KRAS Antibody

Kirsten Rat Sarcoma Viral Oncogene, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

KRAS antibodies are monoclonal or polyclonal immunoglobulin molecules that bind specifically to epitopes on the KRAS protein, a GTPase central to the RAS/MAPK signaling pathway. KRAS mutations (e.g., G12V, G13D) occur in ~25% of human cancers and drive uncontrolled proliferation, metastasis, and therapy resistance . Antibodies targeting KRAS aim to disrupt its oncogenic signaling by:

  • Blocking GTP-binding domains

  • Interfering with membrane localization

  • Promoting immune recognition of mutant KRAS

Mechanisms of Action

KRAS antibodies employ diverse strategies to inhibit oncogenic signaling:

MechanismExample StudyKey Finding
Direct KRAS InhibitionIn vitro colorectal cancer models Anti-KRAS antibodies internalize, aggregate cytoplasmic KRAS, reducing membrane localization and SOX9 expression.
Antibody-Drug ConjugatesXenograft mouse models Anti-EGFR antibody-KRAS siRNA complexes reduce tumor growth in resistant cancers by 60–70%.
Immunotherapy SynergyUCSF/NYU Langone studies Antibodies bind drug-KRAS complexes on cell surfaces, recruiting T cells to eliminate resistant tumors.

Overcoming Drug Resistance

  • Anti-EGFR/KRAS-siRNA complexes restored sensitivity in cetuximab-resistant colon cancers, suppressing clonogenic growth by 85% in vitro .

  • ARS1620-KRAS antibody conjugates eliminated drug-resistant lung cancer cells by enhancing immune recognition .

Immune System Engagement

  • Engineered antibodies targeting KRAS G12V/HLA-A*03:01 complexes induced T-cell cytotoxicity across multiple cancer types .

  • Phosphoproteomic studies revealed antibody treatment alters B-Raf/MEK/PDK1 phosphorylation, disrupting downstream signaling .

Research Reagents

Commercially available KRAS antibodies enable precise detection and functional studies:

ProductHostApplicationsReactivityKey Feature
KRAS-4B (16155-1-AP)RabbitWB, IHC, IF/ICCHuman, MouseValidated in CRC tissues and cell lines
K-Ras (F234)MouseWB, IP, IFHuman, Mouse, RatTargets aa 54–189; 449 citations
#33197 (CST)RabbitWBHuman, Mouse, RatDetects endogenous K-Ras at 21 kDa

Clinical Translation

  • Phase I trials of KRAS G12C inhibitors + antibodies demonstrated 48-hour persistence of immune-recognizable KRAS-drug complexes .

  • Ex vivo patient-derived models confirmed antibody-mediated KRAS mislocalization reduces tumor viability by 40–50% .

Challenges and Future Directions

  • Delivery Limitations: Intracellular KRAS targeting requires advanced delivery systems (e.g., lipid nanoparticles) .

  • Mutation Heterogeneity: Pan-KRAS antibodies remain elusive due to allele-specific epitopes .

  • Combination Therapies: Ongoing trials explore antibodies with KRAS inhibitors (e.g., sotorasib) and checkpoint blockers .

Product Specs

Introduction
Kirsten Rat Sarcoma Viral Oncogene, Isoform 2A (KRAS-2A) is a protein belonging to the small GTPase superfamily. It plays a role in the development of various cancers, including lung adenocarcinoma, mucinous adenoma, pancreatic ductal carcinoma, and colorectal carcinoma. Under normal circumstances, Ras family members, including KRAS-2A, regulate cell growth and differentiation through a signaling system based on subcellular compartmentalization. However, in its oncogenic state, Ras can disrupt these processes, leading to uncontrolled cell proliferation and impaired apoptosis.
Physical Appearance
A clear and colorless solution that has been sterilized through filtration.
Formulation
The solution contains 1mg/ml of KRAS antibody dissolved in a buffer solution of PBS at pH 7.4. It also includes 10% glycerol and 0.02% sodium azide as preservatives.
Storage Procedures
For short-term storage of up to one month, the solution should be kept at a temperature of 4°C. For extended storage periods, it is recommended to store the solution at -20°C. To maintain product integrity, it is crucial to avoid repeated cycles of freezing and thawing.
Stability / Shelf Life
The product has a shelf life of 12 months when stored at -20°C and a shelf life of 1 month when stored at 4°C.
Applications
This KRAS antibody has undergone rigorous testing in various applications, including ELISA, Western blot analysis, Immunocytochemistry/Immunofluorescence (ICC/IF), and Flow cytometry. These tests confirm its specificity and reactivity for its target. As optimal conditions may vary depending on the specific application, it is recommended to perform a titration to determine the most effective concentration for each experiment.
Synonyms
GTPase KRas, K-Ras 2, Ki-Ras, c-K-ras, c-Ki-ras, KRAS, KRAS2, RASK2, C-K-RAS, CFC2, K-RAS2A, K-RAS2B, K-RAS4A, K-RAS4B, KI-RAS, KRAS1, NS, NS3, Kirsten rat sarcoma viral oncogene homolog.
Purification Method
KRAS antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
PAT2F8AT.
Immunogen
Anti-human KRAS mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a recombinant human KRAS protein 1-186 amino acids purified from E. coli.
Ig Subclass
Mouse IgG2b heavy chain and k light chain.

Q&A

What is KRAS and why are KRAS antibodies significant in cancer research?

KRAS is a proto-oncogene GTPase protein belonging to the RAS subfamily of small GTPases. The protein is approximately 21.7 kilodaltons in mass and is encoded by the KRAS gene, which may also be known as KRAS1, K-RAS2A, K-RAS2B, NS3, K-ras, and C-K-RAS . KRAS antibodies are significant in cancer research because RAS is one of the most common oncogenes, responsible for 20-30% of all human cancers, with KRAS specifically being responsible for most of these cases . Among the three major RAS isoforms (KRAS, HRAS, NRAS), KRAS is the most frequently mutated, accounting for approximately 80% of all RAS-driven cancers . KRAS mutations are particularly prevalent in some of the deadliest cancers, present in 77% of pancreatic, 43% of colorectal, and 27% of non-small cell lung cancers .

Methodologically, researchers use these antibodies to detect, quantify, and characterize KRAS proteins in various experimental settings, including tissue samples and cell cultures. The ability to specifically target KRAS is crucial for understanding its role in cancer development and progression, as well as for developing potential therapeutic strategies.

What applications are KRAS antibodies typically used for in research settings?

KRAS antibodies are utilized across multiple experimental techniques in cancer research:

ApplicationPurposeTypical DilutionsSpecial Considerations
Western Blot (WB)Protein detection and quantification1:500-1:2000Detects bands at ~21 kDa
Immunohistochemistry (IHC)Tissue localization studies1:50-1:200Available for paraffin (IHC-p) and frozen (IHC-fr) sections
Immunofluorescence (IF)Subcellular localization1:100-1:500Useful for determining membrane vs. cytoplasmic distribution
Immunoprecipitation (IP)Protein complex isolation20 μg/mLValidated with rat KNRK cells
Flow Cytometry (FCM)Cell population analysis1:100-1:400Used for quantitative assessment
ELISAQuantitative protein detection1:1000-1:5000High-throughput screening applications

For optimal results, researchers should validate each antibody for their specific application and cell/tissue type, as reactivity can vary between human, mouse, and rat samples despite high sequence homology .

How do I select the appropriate KRAS antibody for my experimental needs?

Selecting the appropriate KRAS antibody requires consideration of multiple factors:

  • Specificity requirements: Determine whether you need an antibody that:

    • Recognizes all KRAS isoforms

    • Differentiates between specific KRAS mutations (e.g., G12V)

    • Distinguishes KRAS from other RAS family members (HRAS, NRAS)

  • Application compatibility: Verify the antibody has been validated for your specific application. For example, the mouse monoclonal anti-KRAS antibody (Thermo Fisher Scientific cat# 415700) has confirmed reactivity in western blots with human HeLa and WI-38 cell lysates, rat KNRK and mouse NIH 3T3 cell lysates, and identifies KRAS at approximately 21 kDa .

  • Species reactivity: Confirm cross-reactivity with your experimental model. Some antibodies, like the one described in result , show 100% homology with mouse KRAS, 95% with rat, and 94% with bovine, making them suitable for comparative studies across species .

  • Antibody format: Consider whether you need a conjugated antibody (e.g., fluorophore-labeled for direct detection) or an unconjugated primary antibody depending on your detection system.

  • Validation evidence: Review published literature that has successfully used the antibody in similar experimental contexts.

For cellular localization studies of KRAS, researchers have successfully employed rabbit polyclonal anti-KRAS antibody (Thermo Fisher Scientific, cat# PA5-27234) at 20 μg/mL concentration , demonstrating its utility in distinguishing membrane-localized KRAS in mutant tumors from cytoplasmic KRAS in normal tissue.

What are the common challenges in KRAS antibody-based western blotting and how can they be addressed?

Western blotting with KRAS antibodies presents several technical challenges that can be addressed through methodological refinements:

ChallengeCauseSolution
Poor signalLow KRAS expressionIncrease protein loading (50-80 μg); use enhanced chemiluminescence detection
Multiple bandsNon-specific bindingIncrease blocking time; optimize antibody concentration; use monoclonal antibodies
High backgroundInadequate washingExtend washing steps with PBST; add 0.1% Tween-20 to washing buffer
Inconsistent loadingVariable expressionUse appropriate loading controls (β-actin, GAPDH); normalize to total protein
Membrane vs. cytosolic fraction detectionKRAS localization variancePerform subcellular fractionation before western blotting

When detecting KRAS proteins, it's essential to use the appropriate lysis buffer that can effectively extract membrane-associated KRAS. For KRAS p.Gly12Val mutants, which predominantly localize to the plasma membrane, researchers have observed a distinct "net-like pattern" compared to the diffuse cytoplasmic distribution in wild-type cells . This localization difference may affect protein extraction efficiency and should be considered when optimizing lysis conditions.

How can I verify the specificity of a KRAS antibody against different RAS isoforms?

Verifying KRAS antibody specificity against different RAS isoforms requires a systematic approach:

  • Sequence analysis and epitope mapping:

    • Compare the sequence homology between KRAS, HRAS, and NRAS at the antibody's epitope region

    • Focus on the hypervariable regions which differ most between RAS isoforms

  • Expression system validation:

    • Use recombinant expression systems with tagged versions of each RAS isoform

    • Perform western blot analysis to detect cross-reactivity

    • Quantify relative binding affinity to each isoform

  • Knockout/knockdown controls:

    • Use CRISPR-Cas9 or siRNA to create KRAS-specific knockouts/knockdowns

    • Confirm absence of signal in these models compared to controls

    • Test for residual signal that might indicate cross-reactivity with other isoforms

  • Competitive binding assays:

    • Pre-incubate antibody with purified KRAS, HRAS, or NRAS proteins

    • Assess reduction in signal intensity as a measure of specificity

  • Mass spectrometry validation:

    • Perform immunoprecipitation using the KRAS antibody

    • Analyze the precipitated proteins by mass spectrometry

    • Identify any co-precipitated RAS family members or isoforms

Notably, certain antibodies like the rabbit monoclonal anti-Ras (Cell Signaling Technology cat# E4K9L) may detect multiple RAS family members, while others like mouse monoclonal anti-KRAS (Thermo Fisher Scientific cat# 415700) are more KRAS-specific . Researchers should carefully review the manufacturer's validation data and perform their own validation when isoform specificity is critical.

What methodologies exist for studying KRAS antibody internalization in cancer cells?

Research has demonstrated that anti-KRAS antibodies can be internalized into cancer cells and affect KRAS function. The following methodologies have been employed to study this phenomenon:

  • Ex vivo patient-derived culture systems:

    • Establish transient matched mucosa-tumor primary cultures (viable for approximately 3 days)

    • This minimizes culture adaptation and maintains original biological characteristics

    • The 2-dimensional nature allows clear visualization of antibody internalization via confocal microscopy

  • Antibody internalization protocol:

    • Prepare antibodies at 20 μg/ml in antibody incubation buffer (DMEM + 1% FBS + 25 mM HEPES + 2X Pen-Strep)

    • Place cells on coverslips, wash thoroughly to remove debris

    • Add 80 μl of diluted antibody to culture plates, then place coverslip with cells facing downward

    • Incubate at 33°C, 10% CO2 for 16 hours

  • Colocalization studies:

    • After internalization, fix cells with 4% paraformaldehyde

    • Permeabilize with 0.1% Triton-X-100

    • Use markers like anti-EEA1 antibody to determine if internalized antibodies are in endosomes

    • Analyze using confocal microscopy with appropriate controls

  • Functional assessment:

    • Evaluate changes in KRAS localization (membrane to cytoplasm)

    • Assess impact on downstream effectors (e.g., SOX9 expression levels)

    • Compare effects between wild-type and mutant KRAS-expressing cells

This approach has shown that anti-KRAS antibodies can enter tumor cells, specifically aggregate KRAS in the cytoplasm, and hinder its translocation to the inner plasma membrane where it activates downstream effectors . This methodology provides insights into potential therapeutic strategies for targeting intracellular KRAS.

How can KRAS antibodies be used to distinguish between wild-type and mutant KRAS protein localization?

KRAS antibodies can effectively distinguish between wild-type and mutant KRAS protein localization through carefully designed immunofluorescence and imaging techniques:

  • Subcellular localization mapping:

    • Research has shown distinct localization patterns: KRAS wild-type predominantly localizes to the cytoplasm, while KRAS mutations (e.g., p.Gly12Val) show concentrated localization at the inner plasma membrane in a distinctive "net-like pattern"

    • This differential localization can be used as a visual biomarker for mutation status

  • Dual immunofluorescence methodology:

    • Fix cells with 4% paraformaldehyde in PBS with Ca2+/Mg2+ (PBSCM)

    • Wash cells with PBSCM (5 times, 5 minutes each)

    • Permeabilize with 0.1% Triton-X-100 for 30 minutes

    • Block with 5% goat serum, 5% FBS, 3% BSA in PBSCM for 1 hour

    • Incubate with primary anti-KRAS antibody (20 μg/mL) at 4°C overnight

    • Visualize using appropriate secondary antibodies and confocal microscopy

  • Quantitative analysis techniques:

    • Measure membrane-to-cytoplasm fluorescence intensity ratios

    • Perform line-scan analysis across cell membranes

    • Use computational image analysis to quantify the "net-like pattern" characteristic of mutant KRAS

  • Correlation with functional outcomes:

    • Link localization patterns to downstream signaling activation

    • For example, correlate membrane localization with increased SOX9 expression, which has been shown to be KRAS-mutation-dependent

Using these approaches in ex vivo patient-derived matched mucosa-tumor primary cultures allows researchers to maintain the biological characteristics of the original tumor while minimizing culture adaptation artifacts that might affect KRAS localization patterns .

What are the emerging therapeutic strategies involving KRAS antibodies for cancer treatment?

Several innovative therapeutic strategies involving KRAS antibodies are being developed for cancer treatment:

  • Intracellular antibody delivery systems:

    • Research has demonstrated that anti-KRAS antibodies can enter live mucosa-tumor cells and specifically aggregate KRAS in the cytoplasm

    • This hinders KRAS translocation to the inner plasma membrane, reducing its ability to activate downstream effectors

    • With efficient intracellular antibody delivery systems, this approach could be developed as combinatorial therapeutics for KRAS-driven cancers

  • mRNA vaccine approaches:

    • Moderna has developed mRNA-5671, an mRNA vaccine enclosed in lipid nanoparticles

    • The mRNA codes for peptides containing the most common KRAS mutations

    • This approach aims to prompt the immune system to produce antibodies against mutant KRAS proteins

    • The vaccine is already in clinical trials

  • Peptide-based anti-cancer vaccines:

    • Targovax has created TG01 and TG02 vaccines containing collections of mutated KRAS peptides

    • These are designed to be injected with QS-21, a compound that boosts immune reactions

    • The goal is to induce the body to develop antibodies that destroy mutant KRAS proteins

  • Combination approaches:

    • KRAS antibody therapies may be more effective when combined with other treatment modalities

    • Potential combinations include small molecule inhibitors of downstream pathways, immune checkpoint inhibitors, or conventional chemotherapy

    • These approaches have shown reduced toxicity compared to small-molecule therapies alone

These strategies represent a significant shift from traditional approaches that have struggled to effectively target KRAS, long considered "undruggable" due to its structure . Antibody-based approaches offer new hope for targeting KRAS-driven cancers, which include some of the deadliest forms such as pancreatic (77% KRAS-mutated) and colorectal cancer (43% KRAS-mutated) .

How can KRAS antibodies be optimized for detecting specific KRAS mutations in clinical samples?

Optimizing KRAS antibodies for mutation-specific detection in clinical samples involves several technical considerations:

  • Epitope-specific antibody development:

    • Generate antibodies targeting specific mutation sites (e.g., G12V, G12D, G13D)

    • Use synthetic peptides containing the mutated sequence as immunogens

    • Screen hybridomas or phage display libraries for highly specific binders

    • Validate specificity against wild-type and various mutant KRAS proteins

  • Differential binding protocols:

    • Optimize antibody concentration and incubation conditions specific to each mutation

    • Develop specific blocking strategies to minimize background in clinical samples

    • Consider dual-antibody approaches using one pan-KRAS antibody and one mutation-specific antibody

  • Validation in patient-derived models:

    • Test antibodies on ex vivo patient-derived matched mucosa-tumor primary cultures

    • Correlate antibody staining patterns with genomic mutation status

    • Establish specific staining patterns for different mutations

  • Quantitative assessment techniques:

    • Develop standardized scoring systems for mutation-specific staining

    • Implement digital pathology approaches for objective quantification

    • Consider multiplex staining with downstream effector markers to confirm functional relevance

  • Addressing tumor heterogeneity:

    • Account for inter-tumor and intra-tumor heterogeneity in KRAS mutation status

    • Develop sampling strategies to ensure representative testing

    • Consider using multiple antibodies targeting different epitopes

The availability of samples that are simultaneously cultured and archived enables systematic experimentation that could potentially address inter-tumor heterogeneity and genetic variability of disease subgroups, ultimately contributing to improved therapeutic approaches .

What are the optimal conditions for using KRAS antibodies in immunofluorescence studies?

Optimizing KRAS antibodies for immunofluorescence requires careful attention to fixation, permeabilization, and staining protocols:

Protocol StepRecommended ConditionsRationale
Fixation4% paraformaldehyde in PBSCM for 15-20 minutes at room temperaturePreserves KRAS membrane localization while maintaining epitope accessibility
WashingPBSCM (5 times, 5 minutes each)Thoroughly removes fixative to reduce background
Permeabilization0.1% Triton-X-100 in PBSCM for 30 minutes at room temperatureAllows antibody access to intracellular KRAS while preserving membrane structures
Blocking5% goat serum, 5% FBS, 3% BSA in PBSCM for 1 hour at room temperatureComprehensive blocking reduces non-specific binding
Primary antibodyAnti-KRAS antibody at 20 μg/mL in blocking buffer, overnight at 4°COptimal concentration for specific binding determined experimentally
Secondary antibodySpecies-appropriate fluorophore-conjugated antibody, 1:500 dilution, 1 hour at room temperatureProvides sensitive detection while minimizing background
Nuclear counterstainDAPI (1 μg/mL) for 5 minutesProvides cellular context for KRAS localization

When studying KRAS localization, it's important to note that wild-type KRAS predominantly localizes to the cytoplasm, while mutant KRAS (e.g., p.Gly12Val) shows a distinctive "net-like pattern" at the inner plasma membrane . Proper optimization of the above conditions is critical for reliably visualizing these different localization patterns.

For co-localization studies, such as determining whether internalized anti-KRAS antibodies are in endosomes, additional markers like anti-EEA1 antibody (e.g., Abcam ab70521, 1:200 dilution) can be incorporated into the protocol .

How can I optimize KRAS antibody-based immunoprecipitation protocols?

Optimizing KRAS antibody-based immunoprecipitation (IP) requires careful consideration of several factors:

  • Lysis buffer optimization:

    • Use buffers that effectively solubilize membrane-associated KRAS

    • Recommended formulation: 50 mM Tris-HCl pH 7.5, 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, with protease and phosphatase inhibitors

    • For membrane-localized mutant KRAS (e.g., G12V), consider adding 0.1% SDS to improve extraction efficiency

  • Antibody selection and preparation:

    • Use antibodies validated for IP, such as rabbit monoclonal anti-Ras (Cell Signaling Technology cat# E4K9L) or mouse monoclonal anti-KRAS (Thermo Fisher Scientific cat# 415700)

    • For optimal results, use dialyzed antibodies without preservatives or antibodies in PBS without preservatives

    • Pre-clear lysates with protein A/G beads to reduce non-specific binding

  • Immunoprecipitation protocol:

    • Use 2-5 μg antibody per 500 μg total protein

    • Incubate antibody with lysate overnight at 4°C with gentle rotation

    • Add pre-equilibrated protein A/G beads and incubate for 2-4 hours

    • Perform stringent washing (at least 5 washes) with decreasing salt concentrations

    • Elute using SDS sample buffer at 95°C for 5 minutes

  • Validation strategies:

    • Include isotype control antibodies to assess non-specific binding

    • Perform reverse IP using antibodies against known KRAS-interacting proteins

    • Confirm successful IP by western blotting for KRAS (21 kDa band)

    • Consider mass spectrometry analysis to identify co-precipitated proteins

  • Troubleshooting common issues:

    • Poor KRAS recovery: Increase antibody amount or lysate concentration

    • High background: Increase washing stringency or pre-clear lysates more thoroughly

    • Multiple bands: Optimize lysis conditions or use more specific antibodies

These protocols have been validated with cell lines such as rat KNRK cells , but may require further optimization for patient-derived samples or specific experimental conditions.

What considerations are important when using KRAS antibodies for quantitative analysis of KRAS expression levels?

Quantitative analysis of KRAS expression using antibodies requires attention to several methodological details:

  • Selection of quantitative techniques:

TechniqueAdvantagesLimitationsBest Practices
Western blot quantificationDistinguishes protein size, detects post-translational modificationsSemi-quantitative, requires careful normalizationUse standard curves with recombinant KRAS; normalize to multiple housekeeping proteins
ELISAHigh throughput, more quantitative than western blotMay not distinguish isoforms without specific antibodiesDevelop standard curves with recombinant KRAS of known concentration
Flow cytometrySingle-cell resolution, can analyze heterogeneous populationsRequires cell permeabilization for intracellular KRASUse isotype controls and fluorescence minus one (FMO) controls
Quantitative immunofluorescenceProvides spatial information with quantificationRequires careful image acquisition and analysisUse consistent exposure settings; analyze multiple fields per sample
  • Normalization strategies:

    • For western blots: normalize to total protein (measured by stain-free technology or Ponceau S staining) rather than single housekeeping proteins

    • For cellular assays: normalize to cell number or DNA content

    • Consider using spike-in standards of known KRAS concentration

  • Addressing biological variables:

    • Account for subcellular localization differences between wild-type and mutant KRAS

    • Wild-type KRAS predominantly localizes to the cytoplasm while mutant KRAS (e.g., G12V) shows membrane localization

    • These localization differences may affect extraction efficiency and quantification

  • Technical considerations:

    • Antibody concentration must be within the linear detection range

    • For western blots, use serial dilutions of lysates to ensure quantification in the linear range

    • Account for potential differences in antibody affinity between wild-type and mutant KRAS

    • Consider using recombinant KRAS proteins (wild-type and mutants) as quantification standards

  • Validation approaches:

    • Correlate protein quantification with mRNA levels (qPCR or RNA-seq)

    • Use multiple antibodies targeting different KRAS epitopes

    • Validate findings using genetic manipulation (overexpression or knockdown)

By implementing these methodological considerations, researchers can achieve more reliable quantitative analysis of KRAS expression levels in both research and clinical settings.

How can KRAS antibodies be used to study KRAS-dependent signaling pathways?

KRAS antibodies enable sophisticated analysis of KRAS-dependent signaling networks:

  • Proximity ligation assays (PLA):

    • Use KRAS antibodies in combination with antibodies against potential interacting partners

    • PLA generates fluorescent signals only when proteins are in close proximity (<40 nm)

    • This approach can map KRAS interaction networks in different cellular contexts

    • Particularly useful for studying how mutations affect KRAS protein-protein interactions

  • Chromatin immunoprecipitation (ChIP) studies:

    • Combine KRAS antibodies with ChIP to identify genomic regions where KRAS-regulated transcription factors bind

    • For example, study how KRAS affects ZNF304-dependent transcriptional silencing of tumor suppressor genes in colorectal cancer cells

    • Correlate these findings with expression data to build comprehensive regulatory networks

  • Phosphoproteomic analysis:

    • Use KRAS antibodies to immunoprecipitate KRAS protein complexes

    • Perform phosphoproteomic analysis to identify phosphorylation changes in KRAS-interacting proteins

    • Compare phosphorylation patterns between wild-type and mutant KRAS-expressing cells

    • This allows mapping of KRAS-dependent phosphorylation networks

  • Pathway inhibitor studies:

    • Use KRAS antibodies to monitor pathway activation before and after treatment with inhibitors

    • Track changes in KRAS localization and downstream effector activation

    • For example, evaluate how anti-KRAS antibody treatment affects SOX9 expression, which has been shown to be KRAS-mutation-dependent

  • Live-cell imaging approaches:

    • Develop cell-permeable fluorescently labeled KRAS antibody fragments or mimetics

    • Use these to track KRAS localization and dynamics in living cells

    • Monitor real-time changes in response to stimuli or drug treatments

These approaches can provide insights into the differential signaling properties of wild-type versus mutant KRAS, potentially revealing new therapeutic vulnerabilities in KRAS-driven cancers.

What are the current limitations of KRAS antibodies and how might these be overcome in future research?

Current KRAS antibody limitations and potential future solutions include:

LimitationCurrent ImpactPotential Solutions
Limited mutation specificityDifficulty distinguishing between KRAS variants (G12D, G12V, etc.)Development of highly specific recombinant antibodies or aptamers targeting specific mutations
Intracellular delivery challengesReduced therapeutic potential due to poor cellular penetrationEngineering cell-penetrating antibodies or antibody fragments; development of novel delivery systems like lipid nanoparticles
Cross-reactivity with other RAS isoformsReduced specificity in experimental and clinical applicationsUse of computational design and structural biology to identify truly KRAS-specific epitopes
Inability to target "undruggable" binding sitesLimited ability to block critical protein-protein interactionsDevelopment of intrabodies or nanobodies that can access cryptic binding sites
Lot-to-lot variabilityReproducibility issues in research and clinical applicationsTransition to recombinant antibody technology with defined sequences and production methods

Promising emerging approaches include:

  • Antibody engineering technologies:

    • Development of bispecific antibodies that simultaneously target KRAS and delivery molecules

    • Creation of antibody-drug conjugates targeting cells with high KRAS expression

    • Engineering of smaller antibody formats (nanobodies, single-chain antibodies) with improved tissue penetration

  • Combination therapeutic strategies:

    • Integration of KRAS antibodies with mRNA vaccines like Moderna's mRNA-5671

    • Combining antibody approaches with peptide-based vaccines such as Targovax's TG01 and TG02

    • Development of multi-modal approaches combining antibodies with small molecule inhibitors

  • Advanced delivery systems:

    • Utilization of properties from naturally cell-penetrating autoantibodies

    • Development of lipid nanoparticle formulations similar to those used in mRNA vaccines

    • Engineering of extracellular vesicle-based delivery systems

  • Novel screening platforms:

    • Implementation of high-throughput antibody discovery platforms targeting specific KRAS mutations

    • Use of structural biology and computational design to develop antibodies against previously inaccessible epitopes

These advancements could transform KRAS antibodies from primarily research tools into effective therapeutic agents for KRAS-driven cancers, which have historically been difficult to treat .

How can KRAS antibodies contribute to personalized medicine approaches in cancer treatment?

KRAS antibodies are poised to make significant contributions to personalized cancer medicine through several avenues:

  • Precision diagnostics:

    • Development of KRAS mutation-specific antibodies could enable rapid immunohistochemical screening of tumor samples

    • This approach could complement or potentially replace more expensive and time-consuming genomic testing in some contexts

    • The distinct localization patterns of wild-type versus mutant KRAS (cytoplasmic versus membrane-associated) could serve as visual biomarkers

  • Patient stratification methodologies:

    • Using anti-KRAS antibodies to characterize patient tumors based on:

      • KRAS mutation status

      • KRAS protein expression levels

      • KRAS subcellular localization patterns

      • KRAS-dependent signaling pathway activation

    • This could identify patient subgroups most likely to benefit from specific treatment approaches

  • Ex vivo drug sensitivity testing:

    • Utilizing patient-derived ex vivo culture systems to assess responsiveness to KRAS-targeted therapies

    • This approach minimizes culture adaptation, maintaining original biological characteristics of tumors

    • The transient nature of these cultures (viable for approximately 3 days) allows testing in a clinically relevant timeframe

  • Monitoring treatment response:

    • Developing liquid biopsy approaches using anti-KRAS antibodies to detect circulating tumor cells or exosomes

    • Tracking changes in KRAS expression or localization in sequential biopsies during treatment

    • Correlating these changes with clinical outcomes to guide treatment adjustments

  • Individualized immunotherapy approaches:

    • Using patient-specific KRAS mutation information to develop personalized vaccines

    • Combining with immune checkpoint inhibitors in patients with specific KRAS mutation profiles

    • This approach could be particularly valuable for microsatellite-stable colorectal cancers, which are typically refractory to immune checkpoint blockade therapy alone

The integration of these approaches could significantly improve outcomes for patients with KRAS-driven cancers, which include some of the most aggressive and treatment-resistant malignancies, such as pancreatic cancer (77% KRAS-mutated) and colorectal cancer (43% KRAS-mutated) .

What emerging research directions show the most promise for KRAS antibody applications?

KRAS antibody research is advancing rapidly, with several promising directions emerging at the intersection of basic science and translational medicine:

  • Therapeutic antibody internalization strategies:

    • Building on recent discoveries that anti-KRAS antibodies can enter live tumor cells and alter KRAS function

    • Developing efficient intracellular antibody delivery systems to target previously "undruggable" KRAS

    • This approach shows potential for KRAS-driven cancers that remain difficult to treat with conventional therapies

  • Mutation-specific targeting approaches:

    • Creating highly specific antibodies against common KRAS mutations (G12D, G12V, G13D)

    • Combining these with nucleic acid-based therapeutics for synergistic effects

    • Leveraging the distinct localization patterns of mutant KRAS for selective targeting

  • Integration with immunotherapy platforms:

    • Advancing mRNA vaccine approaches like Moderna's mRNA-5671

    • Further developing peptide-based anti-cancer vaccines such as Targovax's TG01 and TG02

    • These approaches aim to prompt the immune system to develop antibodies against mutant KRAS proteins

  • Patient-derived model systems:

    • Expanding the use of ex vivo patient-derived matched mucosa-tumor primary cultures

    • These minimize culture adaptation while maintaining original biological characteristics

    • Allowing direct testing of antibody effects on patient-specific tumors

  • Advanced imaging technologies:

    • Developing methods to visualize KRAS dynamics in living cells and tissues

    • Creating antibody-based biosensors for KRAS activation state

    • Implementing high-resolution imaging to understand KRAS membrane organization

Product Science Overview

Introduction

The Kirsten Rat Sarcoma Viral Oncogene (KRAS) is a gene that encodes a protein called K-Ras, which is a member of the Ras family of GTPases. This gene plays a crucial role in cell signaling pathways that regulate cell growth, differentiation, and survival. KRAS is of significant interest in cancer research due to its frequent mutations in various human cancers.

Discovery and Nomenclature

KRAS was first identified as an oncogene in the Kirsten Rat Sarcoma virus, which is how it got its name . The oncogene was derived from a cellular genome, and when found in a cellular genome, it is referred to as a proto-oncogene . The KRAS gene is part of the mammalian RAS gene family, which also includes HRAS and NRAS .

Structure and Function

The KRAS gene provides instructions for making the K-Ras protein, which is involved in the RAS/MAPK signaling pathway . This protein acts as a molecular switch, cycling between an active GTP-bound state and an inactive GDP-bound state . When bound to GTP, K-Ras transmits signals that promote cell proliferation and survival. The protein is inactivated when it hydrolyzes GTP to GDP .

There are two protein products of the KRAS gene in mammalian cells, resulting from the use of alternative exon 4: K-Ras4A and K-Ras4B . These proteins have different structures in their C-terminal regions and use different mechanisms to localize to cellular membranes, including the plasma membrane .

Role in Cancer

Mutations in the KRAS gene are common in various cancers, including colorectal, lung, and pancreatic cancers . These mutations often result in a constitutively active K-Ras protein that continuously signals for cell growth and division, leading to uncontrolled cell proliferation and tumor formation . KRAS mutations are present in approximately 40% of colorectal cancers and are associated with resistance to certain therapies .

Therapeutic Approaches

Given the critical role of KRAS in cancer, it has been a target for therapeutic intervention. Various strategies have been explored, including the development of small molecules that inhibit the function of mutant K-Ras proteins and the use of immunotherapies that target KRAS mutations . For example, multi-peptide cancer vaccines targeting KRAS mutants have shown promise in inducing cancer-specific anti-tumor effects .

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 2024 Thebiotek. All Rights Reserved.