphomas (including follicular lymphoma, marginal zone lymphoma)
Chronic lymphocytic leukemia
Patients who have failed standard treatment approaches
Researchers should implement appropriate CD19 expression screening methodologies to identify suitable patients, as expression levels may vary between different B-cell malignancies and potentially influence response rates .
For researchers investigating DI-B4's binding properties, the following methodologies are recommended:
Flow cytometry binding assays: To quantify binding affinity to CD19-expressing cell lines and primary patient samples
Surface plasmon resonance (SPR): For precise measurement of kon/koff rates and KD values
Epitope mapping: To identify the specific binding region on CD19
Competitive binding assays: To compare with other anti-CD19 antibodies
When evaluating antibody binding characteristics, researchers should consider the following parameters:
| Parameter | Technical Approach | Importance |
|---|---|---|
| Binding affinity | SPR, ELISA | Determines potency requirements |
| Epitope specificity | Epitope mapping, mutational analysis | Influences mechanism of action |
| Cross-reactivity | Testing against related proteins | Assesses potential off-target effects |
| Species cross-reactivity | Multi-species binding assays | Informs preclinical model selection |
When investigating DI-B4's immune-recruiting capabilities, researchers should implement comprehensive in vitro and in vivo assays to characterize:
Antibody-dependent cellular cytotoxicity (ADCC): Using NK cells or other effector cells co-cultured with antibody-opsonized target cells
Complement-dependent cytotoxicity (CDC): Measuring complement activation and membrane attack complex formation
Antibody-dependent cellular phagocytosis (ADCP): Assessing macrophage-mediated clearance of antibody-bound cells
Fc receptor binding profiles: Characterizing interactions with various FcγRs to predict immune activation
These functional assays should be conducted using physiologically relevant effector-to-target ratios and with appropriate controls to isolate specific mechanisms of action .
While the current clinical trial is evaluating DI-B4 as an unconjugated antibody, researchers interested in developing DI-B4-based ADCs should consider the following factors:
Conjugation chemistry: The method of attaching payloads significantly affects ADC homogeneity, stability, and efficacy. Options include:
Drug-antibody ratio (DAR): The optimal number of payload molecules per antibody must be determined experimentally, balancing potency with pharmacokinetic properties. Conventional stochastic conjugation methods can produce heterogeneous products with DARs ranging from 0-8, while site-specific methods allow for more precise DAR control .
Linker selection: The choice between cleavable and non-cleavable linkers affects the mechanism of payload release and potentially the bystander effect. Researchers should evaluate:
For developing homogeneous DI-B4 ADCs, researchers should consider these site-specific strategies:
ThioMab technology: Engineering cysteine residues at specific positions (e.g., light chain V110A and heavy chain A114C) to provide controlled conjugation sites. This approach can achieve high homogeneity (>90% with DAR of 2) .
Disulfide re-bridging: Using bis-reactive reagents like bissulfone reagents, next-generation maleimides (NGMs), or pyridazinediones (PDs) to reconnect reduced interchain disulfides while simultaneously introducing payloads .
Unnatural amino acid incorporation: Introducing amino acids like N-acetyl-L-phenylalanine or azido methyl-L-phenylalanine at specific positions to enable bio-orthogonal conjugation chemistry .
Enzymatic approaches: Using formyl glycine-generating enzyme (FGE) or transglutaminase (TG) to modify specific amino acid sequences engineered into the antibody structure .
Glycoengineering: Exploiting the N-glycan at position N297 in the CH2 domain of each heavy chain for payload attachment, which minimizes interference with antigen binding .
Researchers studying DI-B4 pharmacokinetics should implement a comprehensive sampling strategy that accounts for the unique properties of monoclonal antibodies:
Early distribution phase: Samples at 0h (pre-dose), 1h, 6h, and 24h post-infusion to capture distribution kinetics
Elimination phase: Weekly samples to characterize elimination half-life
Anti-drug antibody (ADA) monitoring: Regular screening throughout treatment to assess immunogenicity
Receptor occupancy analysis: Flow cytometry-based assays to determine CD19 saturation on circulating B-cells
Sample processing should include validated assays for:
Total antibody concentration
Functional antibody concentration
Anti-drug antibody detection
Circulating CD19+ B-cell quantification
Effective pharmacodynamic monitoring for DI-B4 should include:
B-cell depletion kinetics: Flow cytometric analysis of peripheral blood to track CD19+ B-cell counts
Tissue penetration assessment: When feasible, analysis of lymph node or bone marrow biopsies to evaluate tissue distribution
Cytokine release monitoring: Measurement of inflammatory cytokines, particularly during initial dosing
Immune cell activation markers: Assessment of NK cell, macrophage, and complement activation
The following biomarkers may be particularly valuable:
| Biomarker Type | Specific Markers | Assessment Method | Clinical Relevance |
|---|---|---|---|
| Target engagement | CD19 occupancy | Flow cytometry | Confirms antibody binding to target |
| Immune activation | CD69, CD25, CD107a | Flow cytometry | Indicates effector cell activation |
| Cytokine response | IL-6, TNF-α, IFN-γ | ELISA/cytometric bead array | Monitors inflammatory response |
| B-cell depletion | CD19+/CD20+ counts | Flow cytometry | Demonstrates biological effect |
| Tumor response | Circulating tumor DNA | Digital PCR | Quantifies tumor burden reduction |
As with other targeted therapies, resistance to DI-B4 may develop through various mechanisms that researchers should proactively investigate:
Target modulation:
CD19 downregulation or internalization
Epitope mutations affecting antibody binding
Alternative splicing of CD19
Shedding of CD19 into circulation
Immune evasion:
Downregulation of complement regulatory proteins
Upregulation of inhibitory Fc receptors
Impaired NK cell or macrophage function
Immunosuppressive tumor microenvironment
Signaling adaptation:
Activation of alternative survival pathways
Upregulation of anti-apoptotic proteins
Metabolic reprogramming
Researchers should develop resistance models through chronic exposure to DI-B4 and characterize molecular changes associated with reduced sensitivity.
Based on the mechanism of action and potential resistance pathways, researchers should consider these combination approaches:
Immune-enhancing combinations:
Checkpoint inhibitors (anti-PD-1/PD-L1)
Immunomodulatory drugs (IMiDs)
Toll-like receptor agonists
Cytokine therapy
Pathway-targeting combinations:
BCR pathway inhibitors (BTK, PI3K inhibitors)
BCL-2 inhibitors
Proteasome inhibitors
Epigenetic modifiers
Multi-targeting antibody combinations:
Anti-CD20 antibodies
Anti-CD22 antibodies
Anti-CD79b antibodies
Researchers should utilize high-throughput screening approaches and mechanism-based rational combinations to identify synergistic interactions with DI-B4.
Researchers should explore advanced applications of DI-B4 beyond its current formulation:
Bispecific adaptations: Engineering DI-B4 into bispecific formats targeting CD19 and:
CD3 for T-cell engagement
CD16 for enhanced NK cell recruitment
Additional B-cell markers for dual-targeting
CAR-T cell development: Incorporating the DI-B4 binding domain into chimeric antigen receptor constructs
Radioimmunotherapy conjugates: Attaching radioisotopes for targeted radiation delivery
Immunocytokine fusions: Creating DI-B4-cytokine fusion proteins to enhance localized immune stimulation
To advance understanding of DI-B4 biology, researchers should incorporate cutting-edge analytical approaches:
Single-cell technologies:
Single-cell RNA sequencing to characterize heterogeneous responses
Mass cytometry (CyTOF) for high-dimensional phenotyping
Imaging mass cytometry for spatial context in tissue samples
Advanced microscopy:
Live-cell imaging to track antibody internalization and trafficking
Super-resolution microscopy to visualize molecular clustering
Intravital microscopy for in vivo dynamics
Systems biology approaches:
Computational modeling of antibody pharmacokinetics
Network analysis of resistance mechanisms
Machine learning for biomarker identification