PBK’s dysregulation is implicated in cancer progression and immune evasion:
High PBK expression correlates with poor survival outcomes in lung adenocarcinoma (LUAD), with a hazard ratio (HR) of 0.47 for disease-specific survival . In pan-cancer analyses, PBK is upregulated in 33 tumor types, including breast, colorectal, and lung cancers .
PBK’s overexpression is associated with immune evasion, as shown by negative correlations with immune infiltration and positive associations with immune checkpoint genes (e.g., PD-L1) .
PBK’s role in mitosis and TP53 destabilization suggests its involvement in bypassing DNA damage checkpoints. During mitosis, phosphorylated PBK binds TP53, reducing G2/M checkpoint efficacy, thereby promoting genomic instability .
PBK expression predicts sensitivity to MEK inhibitors (e.g., trametinib, selumetinib) in preclinical models. High PBK levels correlate with lower IC50 values for these drugs, indicating potential therapeutic targeting .
Drug Class | PBK Correlation | Mechanism |
---|---|---|
MEK Inhibitors | Positive | Enhanced sensitivity in PBK-high tumors |
p38 MAPK Pathway | Phosphorylation | PBK activates p38, modulating lymphoid responses |
Pan-cancer analysis reveals PBK’s differential expression across tissues:
Tissue | PBK Expression (Tumor vs. Normal) | Significance |
---|---|---|
Lung (LUAD) | Upregulated | Poor prognosis, immune evasion |
Breast | Elevated | Correlates with HER2+ subtypes |
Colorectal | High | Linked to MSI-high status |
PBK expression inversely correlates with immune cell infiltration (e.g., CD8+ T cells) and is positively associated with immune suppressive markers (e.g., PD-L1) .
Lymphokine-activated killer T-cell-originated protein kinase (PBK), also known as MAP kinase-interacting serine/threonine kinase 2 (MKNK2), plays a crucial role in cellular processes like cell growth, differentiation, and survival. Primarily found in the placenta and testis, PBK participates in activating lymphoid cells and testicular functions, particularly spermatogenesis. Its activity is closely linked to mitosis, where phosphorylation is essential for its catalytic function. Upon phosphorylation, PBK interacts with the tumor suppressor protein TP53, affecting its stability and potentially influencing the G2/M cell cycle checkpoint, especially during DNA damage responses. The protein's activity is restricted to the mitotic phase. Additionally, PBK interacts with the Dlg protein, a tumor suppressor, through its PDZ domain and PBK's T/SXV motif. Further expanding its role, PBK phosphorylates MAP kinase p38, suggesting its involvement in lymphoid cell activation.
PBK modeling is a computational approach that uses mathematical equations to simulate the absorption, distribution, metabolism, and excretion (ADME) of compounds in humans. These models integrate physiological parameters with chemical-specific data to predict internal exposure levels and potential toxic effects.
In human safety assessment, PBK modeling serves as a crucial component of Next Generation Risk Assessment (NGRA) frameworks. When Threshold of Toxicological Concern (TTC) approaches are insufficient to ensure safety, PBK models offer a more refined estimation of systemic exposure. They translate external exposure (applied dose) to internal exposure metrics such as maximum blood concentration (Cmax) or area under the curve (AUC), which can then be compared to in vitro points of departure to derive bioactivity exposure ratios (BERs) .
The implementation of PBK modeling in safety assessment typically follows a tiered approach:
Level 0: Characterization of exposure scenarios and collection of existing data
Level 1: Predictions using in silico parameterization only
Level 2: PBK modeling based on in vitro parameterization
Level 3: Generation of human pharmacokinetic data for validation and calibration
This structured approach allows risk assessors to employ tools of appropriate complexity for decision-making, progressing to more sophisticated methods only when necessary.
PBK models require three categories of input data for accurate parameterization:
Physiological parameters:
Organ volumes
Blood flow rates
Tissue composition (water, lipid, protein content)
Demographic factors (age, sex, weight)
Anatomical and physiological data:
Cardiac output
Ventilation rates (for inhalation exposure)
Gastrointestinal transit times (for oral exposure)
Skin properties (for dermal exposure)
Chemical-specific parameters:
These parameters can be obtained through various approaches including in vitro experimental measurements, in silico prediction methods, and literature values. The selection of appropriate parameterization methods significantly influences model predictive performance, as demonstrated in studies comparing different approaches for calculating tissue:plasma partition coefficients and fraction unbound in plasma .
The accuracy of PBK models depends largely on the quality of input parameters and the structural appropriateness of the model for the specific compound being studied. Recent comprehensive evaluations provide insights into typical performance metrics:
A systematic study evaluated 38,772 Cmax predictions for 44 compounds using different combinations of in vitro and in silico parameterization approaches. The best performing model configuration achieved:
19 out of 44 compounds (43%) predicted within 2-fold of observed Cmax
34 out of 44 compounds (77%) predicted within 5-fold of observed Cmax
10 compounds (23%) were overestimated by more than 5-fold
Best results were achieved when the hepatic clearance was parameterized based on in vitro (i.e., hepatocytes or liver S9) measured intrinsic clearance values, combined with the method of Rodgers and Rowland for calculating tissue:plasma partition coefficients, and the method of Lobell and Sivarajah for calculating the fraction unbound in plasma .
These findings indicate that while PBK models can provide reasonably accurate predictions for the majority of compounds, there remains a significant subset for which predictions deviate substantially from observed values.
Several methodological approaches can be employed to optimize PBK model performance:
Systematic parameter selection:
Use experimental data for the most sensitive parameters
Apply appropriate in silico methods for physicochemical properties
Incorporate uncertainty analysis for less influential parameters
Tiered implementation strategy:
Validation approaches:
Compare predictions against in vivo human data when available
Use cross-chemical validation within similar chemical spaces
Evaluate consistency across multiple parameterization methods
Structural considerations:
The framework shown in the search results emphasizes an iterative approach where sensitivity analysis identifies the most influential parameters, which then receive the most rigorous experimental or computational attention .
Sensitivity analysis provides a systematic approach to quantify how variation in model inputs affects the outputs, offering crucial insights for model refinement. The methodology involves:
Local sensitivity analysis:
Vary one parameter at a time by a small percentage (e.g., ±10%)
Calculate sensitivity coefficients (ratio of % change in output to % change in input)
Rank parameters by their influence on model predictions
Identify parameters requiring more accurate determination
Global sensitivity analysis:
According to the framework described in the search results, sensitivity analysis is a critical step after initial parameterization to determine the influential parameters that require more accurate determination through in vitro experiments .
This approach ensures that the most critical parameters are characterized with appropriate precision, while less influential parameters can be estimated using simpler methods, optimizing both model performance and resource utilization.
Hepatic clearance is often the most influential parameter in PBK models, and its accurate determination is critical for reliable predictions. Based on systematic comparisons, the following methodological approaches show superior performance:
In vitro measurement systems:
Extrapolation methods:
Well-stirred model for scaling in vitro intrinsic clearance to whole liver clearance
Appropriate correction for binding in the in vitro system and in plasma
Scaling factors based on enzyme content per gram of liver and liver weight
Alternative systems for challenging compounds:
Long-term hepatocyte cultures for slowly metabolized compounds
Hepatocyte co-cultures maintaining metabolic capacity
Microfluidic liver-on-chip systems for improved physiological relevance
The search results specifically indicate that models using in vitro measured intrinsic clearance values from hepatocytes or liver S9 fractions demonstrated superior predictive performance compared to those relying solely on in silico predictions .
Tissue:plasma partition coefficients (Kp values) determine compound distribution in the body and significantly impact PBK model predictions. Based on systematic evaluations, the following methodological approaches provide optimal accuracy:
Selection of calculation methods based on compound properties:
Compound Properties | Recommended Method | Rationale |
---|---|---|
Basic lipophilic compounds | Rodgers and Rowland | Accounts for pH gradients and tissue binding |
Neutral lipophilic compounds | Poulin and Theil (modified) | Better performance for neutral compounds |
Zwitterions | Schmitt | Specifically developed for compounds with multiple charged groups |
Large biologics | Minimal PBPK approaches | Limited tissue distribution requires different approach |
Required input parameters for calculation:
Compound-specific: Lipophilicity (logP), pKa values, fraction unbound in plasma
Tissue-specific: Water content, neutral lipid content, phospholipid content, pH
According to the search results, the method of Rodgers and Rowland for calculating tissue:plasma partition coefficients contributed to more accurate Cmax predictions when combined with appropriate clearance and plasma protein binding parameterization .
Validating PBK models without human in vivo data represents a significant challenge but is increasingly necessary in the shift toward non-animal approaches. Several methodological strategies have emerged:
Cross-chemical validation:
Select a training set of compounds with known human in vivo data
Establish the predictive performance of the model for these compounds
Identify chemical space boundaries where the model performs well
Apply the model only to new compounds within this chemical space
In vitro to in vitro extrapolation:
Conduct in vitro experiments simulating key ADME processes
Compare PBK model predictions to these simplified systems
Validate components of the model individually
Weight-of-evidence approach:
These approaches align with the framework described in the search results, where progression to higher levels of complexity (including human PK data generation) occurs only when necessary for decision-making .
PDZ Binding Kinase (PBK), also known as T-lymphokine-activated killer cell-originated protein kinase (TOPK), is a serine-threonine kinase that belongs to the mitogen-activated protein kinase kinase (MAPKK) family. In normal human cells, PBK plays several critical roles:
Cell cycle regulation:
Phosphorylates mitotic proteins during the mitotic phase
Participates in sister chromatid segregation
Contributes to mitotic checkpoint regulation
DNA damage response:
Involved in the DNA integrity checkpoint
Participates in DNA-dependent DNA replication processes
Responds to cellular stress through pathway activation
Normal tissue expression:
The search results indicate that PBK "is barely expressed in normal tissues" under physiological conditions, suggesting tight regulation of its expression and activation .
PBK expression shows marked differences between normal and cancer tissues, with complex regulatory mechanisms controlling its expression:
In normal tissues:
Tightly controlled expression, predominantly in testis and fetal tissues
Minimal to undetectable expression in most differentiated adult tissues
Transient expression during specific cellular processes (e.g., immune cell activation)
Regulation by cell cycle-dependent transcription factors
In cancer tissues:
Significantly upregulated in most cancer types compared to normal counterparts
Expression correlates with proliferation markers
Associated with tumor stage and progression
The search results specifically note that "PBK expression is relatively high in most cancers compared to their normal counterparts, and this gene is barely expressed in normal tissues."
Data supporting these observations were obtained through comprehensive analysis of RNA-seq data from TCGA and GTEx databases across 33 cancer types and corresponding normal tissues, providing a systematic comparison of expression patterns .
PBK expression shows distinct patterns across different tumor stages, providing insights into its role in cancer progression:
Stage-specific expression:
Early stages (I-II): PBK expression is already elevated compared to normal tissue
Intermediate stages (II-III): Further increased expression in many cancer types
Advanced stages (III-IV): Expression patterns become more complex
The search results specifically state: "PBK expression was correlated with the tumor stage in various cancers. Especially between stage I, II, and III tumors, PBK expression is significantly different."
Cancer-specific patterns:
The search results highlight this unusual pattern: "Interestingly, we found that the PBK expression in COAD and LUSC is downregulated in the advanced stage." This finding suggests cancer-specific roles for PBK at different stages of progression.
Clinical implications:
Early diagnostic potential: Elevated even in stage I tumors
Monitoring: Changes during treatment and progression
Prognostic: Stage-specific relationships with outcomes
Therapeutic: Targeting may be most effective at specific stages
These findings suggest that while PBK overexpression is generally associated with cancer progression, the relationship is not always linear and varies by cancer type.
PBK expression shows significant associations with patient outcomes across multiple cancer types, making it a potential prognostic biomarker:
These findings indicate that PBK expression has significant prognostic value across multiple cancer types, predominantly associated with poorer outcomes, with the potential to enhance current prognostic models in clinical practice.
PBK expression shows complex relationships with immune infiltration in the tumor microenvironment, with distinct patterns across cancer types and immune cell populations:
These findings suggest that PBK may influence tumor progression partly through modulation of the immune microenvironment, with implications for combined targeting strategies and immunotherapy response prediction.
PBK overexpression in cancer is associated with specific molecular pathways that contribute to tumor progression. GSEA analysis revealed several key pathways:
Cell cycle and mitosis pathways:
Sensory perception pathways:
Cancer-specific signaling pathways:
The search results emphasize that "GSEA analysis revealed that PBK participated in a wide range of functions and pathways relevant to the cell cycle and DNA replication... All these terms were enriched in the PBK high-expression side, which suggested that high PBK expression mainly involved these signaling pathways, participated in mitosis and the cell cycle, and may also function in promoting tumor cell proliferation."
These findings collectively suggest that PBK primarily functions in promoting cell cycle progression, DNA replication, and mitosis in cancer cells, consistent with its role as an oncogenic kinase driving proliferation across multiple cancer types.
PBK expression shows significant associations with genomic instability metrics including tumor mutation burden (TMB) and microsatellite instability (MSI) across multiple cancer types:
Association with Tumor Mutation Burden (TMB):
Association with Microsatellite Instability (MSI):
Potential biological mechanisms:
DNA damage response connection: PBK involvement in DNA integrity checkpoint
Cell cycle checkpoint regulation: Mitotic checkpoint functions
Genomic instability resulting from checkpoint dysregulation
These associations between PBK expression and genomic instability markers may have important implications for predicting response to immunotherapy, as high TMB and MSI are established biomarkers for checkpoint inhibitor efficacy in multiple cancer types.
PBK expression has emerging roles in drug response and resistance across different cancer types and drug classes:
Positive correlations with drug effectiveness:
Drug resistance associations:
Potential for therapeutic targeting:
Mechanistic contributions to resistance:
Cell cycle regulation effects: Altered checkpoint responses affecting drug sensitivity
DNA damage response modulation: Enhanced repair capacity after genotoxic stress
Anti-apoptotic signaling: Activation of survival pathways
These findings suggest that PBK modulation could be a valuable strategy for chemosensitization and overcoming resistance, particularly for specific drug classes like MEK inhibitors and platinum compounds in relevant cancer types.
PBK is a protein that plays a crucial role in various cellular processes. It is composed of several structural domains, including the PDZ domain, which is a common protein interaction module. PDZ domains typically recognize short amino acid motifs at the C-termini of target proteins . These interactions are essential for regulating multiple biological processes such as transport, ion channel signaling, and other signal transduction systems .
PBK is expressed predominantly in the testis, specifically in the outer cell layer of seminiferous tubules, as well as in the placenta . It is involved in the activation of lymphoid cells and supports testicular functions, playing a role in spermatogenesis .
PBK functions as a mitotic kinase that phosphorylates MAP kinase p38 and is active during mitosis . When phosphorylated, PBK interacts with the tumor suppressor protein p53 (TP53), leading to the destabilization of TP53 and attenuation of the G2/M checkpoint during doxorubicin-induced DNA damage . This interaction suggests that PBK may play a role in cell cycle regulation and response to DNA damage.
Recombinant human PBK is produced using baculovirus-insect cell expression systems . The recombinant protein is typically purified to a high degree of purity, making it suitable for research purposes. It is often used in studies to understand the kinase’s role in cellular processes and its potential as a therapeutic target.