PI3Kd Human

Phosphoinositide 3-kinase p110d/p85a Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to PI3Kδ Human

PI3Kδ (Phosphoinositide 3-Kinase delta) is a lipid kinase encoded by the PIK3CD gene in humans. It belongs to the Class IA family of PI3Ks and is distinguished by its predominant expression in leukocytes, where it regulates immune cell activation, survival, and migration . PI3Kδ catalyzes the phosphorylation of phosphatidylinositol-4,5-bisphosphate (PIP2) to phosphatidylinositol-3,4,5-trisphosphate (PIP3), a critical second messenger in intracellular signaling pathways . Dysregulation of PI3Kδ is linked to immune disorders, hematologic malignancies, and Activated PI3Kδ Syndrome (APDS), a rare inborn error of immunity .

2.1. Gene and Protein Architecture

  • Gene: PIK3CD (Chromosome 1) encodes the p110δ catalytic subunit .

  • Protein: p110δ forms a heterodimer with regulatory subunits (e.g., p85α, p55α) to stabilize the enzyme and recruit it to activated receptors .

Table 1: Human PI3K Class IA Catalytic Subunits

GeneProteinExpressionAssociated Regulatory Subunits
PIK3CDp110δLeukocytes, CNSp85α, p55α, p50α
PIK3CAp110αUbiquitousp85α, p55α, p50α
PIK3CBp110βUbiquitousp85α, p55α, p50α

2.2. Regulatory Domains

  • SH2 Domains: Present in regulatory subunits (e.g., p85α), enabling binding to phosphorylated tyrosine residues on receptors like CD28 or BCR .

  • Catalytic Domain: Binds ATP and mediates PIP2 phosphorylation .

3.1. Signaling Pathways

  • PIP3 Production: PI3Kδ-generated PIP3 recruits pleckstrin homology (PH) domain-containing proteins (e.g., AKT, PDK1) to the plasma membrane, activating downstream effectors like mTOR and FOXO1 .

  • Immune Cell Regulation:

    • B Cells: Mediates survival and class-switching via BCR signaling .

    • T Cells: Modulates TCR signaling and regulatory T cell (Treg) function .

    • Myeloid Cells: Controls cytokine production (e.g., IL-12, IL-23) .

3.2. Pathogenic Mutations

Gain-of-function mutations in PIK3CD (e.g., E1021K) or PIK3R1 (e.g., splice variants) cause APDS, characterized by lymphoproliferation, immunodeficiency, and autoimmunity .

Table 2: APDS-Associated Mutations

GeneMutationFunctional ImpactClinical Phenotype
PIK3CDE1021KHyperactivated PI3Kδ signalingRecurrent infections, lymphoma
PIK3R1Splice variantsLoss of p85α regulatory functionAutoimmunity, hypogammaglobulinemia

4.1. Role in Disease

  • Cancer: Overexpressed in B-cell malignancies (e.g., CLL, AML) and solid tumors .

  • Immune Dysregulation:

    • APDS patients exhibit reduced memory B cells, elevated IgM, and defective Treg function .

    • PI3Kδ inhibitors (e.g., idelalisib) show efficacy in leukemia but induce immune-related adverse events (e.g., colitis) .

4.2. Targeted Therapies

  • Leniolisib: A selective PI3Kδ inhibitor approved for APDS, normalizing PI3K pathway hyperactivity and improving lymphoproliferation .

  • Combination Strategies: Intermittent dosing of PI3Kδ inhibitors (e.g., AMG319) reduces toxicity while maintaining anti-tumor immunity in solid tumors .

5.1. Preclinical Insights

  • Mouse Models: Pik3cd knockout mice exhibit impaired immune responses and increased susceptibility to infections, mimicking human APDS .

  • Single-Cell Analysis: PI3Kδ inhibition in HNSCC patients reduces intratumoral Tregs and enhances CD8+ T cell cytotoxicity .

5.2. Translational Challenges

  • Toxicity Management: Chronic PI3Kδ inhibition depletes tissue-resident Tregs, triggering colitis and autoimmune sequelae .

  • Biomarker Development: PI3Kδ activity assays and PIK3CD variant profiling are critical for personalized therapy .

Product Specs

Description
The class IA PI3Kd is primarily found in endothelial cells and white blood cells. Studies have shown that inactivating PI3Kd in bone marrow mast cells (BBMCs) can lead to several defects, including reduced stem cell factor-mediated proliferation, adhesion and migration, and impaired allergen-IgE induced degranulation and cytokine release. Additionally, research indicates that PI3Kd plays a role in TNFa-induced signaling in neutrophils, as evidenced by reduced Akt phosphorylation and PDK1 activity when treated with the d-specific inhibitor IC87114. This product features a GST-Tag on the catalytic subunit, making it suitable for GST pull-down assays. The heterodimer was purified using affinity chromatography. The molecular weight of the p110d chain is 120.2 kDa (excluding the Tag), and the p85a chain is 83.5 kDa.
Physical Appearance
Sterile, filtered liquid.
Formulation
Solution of PI3Kd in a buffer containing 25mM Tris-HCl (pH 8.0), 50mM NaCl, 0.5mM MgCl2, and 50% glycerol.
Stability
For short-term storage (2-4 weeks), store at 4°C. For long-term storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE analysis.
Unit Definition
One unit of activity is defined as the amount of enzyme required to catalyze the transfer of one picomole of phosphate to PIP2 per minute.
Specific Activity
3,000 U/mg.
Synonyms
Phosphoinositide 3-kinase p110d/p85a, PI3Kd.
Source
Sf9 insect cells.

Q&A

What is PI3K delta and how does it differ from other PI3K isoforms?

PI3K delta (PI3Kδ) is one of four class I PI3K isoforms that plays a critical role in immune cell development and function. Unlike the more ubiquitously expressed PI3Kα, PI3Kδ expression is largely restricted to leukocytes, making it particularly important in immune system regulation. Structurally, PI3Kδ consists of a catalytic subunit (p110δ, encoded by PIK3CD) and a regulatory subunit (p85α, encoded by PIK3R1). The unique tissue distribution pattern of PI3Kδ makes it an attractive target for developing isoform-selective inhibitors to treat immune dysregulation without affecting other tissues substantially .

How does PI3K delta signaling regulate normal immune function?

PI3Kδ signaling is integral to the development and function of both B and T lymphocytes. In normal immune function, PI3Kδ activation must be carefully balanced to optimize immune responses against pathogens. The enzyme regulates multiple immune processes including:

  • B cell development, survival, and antibody production

  • T cell activation, proliferation, and cytokine production

  • Immune cell migration and trafficking

  • Integration of signals from various cytokine and antigen receptors

Mouse genetic models have demonstrated that proper "dosing" of PI3K signaling is critical, as both insufficient and excessive PI3Kδ activity impair immune function .

What genetic mechanisms lead to PI3K delta dysregulation in humans?

PI3Kδ dysregulation occurs through two primary genetic mechanisms:

  • Gain-of-function (GOF) mutations in PIK3CD (causing APDS1): These include variants such as E1021K, E525K, and N334K that enhance the basal activity and membrane binding of p110δ

  • Loss-of-function (LOF) mutations in PIK3R1 (causing APDS2): These mutations affect the regulatory subunit that normally constrains PI3K activity

Both mechanisms ultimately result in hyperactivation of PI3K signaling, despite arising from seemingly opposite functional changes in the respective genes .

What is the clinical spectrum of Activated PI3K Delta Syndrome?

APDS presents with a heterogeneous clinical picture characterized primarily by:

  • Immunodeficiency: Recurrent respiratory tract infections (particularly with Streptococcus pneumoniae), viral susceptibility (especially to herpesviruses like EBV and CMV), and bronchiectasis

  • Lymphoproliferation: Persistent enlargement of lymph nodes, tonsils, and spleen

  • Autoimmunity: Including autoimmune cytopenias, colitis, arthritis, and granulomatous skin lesions

  • Malignancy risk: Particularly B-cell lymphomas

  • Growth retardation: More common in APDS2 patients

  • Developmental delay: Observed in a subset of patients

Disease severity varies significantly, with some individuals remaining asymptomatic despite carrying pathogenic variants .

What is the epidemiological profile of APDS in human populations?

Survival analysis from a comprehensive literature review of 256 APDS patients (193 with APDS1 and 63 with APDS2) revealed decreased survival rates compared to earlier smaller studies:

  • Conditional survival rate at age 20: 87%

  • Conditional survival rate at age 30: 74%

  • Conditional survival rate at age 40 and 50: 68%

How do APDS1 and APDS2 compare in terms of clinical manifestations?

While APDS1 and APDS2 share many clinical features, some important differences exist:

FeatureAPDS1 (PIK3CD mutations)APDS2 (PIK3R1 mutations)
MechanismGain-of-function in catalytic subunitLoss-of-function in regulatory subunit
Growth impairmentLess commonMore frequently observed
B-cell dysfunctionPresent but variableMore pronounced defects in B-cell expansion and affinity maturation
Syndromic featuresGenerally absentMore common
Intracellular signalingSpecific pattern of activationDistinct signaling abnormalities
Clinical severityVariableVariable but potentially more severe

Both subtypes share susceptibility to infections, lymphoproliferation, and autoimmunity, but the specific mechanisms and degree of immune abnormalities can differ .

How does hyperactivation of PI3K delta disrupt normal lymphocyte development and function?

Hyperactivation of PI3Kδ profoundly affects lymphocyte homeostasis through several mechanisms:

In B cells:

  • Accelerated development with premature exit from the bone marrow

  • Increased rates of apoptosis leading to depletion of naïve B cells

  • Impaired class-switching to IgG antibodies, often resulting in a Hyper-IgM phenotype

  • Shift toward transitional B cells and plasmablasts

  • Preferential expansion of polyclonal and self-reactive B cells at the expense of antigen-specific clones

  • Defective response to T-cell independent vaccine stimuli

In T cells:

  • Decreased CD4+ T cells with normal or increased CD8+ T cells (inverted CD4+/CD8+ ratio)

  • Premature senescence of CD8+ T cells (evidenced by CD57+ expression and shortened telomeres)

  • T cell exhaustion (increased PD-1, CD160, and CD244 expression)

  • Depletion of naïve CD4+ and CD8+ T cell reservoirs

  • Impaired T follicular helper cell function affecting B cell responses

These abnormalities collectively contribute to both immunodeficiency and autoimmunity, as the balance between effective pathogen clearance and self-tolerance is disrupted .

What cellular mechanisms link PI3K delta hyperactivation to lymphoma development?

The increased risk of lymphoma in APDS patients likely stems from multiple PI3Kδ-mediated mechanisms:

  • Chronic antigenic stimulation: Persistent infections, particularly with oncogenic viruses like EBV, provide continuous B-cell stimulation

  • Impaired immune surveillance: Defective T-cell function compromises elimination of malignantly transformed cells

  • Enhanced cell survival signaling: Hyperactive PI3K/AKT pathway directly promotes cell survival and resistance to apoptosis

  • Genomic instability: Aberrant activation-induced cytidine deaminase (AID) activity during defective class-switching increases mutation rates

  • Defective checkpoint controls: Dysregulation of cell cycle control mechanisms

  • Lymphoproliferative environment: Chronic lymphoid expansion creates conditions favoring malignant transformation

Research has demonstrated that PI3K signaling is one of the most frequently aberrantly-activated pathways in cancer more broadly, highlighting its oncogenic potential .

How do distinct mutations in PIK3CD and PIK3R1 produce similar disease phenotypes?

This represents one of the most fascinating aspects of APDS pathogenesis. Despite seemingly opposite functional effects (gain-of-function in PIK3CD versus loss-of-function in PIK3R1), both mutation types converge on increased PI3Kδ activity:

  • PIK3CD mutations (E1021K, E525K, N334K): These directly enhance p110δ catalytic activity and membrane binding, bypassing normal regulatory constraints

  • PIK3R1 mutations: The p85α regulatory subunit normally inhibits the catalytic activity of p110δ. Disruption of p85α through deletion or mutation releases this inhibitory control

This exemplifies how different genetic mechanisms can converge on a common biochemical phenotype. The specific molecular details of each mutation may influence subtle differences in pathway hyperactivation, potentially explaining some of the clinical heterogeneity between APDS1 and APDS2 .

What are the optimal methods for identifying and functionally validating novel PI3K delta variants?

A comprehensive approach to identifying and validating PI3Kδ variants includes:

  • Genetic screening:

    • Targeted gene panels covering PIK3CD and PIK3R1

    • Whole exome sequencing (WES) for broader coverage

    • Whole genome sequencing (WGS) to detect intronic and regulatory variants

  • Variant classification:

    • In silico prediction tools for functional impact

    • Population frequency databases to assess rarity

    • Conservation analysis across species

    • Structural modeling to predict effects on protein function

  • Functional validation:

    • Phospho-flow cytometry to measure AKT phosphorylation

    • In vitro kinase assays to directly measure enzyme activity

    • B and T cell phenotyping to assess cellular impacts

    • Patient-derived cell analysis for ex vivo confirmation

    • CRISPR-engineered cell lines or mouse models carrying specific variants

  • Clinical correlation:

    • Detailed phenotyping of variant carriers

    • Family segregation analysis

    • Response to PI3Kδ inhibitors as functional confirmation

This multifaceted approach is critical because many PIK3CD and PIK3R1 variants have inconclusive functional significance, complicating diagnosis and treatment decisions .

What experimental approaches can distinguish the roles of PI3K delta in different immune cell subsets?

Several sophisticated experimental approaches can delineate PI3Kδ functions across immune cell populations:

  • Conditional knockout models:

    • Cell-type specific Cre-loxP systems to delete PIK3CD or PIK3R1 in specific lineages

    • Inducible systems to control timing of deletion

  • Selective inhibitors with different properties:

    • Isoform-selective PI3Kδ inhibitors at varying concentrations

    • Structurally distinct inhibitors to control for off-target effects

    • Allosteric versus ATP-competitive inhibitors

  • Advanced cellular techniques:

    • Adoptive transfer experiments with labeled cells

    • Mixed bone marrow chimeras to study cell-intrinsic effects

    • Single-cell transcriptomics and proteomics to identify cell-specific responses

    • CRISPR screens to identify cell-type specific dependencies

  • Ex vivo systems:

    • Patient-derived organoids to model tissue-specific responses

    • In vitro differentiation systems from patient stem cells

    • Co-culture systems to examine cell-cell interactions

These approaches have been instrumental in establishing the critical role of PI3Kδ in lymphocyte development and function while preserving most myeloid cell activities .

What are the challenges in developing isoform-selective PI3K inhibitors for research and therapy?

Developing truly selective PI3Kδ inhibitors presents numerous challenges:

  • Structural similarities: The ATP-binding pockets of different PI3K isoforms share significant homology, making absolute selectivity difficult to achieve

  • Dosing challenges: Finding the therapeutic window between efficacy and toxicity is complex:

    • Too little inhibition fails to correct pathologic hyperactivation

    • Too much inhibition suppresses normal immune function

    • Different tissues may require different levels of inhibition

  • Pharmacokinetic considerations:

    • Tissue penetration varies between inhibitors

    • Blood-brain barrier penetration may be relevant for CNS effects

    • Drug-drug interactions must be considered

  • Resistance mechanisms:

    • Compensatory upregulation of other PI3K isoforms

    • Alternative pathway activation

    • Selection for resistant cellular populations

  • Methodological validation challenges:

    • In vitro selectivity may not translate to in vivo specificity

    • Appropriate biomarkers to confirm target engagement

    • Differentiating on-target versus off-target effects

Despite these challenges, the FDA approval of Joenja (leniolisib) in March 2023 marks significant progress in targeting PI3Kδ for APDS treatment .

What diagnostic algorithm should be implemented to identify APDS patients?

An evidence-based diagnostic algorithm for APDS should include:

  • Clinical suspicion triggered by:

    • Recurrent sinopulmonary infections beginning in early childhood

    • Persistent lymphadenopathy, hepatosplenomegaly

    • Bronchiectasis or progressive lung disease

    • Herpesvirus infections (particularly EBV, CMV)

    • Autoimmune manifestations

    • Family history consistent with autosomal dominant inheritance

  • Initial immunological workup:

    • Immunoglobulin levels (often showing hypogammaglobulinemia or hyper-IgM pattern)

    • Lymphocyte subset analysis (looking for decreased naïve B cells, inverted CD4/CD8 ratio)

    • B and T cell functional studies

    • Vaccine response evaluation

  • Genetic testing approach:

    • Targeted sequencing of PIK3CD and PIK3R1 for common variants

    • Progression to whole exome/genome sequencing if targeted testing is negative

    • Functional validation studies for variants of uncertain significance

  • Specialized biomarkers:

    • Phospho-flow cytometry for pAKT levels

    • Analysis of T cell exhaustion markers

    • Measurement of senescent T cell populations

Early diagnosis is crucial, as delayed recognition can lead to irreversible complications like bronchiectasis or malignancy development .

How does the efficacy of targeted PI3K delta inhibitors compare with traditional immunomodulatory approaches in APDS?

The treatment landscape for APDS has evolved from symptomatic management to targeted therapy:

Traditional approaches:

  • Immunoglobulin replacement therapy: Addresses antibody deficiency but not underlying lymphoproliferation

  • Immunosuppressants: Can reduce autoimmunity and lymphoproliferation but may worsen infection risk

  • Antimicrobial prophylaxis: Reduces infection frequency but doesn't address disease mechanism

  • Hematopoietic stem cell transplantation (HSCT): Potentially curative but carries significant risks

Targeted PI3K delta inhibition (leniolisib/Joenja):

  • Directly addresses the underlying molecular pathology

  • Clinical trial results showed significant improvement in:

    • Reduction in lymph node size by day 85

    • 37% improvement in naïve B cell counts compared to placebo

    • Correction of immunophenotype

  • Potentially avoids risks associated with HSCT

  • May simultaneously address both immunodeficiency and lymphoproliferation

What factors should guide the decision between pharmacological management and hematopoietic stem cell transplantation in APDS patients?

This critical treatment decision should consider multiple factors:

Factors favoring pharmacological management:

  • Milder disease phenotype without life-threatening complications

  • Absence of significant structural lung damage

  • Good response to immunoglobulin replacement and antimicrobial prophylaxis

  • Availability of PI3Kδ inhibitors (age ≥12 years, weight ≥45 kg)

  • Patient preference for non-invasive approach

  • Absence of suitable transplant donors

Factors favoring HSCT consideration:

  • Severe or progressive disease despite conventional management

  • Development of malignancy

  • Extensive structural lung damage

  • Severe autoimmunity resistant to treatment

  • Very early disease identification (potentially better transplant outcomes)

  • Young age with suitable donor availability

  • Family preference based on avoiding lifelong medication

Decision-making framework:

  • Comprehensive multidisciplinary evaluation

  • Disease severity risk stratification

  • Assessment of existing irreversible complications

  • Consideration of patient age and donor availability

  • Discussion of risks/benefits with patient and family

  • Individualized decision making with periodic reassessment

The advent of targeted therapies has complicated this decision, and optimal strategies may involve sequential approaches (e.g., stabilization with PI3Kδ inhibitors before proceeding to HSCT in selected cases) .

How might single-cell technologies advance our understanding of PI3K delta biology in rare immunodeficiencies?

Single-cell technologies offer unprecedented opportunities to dissect PI3Kδ biology:

  • Single-cell RNA sequencing applications:

    • Identifying cell-specific transcriptional signatures of PI3Kδ hyperactivation

    • Discovering compensatory mechanisms in different immune compartments

    • Detecting rare cellular populations with distinct responses to PI3Kδ dysregulation

    • Tracking clonal evolution in lymphoproliferative and pre-malignant conditions

  • Single-cell proteomics and phospho-proteomics:

    • Mapping PI3K signaling networks at individual cell resolution

    • Identifying novel downstream targets and feedback mechanisms

    • Characterizing pathway cross-talk unique to specific cell lineages

  • Spatial transcriptomics and proteomics:

    • Understanding tissue-specific consequences of PI3Kδ dysregulation

    • Mapping cellular interactions in lymphoid tissues

    • Characterizing microenvironmental influences on PI3K signaling

  • Integrated multi-omic approaches:

    • Correlating genotype with cell-specific phenotypes

    • Building comprehensive models of PI3K signaling across immune development

    • Identifying biomarkers predictive of disease progression or treatment response

These technologies could reveal how seemingly identical mutations produce heterogeneous clinical phenotypes and identify novel therapeutic targets beyond direct PI3Kδ inhibition .

What therapeutic strategies beyond direct PI3K inhibition might address APDS pathology?

Novel therapeutic approaches may offer advantages over direct PI3Kδ inhibition:

  • Downstream pathway modulation:

    • mTOR inhibitors to target a critical downstream effector

    • AKT inhibitors with potential for more selective effects

    • S6 kinase inhibitors to modulate a specific branch of PI3K signaling

  • Combination approaches:

    • Low-dose PI3Kδ inhibition combined with targeted immunomodulators

    • Sequential therapy regimens to minimize resistance development

    • Pathway-specific combinations tailored to individual patient phenotypes

  • Advanced cellular therapies:

    • Autologous gene-edited stem cell approaches

    • CRISPR-based correction of pathogenic variants

    • CAR-T or engineered T-regulatory cell therapies for autoimmune manifestations

  • Innovative drug delivery systems:

    • Tissue-targeted delivery to minimize systemic toxicity

    • Controlled-release formulations for optimized pharmacokinetics

    • Cell-specific targeting strategies

  • Precision medicine strategies:

    • Biomarker-guided therapy selection

    • Variant-specific therapeutic approaches

    • Integration of artificial intelligence to predict optimal treatment regimens

These approaches may overcome limitations of current PI3Kδ inhibitors while addressing the complex disease biology of APDS .

How might better understanding of PI3K delta in APDS inform therapeutic strategies for more common immune disorders?

The rare disease APDS serves as a valuable "experiment of nature" that can inform broader therapeutic applications:

  • Applications to common variable immunodeficiency (CVID):

    • Identification of CVID subgroups with subclinical PI3K dysregulation

    • Targeted therapies for CVID patients with similar cellular phenotypes

    • Biomarkers derived from APDS research to stratify CVID patients

  • Relevance to autoimmune diseases:

    • Tailored PI3Kδ inhibition in autoimmune cytopenias

    • Application to autoimmune conditions sharing lymphocyte dysregulation patterns

    • Understanding of dose-dependent effects to minimize immunosuppression

  • Cancer immunotherapy implications:

    • Leveraging knowledge of PI3Kδ in T cell exhaustion for checkpoint inhibitor enhancement

    • Optimizing CAR-T manufacturing through PI3K pathway modulation

    • Development of combination strategies for hematologic malignancies

  • Broader implications for precision immunology:

    • Framework for pathway-specific therapeutic targeting

    • Models for understanding gene dosage effects in immune regulation

    • Paradigms for translating rare disease mechanisms to common conditions

The bidirectional knowledge transfer between rare and common diseases represents an efficient path to therapeutic innovation, with APDS research potentially benefiting patients across the immunological disease spectrum .

Product Science Overview

Structure and Function

Class IA PI3Ks consist of a catalytic subunit (p110) and a regulatory subunit (p85). The p110 subunit has three isoforms: p110α, p110β, and p110δ. The p85 subunit also has multiple isoforms, including p85α, p55α, p50α, p85β, and p55γ . The p110δ isoform, in particular, is predominantly expressed in leukocytes and is involved in immune responses .

The p110δ/p85α complex is a heterodimer where the p110δ catalytic subunit interacts with the p85α regulatory subunit. This interaction is crucial for the regulation of the kinase activity of p110δ. The p85α subunit contains SH2 (Src Homology 2) domains that bind to phosphorylated tyrosine residues on receptor tyrosine kinases (RTKs) or other adaptor proteins, leading to the recruitment of the p110δ/p85α complex to the plasma membrane . Upon membrane recruitment, the p110δ subunit phosphorylates phosphatidylinositol (4,5)-bisphosphate (PIP2) to generate phosphatidylinositol (3,4,5)-trisphosphate (PIP3), which acts as a second messenger to activate downstream signaling pathways .

Recombinant Expression

The human recombinant p110δ/p85α complex is typically expressed in baculovirus-infected insect cells. This system allows for the production of high-quality, active enzyme complexes that are suitable for biochemical and structural studies . The recombinant complex is often used to study enzyme kinetics, screen inhibitors, and profile selectivity .

Clinical Relevance

The p110δ isoform is upregulated in certain leukemias, such as chronic lymphocytic leukemia (CLL), and is a target for specific inhibitors like CAL-101 (Idelalisib), which has shown promise in clinical trials . The unique regulatory mechanisms of p110δ, as compared to other isoforms like p110α and p110β, make it an attractive target for therapeutic intervention .

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.