NOG Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Nogo-A Monoclonal Antibodies

Nogo-A is a membrane protein expressed by oligodendrocytes and neurons, primarily inhibiting axonal regeneration and plasticity in the central nervous system (CNS) . Anti-Nogo-A mAbs neutralize its inhibitory activity, promoting neuronal repair.

Mechanism of Action:

  • Neurite Outgrowth Promotion: Antibodies block Nogo-A’s interaction with receptors (e.g., NgR1), enabling neurite sprouting and synaptic plasticity .

  • Neuroprotection: Enhances survival of stressed neurons by modulating L1 cell adhesion molecule and polysialic acid expression .

  • Target-Mediated Drug Disposition (TMDD): High-affinity binding to Nogo-A may influence pharmacokinetics (PK) .

Noggin Monoclonal Antibodies

Noggin binds and inhibits bone morphogenetic proteins (BMPs), regulating developmental pathways. Anti-Noggin mAbs are primarily used as research tools.

Applications:

  • Immunohistochemistry (IHC): Detects Noggin in muscle biopsies or developmental tissues .

  • Western Blotting (WB): Validates Noggin protein expression in experimental models .

Nogo-A Antibodies in Neurological Disorders

IndicationKey FindingsSource
Amyotrophic Lateral Sclerosis (ALS)Ozanezumab (humanized anti-Nogo-A mAb) showed dose-dependent skeletal muscle co-localization with Nogo-A in Phase I/IIa trials.
Ischemic StrokeIntranasal anti-Nogo-A mAb delivery enhanced corticofugal projection growth and functional recovery in rat models.
Spinal Cord InjuryPreclinical studies demonstrated improved axonal regeneration and motor function.

Noggin Antibodies in Research

ApplicationDetailsSource
IHC StainingMouse mAb (clone OTI1C1) detects Noggin in human, mouse, and rat tissues.
WB AnalysisAnti-Noggin mAb (clone 4C9) identifies protein expression at ~23.7 kDa.

Nogo-A Antibodies

  • In Vivo Efficacy:

    • Stroke Models: Daily intranasal anti-Nogo-A mAb (2 weeks) improved forelimb function and corticospinal tract sprouting in rats .

    • ALS Trials: Ozanezumab (0.01–15 mg/kg IV) demonstrated safety and target engagement in skeletal muscle .

  • In Vitro Mechanisms:

    • Neuronal Survival: 5 μg/mL monoclonal anti-Nogo-A increased survival of H₂O₂-stressed cerebellar granule neurons by 25–30% .

    • Signal Transduction: c-Src, c-Fyn, and PKA inhibitors reduced antibody-induced neurite outgrowth, highlighting signaling pathways .

Noggin Antibodies

  • Reactivity:

    • Species Specificity: Mouse mAb (OTI1C1) reacts with human, mouse, and rat Noggin .

    • Epitope: Targets amino acids 28–232 of human Noggin .

Nogo-A Antibodies

  • PK Parameters:

    • Ozanezumab: Linear kinetics observed at doses ≤15 mg/kg; clearance influenced by FcRn binding .

    • Intranasal Delivery: Achieved CNS concentrations comparable to intrathecal administration .

  • Safety Profile:

    • Common AEs: Flu-like symptoms (fatigue, fever), injection-site reactions .

    • Rare Risks: Cytokine release syndrome, capillary leak syndrome .

Comparative Analysis: Nogo-A vs. Noggin Antibodies

ParameterNogo-A AntibodiesNoggin Antibodies
Target FunctionInhibits axonal regenerationInhibits BMP signaling
Therapeutic UseNeurological disorders (ALS, stroke)Research (IHC, WB)
Antibody SourceHumanized (e.g., ozanezumab)Mouse (e.g., clone OTI1C1)
Key Clinical DataPhase I/IIa trials for ALS No therapeutic applications reported

Nogo-A Antibodies

  • Manufacturing: High production costs and scalability issues .

  • Next-Gen mAbs: Dual-targeting antibodies (e.g., Nogo-A + Ephrin) for enhanced CNS penetration .

Noggin Antibodies

  • Research Limitations: Limited to preclinical studies; no human therapeutic applications .

Product Specs

Form
The monoclonal antibody is supplied as a liquid formulation in phosphate-buffered saline (PBS) containing 50% glycerol, 0.5% bovine serum albumin (BSA), and 0.02% sodium azide. The pH is adjusted to 7.3.
Lead Time
Orders are typically dispatched within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
NOG; NOGG_HUMAN; Noggin; SYM 1; SYM1; Symphalangism 1 (proximal); Synostoses (multiple) syndrome 1; SYNS 1; SYNS1.

Q&A

What is the NOG mouse model and why is it significant for monoclonal antibody research?

The NOG (NOD/Shi-scid-IL2Rγnull) mouse is an immunodeficient model lacking functional T cells, B cells, and NK cells due to the scid mutation and IL-2Rγ deficiency. This model is particularly valuable for monoclonal antibody research as it permits successful engraftment of human immune cells and tumors, creating an effective humanized model system.

The severe immunodeficiency of NOG mice provides an ideal platform for evaluating human-specific monoclonal antibodies without interference from the mouse's own immune system. This makes them especially valuable for studying antibody-dependent cellular cytotoxicity (ADCC), a key mechanism for many therapeutic monoclonal antibodies targeting cancer and other diseases .

How do NOG-FcγR−/− mice differ from standard NOG mice for antibody research?

NOG-FcγR−/− mice are engineered to be deficient for both Fcer1g and Fcgr2b genes, resulting in monocytes/macrophages that do not express either activating (FcγRI, III, and IV) or inhibitory (FcγRIIB) Fcγ receptors. This genetic modification creates several significant advantages:

  • ADCC by mouse innate immune cells is strongly reduced in this strain

  • Xenogeneic human tumor growth inhibition by mouse innate cells upon antibody treatment is abrogated

  • Segregation of human NK cell-mediated ADCC from mouse cell-mediated ADCC becomes possible

These characteristics make NOG-FcγR−/− mice particularly valuable for validating the in vivo function of antibody drug candidates, allowing researchers to specifically attribute observed effects to human immune cell activity rather than mouse innate immunity.

What mechanisms do monoclonal antibodies use to target cancer cells?

Monoclonal antibodies employ multiple mechanisms to combat cancer cells:

  • Recognition of specific targets: Monoclonal antibodies are designed to interact with specific targets on cancer cells, functioning as targeted cancer therapy

  • Immune system marking: Some monoclonal antibodies mark cancer cells so the immune system can better recognize and destroy them. For example, rituximab binds to CD20 on B cells and some cancer cells, triggering immune-mediated destruction

  • T cell recruitment: Certain antibodies bring T cells in proximity to cancer cells, facilitating immune response. Blinatumomab (Blincyto®) exemplifies this approach by binding to both CD19 on leukemia cells and CD3 on T cells

  • Direct signaling interference: Antibodies can directly disrupt growth signaling pathways essential for cancer cell survival

  • Antibody-drug conjugates: Some monoclonal antibodies are conjugated with toxins to deliver cytotoxic payloads directly to cancer cells

The specific mechanism employed depends on the antibody design, target antigen, and therapeutic goal.

What are the common side effects of monoclonal antibody therapeutics?

Monoclonal antibodies can cause various side effects, which vary based on individual factors. Common adverse reactions include:

Needle site reactions:

  • Pain, swelling, soreness

  • Redness, itchiness, rash

Flu-like symptoms:

  • Chills, fatigue, fever

  • Muscle aches and pains

  • Nausea, vomiting, diarrhea

More serious adverse events:

  • Mouth and skin sores potentially leading to infections

  • Cardiovascular complications (high blood pressure, congestive heart failure, heart attacks)

  • Inflammatory lung disease

  • Allergic reactions (ranging from mild to severe)

  • Capillary leak syndrome (fluid and proteins leaking from blood vessels)

  • Cytokine release syndrome

Understanding these potential adverse effects is critical for researchers designing preclinical studies and early-phase clinical trials for monoclonal antibody therapeutics.

How does the addition of human IL-15 in NOG mice enhance monoclonal antibody research?

The development of NOG-hIL-15 transgenic mice represents a significant advancement for monoclonal antibody research. These mice express human IL-15, which provides several research advantages:

  • Enables long-term persistence of human NK cells in the mouse model

  • Supports human NK cell functionality, facilitating studies of ADCC mechanisms

  • Permits evaluation of antibody-dependent tumor suppression mediated specifically by human NK cells

  • Creates a more physiologically relevant environment for testing human-specific antibodies

When combined with FcγR deficiency (NOG-FcγR−/−-hIL-15 Tg), these mice provide an even more specialized platform where "specific detection of human NK cell-mediated ADCC is possible" without interference from mouse innate cells . This model is particularly valuable for evaluating antibody candidates that rely on NK cell-mediated mechanisms for their therapeutic effect.

What methodological approaches should be used to detect human NK cell-mediated ADCC in modified NOG models?

Effective detection of human NK cell-mediated ADCC in NOG-FcγR−/−-hIL-15 Tg mice requires sophisticated experimental design:

Experimental setup:

  • Include appropriate control groups (antibody alone, NK cells alone, and the combination)

  • Properly time the administration of antibodies relative to NK cell engraftment

  • Monitor changes in target cell populations over time

Evidence from research:
In experiments with Rituximab (anti-CD20) treatment, researchers observed that only mice receiving both human NK cells and antibody treatment showed protection of bone marrow cells, demonstrating the specificity of this model for detecting human NK cell-mediated ADCC .

Analytical techniques:

  • Flow cytometry to quantify target cell populations

  • Immunohistochemistry to visualize NK cell infiltration and target cell destruction

  • Cytokine profiling to assess NK cell activation status

  • Ex vivo functional assays to confirm NK cell cytotoxic capacity

This methodological approach allows researchers to specifically attribute observed therapeutic effects to human NK cell-mediated mechanisms rather than mouse innate immunity.

What pharmacokinetic factors must be considered when designing monoclonal antibody studies?

Monoclonal antibodies possess complex pharmacokinetic properties that require special consideration:

Key PK characteristics:

  • Poor bioavailability necessitating parenteral administration

  • Slow distribution throughout tissues

  • Both linear and non-linear elimination processes

  • Long persistence in the body compared to small molecules

Critical design considerations:

  • Selection of appropriate dosing regimens accounting for slow distribution

  • Extended sampling schedules to capture complete PK profiles

  • Monitoring for target-mediated drug disposition effects

  • Assessment of potential immunogenicity

  • Use of sentinel dosing approach in first-in-human studies

As noted by experts: "The PK and PD of mAbs are complex and differ from those of non-mAb drugs. There are numerous PK factors that should be taken into account when designing and running an early phase clinical trial, especially if an antibody has a novel mechanism of action."

How can alternative delivery methods improve monoclonal antibody efficacy in targeting specific tissues?

Conventional systemic administration of monoclonal antibodies faces significant challenges, particularly when targeting the central nervous system (CNS). Research has demonstrated that alternative delivery approaches can significantly improve antibody delivery to difficult-to-reach tissues:

Intranasal delivery advantages:

  • Provides "rapid transfer of significant amounts of antibody to the brain and spinal cord in intact adult rats"

  • Bypasses the blood-brain barrier that typically restricts antibody access

  • Offers a non-invasive alternative to intrathecal administration

  • Can achieve therapeutic concentrations sufficient to enhance functional recovery in disease models

Quantitative comparison of delivery methods:
Research comparing intranasal and intrathecal delivery of anti-Nogo-A antibody demonstrated detectible antibody levels throughout the CNS, including cervical, thoracic, and lumbar spinal cord after 7 days of daily intranasal application (1 mg/day) . This finding suggests that intranasal delivery represents a viable alternative to more invasive methods for CNS-targeted antibody therapies.

What safety monitoring protocols are essential for first-in-human trials of novel monoclonal antibodies?

First-in-human (FIH) studies with monoclonal antibodies require rigorous safety protocols:

Sentinel group approach:
"Regulatory agencies and research ethics committees rightly insist on the use of a 'sentinel' group approach in FIH, particularly in the single dose part, comprising one active-treated and one placebo-treated subject at the start of the study, and at each dose increment."

Essential safety monitoring elements:

  • Staggered dosing to identify adverse events before exposing entire cohorts

  • Safety review committees to evaluate emerging data

  • Comprehensive PK/PD data review at interim stages

  • Extended follow-up periods to capture delayed adverse effects

  • Specific monitoring for cytokine release syndrome and other antibody-specific reactions

Dose selection methodology:
The "growing shift from NOAEL to MABEL" (Minimum Anticipated Biological Effect Level) approach for selecting starting doses has potential to reduce risks to trial subjects being dosed with novel monoclonal antibodies for the first time .

This comprehensive safety approach is particularly crucial for antibodies with novel mechanisms of action, as demonstrated by past adverse events in clinical trials.

How can NOG mouse models be optimized for evaluating specific monoclonal antibody mechanisms?

Optimization of NOG mouse models requires tailored modifications based on the specific mechanism being studied:

For studying ADCC mechanisms:

  • NOG-FcγR−/− mice eliminate mouse innate cell interference

  • NOG-FcγR−/−-hIL-15 Tg mice support human NK cell function for extended periods

For evaluating antibody biodistribution:

  • Combining multiple imaging modalities with tissue sampling

  • Quantitative ELISA measurement of antibody concentrations in target tissues

For assessing on-target/off-tumor effects:

  • Humanization of relevant target antigens through knock-in approaches

  • More complete immune system reconstitution using CD34+ stem cells

These optimizations create "useful models for validating the in vivo function of antibody drug candidates" with greater translational relevance.

What are the key differences between monoclonal antibody pharmacology in NOG models versus human patients?

Several important pharmacological differences must be considered when translating findings from NOG models to humans:

ParameterNOG Mouse ModelsHuman PatientsImplications
Antibody half-lifeGenerally shorterTypically longerMay underestimate duration of effect
Target antigen expressionMay differ in distribution/densityHuman-specific patternsCould affect targeting efficiency
Immune effector functionsLimited to engrafted human cellsComplete native systemMay underestimate full therapeutic effect
Immunogenicity riskLimited assessment due to immunodeficiencyCritical clinical concernAdditional testing needed
BiodistributionMore rapid equilibration in smaller organismSlower tissue penetrationDosing adjustments required

These differences highlight why "Careful trial design, informed by knowledge of an antibody's PK peculiarities, is essential if the study is to run both smoothly and safely."

How can researchers minimize microglial activation when studying CNS-targeted monoclonal antibodies?

CNS-targeted monoclonal antibody research must address potential microglial activation, which can confound interpretation of results:

Research findings:
Studies examining intranasal delivery of anti-Nogo-A antibody (11C7) found "No difference in local microglial activity was observed compared with the untreated corresponding brain tissue" after both single (24h) and repeated (72h) intranasal application .

Methodological approaches to minimize activation:

  • Careful antibody formulation to reduce immunogenicity

  • Use of appropriate isotype controls to distinguish specific from non-specific effects

  • Monitoring microglial activation markers in treated tissues

  • Selection of administration routes that minimize neuroinflammation

  • Quantitative assessment of microglial morphology and activation state

These approaches help ensure that observed therapeutic effects are due to the antibody's mechanism of action rather than non-specific inflammatory responses.

What experimental approaches can distinguish between direct antibody effects and immune-mediated mechanisms?

Distinguishing between direct and immune-mediated antibody effects requires sophisticated experimental design:

Comparative studies in different NOG variants:

  • Standard NOG mice (residual mouse innate immunity)

  • NOG-FcγR−/− mice (reduced mouse ADCC)

  • NOG-FcγR−/−-hIL-15 Tg mice (human NK-mediated ADCC)

Experimental controls:

  • F(ab')2 fragments (lacking Fc region) to isolate direct binding effects

  • Fc-mutated antibodies with reduced effector function

  • Depletion of specific effector cell populations

Functional readouts:

  • Target phosphorylation/signaling studies to detect direct effects

  • Immune cell infiltration and activation markers for immune-mediated mechanisms

  • Temporal analysis to separate rapid direct effects from delayed immune responses

These approaches help researchers determine "the full PD pathways" of a therapeutic antibody, which is critical for "selecting the most appropriate animal species from both PK/PD and safety considerations" before advancing to clinical studies .

What are the most important considerations when planning NOG mouse studies for monoclonal antibody development?

Effective planning of NOG mouse studies for monoclonal antibody development requires attention to several critical factors:

Key experimental design considerations:

  • Selection of appropriate NOG variant based on mechanism of interest

  • Adequate sample size for statistical power

  • Appropriate dosing schedule accounting for antibody pharmacokinetics

  • Comprehensive endpoint assessment (efficacy, safety, PK/PD)

  • Inclusion of relevant control groups and antibodies

Technical factors:

  • Standardization of human cell engraftment protocols

  • Validation of analytical methods for detecting human cells and antibodies

  • Quality control of antibody preparations to ensure consistency

Translational considerations:

  • Selection of clinically relevant dosing regimens

  • Incorporation of biomarkers applicable to human studies

  • Design of experiments that address specific regulatory requirements

As noted in the research: "Before planning a first-in-human (FIH) study, robust preclinical data should be available providing sufficient insight into the full PD pathways, and used to select the most appropriate animal species from both PK/PD and safety considerations."

How might newer generation NOG models further enhance monoclonal antibody research?

Next-generation NOG models are being developed with additional modifications that could significantly advance monoclonal antibody research:

Emerging NOG platforms:

  • Models with humanized target antigens (knock-in approaches)

  • Further refined human cytokine expression profiles

  • NOG variants expressing human Fc receptors

  • Models supporting more complete human immune system reconstitution

Potential research applications:

  • More accurate prediction of therapeutic index

  • Better assessment of on-target/off-tumor effects

  • Enhanced evaluation of combination therapies

  • Improved modeling of antibody dosing regimens

These advanced models will likely provide "a proof of concept for academic research" with increased translational relevance, addressing some of the current limitations in predicting human responses .

What are the emerging delivery strategies for improving monoclonal antibody efficacy?

Novel delivery approaches are addressing traditional limitations of monoclonal antibody therapy:

Innovative delivery methods:

  • Intranasal delivery has shown promise for CNS targeting, with research demonstrating that "intranasally applied therapeutic monoclonal antibody was sufficient to enhance functional recovery in a model of ischemic stroke"

  • Advances in formulation science to enhance tissue penetration

  • Engineered antibody fragments with improved biodistribution properties

  • Site-specific delivery systems to increase local concentration while minimizing systemic exposure

These approaches aim to overcome the inherent challenges of monoclonal antibody therapy, including poor bioavailability and limited tissue penetration, potentially expanding the therapeutic applications of these agents.

How can combination therapies be effectively evaluated using NOG mouse models?

Evaluation of combination therapies requires specialized approaches in NOG mouse models:

Methodological considerations:

  • Staggered introduction of therapeutic agents to assess sequence-dependent effects

  • Comprehensive assessment of pharmacokinetic interactions

  • Monitoring for unexpected toxicities from combination therapy

  • Evaluation of multiple dosing ratios to identify optimal combinations

Experimental design:

  • Inclusion of single-agent control groups for each component

  • Detailed analysis of mechanism-based biomarkers

  • Assessment of potential antagonistic interactions

  • Evaluation of immune cell phenotypes and functionality changes

NOG models are particularly valuable for evaluating combinations of antibody therapeutics with other modalities, including cellular therapies, small molecules, and additional antibodies targeting complementary pathways.

What role will advanced imaging play in monoclonal antibody development using NOG models?

Advanced imaging techniques are becoming increasingly important in monoclonal antibody research:

Key imaging applications:

  • Real-time tracking of antibody biodistribution

  • Monitoring target engagement in living animals

  • Assessment of pharmacodynamic responses

  • Visualization of immune cell trafficking and interaction with target cells

Emerging technologies:

  • Intravital microscopy for dynamic cellular interactions

  • PET imaging with radiolabeled antibodies for whole-body distribution

  • Optical imaging using fluorescently labeled antibodies

  • Multimodal approaches combining anatomical and functional imaging

These techniques will provide deeper insights into antibody mechanisms and help optimize dosing regimens, potentially accelerating the translation of findings from NOG models to clinical applications.

How can machine learning approaches improve translation of NOG model findings to human applications?

Machine learning offers promising approaches to enhance translation of NOG model data:

Potential applications:

  • Integration of preclinical PK/PD data with human in vitro findings to predict clinical outcomes

  • Pattern recognition across multiple NOG model variants to identify key predictive parameters

  • Development of algorithms to optimize antibody properties based on in vivo performance

  • Creation of translational models that account for species differences in target expression and immune function

By leveraging the growing body of data from NOG models alongside emerging clinical results, machine learning approaches may help address the current limitations in translating preclinical findings to human applications, ultimately improving prediction of clinical efficacy and safety.

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.