Fact-checked by Grok 2 weeks ago

Wee1

Wee1 (WEE1) is a nuclear tyrosine kinase that serves as a critical regulator of the eukaryotic cell cycle, primarily enforcing the G2/M checkpoint to prevent mitotic entry in the presence of DNA damage or replication stress. By phosphorylating cyclin-dependent kinase 1 (CDK1) on tyrosine residue 15, Wee1 inhibits the CDK1-cyclin B complex (also known as mitosis-promoting factor), thereby halting cell cycle progression and providing time for DNA repair mechanisms to operate. This inhibitory phosphorylation is activated through upstream signaling pathways, including ATM/ATR kinases and their downstream effector CHK1, which respond to genotoxic insults. Beyond the G2/M transition, Wee1 contributes to intra-S phase checkpoint control and safeguards replication fork integrity by modulating endonuclease activity, such as inhibiting MUS81-EME1/2 to prevent excessive DNA cleavage during replication stress. In cancer biology, Wee1 is frequently overexpressed in diverse malignancies—including breast, ovarian, colorectal, hematological, and solid tumors like non-small cell lung cancer and glioblastoma—where it supports unchecked proliferation, genomic instability, and therapeutic resistance, particularly in p53-deficient cells that rely on Wee1 for survival post-DNA damage. As a result, Wee1 inhibition has become a focal point in oncology, with selective small-molecule inhibitors like adavosertib (AZD1775, MK-1775) demonstrating preclinical synergy with DNA-damaging agents such as chemotherapy and radiation, and advancing through clinical trials to exploit synthetic lethality in tumor cells. Wee1's regulation involves post-translational modifications, including phosphorylation at sites like Ser123 and ubiquitin-mediated degradation, ensuring precise temporal control during cell division.

Background and Discovery

Discovery

Wee1 was originally discovered in the mid-1970s through genetic screens for temperature-sensitive mutants in the fission yeast , conducted by and colleagues as part of efforts to elucidate controls coordinating cell growth and division. These screens identified mutants that altered cell size and the timing of , with the wee1 mutants exhibiting a distinctive small-cell ("wee") phenotype due to premature mitotic entry at reduced cell volumes compared to wild-type cells. The wee1 mutants were isolated alongside other cell division cycle (cdc) mutants, mapping to a single locus that delayed or advanced the G2/M transition, thereby linking cell size to mitotic onset. The naming of the wee1 gene stems from the Scottish word "wee," meaning small, which aptly described the compact of the mutant cells resulting from accelerated without proportional growth. Early phenotypic analyses revealed that wee1 mutants were viable but hypersensitive to conditions promoting premature , underscoring Wee1's role as a negative regulator essential for proper timing in . This discovery laid foundational insights into size-dependent controls in eukaryotic . In the 1980s, the wee1+ gene was cloned by complementation in S. pombe strains overexpressing the mitotic inducer cdc25+, allowing isolation of the wild-type gene that suppressed the elongated phenotype. Sequencing by Paul Russell and Paul Nurse in 1987 demonstrated that wee1+ encodes a protein kinase homolog with a conserved tyrosine kinase domain in its C-terminus, marking the first identification of a dose-dependent mitotic inhibitor at the molecular level. Overexpression of wee1+ delayed mitosis, while loss-of-function advanced it, confirming its inhibitory function upstream of the cdc2 kinase. The homolog, WEE1, was first identified in 1991 via functional complementation screens in fission yeast for genes interacting with CDK1 (p34cdc2), the key mitotic . A human cDNA (WEE1Hu) was isolated by et al. that, when expressed in S. pombe, generated elongated cells by blocking the G2/M transition, indicating conserved inhibitory activity. Subsequent biochemical studies in 1993 verified that human WEE1 specifically phosphorylates CDK1 on tyrosine 15, a modification that inhibits CDK1 activation and ensures mitotic delay. Key milestones in the 1990s further connected Wee1 to DNA damage responses, establishing its central role in the G2/M checkpoint. Pioneering work showed that upon DNA damage, the checkpoint kinase Chk1 phosphorylates Wee1, enhancing its stability and activity to sustain inhibitory phosphorylation of CDK1, thereby preventing mitosis until repair occurs. These findings, including studies in fission yeast demonstrating Chk1-dependent Wee1 activation, highlighted Wee1 as a critical effector in damage-induced cell cycle arrest across eukaryotes.

Structure

Human Wee1 is a 646-amino acid protein encoded by the WEE1 gene located on 11p15.4. The protein features a conserved N-terminal regulatory domain spanning residues 1–290 and a C-terminal domain encompassing residues 291–646. The domain exhibits a bilobal typical of protein , with an N-terminal lobe composed primarily of a five-stranded antiparallel β-sheet and a glycine-rich loop (residues 306–311), and a C-terminal lobe dominated by α-helices forming a four-helix bundle. These lobes are connected by a hinge region (residues 377–381) that facilitates conformational flexibility. The ATP-binding cleft lies at the interface between the lobes, bounded by elements from the β-sheet, glycine-rich loop, hinge, and catalytic loop. The activation loop, spanning residues 462–486, adopts an ordered active conformation in the C-terminal lobe, stabilized by secondary structures and side-chain interactions such as the Asp479–Arg481 , without requiring for kinase activity. High-resolution crystal structures, including that of the kinase domain complexed with inhibitor PD0407824 (PDB: 1X8B at 1.8 Å resolution), illustrate these structural elements and highlight Wee1-specific residues in the that confer specificity for phosphorylating Tyr15 on the CDK1 . Additional structures, such as PDB: 3BI6, confirm the conservation of this fold with bound inhibitors. Structural studies predominantly utilize truncated constructs of the kinase domain (residues 291–575), omitting the N-terminal regulatory domain, which contains elements like nuclear localization signals that direct to the during . This truncation simplifies crystallization but excludes regulatory features unique to the full-length protein. Post-translational modification sites integral to the include at Ser642 in the C-terminal region, which modulates activity.

Function and Mechanism

Core Function

Wee1 functions as a dual-specificity that inhibits progression by phosphorylating (CDK1) on the 15 (Tyr15) residue, thereby preventing premature entry into during the G2/M transition. This inhibitory maintains CDK1 in an inactive state until appropriate cellular conditions are met, ensuring orderly progression through the . In cells, Wee1 accounts for the majority of Tyr15 activity on CDK1, as demonstrated by depletion studies . The core mechanism of Wee1 involves direct binding to the CDK1-cyclin B complex, followed by the catalytic transfer of a group from ATP to CDK1's Tyr15. This modification disrupts CDK1's ATP-binding affinity and catalytic efficiency, substantially reducing its capacity to phosphorylate downstream mitotic substrates such as lamin B and nuclear lamins, which are essential for nuclear envelope breakdown and chromosome condensation. The kinase reaction proceeds as follows: \text{Wee1} + \text{ATP} + \text{CDK1-Tyr15} \to \text{Wee1} + \text{[ADP](/page/ADP)} + \text{CDK1-pTyr15} This precise inhibition allows Wee1 to act as a , delaying mitotic onset until is complete or damage is repaired, thereby safeguarding genomic integrity against replication errors or lesions that could lead to chromosomal instability. Wee1 also contributes to S-phase by phosphorylating CDK2 on Tyr15, helping maintain replication stability under stress. Wee1's subcellular localization further supports its core function, with the kinase predominantly nuclear during interphase to target nuclear-localized CDK1-cyclin B complexes and prevent ectopic activation. Upon mitotic entry, Wee1 relocalizes to the cytoplasm, consistent with the activation of CDK1 and progression through mitosis. This dynamic partitioning ensures spatially restricted inhibition, optimizing control over the G2/M boundary.

Regulation

Wee1 activity is tightly controlled through multiple post-translational modifications, primarily events that modulate its stability, localization, and enzymatic function. Positive regulation occurs via phosphorylation by checkpoint kinase 1 (Chk1) at serine 642 (Ser642), which promotes binding to 14-3-3 proteins and enhances Wee1's nuclear retention, thereby stabilizing the protein and augmenting its inhibitory effect on (CDK1) during . This phosphorylation is particularly important in maintaining the G2/M checkpoint under normal conditions. In the context of the DNA damage response, ataxia-telangiectasia mutated () and ATM- and Rad3-related (ATR) kinases detect DNA lesions and activate Chk1, which in turn phosphorylates Wee1 at Ser642 to reinforce nuclear localization and activity, allowing time for before mitotic entry. This pathway is crucial in cells lacking functional , where the G1 checkpoint is compromised, making Wee1 a key guardian of genomic integrity. Negative regulation of Wee1 is mediated by CDK1 phosphorylation at multiple sites, including Ser123, which creates motifs for polo-like kinase 1 () and casein kinase 2 (CK2), ultimately leading to recruitment of the E3 ubiquitin ligase β-TrCP. This multi-step process triggers ubiquitination of Wee1 and its subsequent proteasomal degradation, reducing Wee1 levels at the G2/M transition to permit mitotic progression. further contributes by Wee1 at Ser53, accelerating this degradation cascade. Transcriptional control of Wee1 expression responds to cellular stress, with evidence indicating upregulation of WEE1 mRNA in contexts of genomic instability, though direct dependence remains context-specific and is often observed in p53-deficient states where Wee1 compensates for checkpoint loss. A key feedback loop exists between Wee1 and CDK1: Wee1 inhibits CDK1 by phosphorylating it at tyrosine 15 (Tyr15), preventing premature mitotic entry, while active CDK1 reciprocates by phosphorylating Wee1 to promote its inactivation and , ensuring timely release from arrest. This reciprocal regulation fine-tunes the G2/M transition and integrates upstream signals for fidelity.

Homologues and Evolution

Homologues Across Species

The Wee1 kinase was first discovered in the fission yeast Schizosaccharomyces pombe as a key negative regulator of the G2/M transition, where it enforces cell cycle arrest by phosphorylating Cdc2 on tyrosine 15 to inhibit premature mitosis entry. The S. pombe Wee1 protein comprises 877 amino acids, including an N-terminal regulatory domain of approximately 550 amino acids and a C-terminal catalytic kinase domain of about 350 amino acids, making it essential for maintaining genomic integrity during replication stress-induced G2 arrest. In humans, the primary functional homolog is WEE1 (also designated WEE1A), encoded by the WEE1 gene on chromosome 11q13.3 and ubiquitously expressed across somatic tissues to coordinate nuclear G2 checkpoint responses. This 646-amino-acid protein features a conserved domain responsible for inhibitory of CDK1. A secondary homolog, WEE1B (also known as WEE2), is restricted to expression and contributes to meiotic arrest by phosphorylating CDK1. Among mammals, the Wee1 ortholog exhibits high sequence conservation with its counterpart, achieving approximately 85-90% overall identity, though the N-terminal regulatory shows greater divergence that may influence species-specific modulation. In other eukaryotes, such as the Drosophila melanogaster, Wee1 has a direct homolog alongside the related but distinct Myt1 , which shares significant similarity in the (around 50% identity to Wee1) while differing in association and dual-site capability on CDK1. In plants like , the Wee1 homolog localizes predominantly to the and responds to inhibition, displaying sequence similarity to metazoan Wee1 primarily within the despite overall architectural differences. Sequence conservation of Wee1 is particularly pronounced in the kinase domain across metazoans, exceeding 70% identity, which underscores its core catalytic role in CDK inhibition, whereas the N-terminal regions exhibit more variability linked to regulatory phosphorylation sites and subcellular targeting.

Evolutionary Conservation

The Wee1 domain originated in the last eukaryotic common ancestor (LECA), where it functioned as part of the core regulatory machinery alongside Cdc25 phosphatases and Cdk1. Ancestral state reconstructions indicate that Wee1 was already present in LECA, contributing to inhibitory of cyclin-dependent kinases to prevent premature mitotic entry. In non-opisthokont lineages, such as , Wee1 homologs primarily exhibit serine/ kinase activity, but specificity evolved specifically within , enabling precise regulation of CDK1 at residues. This evolutionary shift underscores Wee1's adaptation to more complex checkpoint mechanisms in animal lineages. The CDK1 Tyr15 phosphorylation motif targeted by Wee1 is strictly conserved across eukaryotes, from fission yeast (Schizosaccharomyces pombe) to humans, reflecting strong purifying selection to maintain G2/M checkpoint integrity. This conservation ensures that Wee1-mediated inhibition of CDK1 activity remains a universal brake on mitotic progression, preventing genome instability in diverse species. Comparative genomics analyses reveal that catalytic residues in the Wee1 kinase domain exhibit over 80% sequence identity in alignments spanning yeast, invertebrates, and vertebrates, highlighting the essential nature of these sites for substrate recognition and phosphotransfer. Such high conservation in key motifs attests to the adaptive pressures favoring Wee1's role in DNA damage response and replication fidelity. In vertebrates, Wee1 underwent adaptations including the acquisition of nuclear export signals (), which facilitate CRM1-dependent shuttling between and for enhanced spatiotemporal control of checkpoints. These signals allow dynamic relocalization of Wee1 in response to DNA damage, enabling finer tuning of CDK1 inhibition during S and G2 phases compared to invertebrate homologs. Phylogenetic analyses indicate that events following metazoan divergence gave rise to the expanded WEE1/MYT1 family, with MYT1 emerging as a membrane-associated paralog specializing in / dual .

Cellular Roles and Phenotypes

Role in Cell Cycle Checkpoints

Wee1 serves as a key regulator in the G2/M cell cycle checkpoint, primarily by phosphorylating cyclin-dependent kinase 1 (CDK1) on tyrosine 15 to inhibit its activity and prevent premature entry into mitosis when DNA damage, replication stress, or incomplete S-phase replication is detected. This inhibitory phosphorylation maintains CDK1 in an inactive state complexed with cyclin B1, allowing sufficient time for DNA repair mechanisms to operate and safeguarding genomic integrity before chromosome segregation. Activation of Wee1 in this context is mediated by upstream checkpoint kinases, such as Chk1, which directly phosphorylates Wee1 to enhance its function in response to genotoxic stress. Wee1 integrates with other cell cycle checkpoints to provide coordinated control, including interplay with the G1/S checkpoint via the p53-Wee1 axis, where p53 orchestrates early arrest in G1 phase while Wee1 reinforces late-stage surveillance at G2/M to ensure comprehensive DNA integrity assessment. In the replication stress response, the ATR-Chk1-Wee1 pathway further coordinates Wee1 activation; ATR senses single-stranded DNA at stalled replication forks and phosphorylates Chk1, which then stabilizes Wee1 to suppress CDK activity and prevent collapse of replication forks or untimely mitotic progression. This multi-checkpoint coordination ensures robust protection against replication errors that could propagate to mitosis. The inhibition of mitotic entry by Wee1 operates through a governed by the balance between Wee1 activity and the counteracting Cdc25, where a critical must shift to fully activate CDK1 and trigger only after checkpoint satisfaction. In normal cells, this Wee1-mediated control promotes accurate during by averting entry with unresolved issues, and disruptions in Wee1 function can result in due to missegregation events. Experimental evidence supports these roles: overexpression of Wee1 in delays mitotic onset, enabling cells to achieve larger size before division, while knockdown in mammalian synchronized cells accelerates mitotic entry, often leading to aberrant division timing.

Mutant Phenotypes

In the fission yeast , deletion of wee1 (wee1Δ) results in viable cells that exhibit a characteristic "" phenotype, characterized by reduced cell size at division due to premature entry into before sufficient growth has occurred. These mutants display an elongated morphology as small daughter cells elongate to attempt size compensation prior to dividing again, leading to ongoing premature without lethality under standard conditions. Suppressor mutations in the cdc2 gene, which encodes the , can partially restore normal cell size and division timing by reducing CDK activity, thereby counteracting the accelerated mitotic entry. In mammals, complete knockout of Wee1 in mice leads to embryonic lethality around implantation, accompanied by accumulation of DNA damage and chromosomal abnormalities in pre-implantation embryos. Conditional knockouts in somatic tissues, such as mammary glands, reveal genomic instability, including increased , chromosome fragmentation, and elevated DNA double-strand breaks, highlighting Wee1's role in maintaining chromosomal integrity beyond embryogenesis. Overexpression of Wee1 in S. pombe produces the opposite phenotype to loss-of-function, resulting in enlarged, highly elongated cells due to prolonged G2 phase arrest and delayed mitotic entry. In mammalian cells, Wee1 overexpression similarly extends G2 duration, leading to larger cell size, and confers hypersensitivity to DNA-damaging agents such as UV radiation or methyl methanesulfonate (MMS) by overly inhibiting CDK1 and impairing adaptive checkpoint responses. In human cell lines, siRNA-mediated knockdown of Wee1 induces premature mitotic entry, particularly under replication stress, culminating in characterized by aberrant segregation and failure. This is exacerbated by DNA-damaging conditions, promoting increased through activation of pathways and DNA damage signaling. Rescue experiments in S. pombe demonstrate that wild-type Wee1 expression fully complements the wee1Δ , restoring normal size and division timing, whereas expression of a kinase-dead Wee1 (e.g., K345R) fails to do so, confirming the requirement for catalytic activity in inhibitory phosphorylation of Cdc2.

Role in Cancer

Expression and Dysregulation in Tumors

Wee1 is frequently overexpressed in a variety of solid tumors, including , ovarian, and . In and low-grade , hypomethylation of the WEE1 promoter leads to elevated expression, contributing to tumor progression. Similarly, in ovarian and cancers, WEE1 mRNA and protein levels are heightened, often linked to chromosomal rather than direct of the WEE1 locus. High WEE1 expression correlates with poor clinical outcomes across multiple cancer types, including advanced tumor stages and increased metastasis risk, as evidenced by analyses of (TCGA) datasets. In and colorectal cancers, elevated WEE1 mRNA levels are independently associated with reduced overall survival and progression. TCGA data further reveal that high WEE1 expression predicts worse in gliomas, particularly when combined with markers of replication stress. In the context of TP53 mutations, which impair the G1/S checkpoint, Wee1 is upregulated as a compensatory to reinforce the G2/M checkpoint and prevent in tumor cells. This synthetic lethality dynamic is prominent in p53-mutant cancers, where elevated Wee1 expression sustains survival amid genomic instability. Wee1 is overexpressed in hematological malignancies, consistent with its oncogenic role in promoting survival under stress. Recent studies from 2024-2025 have linked elevated Wee1 expression to immunotherapy resistance, with non-responders to PD-1 blockade showing significantly higher levels, associated with poor prognosis and reduced T-cell infiltration.

Contribution to Cancer Progression

Wee1 overexpression reinforces the G2/M checkpoint, enabling cancer cells to survive DNA damage by delaying mitotic entry and allowing time for repair, which promotes checkpoint adaptation and fosters genomic instability essential for tumorigenesis. This adaptation permits the accumulation of mutations, driving tumor evolution, as evidenced in models where elevated Wee1 activity sustains proliferation despite unresolved DNA lesions. In p53-deficient cancers, which often lack the G1/S checkpoint, Wee1 becomes a critical dependency for maintaining viability amid replication stress. Wee1 contributes to therapy resistance by enhancing mechanisms in response to and radiotherapy, thereby reducing treatment-induced . Recent studies from 2025 have further linked cytoplasmic Wee1 to PD-1 blockade resistance in NANOG-high tumors, where it hyperactivates the HSP90A/TCL1/AKT signaling axis, promoting tumor proliferation and immune evasion. In preclinical models, xenografts from high-Wee1-expressing cells exhibit reduced and slower tumor regression when treated with , underscoring Wee1's role in chemoprotection. Conversely, CRISPR-mediated Wee1 knockout sensitizes malignant pleural cells to standard , enhancing and cell death. Emerging 2024-2025 research highlights Wee1's involvement in resistance within ARID1A-mutant EGFR-driven cancers, where Wee1 upregulation drives adaptive survival pathways that counteract inhibition.

Therapeutic Targeting

Wee1 Inhibitors

Wee1 inhibitors are small-molecule compounds designed to block the activity of Wee1, a critical regulator of the G2/M . By inhibiting Wee1, these agents prevent the of CDK1 at tyrosine 15 (Tyr15), leading to premature activation of CDK1 and unscheduled entry into . This mechanism induces and exacerbates replication stress, particularly in p53-deficient cancer cells that lack robust G1 checkpoint control and are unable to repair DNA damage effectively. First-generation Wee1 inhibitors, such as adavosertib (AZD1775 or MK-1775), are ATP-competitive agents that bind to the hinge region of the Wee1 . Adavosertib exhibits potent inhibition with an of approximately 5 nM in cell-free assays and demonstrates selectivity against a panel of kinases, though it shows some off-target activity. This compound has been extensively studied in preclinical models for its ability to sensitize tumors to DNA-damaging therapies by abrogating checkpoint arrest. However, its development was discontinued in 2022 due to toxicity concerns. Second-generation inhibitors aim to enhance selectivity and reduce associated with off-target effects observed in earlier compounds. Azenosertib (ZN-c3) represents a key example, with an of 3.8 nM for Wee1 and improved kinase selectivity, including over 60-fold preference over ( 227 nM), minimizing unintended inhibition of polo-like kinases that contribute to myelosuppression. This optimization supports better tolerability through intermittent dosing schedules while maintaining robust antitumor activity via CDK1 derepression and mitotic induction. Emerging Wee1 inhibitors focus on further refining selectivity and pharmacokinetic properties to expand therapeutic windows. APR-1051, developed by Aprea Therapeutics, is an orally bioavailable agent with high selectivity for Wee1, designed to limit off-target effects and associated myelosuppression, as evidenced by low rates of in early dosing cohorts. It entered phase 1 evaluation in 2024, with updates in 2025 from the ACESOT-1051 trial in advanced solid tumors showing promising anti-proliferative activity in preclinical models. Similarly, Debio 0123 (zedorosertib) is a highly selective oral ( 0.8 nM for Wee1) with no detectable activity against or PLK2, and notable brain penetration (brain-to-plasma AUC ratio of 0.49–0.60), enabling potential use in malignancies. Preclinical studies have highlighted synergies between Wee1 inhibitors and other targeted agents, enhancing efficacy in replication-stressed tumors. Combinations with , such as , promote by amplifying DNA damage and defects, as demonstrated in models where dual inhibition controlled tumor growth more effectively than monotherapy. Synergy with CHK1 inhibitors arises from complementary checkpoint abrogation, leading to increased S-phase DNA damage and cell death in and cells. Wee1 inhibitors also cooperate with statins, which disrupt the and indirectly sensitize p53-mutant cancers to checkpoint loss, showing additive antiproliferative effects in gynecologic malignancies. Recent 2025 investigations into PKMYT1 co-inhibition, including pairings like Debio 0123 with lunresertib, reveal synergistic eradication of ovarian and organoids at low doses by dual blockade of CDK1 regulators.

Clinical Development and Trials

Adavosertib (AZD1775), the first Wee1 inhibitor to enter clinical development, has been evaluated in multiple phase II trials, including combinations with in gynecologic cancers. In a phase II study of adavosertib monotherapy in recurrent reported in 2025, an objective response rate (ORR) of 29.4% was observed, with activity noted in TP53-mutated cases, though tolerability was limited at higher doses. A separate phase II trial combining adavosertib with in refractory non-small cell reported poorer tolerability compared to docetaxel alone, highlighting challenges in combination regimens. Despite a phase II failure in SETD2-altered clear cell where no objective responses were seen, results from these trials have informed the development of subsequent Wee1 inhibitors. Azenosertib (ZN-c3), a selective , has progressed through 1/2 trials in advanced solid tumors, demonstrating preliminary antitumor activity. In the ZN-c3-001 1/2 , monotherapy at therapeutic doses showed clinical responses in heavily pretreated patients, including an ORR of 34.9% in E1-positive, platinum-resistant . Updated 2025 data from ongoing trials, including ASCO presentations, indicate manageable safety with signals of efficacy in BRCA-mutated subsets, though exact maximum tolerated doses (MTD) vary by regimen. Combination studies with are ongoing in pancreatic and cohorts, showing improved event-free survival compared to historical controls. APR-1051, a next-generation Wee1 inhibitor designed for improved selectivity, reported phase 1 updates in October 2025 from the ACESOT-1051 trial in advanced solid tumors with cancer-associated gene alterations. Early data at the 100 mg dose level showed stable disease in 3 of 4 patients per RECIST v1.1 criteria, with no myelosuppression observed in initial cohorts, suggesting reduced toxicity relative to first-generation inhibitors like AZD1775. Activity signals were noted in DNA damage response-deficient tumors, including ATM alterations, supporting further evaluation in biomarker-enriched populations. Debio 0123, a brain-penetrant , is in phase 1 dose escalation for advanced solid tumors, including . The ongoing phase 1/2 trial (NCT05765812) in combination with focuses on identifying dose-limiting toxicities in recurrent . As of November 2025, the trial remains active with no published efficacy data yet. Clinical development of faces common challenges, including myelosuppression as a dose-limiting toxicity in many regimens, though newer agents like APR-1051 show reduced hematologic effects. Emerging 2025 data highlight synergies with , such as enhancing anti-PD-1 responses in PD-1-resistant by promoting immune activation in high-NANOG tumors. Biomarkers like deficiency (HRD) scores and CCNE1 amplification are increasingly used for patient selection, correlating with improved responses in ovarian and cancers.

References

  1. [1]
    Molecular Pathways: Targeting the Protein Kinase Wee1 in Cancer
    Wee1 is a protein kinase that regulates the G2 checkpoint and prevents entry into mitosis in response to DNA damage (Fig. 1; ref. 1). The cell cycle is a highly ...Abstract · Background · Clinical–Translational Advances · Toxicity of Wee1 Inhibitors
  2. [2]
    A WEE1 family business: regulation of mitosis, cancer progression ...
    Sep 21, 2020 · This review recapitulates and discusses the most recent findings on the biological function of WEE1/PKMYT1 during the cell cycle and in the DNA damage repair.
  3. [3]
    [PDF] Paul M. Nurse - Nobel Lecture
    Murdoch used fission yeast to study how cells grow during the cell cycle, devising procedures for physiological analysis and to synchronise cells so they ...
  4. [4]
    Wee1 + -like gene in human cells - Nature
    Sep 5, 1991 · We report here that overexpression of WEE1Hu in fission yeast generates very elongated cells as a result of inhibition of the G2-M transition in the cell cycle.Missing: paper | Show results with:paper
  5. [5]
    WEE1 Gene - GeneCards | WEE1 Protein | WEE1 Antibody
    This gene encodes a nuclear protein, which is a tyrosine kinase belonging to the Ser/Thr family of protein kinases. This protein catalyzes the inhibitory ...Missing: identification 1991<|separator|>
  6. [6]
    WEE1 inhibition in cancer therapy: Mechanisms, synergies ...
    WEE1 kinase plays a central role in maintaining genomic stability by regulating critical cell cycle checkpoints, DNA damage repair, and replication processes( ...
  7. [7]
  8. [8]
  9. [9]
  10. [10]
    Subcellular localisation of human wee1 kinase is regulated during ...
    We demonstrate that wee1: (i) is localised in the nucleus during interphase; (ii) is associated with unidentified nuclear structures at G1/S and in prophase; ( ...
  11. [11]
    Wee1-like protein kinase - Homo sapiens (Human) - UniProt
    Feb 1, 1996 · Acts as a negative regulator of entry into mitosis (G2 to M transition) by protecting the nucleus from cytoplasmically activated cyclin B1-complexed CDK1.Missing: homolog 1991
  12. [12]
  13. [13]
  14. [14]
    Wee1 controls genomic stability during replication by regulating the ...
    Aug 22, 2011 · Wee1 is essential for normal DNA replication and for genomic stability, at least in part by inhibiting a general DNA damage response induced ...Wee1 Down-Regulation... · Wee1 Controls Replication... · Codepletion Of Wee1 And...
  15. [15]
    Spatiotemporal regulations of Wee1 at the G2/M transition
    Jan 13, 2011 · To study the role of Wee1 accumulated at the SPB, we aimed to determine to which face of the SPB Wee1 is localized during the G2/M transition.
  16. [16]
    Cyclin-dependent kinase (CDK) phosphorylation destabilizes ...
    Wee1 family protein kinases that inhibit Cdc2 during the G2 phase of the cell cycle must be down-regulated at the onset of mitosis. At that time, somatic Wee1 ...Missing: PDB | Show results with:PDB
  17. [17]
  18. [18]
    Regulation of Schizosaccharomyces pombe Wee1 tyrosine kinase
    May 16, 1997 · Wee1 tyrosine kinase regulates mitosis by carrying out the inhibitory tyrosine 15 phosphorylation of Cdc2 M-phase inducing kinase.Missing: cloning 1980s
  19. [19]
    Regulation of Schizosaccharomyces pombe Wee1 Tyrosine Kinase
    Dec 13, 1996 · Wee1 tyrosine kinase regulates mitosis by carrying out the inhibitory tyrosine 15 phosphorylation of Cdc2. M-phase inducing kinase.
  20. [20]
    Glucose restriction induces transient G2 cell cycle arrest extending ...
    Jan 25, 2016 · Taken together, these observations indicated that Wee1 is essential for G2 cell cycle arrest in response to glucose restriction. ... pombe) – ...
  21. [21]
    Structural Basis of Wee Kinases Functionality and Inactivation by ...
    Wee kinases negatively regulate the cell cycle by inhibiting CDK1. Wee1 and Wee2 have unique structural features that distinguish them from Myt1.Missing: review | Show results with:review
  22. [22]
    Identification and characterization of human Wee1B, a new member ...
    Oct 9, 2008 · We identified human Wee1B as a novel Cdk1-inhibitory kinase. The identification of this new member of the Wee1 family suggests that inhibition of Cdk1 is ...
  23. [23]
    Wee1B Is an Oocyte-Specific Kinase Involved in the Control of ...
    Sep 20, 2005 · mWee1B is a key MPF inhibitory kinase in mouse oocytes, functions downstream of PKA, and is required for maintaining meiotic arrest.
  24. [24]
    Comparative analysis of 1196 orthologous mouse and human full ...
    Protein sequence conservation varied between 36% and 100% identity, with an average value of 85%. The average degree of nucleotide sequence identity for the ...
  25. [25]
  26. [26]
    Characterization of maize (Zea mays L.) Wee1 and its activity in ...
    In contrast, residues 179–403 of ZmWee1 show extensive similarity to the protein kinase domain of Wee1 from Drosophila, humans, S. pombe, and S. cerevisiae.Sign Up For Pnas Alerts · Results · Molecular Cloning Of Zmwee1<|separator|>
  27. [27]
    Systematic Localization of the Arabidopsis Core Cell Cycle Proteins ...
    RBR1 and WEE1 proteins localized exclusively to the nucleus (Fig. 2, Ee and Ef). Subcellular Localization during Cell Division in BY2 Cells. To build a more ...Results · Systematic Gfp Tagging Of... · DiscussionMissing: similarity | Show results with:similarity
  28. [28]
    Arabidopsis WEE1 Kinase Controls Cell Cycle Arrest in Response to ...
    We demonstrate that the cell cycle regulatory WEE1 kinase gene of Arabidopsis thaliana is transcriptionally activated upon the cessation of DNA replication or ...Missing: similarity | Show results with:similarity
  29. [29]
    Structure and Inhibition of the Human Cell Cycle Checkpoint Kinase ...
    We have determined the crystal structure of the catalytic domain of human somatic Wee1 (Wee1A) complexed with an active-site inhibitor at 1.8 Å resolution.
  30. [30]
    Structural conservation of WEE1 and its role in cell cycle regulation ...
    Dec 13, 2021 · The WEE1 kinase is ubiquitous in plant development and negatively regulates the cell cycle through phosphorylations.
  31. [31]
    Evolution of opposing regulatory interactions underlies the ... - Nature
    May 27, 2021 · Our results show that this reversed action of a kinase-phosphatase pair, Wee1 and Cdc25, on CDK is particularly suited to establish a stable G2 phase and to ...
  32. [32]
    Gauchos and ochos: a Wee1-Cdk tango regulating mitotic entry - PMC
    May 13, 2010 · The region in human somatic Wee1 that encompasses RxL1 also binds Crm1, directing Wee1 export from the nucleus. These studies have illuminated ...
  33. [33]
    Genome-wide characterization of the sunflower kinome - NIH
    Dec 2, 2024 · ... (Wee1, Wee2, and Myt1 kinases), and WNK (with no lysine-K) ... family that likely underwent numerous duplication events during evolution.
  34. [34]
    The G2 DNA damage checkpoint targets both Wee1 and Cdc25
    May 15, 2000 · We propose therefore that in fission yeast, the G2 DNA damage checkpoint promotes cell cycle delay through a double-lock mechanism, ...Results · Chk1 Overexpression In The... · Lack Of Checkpoint Control...
  35. [35]
    the checkpoint kinases ATR, CHK1 and WEE1 restrain CDK activity ...
    Sep 21, 2011 · Similarly to CHK1, WEE1 depletion causes increased ssDNA and RPA loading indicative of replication stress ( 26 ). Drug-based WEE1 inhibition ...
  36. [36]
    Wee1 and Chk1 – crosstalk between key players in replicative stress
    May 20, 2015 · Replicative stress is a tumor cell-associated feature that includes the accumulation of stalled or collapsed replication forks.
  37. [37]
    Modeling the fission yeast cell cycle: Quantized cycle times in wee1
    When MPF activity in the nucleus reaches a threshold value, it inactivates Wee1 (and Mik1) and activates Cdc25. As a result, preMPF is converted ...
  38. [38]
    Regulators of cyclin-dependent kinases are crucial for maintaining ...
    Mar 1, 2010 · WEE1 depletion inhibited the phosphorylation of CDK1 on Tyr15, which is consistent with the published function of WEE1 (Fig. S1 D; Coleman ...Missing: core | Show results with:core
  39. [39]
    Negative regulation of mitosis in fission yeast by the Shk1interacting ...
    ... protein truncated by the deletion of the N-terminal 118 amino acids of the full-length Shk1 protein. This N-terminal truncated Shk1 mutant is nonetheless ...<|separator|>
  40. [40]
    Epistatic gene interactions in the control of division in fission yeast
    May 1, 1979 · I report here that the mitotic defect caused by a defective cdc25 allele is suppressed in wee mutants. Suppression by wee1 mutants is almost complete.<|control11|><|separator|>
  41. [41]
    Expression of Arabidopsis WEE1 in tobacco induces unexpected ...
    Jun 18, 2019 · This phenotype is similar to expression of Spcdc25 and is consistent with a dominant negative effect on WEE1 action. Consistent with this ...Missing: localization | Show results with:localization
  42. [42]
    S-phase-specific activation of Cds1 kinase defines a subpathway of ...
    We show that the Cds1 kinase is required to slow S phase in the presence of DNA-damaging agents. Cds1 is phosphorylated and activated by S-phase arrest.
  43. [43]
    Forced Mitotic Entry: Therapeutic Strategy by WEE1 Inhibition
    We show that WEE1 inhibition forces S-phase–arrested cells directly into mitosis without completing DNA synthesis, resulting in highly abnormal mitoses ...
  44. [44]
    Identification of DNA methylation-regulated WEE1 with potential ...
    WEE1 was upregulated in LGG and glioblastoma (GBM), but it had a more significant prognostic impact in LGG compared to other cancers.
  45. [45]
    PKMYT1, exacerbating the progression of clear cell renal cell ...
    The relationships between clinical variables and the expression levels among WEE family kinases were evaluated in the TCGA dataset. Expression levels of WEE1 ...
  46. [46]
    WEE1 Inhibitor Adavosertib Exerts Antitumor Effects on Colorectal ...
    Sep 12, 2024 · Our findings indicate that WEE1 inhibitors can be used for treating CRC, particularly in cases with p53 mutations.<|control11|><|separator|>
  47. [47]
    Targeting WEE1 Inhibits Growth of Breast Cancer Cells That Are ...
    G1 checkpoint can be deregulated in cancer cells with p53 mutation and these cells show increased sensitivity to WEE1 inhibition by prematurely entering G2 and ...Missing: elevated | Show results with:elevated<|separator|>
  48. [48]
    Wee1 Kinase: A Potential Target to Overcome Tumor Resistance to ...
    Wee1 is the principal gatekeeper for both G2/M and S-phase checkpoints, where it plays a key role in cell cycle regulation and DNA damage repair.
  49. [49]
    Enhancement of chemosensitivity by WEE1 inhibition in EGFR-TKIs ...
    WEE1 kinase, a G2/M checkpoint regulator, was recently considered as a putative biomarker for the platinum-based chemo-response. The aim of this study is to ...
  50. [50]
    [PDF] Clinical efficacy and molecular response correlates of the WEE1 ...
    Aug 15, 2021 · Tumor immunostaining studies were undertaken to explore the effects of WEE1 inhibitor therapy on immune infiltration and DNA damage markers.
  51. [51]
    Researchers reveal how WEE1 drives cancer resistance ... - ecancer
    Jul 17, 2025 · The team found that WEE1 expression was significantly elevated in non-responders, correlating with poor prognosis, high tumour proliferation, ...
  52. [52]
    Cytoplasmic WEE1 Promotes Resistance to PD-1 Blockade Through ...
    WEE1 is transcriptionally upregulated by stemness factor NANOG and predominantly localized in the cytoplasm, not the nucleus, following AKT-dependent S642 ...Missing: thaliana | Show results with:thaliana
  53. [53]
    Targeting the DNA Damage Response for Cancer Therapy by ...
    As most cancer therapies aim to induce lethal amounts of DNA damage in cancer cells, Wee1 overexpression promotes cancer cell survival (and resistance) by ...Missing: enlarged MMS
  54. [54]
    WEE1 expression linked to poor survival in multiple myeloma
    Feb 20, 2025 · WEE1 inhibition has been shown to dysregulate the cellular machinery associated with the first stage of mitosis in the G1-S transition [22] and ...Results · Wee1 Expression Has... · Wee1-High Cohort Is 3.2×...
  55. [55]
    Wee1 Kinase: A Potential Target to Overcome Tumor Resistance to ...
    Wee1 kinase inhibition causes a significant reduction in phospho- CDK1 (Tyr15), thus promoting the accumulation of the active CDK1-cyclin B1 complex and ...
  56. [56]
    MYC and therapy resistance in cancer: risks and opportunities - PMC
    Its pharmacological inhibition revealed a noncanonical role in preventing MYC‐driven senescence [229] and mediating the cooperation between MYC and RAS ...
  57. [57]
    Inhibiting WEE1 Augments the Antitumor Efficacy of Cisplatin in ...
    May 25, 2023 · We confirmed that AZD-1775 combined with cisplatin reduced tumor volume and proliferation activity and increased the markers of cell apoptosis ...
  58. [58]
    (PDF) CRISPR Screening Identifies WEE1 as a Combination Target ...
    Jun 12, 2025 · In this study, we reported on a kinome CRISPR/Cas9 knockout screen that identified several G2–M checkpoint kinases, including WEE1, whose loss ...
  59. [59]
    Targeting WEE1 to Overcome ARID1A Mutation-Driven Osimertinib ...
    This study aimed to investigate the mechanisms driving osimertinib resistance and identify therapeutic strategies.
  60. [60]
    Small-molecule inhibition of Wee1 kinase by MK-1775 selectively ...
    Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumor cells to DNA-damaging agents.
  61. [61]
    Azenosertib Is a Potent and Selective WEE1 Kinase Inhibitor with ...
    These observations have made WEE1 a promising anticancer therapeutic target. Azenosertib (ZN-c3) is a novel, selective, and orally bioavailable WEE1 inhibitor.Missing: IC50 | Show results with:IC50
  62. [62]
    [PDF] Early safety and efficacy of APR-1051, a novel WEE1 inhibitor, in ...
    Oct 26, 2025 · ... APR-1051, a novel WEE1 inhibitor, in patients with cancer-associated gene alterations: Updated data from ACESOT-1051 phase 1 trial. B011.
  63. [63]
    [PDF] debio 0123 is a selective wee1 inhibitor that effectively penetrates ...
    Debio 0123 is an investigational, orally bioavailable, highly selective, adenosine triphosphate. (ATP)-competitive inhibitor of the WEE1 tyrosine kinase. WEE1 ...
  64. [64]
    Combined PARP and WEE1 inhibition triggers anti-tumor immune ...
    Aug 15, 2023 · We demonstrate that combined PARP and WEE1 inhibition are synergistic in controlling tumour growth in BRCA1/2 wild-type TNBC preclinical models.
  65. [65]
    Combined inhibition of Wee1 and Chk1 as a therapeutic strategy in ...
    Dec 5, 2023 · Figure 2 High CHK1 and WEE1 expression is associated with a poor outcome in MM. Correlation between CHK1 and WEE1 expression and overall ...Analysis Of Chk1 And Wee1... · Proliferation Assays And... · Results<|separator|>
  66. [66]
    Recent Advances of WEE1 Inhibitors and Statins in Cancers ... - NIH
    Oct 4, 2021 · In this review, we summarize recent advances with Wee1 inhibitors, statins, and mevalonate pathway inhibitors in cancers with p53 mutations.Wee1 Inhibitors · Azd1775 · Mutant P53 And Mevalonate...<|separator|>
  67. [67]
    The WEE1 inhibitor Debio 0123 is synergistic with the PKMYT1 ...
    The WEE1 inhibitor Debio 0123 is synergistic with the PKMYT1 inhibitor lunresertib in preclinical models of ovarian and breast cancer. April 28, 2025 ...Missing: PARP CHK1 statins
  68. [68]
    WEE1 inhibitor adavosertib in combination with carboplatin in ...
    Aug 9, 2025 · A phase II study of the WEE1 inhibitor, adavosertib, (AZD1775) showed an objective response rate of 29.4% in recurrent uterine serous ...
  69. [69]
  70. [70]
    A Phase II Trial of the WEE1 Inhibitor Adavosertib in SETD2-Altered ...
    Adavosertib failed to exhibit objective responses in SETD2-altered ccRCC and other solid tumor malignancies although prolonged SD was observed ...Missing: AZD1775 combos
  71. [71]
    Azenosertib Shows Promising Efficacy in Platinum-Resistant ...
    Feb 4, 2025 · The ZN-c3-001 and MAMMOTH trials included patients with ovarian cancer and other solid tumors. Two treatment-related grade 5 adverse effects ...Missing: ASCO MTD BID BRCA-<|separator|>
  72. [72]
    Azenosertib Is a Potent and Selective WEE1 Kinase Inhibitor with ...
    Aug 1, 2025 · Azenosertib (ZN-c3) is a novel, selective, and orally bioavailable WEE1 inhibitor. The antiproliferative activity of azenosertib on cancer ...Missing: IC50 PLK1
  73. [73]
    Phase 1 results of the WEE1 inhibitor, azenosertib, in combination ...
    May 29, 2024 · Azenosertib + gem was well tolerated at the MTD and provided a greater EFS than historical control cohorts of salvage therapy in pts with R/R osteosarcoma.
  74. [74]
    Aprea Therapeutics Provides Clinical Update from ACESOT-1051 ...
    Oct 24, 2025 · The latest results show that, at the 100 mg APR-1051 dose level, 3 out of 4 patients achieved stable disease, as measured using RECIST v1.1 ...
  75. [75]
    The WEE1 Inhibitor APR-1051 Shows Early Safety and Tolerability ...
    Oct 25, 2024 · Treatment with APR-1051 appears safe and well tolerated in patients with advanced solid tumors and select cancer-associated gene alterations.
  76. [76]
    Debio 0123, a highly selective WEE1 inhibitor in adult patients with ...
    During the recently completed dose escalation, Debio 0123 was given once daily over a 21-day cycle and had a manageable safety profile with dose proportional ...Missing: 120 glioblastoma<|separator|>
  77. [77]
  78. [78]
    Aprea Therapeutics Reports Second Quarter 2024 Financial Results ...
    Aug 12, 2024 · Enrollment commenced in the ACESOT-1051 Phase 1 trial evaluating APR-1051 – no myelosuppression observed in the first of eight planned ...
  79. [79]
    Genomic instability and CCNE1 amplification as emerging ...
    Aug 5, 2025 · Our real-world study supports the clinical utility of the GIM metric and the analytical validity of CCNE1 amplification, a new promising biomarker for ...