Skip to main content
Erschienen in: Journal of Hematology & Oncology 1/2020

Open Access 01.12.2020 | Review

A WEE1 family business: regulation of mitosis, cancer progression, and therapeutic target

verfasst von: Andrea Ghelli Luserna di Rorà, Claudio Cerchione, Giovanni Martinelli, Giorgia Simonetti

Erschienen in: Journal of Hematology & Oncology | Ausgabe 1/2020

Abstract

The inhibition of the DNA damage response (DDR) pathway in the treatment of cancer has recently gained interest, and different DDR inhibitors have been developed. Among them, the most promising ones target the WEE1 kinase family, which has a crucial role in cell cycle regulation and DNA damage identification and repair in both nonmalignant and cancer cells. 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, with a focus on their dual role as tumor suppressors in nonmalignant cells and pseudo-oncogenes in cancer cells. We here report the available data on the molecular and functional alterations of WEE1/PKMYT1 kinases in both hematological and solid tumors. Moreover, we summarize the preclinical information on 36 chemo/radiotherapy agents, and in particular their effect on cell cycle checkpoints and on the cellular WEE1/PKMYT1-dependent response. Finally, this review outlines the most important pre-clinical and clinical data available on the efficacy of WEE1/PKMYT1 inhibitors in monotherapy and in combination with chemo/radiotherapy agents or with other selective inhibitors currently used or under evaluation for the treatment of cancer patients.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ALL
Acute lymphoblastic leukemia
AML
Acute myeloid leukemia
BC
Breast cancer
C
Chemotherapy
CC
Colorectal cancer
CN
Copy number
CNA
Copy number alteration
CLL
Chronic lymphocyte leukemia
CML
Chronic myeloid leukemia
DDR
DNA damage response
DLBCL
Diffuse large B cell lymphoma
ES
Esophageal cancer
GC
Gastric cancer
GL
Gliomas
GLB
Glioblastoma
HC
Hepatocellular carcinoma
HR
Homologous recombination
HNSCC
Head and neck squamous cell carcinoma
LC
Lung cancer
M
Monotherapy
MaM
Malignant melanoma
MCL
Mantle cell lymphoma
MM
Multiple myeloma
MPF
Mitotic promoting factor
N
Neuroblastoma
NSCLC
Non-small-cell lung cancer
OC
Ovarian cancer
PC
Pancreatic cancer
R
Radiotherapy
S
Sarcomas
SAC
Spindle assembly checkpoint
TNBC
Triple-negative breast cancer

Background

The WEE1 kinase family consists of three serine/threonine kinases sharing conserved molecular structures and encoded by the following genes: WEE1 (WEE1 G2 Checkpoint Kinase), PKMYT1 (membrane-associated tyrosine- and threonine-specific cdc2-inhibitory kinase), and WEE1B (WEE2 oocyte meiosis inhibiting kinase). In eukaryotic somatic cells, WEE1 and PKMYT1 play a key role in cell cycle regulation and, in particular, they are involved in the entry into mitosis [1]. Their role as regulators is crucial during normal cell cycle progression and in response to DNA damages, as part of the DNA damage response (DDR) pathways. Similarly, WEE2 regulates cell cycle progression and, in particular, meiosis [2]. Briefly, WEE2 plays a dual regulatory role in oocyte meiosis by preventing premature restart prior to ovulation and permitting metaphase II exit at fertilization [3]. Despite the identification of WEE2 somatic mutations (1.9% of cases) and copy number (CN) alterations (22.5% of patients with CN loss and 22.5% with CN gain) across several cancer types (https://​portal.​gdc.​cancer.​gov), they have not been functionally linked to tumor development so far. Therefore, the following sections will be focused on WEE1 and PKMYT1 kinases that have a well-recognized role in oncology and hemato-oncology.

WEE1 and PKMYT1 in cell cycle regulation

WEE1 and PKMYT1 act as tumor suppressors in non-malignant eukaryotic somatic cells. Similarly to other DDR-related kinases, their main biological function is to prevent replication of cells with altered DNA. The main downstream target of WEE1 family kinases is the cyclin-dependent kinase 1 (CDK1)-cyclin B1 complex, also known as mitotic-promoting factor (MPF). WEE1 phosphorylates CDK1 on Tyr15 while PKMYT1 has a dual activity on Tyr15 and Thr14 [4] (Fig. 1a). The phosphorylation of those residues keeps the MPF complex inhibited until the cell approaches mitosis. WEE1 is located in the nucleus, while PKMYT1 is associated with the endoplasmic reticulum and Golgi apparatus [5, 6], and regulates Golgi membrane reassembly following mitosis [7]. Together, WEE1 and PKMYT1 ensure that CDK1 remains inactive as it shuttles into and out of the nucleus [8]. Through its extra-nuclear localization, PKMYT1 can also promote CDK1 cytosolic segregation. At the G2/M border, if no DNA damage has been detected, CDK1 phosphorylation on Tyr15 and Thr14 is rapidly removed by CDC25C phosphatase. In the nucleus, the CDK-activating kinase (CAK) complex composed by cyclin-dependent kinase 7 (CDK7), cyclin H1, and MAT1 promotes MPF complex activation through the phosphorylation of CDK1(Thr161) [9, 10]. The active MPF complex is then imported into the nucleus through phosphorylation of cyclin B1 (Ser126, Ser128, Ser133, and Ser147) [11]. This event is required to enter mitosis. The relevance of WEE1 and PKMYT1 regulation of CDK1 has been recently confirmed by in vivo studies. Indeed, the replacement of the CDK1 inhibitory phosphorylation sites with non-phosphorylatable amino acids (CDK1T14A/Y15F) was embryonic lethal in mice [12]. Once activated, the MPF complex can phosphorylate WEE1 and PKMYT1 to promote their inactivation via different cascades [5, 13, 14]. WEE1 is phosphorylated (Ser123) by CDK1 at the onset of mitosis, thereby generating a binding motif for polo like kinase 1 (PLK1) and casein kinase 2 (CK2), that in turn phosphorylate WEE1 (Ser53 and Ser121, respectively) [14, 15]. Together, the phosphorylation of the three Ser residues serves as a tag for the degradation of WEE1 by the ubiquitin ligase SCFβ-TrCP [13]. PKMYT1 is also phosphorylated by CDK1 and PLK1 and this event promotes its degradation [16]. In addition to the checkpoint function at the G2/M border, recent findings highlighted a role of WEE1 in the regulation of replication dynamics during S phase (intra S phase checkpoint). When cells reach the S phase, replication is initiated from a large number of replication origins triggered through the activation of the pre-replication complex [17] and following the activation of S phase specific CDK, primarily CDK2 [18, 19]. Similarly to CDK1, CDK2 regulation is controlled through Tyr15 phosphorylation status, that is balanced by WEE1 (Fig. 1a) and cell division cycle 25A (CDC25A) activity [20]. Both WEE1 and CDC25A/C have been shown to modulate unperturbed replication through regulating CDK1/CDK2 activity. Monoallelic expression of CDK1T14A/Y15F induced replication stress and S phase arrest in mouse embryonic fibroblasts (MEFs), with substantial increase of γH2AX levels, chromosomal fragmentation, and DDR activation, as a consequence of intra-S phase DNA damage [12]. Moreover, unscheduled origin firing due to loss of WEE1 leads to exhaustion of the replication protein A1 (RPA1) pool and, as a consequence, to death during DNA replication (replication catastrophe). The intra S phase activity of WEE1 is independent from PKMYT1 that is unable to phosphorylate CDK2 [5]. In addition, WEE1, but not PKMYT1, contributes to the control of mitosis exit. Indeed, Wee1-deficient MEFs showed mitotic defects (e.g., in the number and position of centrosomes) that induce arrest in mitosis or, in the majority of cells, mitotic slippage [21, 22]. At the end of mitosis, WEE1 inhibits CDK1 through phosphorylation of its Tyr15 residue (Fig. 1a). This event is dependent on the activation of the CTD phosphatase subunit 1 (FCP1) that dephosphorylates and activates WEE1 and other crucial component of the spindle assembly checkpoint (SAC) complex [23]. Although the precise mechanisms that regulate FCP1 activity is still unknown, it has been showed that FCP1 promotes the dephosphorylation of crucial SAC components, including cell division cycle 20 (CDC20) and ubiquitin specific peptidase 44 (USP44), thus promoting APC/CCdc20 activation and chromosome segregation [2426]. Moreover, WEE1 directly interacts with APC/C components, including fizzy and cell division cycle 20 related 1 (CDH1), CDC20, cell division cycle 27 (CDC27), and its deletion enforced APC/C activity, resulting in alterations of the level of APC/C substrates and mitosis progression at the expense of genomic stability [21].

WEE1 regulates replication forks and genome stability

The activity of WEE1 through the cell cycle can explain its tumor suppressor function, at least in nonmalignant cells. This observation was confirmed and disentangled in preclinical studies. Indeed, conditional Wee1 heterozygous deletion in the murine mammary epithelium caused enhanced proliferation, with cells progressing into mitosis while still undergoing DNA replication, and consequent accumulation of DNA damage, resulting in genomic instability and, ultimately, in tumor development [21]. Biological processes such as DNA replication and homologous recombination involve the formation of branched DNA structures that physically link chromosomes. Such DNA structures needs to be disengaged prior to entry into mitosis, in order to ensure proper chromosome segregation. Eukaryotic cells evolved different mechanisms to identify and process branched DNA structures (Y-shape DNA) and the most important one involves the structure-selective endonuclease MUS81. MUS81 forms heterodimeric complexes with the non-catalytic subunits EME1 or EME2 and recognizes Y-shape DNA structures during DNA replication or during mitosis (homologous recombination). The activity of MUS81-EME1/2 complex is crucial to recover stalled replication forks, during prolonged S phase arrest, and to reset DNA junction between twin chromatids during homologous recombination [27]. In unperturbed cells, WEE1 protects replication forks and prevents the generation DNA damages and chromosome pulverization through an indirect inhibition of MUS81 functionality [28]. Indeed, WEE1 phosphorylates CDK1 and CDK2, thus preventing the CDK-mediated phosphorylation and activation of MUS81-EME1/2 complexes [29]. Lack of WEE1-dependent regulation of MUS81-EME1/2 endonucleases may lead to cleavage of unwanted DNA structure (excessive replication forks), which would slow down replication progression and increase genomic instability [27, 28] (Fig. 1b).

WEE1 and PKMYT1 deregulation in cancer cells

WEE1 and PKMYT1 act like oncogenes

The biological role of WEE1 and PKMYT1 in cancer cells is not fully understood. Reduced WEE1 expression has been detected in breast cancer compared with normal tissues, independently of the tumor grade [21]. However, most findings suggest that both kinases act like oncogenes rather than tumor suppressors. Indeed, they are frequently overexpressed in both solid and hematological tumors and a genome-wide CRISPR screen of 563 cancer cell lines, showed that they are essential for the cell viability of almost all cell lines [30]. The dependency of cancer cells on WEE1 family proteins may be linked to the following mechanisms (Fig. 2): (i) the high proliferation rate of cancer cells that follows the activation of driver oncogenes (e.g. RAS, MYC) needs to be sustained by a strong cell cycle regulation machinery; (ii) cancer cells frequently inactivate p53, which is a key gatekeeper of G0/G1 and S phases and, as a consequence, the regulation of cell cycle is sustained entirely by the G2/M checkpoint; (iii) the over-expression of DDR-related kinases is fundamental to maintain a tolerable level of genetic instability, an intrinsic feature of cancer cells [31, 32]. Therefore, we can speculate that, once the malignant transformation process has been induced, WEE1 upregulation exerts a pro-tumorigenic functions by securing a tolerable level of genomic instability to cancer cells. The following sections summarize the current knowledge on the molecular and functional alterations of WEE1 and PKMYT1 in hematological and solid tumors.

WEE1 and PKMYT1 genetic lesions in cancer

WEE1 and PKMYT1 are rarely mutated in cancer patients, with an overall mutation frequency of 1.2% and 0.2%, respectively (https://​portal.​gdc.​cancer.​gov). The distribution of somatic mutations is highly heterogeneous across cancer types (WEE1: 0.2–7.6%; PKMYT1:0.1–3.6%), with a higher frequency in uterine corpus endometrial carcinoma (UCEC) and tumors of the gastrointestinal tract (stomach and colon adenocarcinoma, Fig. 3a, b). In particular, WEE1 mutations have been reported in 7.6% of UCEC cases. Moreover, PKMYT1 lesions have been also detected in 2.7% of diffuse large B cell lymphoma (DLBC). Conversely, both kinase genes are rarely mutated in brain lower grade glioma (LGG), ovarian serous cystadenocarcinoma (OV), prostate adenocarcinoma (PRAD), and sarcoma (SARC), with a frequency lower than 0.5%. Both genes are mainly targeted by missense mutations that preferentially cluster in the region encoding the WEE1 kinase domain and its surroundings (Fig. 3c), suggesting a potential gain of function effect of the kinase activity. Conversely, the mutations are scattered throughout the PKMYT1 sequence (Fig. 3d). Little is known about the functional consequences of WEE1 and PKMYT1 mutations. In the majority of cancer types, the transcript expression in the mutated cases is higher than the median value of the entire cohort (https://​www.​cbioportal.​org), supporting once more an oncogenic function. In pancreatic adenocarcinoma (PA) patients and cell lines, an insertion was identified in the WEE1 poly-T track, which contains the binding site of the HuR RNA binding protein [33]. The insertion resulted in decreased WEE1 expression upon mitomycin-induced DNA damage, which would argue against a protective effect of the mutation. Copy number alterations (CNAs) represent a more frequent event compared with mutations, with the WEE1 gene being predominantly involved in CN loss (23.7% of cases versus 7.8% of patients with CN gains), while PKMYT1 showing a higher percentage of CN gain (15.9% versus 12.0% of CN loss, Fig. 3e, f). The predominance of WEE1 deletion events (6.3% versus 3.25% of cases with amplification) was also observed in breast cancer, in line with its reduced expression, as mentioned above [21]. Overall, cancer types showing the highest recurrence (> 10%) of CNAs were OV (27.7%), lung squamous cell carcinoma (LUSC, 14.8%), uterine carcinosarcoma (UCS, 12.5%), and SARC (11.2%) for WEE1 and OV (18.8%), bladder urothelial carcinoma (BLCA, 13.7%), and esophageal carcinoma (ESCA, 10.3%) for PKMYT1. Of note, OV and LUSC have been classified as tumors with multiple recurrent chromosomal gains and losses [34], which may suggest a bystander effect related to chromosomal instability in these tumor types, especially in the case of WEE1 deletion, that is unexpected, based on the general oncogenic function exerted by the kinase.

WEE1 and PKMYT1 functional role in hematological and solid tumors

Few studies have analyzed WEE1 and PKMYT1 expression in hematological malignancies. Our group showed that WEE1 kinase is highly expressed in acute lymphoblastic leukemia (ALL) cell lines and primary cells in comparison with normal hematopoietic cells, and that PKMYT1 is upregulated in relapsed ALL samples compared with nonmalignant hematopoietic cells [35]. Moreover, we demonstrated that ALL cells are dependent on WEE1 functionality for their survival and proliferation and that PKMYT1 levels may influence the sensitivity to the WEE1 inhibitor AZD-1775 [35]. Similar results on the role of WEE1 were obtained in multiple myeloma (MM), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), and chronic lymphocyte leukemia (CLL) [3639]. In AML cells, WEE1 and PKMYT1 are key gene discriminating between FLT3-ITD, FLT3-TKD, and NRAS-mutated samples. They were expressed at lower levels selectively in FLT3-ITD specimens in comparison with wild-type cells, suggesting either a tumor suppressor role in the leukemogenic process or a potential vulnerability n this AML subtype [40]. Pharmacological WEE1 inhibition alone or in combination with histone deacetylase inhibitors showed therapeutic potential in FLT3-ITD AML, confirming their dependency on WEE1 activity [41]. Since FLT3-ITD AML have intrinsic homologous recombination repair defects [42]. WEE1 inhibition may exacerbate the cell genotoxic stress by disrupting multiple cell cycle checkpoints. WEE1 has been showed to be a valuable target also for lymphoma patients [43]. In parallel, PKMYT1 proved to be essential for MM cell line viability, since its downregulation strongly decreased cell growth, while inducing apoptosis [44].
WEE1 and PKMYT1 are also over-expressed in solid tumors, including hepatocellular carcinoma, colon cancer, glioblastoma, non-small-cell lung cancer (NSCLS), neuroblastoma, and gastric cancers [31, 32, 4547]. High WEE1 expression has been associated with negative prognostic factors including lymph node involvement, induction of metastasis, increased biomarkers of proliferation (CCND1, Ki67, or CCNA1) and resistance to treatments (radiotherapy or chemotherapy) [4851]. Elevated PKMYT1 levels have been associated with tumor progression, a more aggressive disease, the induction of metastasis at least in NSCLS patients [45] and, generally, with poor prognosis. Depending on the cancer subtype, the expression of WEE1 and PKMYT1 has been linked with the activation of cellular pathways crucial for the specific disease. In melanoma cells, WEE1 silencing caused an increase of phospho p38 protein levels, indicating a role in the regulation of p38/MAPK pathway activation during p53-independent DNA damage response [49]. In hepatocellular carcinoma and colorectal cancers, PKMYT1 regulates epithelial-mesenchymal transition (EMT), a process relevant to tumor progression, invasion, metastasis, and drug resistance, through the activation of the beta-catenin/TCF signaling [32, 46], while PKMYT1 has been reported to control Notch pathway in NSCLC [45]. In particular, crucial component of the pathway, including NOTCH1, p21, and HES1 are downregulated by chemical inhibition of PKMYT1 [45]. In neuroblastic tumors, PKMYT1 is required to stabilize MYCN protein, which is a crucial proto-oncogene for this cancer types [52]. Moreover, in esophageal squamous cell carcinoma (ESCC) cell lines and primary cells, the expression of PKMYT1 is associated with and regulates the activation of the AKY/mTOR pathway [53] (Table 1). Taken together, this evidence suggests a broad role of WEE1/PKMYT1 besides the DNA damage response pathway that may increase the interest towards its therapeutic targeting.
Table 1
WEE1 and PKMYT1 molecular alterations in hematological and solid tumors according to literature
Gene
Genetic alteration
Disease
Effect/prognostic value
Reference
Hematological tumors
WEE1
Over-expression
ALL; AML; MM; CML; CLL; DLBCL
Crucial for cell viability of cancer cells (experimentally proven).
[3540, 43, 54]
Copy number Gain
AML
Biological effect or prognostic value unknown
[55]
PKMYT1
Over-expression
ALL; MM
Crucial for cell viability of cancer cells (experimentally proven).
[35, 44]
Solid tumors
WEE1
Over-expression
GC; MaM; GL; OC; CC
Associated with lymph node involvement, induction of metastasis, increased biomarkers of proliferation (CCND1, Ki67 or CCNA1), resistance to treatment and poor overall survival.
[4851, 5660]
Mutation
PA
Insertion causing decrease WEE1 expression upon DNA damage
[33]
PKMYT1
Over-expression
HC; CC; GLB; NSCLC; N; GS
Associated with tumor progression, aggressive disease and poor overall survival.
[31, 32, 4547]
Mutation
N
Biological effect or prognostic value unknown
[61]
ALL acute lymphoblastic leukemia, AML acute myeloid leukemia, MM multiple myeloma, CML chronic myeloid leukemia, CLL chronic lymphocyte leukemia, DLBCL diffuse large B cell lymphoma, GC gastric cancer, MaM malignant melanoma, GL gliomas, OC ovarian cancer, CC colorectal cancer, PA pancreatic adenocarcinoma, HC hepatocellular carcinoma, GLB glioblastoma, NSCLC non-small-cell lung cancer, N neuroblastoma

Development of WEE1 and PKMYT1 inhibitors

WEE1 and PKMYT1 inhibitors have single agent and chemo-sensitizer effects

Due to their potential oncogenic role, WEE1 and PKMYT1 have been investigated as therapeutic targets for hematological and solid tumors. Several pharmacological inhibitors have been designed and subsequently validated in different cancer models. The available literature highlights a common mechanism of action of WEE1/PKMYT1 inhibitors in cancer cells either in single agent or in combination with DNA damaging agents (chemotherapy/radiotherapy). WEE1/PKMYT1 kinase inhibition causes G2/M cell cycle checkpoint override, premature mitotic entry, and cell death during mitosis, through a mechanism generally known as mitotic catastrophe (Fig. 4a). From a biological point of view, the inhibition of WEE1 kinase causes a significant reduction of phospho-CDK1 (Tyr15), thus promoting the accumulation of active CDK1-cyclin B1 complex and, consequently, mitotic entry. The beginning of mitosis is also associated with a progressive accumulation of DNA damages and the degeneration in mitotic catastrophe. The sensitivity to WEE1 kinase inhibitors in relation to TP53 mutational status remains controversial. Indeed, some studies reported increased sensitivity of TP53 mutant cell lines to WEE1 inhibitors in comparison to TP53 wild-type ones [62, 63], while others showed no association between p53 functionality and the effectiveness of WEE1 inhibition [35, 64]. These discrepancies may be linked to the intrinsic chromosomal instability of the analyzed tumors and to additional alterations deregulating the G1 checkpoint in TP53 wild-type cases that may enhance the sensitivity to WEE1 targeting.
Regarding the role of WEE1 inhibitors as chemo-sensitizer agents, a large number of studies demonstrated a synergistic activity between DNA damaging agents (chemotherapy including doxorubicin, cytarabine, methotrexate, cisplatin, clofarabine, etoposide, 5-fluorouracil, and radiotherapy) and different WEE1/PKMYT1 inhibitors in preclinical models [48, 56, 6569]. The mechanism of action of the combination is based on the inhibition of the DDR pathway following induction of DNA damage induced by the chemotherapy or radiotherapy agents. In this scenario, cancer cells with damaged DNA fail to arrest cell cycle, continue to proliferate, and accumulate massive DNA damage until a point of no return (Fig. 4b). Indeed, several DNA damaging agent promote the indirect activation of WEE1 and PKMYT1 kinases, as showed mostly by the activation of cell cycle checkpoints (S and G2/M checkpoints) in cancer cells. We summarized in Table 2 the results of preclinical studies in which the effect of different chemotherapy agents or radiotherapy has been evaluated in terms of cell cycle perturbation and altered expression of WEE1 or PKMYT1 following in vitro or in vivo treatment. Taken together, the abovementioned data prove that WEE1 and PKYMYT1 are ideal targets to override cell cycle checkpoint regulation and to improve the efficacy of DNA-damaging agents. In particular, tumors with a high level of chromosomal instability may respond to WEE1/PKMYT1 inhibition per se, while cases with a more stable genomic asset may benefit of the combination between DNA-damaging agents and WEE1 family kinase inhibitors. The following sections reports the main preclinical and clinical findings obtained using small molecules inhibitors of WEE1 and PKMYT1 kinases.
Table 2
Effects of standard of care chemo/radiotherapy agents on cell cycle checkpoints activation
Chemotherapy agents/radiotherapy
Intra S checkpoint
G2/M checkpoint
WEE1 and/or PKMYT1 experimentally proven involvement in cancer model
Actinomycin
No
Yes [70, 71]
WEE1 upregulation [71]
Azacitidine
No
Yes [72]
NA
Bleomycin
No
Yes [73]
NA
Carboplatin
No
Yes [74]
NA
Cisplatin
No
Yes [75, 76]
WEE1 inhibition enhanced cytotoxicity [76, 77]
Cyclophosphamide
NA
NA
WEE1 upregulation [78]
Cytarabine
Yes [79, 80]
Yes [79, 80]
WEE1 upregulation [38, 81]
Clofarabine
Yes [35]
No
WEE1 inhibition enhanced cytotoxicity [35]
Daunorubicin
Yes [82]
Yes [82]
NA
Decitabine
No
Yes [83]
NA
Docetaxel
No
Yes [84, 85]
NA
Doxorubicin
No
Yes [86]
WEE1 upregulation [86]; WEE1 inhibition enhanced cytotoxicity [35]
Epirubicin
No
Yes [87, 88]
WEE1 inhibition enhanced cytotoxicity [89]
Epothilone
No
Yes [90]
NA
Etoposide
No
Yes [91]
WEE1 inhibition enhanced cytotoxicity [67]
Fluorouracil
Yes [92]
No
WEE1 inhibition enhanced cytotoxicity [92]
Fludarabine
Yes [80]
No
NA
Gemcitabine
Yes [80]
No
WEE1 upregulation [93]; WEE1 inhibition enhanced cytotoxicity [94]
Hydroxyurea
Yes [95]
No
WEE1 inhibition enhanced cytotoxicity [96]
Idarubicin
No
Yes [97]
NA
Irinotecan
No
Yes [98]
WEE1 inhibition enhanced cytotoxicity [99]
Mechlorethamine
Yes [100]
Yes [100]
NA
Mercaptopurine
Yes [101]
No
NA
Methotrexate
Yes [102]
No
WEE1 inhibition enhanced cytotoxicity [103]
Mitoxantrone
No
Yes [104]
WEE1 inhibition enhanced cytotoxicity [78]
Oxaliplatin
No
Yes [105]
WEE1 inhibition enhanced cytotoxicity [106]
Paclitaxel
No
Yes [107]
WEE1 inhibition enhanced cytotoxicity [108]
Pemetrexed
Yes [109]
No
WEE1 inhibition enhanced cytotoxicity [110]
Radiotherapy (ionizing radiation)
No
Yes [111]
WEE1 inhibition enhanced cytotoxicity [68]
Teniposide
Yes [112]
Yes [112]
NA
Thioguanine
Yes [113]
Yes [113]
WEE1 inhibition enhanced cytotoxicity [65]
Topotecan
No
Yes [114]
WEE1 inhibition enhanced cytotoxicity [115]
Vinblastine
No
Yes [116]
NA
Vincristine
No
Yes [117]
WEE1 inhibition enhanced cytotoxicity [118]

Preclinical studies of WEE1 and PKMYT1 inhibitors

Several targeted compounds showed an inhibitory activity on WEE1 and PKMYT1 kinases and their efficacy was proven in a number of tumor types. Table 3 shows the main preclinical studies that used WEE1/PKMYT1 inhibitors in single agent or in combination with chemo/radiotherapy agents in different tumor types.
Table 3
Preclinical studies evaluating the effect of WEE1 inhibitors in monotherapy or in combination with chemotherapy/radiotherapy in cancer
Inhibitor
Treatment
Cancer model
Main biological effect
References
PD0166285
M
GBM-astrocytoma
-G2/M checkpoint override
-Forced mitotic entry
[51]
Adavosertib
M
MM, ALL, AML TNBC, DLBCL, MCL
-G2/M checkpoint override
-Forced mitotic entry
-Mitotic catastrophe
-Replicative catastrophe
[35, 99, 119122]
PD0166285
+R
GBM-astrocytoma
-Mitotic catastrophe
-Inhibition of DNA repair
[51]
Adavosertib
+R
CC, LC, BC, PC, OC, DLBCL, ES
-Increased DNA damage
-Induction of apoptosis
-Mitotic catastrophe
[78, 99, 123126]
Adavosertib
+C
AML, ALL, MM, BC, CC, GC, DLBCL
-S or G2/M checkpoint override
-Increased DNA damaged
-Induction of apoptosis
[35, 37, 38, 76, 92, 99, 121, 127129]
Adavosertib
+HDAC i
AML, HNSCC
-Replication stress
-Replicative catastrophe
-Increased DNA damage
-Inhibition of DNA repair
[41, 130, 131]
Adavosertib
+ATR i
AML, DLBCL, MCL, BC
-Replication stress
-Replicative catastrophe
-Increased DNA damaged
-Inhibition of DNA repair
[132135]
Adavosertib
+mTOR i
AML, ALL, OC, NSCLC
-Inhibition of DNA repair
[136139]
Adavosertib
+CHK1 i
MCL, DLBCL, ALL, AML
-Replication stress
-Increased DNA damage
-Replicative catastrophe
[103, 140142]
Adavosertib
+BCL2i/MCL-1 i
DLBCL
-Force mitotic entry
-Increase DNA damage
-INDUCTION of apoptosis
[143]
Adavosertib
+PARP1 i
NSCLC, AML, ALL
-G2/M checkpoint override
-Replication stress
-Increased DNA damage
-Inhibition of DNA repair
[126, 144146]
Adavosertib
+AURORA A i
HNSCC
-Forced mitotic entry
-Mitotic catastrophe
[147]
Adavosertib
+CDK2 i
BC
-Replication stress
-Replicative catastrophe
[89]
Adavosertib
+SIRT1 i
LC
-Inhibition of DNA repair
[148]
Adavosertib
+CDK4-6 i
S
-Replication stress
[149]
Adavosertib
+BCR-ABL1 i
ALL
-Inhibition of DNA repair -G2/M checkpoint override
[35]
Adavosertib
+Proteasome i
MM
-G2/M checkpoint override
-Forced mitotic entry
-Inhibition of DNA repair
[36]
Adavosertib
+BET i
NSCLC
-Inhibition of DNA repair
-Forced mitotic entry
-Mitotic catastrophe
[150]
M monotherapy, R radiotherapy, C chemotherapy, ALL acute lymphoblastic leukemia, AML acute myeloid leukemia, MM multiple myeloma, DLBCL diffuse large B cell lymphoma, MCL mantle cell lymphoma, GC gastric cancer, GL gliomas, OC ovarian cancer, CC colorectal cancer, PC pancreatic cancer, ES esophageal cancer, HC hepatocellular carcinoma, GLB glioblastoma, NSCLC non-small-cell lung cancer, N neuroblastoma, S sarcomas, LC lung cancer, BC breast cancer, HNSCC head and neck squamous cell carcinoma, TNBC triple negative breast cancer
PD0166285 is the first reported drug, with an inhibitory activity against WEE1, PKMYT1, and a range of other kinases including c-Src, EGFR, FGFR1, CHK1, and PDGFRb [151].
Adavosertib (AZD-1775) is the first highly potent and selective WEE1 inhibitor. A large number of preclinical studies evaluated its efficacy in single agent and in combinatory approaches. Regarding the mechanism of action, adavosertib induces S and/or G2/M cell cycle checkpoints override, depending on cancer types, when used in monotherapy. Cell cycle perturbation is associated with a progressive accumulation of DNA damages and by the induction of apoptosis [35, 99, 119122]. This last event is cell cycle phase-dependent and can occur (i) as a consequence of S phase checkpoint override, when cancer cells start DNA replication even in the presence of DNA damages (replicative catastrophe); (ii) following G2/M phase checkpoint override, that results in forced entry into mitosis, even in the presence of DNA damages (mitotic catastrophe).
In combination strategies, adavosertib was able to enhance the cytotoxicity of chemo/radiotherapy agents, by inducing cell cycle checkpoint override, inhibition of DNA damage repair, and induction of apoptosis [35, 37, 38, 92, 121, 127129]. The chemo-sensitizer efficacy of DDR inhibitors has been linked to drug scheduling [94, 152, 153]. Recently in pancreatic adenocarcinoma cells, it has been reported that the efficacy of a triple regimen combining gemcitabine, CHK1, and WEE1 inhibitors is strictly dependent on the timing of drug administration. Indeed, the maximum effect of the combination is obtained when gemcitabine and CHK1 inhibitors are administered simultaneously (thus inducing replicative stress) and adavosertib is added at a later time [94].
Moreover, strong synergism has been observed by combining adavosertib with small molecules, including DDR-related inhibitors (CDK2 [89], CDK4-6 [149], CHK1 [103, 140142], ATM [132135], AURORA A [147], PARP1 [144], SIRT1 [148] inhibitors), histone deacetylase (HDAC) inhibitors [41, 130, 131], tyrosine kinase inhibitors (BCR-ABL1 inhibitors [35]), anti-apoptotic protein inhibitors (BCL2 and MCL1 inhibitors [143]), mTOR inhibitor [136139], and proteasome inhibitors [36].
We have recently reported synergistic effects of adavosertib in combination with different tyrosine kinase inhibitors in both BCR-ABL1-positive and -negative ALL cell lines and primary cells. Interestingly, strong synergism was found in BCR-ABL1-negative ALL cell lines treated with adavosertib in combination with bosutinib isomer. In the study, we speculated that the strong cytotoxic effect of the combination was due to the concomitant inhibition of WEE1 and PKMYT1 kinases [35]. Indeed, no selective inhibitor has been currently developed to target its functionality. However, several known tyrosine kinase inhibitors have an inhibitory off-target effect on PKMYT1. Among them, compounds commonly used for the treatment of BCR-ABL1-positive CML and ALL, as dasatinib and bosutinib (and a structural isomer of bosutinib [154, 155]) were shown to inhibit PKMYT1 activity.
Overall, the data suggest that WEE1/PKMYT1 inhibition is a suitable pharmacological target for combination strategies in cancer. The broad spectrum of activities exerted by the two kinases, and especially by WEE1, across the cell cycle, makes them good candidates for a number of diverse therapeutic combinations.

WEE1 inhibitors from bench to bedside

Several clinical studies are currently evaluating the efficacy of adavosertib on different aggressive and advanced tumors (Table 4).
Table 4
Clinical trials evaluating WEE1/PKMYT1 inhibitor in monotherapy or in combination for cancer therapy
Study ID
Study title
Tumor
Interventions
Status
Phase
NCT02610075
Phase Ib Study to Determine MTD of AZD1775 Monotherapy in Patients With Locally Advanced or Metastatic Solid Tumours.
S
AZD1775
C
1
NCT03668340
AZD1775 in Women With Recurrent or Persistent Uterine Serous Carcinoma
S
AZD1775
R
2
NCT02482311
Safety, Tolerance, PK, and Anti-tumour Activity of AZD1775 Monotherapy in Patients With Advanced Solid Tumours
S
AZD 1775
C
1
NCT02207010
A Phase 0 Study of AZD1775 in Recurrent GBM Patients
S
AZD1775
NA
1
NCT03315091
Phase I Study to Assess the Effect of Food on AZD1775 Pharmacokinetics in Patients With Advanced Solid Tumours
S
AZD1775
C
1
NCT01748825
AZD1775 for Advanced Solid Tumors
S/H
AZD1775
ANR
1
NCT02511795
AZD1775 Combined With Olaparib in Patients With Refractory Solid Tumors
S
AZD1775 + Olaparib
C
1
NCT03313557
AZD1775 Continued Access Study to Assess Safety and Tolerability for Patients Enrolled in AZD1775 Clinical Pharmacology Studies
S
AZD1775
C
1
NCT02593019
Phase II, Single-arm Study of AZD1775 Monotherapy in Relapsed Small Cell Lung Cancer Patients
S
AZD1775
NA
2
NCT02688907
Phase II, Single-arm Study of AZD1775 Monotherapy in Relapsed Small Cell Lung Cancer Patients With MYC Family Amplification or CDKN2A Mutation Combined With TP53 Mutation
S
AZD1775
T
2
NCT02087176
A Placebo Controlled Study Comparing AZD1775 + Docetaxel Versus Placebo + Docetaxel to Treat Lung Cancer
S
AZD1775 + Docetaxel
T
2
NCT03012477
CISPLATIN + AZD-1775 In Breast Cancer
S
AZD1775 + Cisplatin
ANR
2
NCT02341456
Phase Ib Study AZD1775 in Combination With Carboplatin and Paclitaxel in Adult Asian Patients With Solid Tumours
S
AZD1775 + Carboplatin or Paclitaxel
C
1
NCT02791919
Wee1 Kinase Inhibitor AZD1775 and Combination Chemotherapy in Treating Children, Adolescents and Young Adults With Relapsed or Refractory Acute Myeloid Leukemia
H
AZD1775 + Cytarabine or Filgrastim or Fludarabine Phosphate
W
1
NCT02513563
AZD1775 Plus Carboplatin-Paclitaxel in Squamous Cell Lung Cancer
S
AZD1775 + Carboplatin or Paclitaxel
R
2
NCT03718143
AZD1775 in Advanced Acute Myeloid Leukemia, Myelodysplastic Syndrome and Myelofibrosis
H
AZD1775 + Cytarabine
T
2
NCT02585973
Dose-escalating AZD1775 + Concurrent Radiation + Cisplatin for Intermediate/High Risk HNSCC
S
AZD1775 + Cisplatin + Radiation
R
1
NCT02087241
Ph II Trial of Carboplatin and Pemetrexed With or Without AZD1775 for Untreated Lung Cancer
S
AZD1775 + pemetrexed or carboplatin
T
2
NCT02381548
Phase I Trial of AZD1775 and Belinostat in Treating Patients With Relapsed or Refractory Myeloid Malignancies or Untreated Acute Myeloid Leukemia
H
AZD1775 + Belinostat
T
1
NCT03333824
Effects of AZD1775 on the PK Substrates for CYP3A, CYP2C19, CYP1A2 and on QT Interval in Patients With Advanced Cancer
S
AZD1775
C
1
NCT02906059
Study of Irinotecan and AZD1775, a Selective Wee 1 Inhibitor, in RAS or BRAF Mutated, Second-line Metastatic Colorectal Cancer
S
AZD1775 + Irinotecan
R
1
NCT02037230
Dose Escalation Trial of AZD1775 and Gemcitabine (+Radiation) for Unresectable Adenocarcinoma of the Pancreas
S
AZD1775 + Gemcitabine+ Radiation Therapy
C
1,2
NCT02617277
Safety, Tolerability and Pharmacokinetics of AZD1775 (Adavosertib) Plus MEDI4736 (Durvalumab) in Patients With Advanced Solid Tumours
S
AZD1775 + Durvalumab
ANR
1
NCT02666950
WEE1 Inhibitor AZD1775 With or Without Cytarabine in Treating Patients With Advanced Acute Myeloid Leukemia or Myelodysplastic Syndrome
H
AZD1775 + Cytarabine
C
2
NCT01047007
A Dose Escalation Study of MK1775 in Combination With 5-FU or 5-FU/CDDP in Patients With Advanced Solid Tumor (1775-005)
S
AZD1775 + 5-FU or 5-FU/CDDP
T
1
NCT01164995
Study With Wee-1 Inhibitor MK-1775 and Carboplatin to Treat p53 Mutated Refractory and Resistant Ovarian Cancer
S
AZD1775 + carboplatin
NA
2
NCT02448329
Study of AZD1775 in Combination With Paclitaxel, in Advanced Gastric Adenocarcinoma Patients Harboring TP53 Mutation as a Second-line Chemotherapy
S
AZD1775 + paclitaxel
R
2
NCT02508246
WEE1 Inhibitor MK-1775, Docetaxel, and Cisplatin Before Surgery in Treating Patients With Borderline Resectable Stage III-IVB Squamous Cell Carcinoma of the Head and Neck
S
AZD1775 + Cisplatin + Docetaxel
C
1
NCT03253679
AZD1775 in Treating Patients With Advanced Refractory Solid Tumors With CCNE1 Amplification
S
AZD1775
R
2
NCT01076400
A Study of MK-1775 in Combination With Topotecan/Cisplatin in Participants With Cervical Cancer (MK-1775-008)
S
AZD1775 + Topotecan or Cisplatin
T
1,2
NCT02196168
Cisplatin With or Without WEE1 Inhibitor MK-1775 in Treating Patients With Recurrent or Metastatic Head and Neck Cancer
S
AZD1775 +Cisplatin
T
2
NCT02101775
Gemcitabine Hydrochloride With or Without WEE1 Inhibitor MK-1775 in Treating Patients With Recurrent Ovarian, Primary Peritoneal, or Fallopian Tube Cancer
S
AZD1775 + Gemcitabine
ANR
2
NCT03028766
WEE1 Inhibitor With Cisplatin and Radiotherapy: A Trial in Head and Neck Cancer
S
AZD1775 + Cisplatin + Radio therapy
ANR
1
NCT01357161
A Study of MK-1775 in Combination With Paclitaxel and Carboplatin Versus Paclitaxel and Carboplatin Alone for Participants With Platinum-Sensitive Ovarian Tumors With the P53 Gene Mutation (MK-1775-004)
S
AZD1775 + paclitaxel + carboplation
C
2
NCT03284385
Testing AZD1775 in Advanced Solid Tumors That Have a Mutation Called SETD2
S
AZD1775
R
2
NCT00648648
A Dose Escalation Study of MK-1775 in Combination With Either Gemcitabine, Cisplatin, or Carboplatin in Adults With Advanced Solid Tumors (MK-1775-001)
S
AZD1775 + Gemcitabine or Cisplatin or Carboplatin
C
1
NCT02194829
Paclitaxel Albumin-Stabilized Nanoparticle Formulation and Gemcitabine Hydrochloride With or Without WEE1 Inhibitor MK-1775 in Treating Patients With Previously Untreated Pancreatic Cancer That Is Metastatic or Cannot Be Removed by Surgery
S
AZD-1775 + Gemcitabine + paclitaxel
ANR
1,2
NCT02576444
Olaparib Combinations
S
AZD1775 + olaparib
ANR
2
NCT04197713
Testing the Sequential Combination of the Anti-cancer Drugs Olaparib Followed by Adavosertib (AZD1775) in Patients With Advanced Solid Tumors With Selected Mutations and PARP Resistance, STAR Study
S
AZD1775 + olaparib
ANR
1
NCT01922076
Adavosertib and Local Radiation Therapy in Treating Children With Newly Diagnosed Diffuse Intrinsic Pontine Gliomas
S
AZD1775 + Radiation Therapy
ANR
1
NCT03579316
Adavosertib With or Without Olaparib in Treating Patients With Recurrent Ovarian, Primary Peritoneal, or Fallopian Tube Cancer
S
AZD1775 + olaparib
R
2
NCT02095132
Adavosertib and Irinotecan Hydrochloride in Treating Younger Patients With Relapsed or Refractory Solid Tumors
S
AZD1775 + Irinotecan or Irinotecan Hydrochloride
R
1,2
NCT03345784
Adavosertib, External Beam Radiation Therapy, and Cisplatin in Treating Patients With Cervical, Vaginal, or Uterine Cancer
S
AZD1775 +Cisplatin + Radiation (External Beam Radiation Therapy)
R
1
NCT01849146
Adavosertib, Radiation Therapy, and Temozolomide in Treating Patients With Newly Diagnosed or Recurrent Glioblastoma
S
AZD1775 + Radiation Therapy + Temozolomide
R
1
NCT02937818
A Phase II, Study to Determine the Preliminary Efficacy of Novel Combinations of Treatment in Patients With Platinum Refractory Extensive-Stage Small-Cell Lung Cancer
S
AZD1775 + carboplatin
ANR
2
NCT02546661
Open-Label, Randomised, Multi-Drug, Biomarker-Directed, Phase 1b Study in Pts w/ Muscle Invasive Bladder Cancer
S
AZD1775 + Durvalumab
ANR
1
NCT02659241
Adavosertib Before Surgery in Treating Patients With Advanced High Grade Ovarian, Fallopian Tube, or Primary Peritoneal Cancer
S
AZD1775
R
1
NCT02272790
Adavosertib Plus Chemotherapy in Platinum-Resistant Epithelial Ovarian, Fallopian Tube, or Primary Peritoneal Cancer
S
AZD1775 + Paclitaxel or Carboplatin or Gemcitabine or pegylated liposomal doxorubicin
ANR
2
NCT02813135
European Proof-of-Concept Therapeutic Stratification Trial of Molecular Anomalies in Relapsed or Refractory Tumors
S/H
AZD1775 + carboplatin
R
1,2
NCT03330847
To Assess Safety and Efficacy of Agents Targeting DNA Damage Repair With Olaparib Versus Olaparib Monotherapy.
S
AZD1775 + olaparib
R
2
NCT01827384
MPACT Study to Compare Effects of Targeted Drugs on Tumor Gene Variations
S
AZD1775 + carboplatin
R
2
NCT02465060
Targeted Therapy Directed by Genetic Testing in Treating Patients With Advanced Refractory Solid Tumors, Lymphomas, or Multiple Myeloma (The MATCH Screening Trial)
S/H
AZD1775
R
2
S solid tumor, H hematological tumor, C completed, R recruiting, W withdraw, ANR active not recruiting, T terminated, NA status unknown (last update 04/22/2020)
The results of phase I trials showed that adavosertib is well tolerated both in single agent and in combination. Depending on the study, the maximum tolerated dose (MTD) was established between 150 and 225 mg orally twice per day for 2.5 days per 2 weeks [156158]. The most common adverse events reported in the abovementioned studies were fatigue, nausea, vomiting, diarrhea, and hematologic toxicity. Moreover, correlative studies performed on tumor biopsies confirmed in vivo the mechanism of action of adavosertib. Indeed, immunohistochemistry analyses showed a reduction of phospho-CDK1 (Tyr15) and an increase of DNA damages (phospho-γH2AX) in cancer cells [156, 157].
The phase II studies confirmed that adavosertib sensitizes cancer patients to different chemotherapy agents. Interestingly, adavosertib showed efficacy when combined with carboplatin in TP53-mutated ovarian cancer patients, refractory or resistant to first-line platinum-based chemotherapy [159]. Similar results were reported in platinum-resistant primary ovarian cancer patients after treatment with the combination of adavosertib and a single chemotherapeutic agent (carboplatin, paclitaxel, gemcitabine, or pegylated liposomal doxorubicin) [160].

Primary resistance and predictive markers of response to WEE1/PKMYT1 based therapies

Several DDR inhibitors have proved their efficacy against different cancer types in the preclinical and clinical settings [161165]. Among them, WEE1 inhibitor seems to be the most effective ones, also favored by a relative low off-target toxicity. However, despite the number of studies and the promising results, few predictive markers of response have been identified. Recently, cyclin E level has been linked to the efficacy of adavosertib in breast cancer models [89], with cyclin E-high cells, that generally show elevated chromosome instability, being more sensitive compared with cyclin E-low ones. Despite the reported low levels of WEE1 expression in breast cancer, chromosome instability, that has also prognostic potential mainly in grade 2 tumors [89], may explain the effectiveness of WEE1 inhibitors, as supported by the predictive role of cyclin E. Our group and others showed that high PKMYT1 expression associates with reduced sensitivity to adavosertib, indicating a potential compensatory effect [35, 166]. Moreover, high-throughput proteomic profiling demonstrated that small cell lung cancer and ovarian cancer models with primary resistance to adavosertib express high levels of AKT/mTOR pathway molecules and phosphorylated S6 ribosomal protein [137, 138]. In acute leukemia models, the sensitivity to adavosertib has been recently linked to HDAC and MYC regulation. Indeed, by generating adavosertib-resistant models, the researchers found that resistant acute leukemia cell lines are dependent on increased HDAC activity for their survival, partly due to increased KDM5A function. In addition, gene expression analyses demonstrated a HDAC-dependent expression of MYC in the adavosertib-resistant cell lines [167]. These observations support the success of preclinical studies combining WEE1 and HDAC [41, 130, 131] or bromodomain inhibitors [150].

Conclusion

Thanks to a constantly growing amount of preclinical and clinical data, our knowledge on cancer biology is increasing and, consequently, the list of cancer hallmarks has been progressively expanding. Recent findings demonstrated that cancer cells are characterized by functional and molecular alterations in crucial genes involved in the DDR pathway, which is fundamental for cell cycle regulation, DNA damages recognition, and repair. Functional alterations of DDR-gene have a deep impact on tumor progression and on the clinical outcome of cancer patients. Indeed, the efficacy of standard of care chemo/radiotherapy regimens depends on the generation of DNA damages in proliferating malignant cells. In this scenario, the overexpression or uncontrolled activation of DDR pathways has been showed to protect cancer cells from the therapeutic effect of DNA damaging agents. Moreover, a large number of preclinical studies highlighted that cancer cells depend on the functionality of DDR pathways in order to survive, to tolerate the replicative stress induced by the high proliferative rate and to sustain the intrinsic genetic instability. For these reasons, selective inhibitors have been developed in order to exploit cancer cells’ dependency on DDR-gene functionality. Pre-clinical data has proven the efficacy of DDR inhibition in different kinds of hematological and solid tumors, both as monotherapy and in combination with a wide number of DNA damaging agents. Among DDR inhibitors, the most effective once are those targeting PARP1 and WEE1 family kinases. The effectiveness of PARP1 inhibitors is however dependent on homologous recombination (HR) repair deficiency while WEE1 family kinases inhibitors seems to have a widespread efficacy independently from a specific the genetic background. Indeed, cancer cells seem to be strictly dependent on the functionality of WEE1/PKMYT1 kinases to survive, especially those with alterations targeting the G1 checkpoint. WEE1/PKMYT1 kinases are involved in different biological processes and they seem to play diverse roles in nonmalignant and in cancer cells. Indeed, they control cell cycle regulation and genetic stability in nonmalignant cells and for these reasons act as tumor suppressor genes. Conversely, their ability of promote DNA damages repair and cell cycle control makes them act as pseudo-oncogenes in cancer cells. Several molecular studies showed that malignant cells have high expression level of WEE1 and PKMYT1, which has become a good prognostic biomarker for chemo/radiotherapy regimens. However, we currently lack information regarding predictive markers of response to WEE1/PKMYT1 inhibitors. Large preclinical and clinical studies should be conducted in order to identify specific molecular backgrounds in which the use of WEE1/PKYMT1 inhibitors may be recommended. The identification of molecular vulnerabilities in cancer patients will be fundamental to design novel therapeutic regimens using WEE1/PKMYT1 inhibitors in a chemo/radiotherapy-free, synthetic lethality-based approach.

Acknowledgements

Not applicable.
Not applicable.
All authors read and approved the final manuscript.

Competing interests

GM has competing interests with Novartis, BMS, Roche, Pfizer, ARIAD, and MSD. The other authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Schmidt M, Rohe A, Platzer C, et al. Regulation of G2/M transition by inhibition of WEE1 and PKMYT1 Kinases. Molecules. 2017;22:2045.PubMedCentralCrossRef Schmidt M, Rohe A, Platzer C, et al. Regulation of G2/M transition by inhibition of WEE1 and PKMYT1 Kinases. Molecules. 2017;22:2045.PubMedCentralCrossRef
2.
Zurück zum Zitat Solc P, Schultz RM, Motlik J. Prophase I arrest and progression to metaphase I in mouse oocytes: Comparison of resumption of meiosis and recovery from G2-arrest in somatic cells. Mol Hum Reprod. 2010;16:654–64.PubMedPubMedCentralCrossRef Solc P, Schultz RM, Motlik J. Prophase I arrest and progression to metaphase I in mouse oocytes: Comparison of resumption of meiosis and recovery from G2-arrest in somatic cells. Mol Hum Reprod. 2010;16:654–64.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Nakanishi M, Ando H, Watanabe N, et al. Identification and characterization of human Wee1B, a new member of the Wee1 family of Cdk-inhibitory kinases. Genes Cells. 2000;5(10):839–47.PubMedCrossRef Nakanishi M, Ando H, Watanabe N, et al. Identification and characterization of human Wee1B, a new member of the Wee1 family of Cdk-inhibitory kinases. Genes Cells. 2000;5(10):839–47.PubMedCrossRef
4.
Zurück zum Zitat Mueller PR, Coleman TR, Kumagai A, Dunphy WG. Myt1: A membrane-associated inhibitory kinase that phosphorylates Cdc2 on both threonine-14 and tyrosine-15. Science. 1995;270(5233):86–90.PubMedCrossRef Mueller PR, Coleman TR, Kumagai A, Dunphy WG. Myt1: A membrane-associated inhibitory kinase that phosphorylates Cdc2 on both threonine-14 and tyrosine-15. Science. 1995;270(5233):86–90.PubMedCrossRef
5.
Zurück zum Zitat Booher RN, Holman PS, Fattaey A. Human Myt1 is a cell cycle-regulated kinase that inhibits Cdc2 but not Cdk2 activity. J Biol Chem. 1997;272(35):22300–6.PubMedCrossRef Booher RN, Holman PS, Fattaey A. Human Myt1 is a cell cycle-regulated kinase that inhibits Cdc2 but not Cdk2 activity. J Biol Chem. 1997;272(35):22300–6.PubMedCrossRef
6.
Zurück zum Zitat Liu F, Stanton JJ, Wu Z, Piwnica-Worms H. The human Myt1 kinase preferentially phosphorylates Cdc2 on threonine 14 and localizes to the endoplasmic reticulum and Golgi complex. Mol Cell Biol. 1997;17(2):571–83.PubMedPubMedCentralCrossRef Liu F, Stanton JJ, Wu Z, Piwnica-Worms H. The human Myt1 kinase preferentially phosphorylates Cdc2 on threonine 14 and localizes to the endoplasmic reticulum and Golgi complex. Mol Cell Biol. 1997;17(2):571–83.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Nakajima H, Yonemura S, Murata M, et al. Myt1 protein kinase is essential for Golgi and ER assembly during mitotic exit. J Cell Biol. 2008;181(1):89–103.PubMedPubMedCentralCrossRef Nakajima H, Yonemura S, Murata M, et al. Myt1 protein kinase is essential for Golgi and ER assembly during mitotic exit. J Cell Biol. 2008;181(1):89–103.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Chow JPH, Poon RYC, Ma HT. Inhibitory phosphorylation of cyclin-dependent kinase 1 as a compensatory mechanism for mitosis exit. Mol Cell Biol. 2011;31(7):1478–91.PubMedPubMedCentralCrossRef Chow JPH, Poon RYC, Ma HT. Inhibitory phosphorylation of cyclin-dependent kinase 1 as a compensatory mechanism for mitosis exit. Mol Cell Biol. 2011;31(7):1478–91.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Solomon MJ, Harper JW, Shuttleworth J. CAK, the p34cdc2 activating kinase, contains a protein identical or closely related to p40MO15. EMBO J. 1993;12(8):3133–42.PubMedPubMedCentralCrossRef Solomon MJ, Harper JW, Shuttleworth J. CAK, the p34cdc2 activating kinase, contains a protein identical or closely related to p40MO15. EMBO J. 1993;12(8):3133–42.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Lolli G, Johnson LN. CAK-Cyclin-dependent activating kinase: a key kinase in cell cycle control and a target for Drugs? Cell Cycle. 2005;4(4):572–7.PubMedCrossRef Lolli G, Johnson LN. CAK-Cyclin-dependent activating kinase: a key kinase in cell cycle control and a target for Drugs? Cell Cycle. 2005;4(4):572–7.PubMedCrossRef
11.
Zurück zum Zitat Walsh S, Margolis SS, Kornbluth S. Phosphorylation of the cyclin B1 cytoplasmic retention sequence by mitogen-activated protein kinase and Plx. Mol Cancer Res. 2003;1(4):280–9.PubMed Walsh S, Margolis SS, Kornbluth S. Phosphorylation of the cyclin B1 cytoplasmic retention sequence by mitogen-activated protein kinase and Plx. Mol Cancer Res. 2003;1(4):280–9.PubMed
12.
Zurück zum Zitat Szmyd R, Niska-Blakie J, Diril MK, et al. Premature activation of Cdk1 leads to mitotic events in S phase and embryonic lethality. Oncogene. 2019;38(7):998–1018.PubMedCrossRef Szmyd R, Niska-Blakie J, Diril MK, et al. Premature activation of Cdk1 leads to mitotic events in S phase and embryonic lethality. Oncogene. 2019;38(7):998–1018.PubMedCrossRef
13.
Zurück zum Zitat Watanabe N, Arai H, Nishihara Y, et al. M-phase kinases induce phospho-dependent ubiquitination of somatic Wee1 by SCFbeta-TrCP. Proc Natl Acad Sci U S A. 2004;101(13):4419–24.PubMedPubMedCentralCrossRef Watanabe N, Arai H, Nishihara Y, et al. M-phase kinases induce phospho-dependent ubiquitination of somatic Wee1 by SCFbeta-TrCP. Proc Natl Acad Sci U S A. 2004;101(13):4419–24.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Toyoshima-Morimoto F, Taniguchi E, Shinya N, et al. Polo-like kinase 1 phosphorylates cyclin B1 and targets it to the nucleus during prophase. Nature. 2001;410(6825):215–20.PubMedCrossRef Toyoshima-Morimoto F, Taniguchi E, Shinya N, et al. Polo-like kinase 1 phosphorylates cyclin B1 and targets it to the nucleus during prophase. Nature. 2001;410(6825):215–20.PubMedCrossRef
15.
Zurück zum Zitat Van Vugt MATM, Brás A, Medema RH. Polo-like kinase-1 controls recovery from a G2 DNA damage-induced arrest in mammalian cells. Mol Cell. 2004;15(5):799–811.PubMedCrossRef Van Vugt MATM, Brás A, Medema RH. Polo-like kinase-1 controls recovery from a G2 DNA damage-induced arrest in mammalian cells. Mol Cell. 2004;15(5):799–811.PubMedCrossRef
16.
Zurück zum Zitat Nakojima H, Toyoshima-Morimoto F, Taniguchi E, Nishida E. Identification of a consensus motif for PlK (Polo-like kinase) phosphorylation reveals Myt1 as a Plk1 substrate. J Biol Chem. 2003;278(28):25277–80.CrossRef Nakojima H, Toyoshima-Morimoto F, Taniguchi E, Nishida E. Identification of a consensus motif for PlK (Polo-like kinase) phosphorylation reveals Myt1 as a Plk1 substrate. J Biol Chem. 2003;278(28):25277–80.CrossRef
17.
Zurück zum Zitat Takisawa H, Mimura S, Kubota Y. Eukaryotic DNA replication: from pre-replication complex to initiation complex. Curr Opin Cell Biol. 2000;12(6):690–6.PubMedCrossRef Takisawa H, Mimura S, Kubota Y. Eukaryotic DNA replication: from pre-replication complex to initiation complex. Curr Opin Cell Biol. 2000;12(6):690–6.PubMedCrossRef
18.
Zurück zum Zitat Heller RC, Kang S, Lam WM, et al. Eukaryotic origin-dependent DNA replication in vitro reveals sequential action of DDK and S-CDK kinases. Cell. 2011;146(1):80–91.PubMedPubMedCentralCrossRef Heller RC, Kang S, Lam WM, et al. Eukaryotic origin-dependent DNA replication in vitro reveals sequential action of DDK and S-CDK kinases. Cell. 2011;146(1):80–91.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Labib K. How do Cdc7 and cyclin-dependent kinases trigger the initiation of chromosome replication in eukaryotic cells? Genes Dev. 2010;24(12):1208–19.PubMedPubMedCentralCrossRef Labib K. How do Cdc7 and cyclin-dependent kinases trigger the initiation of chromosome replication in eukaryotic cells? Genes Dev. 2010;24(12):1208–19.PubMedPubMedCentralCrossRef
20.
21.
Zurück zum Zitat Vassilopoulos A, Tominaga Y, Kim HS, et al. WEE1 murine deficiency induces hyper-activation of APC/C and results in genomic instability and carcinogenesis. Oncogene. 2015;34(23):3023–35.PubMedCrossRef Vassilopoulos A, Tominaga Y, Kim HS, et al. WEE1 murine deficiency induces hyper-activation of APC/C and results in genomic instability and carcinogenesis. Oncogene. 2015;34(23):3023–35.PubMedCrossRef
22.
Zurück zum Zitat Ghelli Luserna Di Rorà A, Martinelli G, Simonetti G. The balance between mitotic death and mitotic slippage in acute leukemia: a new therapeutic window? J Hematol Oncol. 2019;12(1):123.PubMedPubMedCentralCrossRef Ghelli Luserna Di Rorà A, Martinelli G, Simonetti G. The balance between mitotic death and mitotic slippage in acute leukemia: a new therapeutic window? J Hematol Oncol. 2019;12(1):123.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Visconti R, Grieco D. Fighting tubulin-targeting anticancer drug toxicity and resistance. Endocr Relat Cancer. 2017;24(9):T107–17.PubMedCrossRef Visconti R, Grieco D. Fighting tubulin-targeting anticancer drug toxicity and resistance. Endocr Relat Cancer. 2017;24(9):T107–17.PubMedCrossRef
24.
Zurück zum Zitat Visconti R, Palazzo L, Della Monica R, Grieco D. Fcp1-dependent dephosphorylation is required for M-phase-promoting factor inactivation at mitosis exit. Nat Commun. 2012;3:894.PubMedCrossRef Visconti R, Palazzo L, Della Monica R, Grieco D. Fcp1-dependent dephosphorylation is required for M-phase-promoting factor inactivation at mitosis exit. Nat Commun. 2012;3:894.PubMedCrossRef
25.
Zurück zum Zitat Musacchio A, Salmon ED. The spindle-assembly checkpoint in space and time. Nat Rev Mol Cell Biol. 2007;8(5):379–93.PubMedCrossRef Musacchio A, Salmon ED. The spindle-assembly checkpoint in space and time. Nat Rev Mol Cell Biol. 2007;8(5):379–93.PubMedCrossRef
27.
28.
Zurück zum Zitat Domínguez-Kelly R, Martín Y, Koundrioukoff S, et al. Wee1 controls genomic stability during replication by regulating the Mus81-Eme1 endonuclease. J Cell Biol. 2011;194(4):567–79.PubMedPubMedCentralCrossRef Domínguez-Kelly R, Martín Y, Koundrioukoff S, et al. Wee1 controls genomic stability during replication by regulating the Mus81-Eme1 endonuclease. J Cell Biol. 2011;194(4):567–79.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Duda H, Arter M, Gloggnitzer J, et al. A mechanism for controlled breakage of under-replicated chromosomes during mitosis. Dev Cell. 2016;39(6):740–55.PubMedCrossRef Duda H, Arter M, Gloggnitzer J, et al. A mechanism for controlled breakage of under-replicated chromosomes during mitosis. Dev Cell. 2016;39(6):740–55.PubMedCrossRef
30.
Zurück zum Zitat Asquith CRM, Laitinen T, East MP. PKMYT1: a forgotten member of the WEE1 family. Nat Rev Drug Discov. 2020;19(3):157.PubMedCrossRef Asquith CRM, Laitinen T, East MP. PKMYT1: a forgotten member of the WEE1 family. Nat Rev Drug Discov. 2020;19(3):157.PubMedCrossRef
31.
Zurück zum Zitat Liu Y, Qi J, Dou Z, et al. Systematic expression analysis of WEE family kinases reveals the importance of PKMYT1 in breast carcinogenesis. Cell Prolif. 2020;53(2):e12741.PubMedCrossRef Liu Y, Qi J, Dou Z, et al. Systematic expression analysis of WEE family kinases reveals the importance of PKMYT1 in breast carcinogenesis. Cell Prolif. 2020;53(2):e12741.PubMedCrossRef
32.
Zurück zum Zitat Jeong D, Kim H, Kim D, et al. Protein kinase, membrane-associated tyrosine/threonine 1 is associated with the progression of colorectal cancer. Oncol Rep. 2018;39(6):2829–36.PubMed Jeong D, Kim H, Kim D, et al. Protein kinase, membrane-associated tyrosine/threonine 1 is associated with the progression of colorectal cancer. Oncol Rep. 2018;39(6):2829–36.PubMed
33.
Zurück zum Zitat Lal S, Cozzitorto JA, Blanco F, et al. 988 Sequence alterations in the WEE1 non-coding region is a facilitator and marker for pancreatic tumorigenesis. Gastroenterology. 2014;S-1034. Lal S, Cozzitorto JA, Blanco F, et al. 988 Sequence alterations in the WEE1 non-coding region is a facilitator and marker for pancreatic tumorigenesis. Gastroenterology. 2014;S-1034.
34.
35.
Zurück zum Zitat Ghelli Luserna Di Rorà A, Beeharry N, Imbrogno E, et al. Targeting WEE1 to enhance conventional therapies for acute lymphoblastic leukemia. J Hematol Oncol. 2018;11(1):99.PubMedPubMedCentralCrossRef Ghelli Luserna Di Rorà A, Beeharry N, Imbrogno E, et al. Targeting WEE1 to enhance conventional therapies for acute lymphoblastic leukemia. J Hematol Oncol. 2018;11(1):99.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Barbosa RSS, Dantonio PM, Guimarães T, et al. Sequential combination of bortezomib and WEE1 inhibitor, MK-1775, induced apoptosis in multiple myeloma cell lines. Biochem Biophys Res Commun. 2019;519(3):597–604.PubMedCrossRef Barbosa RSS, Dantonio PM, Guimarães T, et al. Sequential combination of bortezomib and WEE1 inhibitor, MK-1775, induced apoptosis in multiple myeloma cell lines. Biochem Biophys Res Commun. 2019;519(3):597–604.PubMedCrossRef
37.
Zurück zum Zitat Van Linden AA, Baturin D, Ford JB, et al. Inhibition of Wee1 sensitizes cancer cells to antimetabolite chemotherapeutics in vitro and in vivo, independent of p53 functionality. Mol Cancer Ther. 2013;12(12):2675–84.PubMedPubMedCentralCrossRef Van Linden AA, Baturin D, Ford JB, et al. Inhibition of Wee1 sensitizes cancer cells to antimetabolite chemotherapeutics in vitro and in vivo, independent of p53 functionality. Mol Cancer Ther. 2013;12(12):2675–84.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Porter CC, Kim J, Fosmire S, et al. Integrated genomic analyses identify WEE1 as a critical mediator of cell fate and a novel therapeutic target in acute myeloid leukemia. Leukemia. 2012;26(6):1266–76.PubMedPubMedCentralCrossRef Porter CC, Kim J, Fosmire S, et al. Integrated genomic analyses identify WEE1 as a critical mediator of cell fate and a novel therapeutic target in acute myeloid leukemia. Leukemia. 2012;26(6):1266–76.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Johnston HE, Carter MJ, Larrayoz M, et al. Proteomics profiling of CLL versus healthy B-cells identifies putative therapeutic targets and a subtype-independent signature of spliceosome dysregulation. Mol Cell Proteomics. 2018;17(4):776–91.PubMedPubMedCentralCrossRef Johnston HE, Carter MJ, Larrayoz M, et al. Proteomics profiling of CLL versus healthy B-cells identifies putative therapeutic targets and a subtype-independent signature of spliceosome dysregulation. Mol Cell Proteomics. 2018;17(4):776–91.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Neben K, Schnittger S, Brors B, et al. Distinct gene expression patterns associated with FLT3- and NRAS-activating mutations in acute myeloid leukemia with normal karyotype. Oncogene. 2005;24(9):1580–8.PubMedCrossRef Neben K, Schnittger S, Brors B, et al. Distinct gene expression patterns associated with FLT3- and NRAS-activating mutations in acute myeloid leukemia with normal karyotype. Oncogene. 2005;24(9):1580–8.PubMedCrossRef
41.
Zurück zum Zitat Zhou L, Zhang Y, Chen S, et al. A regimen combining the Wee1 inhibitor AZD1775 with HDAC inhibitors targets human acute myeloid leukemia cells harboring various genetic mutations. Leukemia. 2015;29(4):807–18.PubMedCrossRef Zhou L, Zhang Y, Chen S, et al. A regimen combining the Wee1 inhibitor AZD1775 with HDAC inhibitors targets human acute myeloid leukemia cells harboring various genetic mutations. Leukemia. 2015;29(4):807–18.PubMedCrossRef
42.
Zurück zum Zitat Fan J, Li L, Small D, Rassool F. Cells expressing FLT3/ITD mutations exhibit elevated repair errors generated through alternative NHEJ pathways: implications for genomic instability and therapy. Blood. 2010;116(24):5298–305.PubMedPubMedCentralCrossRef Fan J, Li L, Small D, Rassool F. Cells expressing FLT3/ITD mutations exhibit elevated repair errors generated through alternative NHEJ pathways: implications for genomic instability and therapy. Blood. 2010;116(24):5298–305.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat De Jong MRW, Visser L, Huls G, et al. Identification of relevant drugable targets in diffuse large B-cell lymphoma using a genome-wide unbiased CD20 guilt-by association approach. PLoS One. 2018;13(2):e0193098.PubMedPubMedCentralCrossRef De Jong MRW, Visser L, Huls G, et al. Identification of relevant drugable targets in diffuse large B-cell lymphoma using a genome-wide unbiased CD20 guilt-by association approach. PLoS One. 2018;13(2):e0193098.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Bolomsky A, Gruber F, Stangelberger K, et al. Preclinical validation studies support causal machine learning based identification of novel drug targets for high-risk multiple myeloma. Blood. 2018;132(Supplement 1):3210.CrossRef Bolomsky A, Gruber F, Stangelberger K, et al. Preclinical validation studies support causal machine learning based identification of novel drug targets for high-risk multiple myeloma. Blood. 2018;132(Supplement 1):3210.CrossRef
45.
Zurück zum Zitat Sun QS, Luo M, Zhao HM, Sun H. Overexpression of PKMYT1 indicates the poor prognosis and enhances proliferation and tumorigenesis in non-small cell lung cancer via activation of Notch signal pathway. Eur Rev Med Pharmacol Sci. 2019;23(10):4210–9.PubMed Sun QS, Luo M, Zhao HM, Sun H. Overexpression of PKMYT1 indicates the poor prognosis and enhances proliferation and tumorigenesis in non-small cell lung cancer via activation of Notch signal pathway. Eur Rev Med Pharmacol Sci. 2019;23(10):4210–9.PubMed
46.
Zurück zum Zitat Liu L, Wu J, Wang S, et al. PKMYT1 promoted the growth and motility of hepatocellular carcinoma cells by activating beta-catenin/TCF signaling. Exp Cell Res. 2017;358(2):209–16.PubMedCrossRef Liu L, Wu J, Wang S, et al. PKMYT1 promoted the growth and motility of hepatocellular carcinoma cells by activating beta-catenin/TCF signaling. Exp Cell Res. 2017;358(2):209–16.PubMedCrossRef
47.
Zurück zum Zitat Wang XM, Li QY, Ren LL, et al. Effects of MCRS1 on proliferation, migration, invasion, and epithelial mesenchymal transition of gastric cancer cells by interacting with Pkmyt1 protein kinase. Cell Signal. 2019;59:171–81.PubMedCrossRef Wang XM, Li QY, Ren LL, et al. Effects of MCRS1 on proliferation, migration, invasion, and epithelial mesenchymal transition of gastric cancer cells by interacting with Pkmyt1 protein kinase. Cell Signal. 2019;59:171–81.PubMedCrossRef
48.
49.
Zurück zum Zitat Magnussen GI, Holm R, Emilsen E, et al. High expression of Wee1 is associated with poor disease-free survival in Malignant Melanoma: Potential for targeted therapy. PLoS One. 2012;7(6):e38254.PubMedPubMedCentralCrossRef Magnussen GI, Holm R, Emilsen E, et al. High expression of Wee1 is associated with poor disease-free survival in Malignant Melanoma: Potential for targeted therapy. PLoS One. 2012;7(6):e38254.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Mueller S, Hashizume R, Yang X, et al. Targeting wee1 for the treatment of pediatric high-grade gliomas. Neuro Oncol. 2014;16(3):352–60.PubMedCrossRef Mueller S, Hashizume R, Yang X, et al. Targeting wee1 for the treatment of pediatric high-grade gliomas. Neuro Oncol. 2014;16(3):352–60.PubMedCrossRef
51.
Zurück zum Zitat Mir SE, De Witt Hamer PC, Krawczyk PM, et al. In silico analysis of kinase expression identifies WEE1 as a gatekeeper against mitotic catastrophe in glioblastoma. Cancer Cell. 2010;18(3):244–57.PubMedPubMedCentralCrossRef Mir SE, De Witt Hamer PC, Krawczyk PM, et al. In silico analysis of kinase expression identifies WEE1 as a gatekeeper against mitotic catastrophe in glioblastoma. Cancer Cell. 2010;18(3):244–57.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Chayka O, D’Acunto CW, Middleton O, et al. Identification and pharmacological inactivation of the MYCN gene network as a therapeutic strategy for neuroblastic tumor cells. J Biol Chem. 2015;290(4):2198–212.PubMedCrossRef Chayka O, D’Acunto CW, Middleton O, et al. Identification and pharmacological inactivation of the MYCN gene network as a therapeutic strategy for neuroblastic tumor cells. J Biol Chem. 2015;290(4):2198–212.PubMedCrossRef
53.
Zurück zum Zitat Zhang Q, Zhao X, Zhang C, et al. Overexpressed PKMYT1 promotes tumor progression and associates with poor survival in esophageal squamous cell carcinoma. Cancer Manag Res. 2019;11:7813–24.PubMedPubMedCentralCrossRef Zhang Q, Zhao X, Zhang C, et al. Overexpressed PKMYT1 promotes tumor progression and associates with poor survival in esophageal squamous cell carcinoma. Cancer Manag Res. 2019;11:7813–24.PubMedPubMedCentralCrossRef
54.
55.
Zurück zum Zitat Simonetti G, Padella A, do Valle IF, et al. Aneuploid acute myeloid leukemia exhibits a signature of genomic alterations in the cell cycle and protein degradation machinery. Cancer. 2018;125:1–14. Simonetti G, Padella A, do Valle IF, et al. Aneuploid acute myeloid leukemia exhibits a signature of genomic alterations in the cell cycle and protein degradation machinery. Cancer. 2018;125:1–14.
56.
Zurück zum Zitat Caretti V, Hiddingh L, Lagerweij T, et al. WEE1 kinase inhibition enhances the radiation response of diffuse intrinsic pontine gliomas. Mol Cancer Ther. 2013;12(2):141–50.PubMedCrossRef Caretti V, Hiddingh L, Lagerweij T, et al. WEE1 kinase inhibition enhances the radiation response of diffuse intrinsic pontine gliomas. Mol Cancer Ther. 2013;12(2):141–50.PubMedCrossRef
57.
Zurück zum Zitat Music D, Dahlrot RH, Hermansen SK, et al. Expression and prognostic value of the WEE1 kinase in gliomas. J Neurooncol. 2016;127(2):381–9.PubMedCrossRef Music D, Dahlrot RH, Hermansen SK, et al. Expression and prognostic value of the WEE1 kinase in gliomas. J Neurooncol. 2016;127(2):381–9.PubMedCrossRef
58.
Zurück zum Zitat Egeland EV, Flatmark K, Nesland JM, et al. Expression and clinical significance of Wee1 in colorectal cancer. Tumor Biol. 2016;37(9):12133–40.CrossRef Egeland EV, Flatmark K, Nesland JM, et al. Expression and clinical significance of Wee1 in colorectal cancer. Tumor Biol. 2016;37(9):12133–40.CrossRef
59.
Zurück zum Zitat Slipicevic A, Holth A, Hellesylt E, et al. Wee1 is a novel independent prognostic marker of poor survival in post-chemotherapy ovarian carcinoma effusions. Gynecol Oncol. 2014;135(1):118–24.PubMedCrossRef Slipicevic A, Holth A, Hellesylt E, et al. Wee1 is a novel independent prognostic marker of poor survival in post-chemotherapy ovarian carcinoma effusions. Gynecol Oncol. 2014;135(1):118–24.PubMedCrossRef
60.
Zurück zum Zitat Shu C, Wang Q, Yan X, Wang J. Whole-genome expression microarray combined with machine learning to identify prognostic biomarkers for high-grade glioma. J Mol Neurosci. 2018;64(4):491–500.PubMedCrossRef Shu C, Wang Q, Yan X, Wang J. Whole-genome expression microarray combined with machine learning to identify prognostic biomarkers for high-grade glioma. J Mol Neurosci. 2018;64(4):491–500.PubMedCrossRef
61.
Zurück zum Zitat Novak EM, Halley NS, Gimenez TM, et al. BLM germline and somatic PKMYT1 and AHCY mutations: genetic variations beyond MYCN and prognosis in neuroblastoma. Med Hypotheses. 2016;97:22–5.PubMedCrossRef Novak EM, Halley NS, Gimenez TM, et al. BLM germline and somatic PKMYT1 and AHCY mutations: genetic variations beyond MYCN and prognosis in neuroblastoma. Med Hypotheses. 2016;97:22–5.PubMedCrossRef
62.
Zurück zum Zitat Ku BM, Bae Y-H, Koh J, et al. Mutational status of TP53 defines the efficacy of Wee1 inhibitor AZD1775 in KRAS -mutant non-small cell lung cancer. Oncotarget. 2017;8(40):67526–37.PubMedPubMedCentralCrossRef Ku BM, Bae Y-H, Koh J, et al. Mutational status of TP53 defines the efficacy of Wee1 inhibitor AZD1775 in KRAS -mutant non-small cell lung cancer. Oncotarget. 2017;8(40):67526–37.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Kreahling JM, Foroutan P, Reed D, et al. Wee1 inhibition by MK-1775 leads to tumor inhibition and enhances efficacy of gemcitabine in human sarcomas. PLoS One. 2013;8(3):e57523.PubMedPubMedCentralCrossRef Kreahling JM, Foroutan P, Reed D, et al. Wee1 inhibition by MK-1775 leads to tumor inhibition and enhances efficacy of gemcitabine in human sarcomas. PLoS One. 2013;8(3):e57523.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Ford JB, Baturin D, Burleson TM, et al. AZD1775 sensitizes T cell acute lymphoblastic leukemia cells to cytarabine by promoting apoptosis over DNA repair. Oncotarget. 2015;6(29):28001–10.PubMedPubMedCentralCrossRef Ford JB, Baturin D, Burleson TM, et al. AZD1775 sensitizes T cell acute lymphoblastic leukemia cells to cytarabine by promoting apoptosis over DNA repair. Oncotarget. 2015;6(29):28001–10.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Webster PJ, Littlejohns AT, Gaunt HJ, et al. AZD1775 induces toxicity through double-stranded DNA breaks independently of chemotherapeutic agents in p53-mutated colorectal cancer cells. Cell Cycle. 2017;16(22):2176–82.PubMedPubMedCentralCrossRef Webster PJ, Littlejohns AT, Gaunt HJ, et al. AZD1775 induces toxicity through double-stranded DNA breaks independently of chemotherapeutic agents in p53-mutated colorectal cancer cells. Cell Cycle. 2017;16(22):2176–82.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Kahen E, Yu D, Harrison DJ, et al. Identification of clinically achievable combination therapies in childhood rhabdomyosarcoma. Cancer Chemother Pharmacol. 2016;78(2):313–23.PubMedPubMedCentralCrossRef Kahen E, Yu D, Harrison DJ, et al. Identification of clinically achievable combination therapies in childhood rhabdomyosarcoma. Cancer Chemother Pharmacol. 2016;78(2):313–23.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Lee YY, Cho YJ, Shin SW, et al. Anti-tumor effects of Wee1 kinase inhibitor with radiotherapy in human cervical cancer. Sci Rep. 2019;9(1):15394.PubMedPubMedCentralCrossRef Lee YY, Cho YJ, Shin SW, et al. Anti-tumor effects of Wee1 kinase inhibitor with radiotherapy in human cervical cancer. Sci Rep. 2019;9(1):15394.PubMedPubMedCentralCrossRef
69.
70.
Zurück zum Zitat Xu H, Krystal GW. Actinomycin D decreases Mcl-1 expression and acts synergistically with ABT-737 against small cell lung cancer cell lines. Clin Cancer Res. 2010;16(17):4392–400.PubMedCrossRef Xu H, Krystal GW. Actinomycin D decreases Mcl-1 expression and acts synergistically with ABT-737 against small cell lung cancer cell lines. Clin Cancer Res. 2010;16(17):4392–400.PubMedCrossRef
71.
Zurück zum Zitat Hayashi Y, Fujimura A, Kato K, et al. Nucleolar integrity during interphase supports faithful Cdk1 activation and mitotic entry. Sci Adv. 2018;4(6):eaap7777.PubMedPubMedCentralCrossRef Hayashi Y, Fujimura A, Kato K, et al. Nucleolar integrity during interphase supports faithful Cdk1 activation and mitotic entry. Sci Adv. 2018;4(6):eaap7777.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Alexander VM, Roy M, Steffens KA, et al. Azacytidine induces cell cycle arrest and suppression of neuroendocrine markers in carcinoids. Int J Clin Exp Med. 2010;3(2):95–102.PubMedPubMedCentral Alexander VM, Roy M, Steffens KA, et al. Azacytidine induces cell cycle arrest and suppression of neuroendocrine markers in carcinoids. Int J Clin Exp Med. 2010;3(2):95–102.PubMedPubMedCentral
73.
Zurück zum Zitat Uchida R, Yokota S, Matsuda D, et al. Habiterpenol, a novel abrogator of bleomycin-induced G2 arrest in Jurkat cells, produced by Phytohabitans suffuscus 3787-5. J Antibiot (Tokyo). 2014;67(11):777–81.CrossRef Uchida R, Yokota S, Matsuda D, et al. Habiterpenol, a novel abrogator of bleomycin-induced G2 arrest in Jurkat cells, produced by Phytohabitans suffuscus 3787-5. J Antibiot (Tokyo). 2014;67(11):777–81.CrossRef
74.
Zurück zum Zitat Zhang Z, Zhang H, Hu Z, et al. Synergy of 1,25-dihydroxyvitamin D3 and carboplatin in growth suppression of SKOV-3 cells. Oncol Lett. 2014;8(3):1348–54.PubMedPubMedCentralCrossRef Zhang Z, Zhang H, Hu Z, et al. Synergy of 1,25-dihydroxyvitamin D3 and carboplatin in growth suppression of SKOV-3 cells. Oncol Lett. 2014;8(3):1348–54.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Sarin N, Engel F, Kalayda GV, et al. Cisplatin resistance in non-small cell lung cancer cells is associated with an abrogation of cisplatin-induced G2/M cell cycle arrest. PLoS One. 2017;12(7):e0181081.PubMedPubMedCentralCrossRef Sarin N, Engel F, Kalayda GV, et al. Cisplatin resistance in non-small cell lung cancer cells is associated with an abrogation of cisplatin-induced G2/M cell cycle arrest. PLoS One. 2017;12(7):e0181081.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Chen D, Lin X, Gao J, et al. Wee1 Inhibitor AZD1775 Combined with cisplatin potentiates anticancer activity against gastric cancer by increasing DNA damage and cell apoptosis. Biomed Res Int. 2018;2018:5813292.PubMedPubMedCentral Chen D, Lin X, Gao J, et al. Wee1 Inhibitor AZD1775 Combined with cisplatin potentiates anticancer activity against gastric cancer by increasing DNA damage and cell apoptosis. Biomed Res Int. 2018;2018:5813292.PubMedPubMedCentral
77.
Zurück zum Zitat Zheng H, Shao F, Martin S, et al. WEE1 inhibition targets cell cycle checkpoints for triple negative breast cancers to overcome cisplatin resistance. Sci Rep. 2017;7:43517.PubMedPubMedCentralCrossRef Zheng H, Shao F, Martin S, et al. WEE1 inhibition targets cell cycle checkpoints for triple negative breast cancers to overcome cisplatin resistance. Sci Rep. 2017;7:43517.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat de Jong MRW, Langendonk M, Reitsma B, et al. WEE1 inhibition synergizes with CHOP chemotherapy and radiation therapy through induction of premature mitotic entry and DNA damage in diffuse large B-cell lymphoma. Ther Adv Hematol. 2020;11:2040620719898373.PubMedPubMedCentral de Jong MRW, Langendonk M, Reitsma B, et al. WEE1 inhibition synergizes with CHOP chemotherapy and radiation therapy through induction of premature mitotic entry and DNA damage in diffuse large B-cell lymphoma. Ther Adv Hematol. 2020;11:2040620719898373.PubMedPubMedCentral
79.
Zurück zum Zitat Ma J, Li X, Su Y, et al. Mechanisms responsible for the synergistic antileukemic interactions between ATR inhibition and cytarabine in acute myeloid leukemia cells. Sci Rep. 2017;7:41950.PubMedPubMedCentralCrossRef Ma J, Li X, Su Y, et al. Mechanisms responsible for the synergistic antileukemic interactions between ATR inhibition and cytarabine in acute myeloid leukemia cells. Sci Rep. 2017;7:41950.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Shi Z, Azuma A, Sampath D, et al. S-phase arrest by nucleoside analogues and abrogation of survival without cell cycle progression by 7-hydroxystaurosporine. Cancer Res. 2001;61(3):1065–72.PubMed Shi Z, Azuma A, Sampath D, et al. S-phase arrest by nucleoside analogues and abrogation of survival without cell cycle progression by 7-hydroxystaurosporine. Cancer Res. 2001;61(3):1065–72.PubMed
81.
Zurück zum Zitat Garcia TB, Fosmire SP, Porter CC. Increased activity of both CDK1 and CDK2 is necessary for the combinatorial activity of WEE1 inhibition and cytarabine. Leuk Res. 2018;64:30–3.PubMedCrossRef Garcia TB, Fosmire SP, Porter CC. Increased activity of both CDK1 and CDK2 is necessary for the combinatorial activity of WEE1 inhibition and cytarabine. Leuk Res. 2018;64:30–3.PubMedCrossRef
82.
Zurück zum Zitat Al-Aamri HM, Ku H, Irving HR, et al. Time dependent response of daunorubicin on cytotoxicity, cell cycle and DNA repair in acute lymphoblastic leukaemia. BMC Cancer. 2019;19(1):179.PubMedPubMedCentralCrossRef Al-Aamri HM, Ku H, Irving HR, et al. Time dependent response of daunorubicin on cytotoxicity, cell cycle and DNA repair in acute lymphoblastic leukaemia. BMC Cancer. 2019;19(1):179.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Shang D, Han T, Xu X, Liu Y. Decitabine induces G2/M cell cycle arrest by suppressing p38/NF-κB signaling in human renal clear cell carcinoma. Int J Clin Exp Pathol. 2015;8(9):11140–8.PubMedPubMedCentral Shang D, Han T, Xu X, Liu Y. Decitabine induces G2/M cell cycle arrest by suppressing p38/NF-κB signaling in human renal clear cell carcinoma. Int J Clin Exp Pathol. 2015;8(9):11140–8.PubMedPubMedCentral
84.
Zurück zum Zitat Singh SK, Banerjee S, Acosta EP, et al. Resveratrol induces cell cycle arrest and apoptosis with docetaxel in prostate cancer cells via a p53/p21WAF1/CIP1 and p27KIP1 pathway. Oncotarget. 2017;8(10):17216–28.PubMedPubMedCentralCrossRef Singh SK, Banerjee S, Acosta EP, et al. Resveratrol induces cell cycle arrest and apoptosis with docetaxel in prostate cancer cells via a p53/p21WAF1/CIP1 and p27KIP1 pathway. Oncotarget. 2017;8(10):17216–28.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Morse DL, Gray H, Payne CM, Gillies RJ. Docetaxel induces cell death through mitotic catastrophe in human breast cancer cells. Mol Cancer Ther. 2005;4(10):1495–504.PubMedCrossRef Morse DL, Gray H, Payne CM, Gillies RJ. Docetaxel induces cell death through mitotic catastrophe in human breast cancer cells. Mol Cancer Ther. 2005;4(10):1495–504.PubMedCrossRef
86.
Zurück zum Zitat Vera J, Raatz Y, Wolkenhauer O, et al. Chk1 and Wee1 control genotoxic-stress induced G2-M arrest in melanoma cells. Cell Signal. 2015;27(5):951–60.PubMedCrossRef Vera J, Raatz Y, Wolkenhauer O, et al. Chk1 and Wee1 control genotoxic-stress induced G2-M arrest in melanoma cells. Cell Signal. 2015;27(5):951–60.PubMedCrossRef
87.
Zurück zum Zitat Wu CL, Ping SY, Yu CP, Yu DS. Tyrosine kinase receptor inhibitor-targeted combined chemotherapy for metastatic bladder cancer. Kaohsiung J Med Sci. 2012;28(4):194–203.PubMedCrossRef Wu CL, Ping SY, Yu CP, Yu DS. Tyrosine kinase receptor inhibitor-targeted combined chemotherapy for metastatic bladder cancer. Kaohsiung J Med Sci. 2012;28(4):194–203.PubMedCrossRef
88.
Zurück zum Zitat Senthebane DA, Jonker T, Rowe A, et al. The role of tumor microenvironment in chemoresistance: 3D extracellular matrices as accomplices. Int J Mol Sci. 2018;19(10):2861.PubMedCentralCrossRef Senthebane DA, Jonker T, Rowe A, et al. The role of tumor microenvironment in chemoresistance: 3D extracellular matrices as accomplices. Int J Mol Sci. 2018;19(10):2861.PubMedCentralCrossRef
89.
Zurück zum Zitat Chen X, Low KH, Alexander A, et al. Cyclin E overexpression sensitizes triple-negative breast cancer to Wee1 kinase inhibition. Clin Cancer Res. 2018;24(24):6594–610.PubMedPubMedCentralCrossRef Chen X, Low KH, Alexander A, et al. Cyclin E overexpression sensitizes triple-negative breast cancer to Wee1 kinase inhibition. Clin Cancer Res. 2018;24(24):6594–610.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Seung HL, Seung MS, Dong JS, et al. Epothilones induce human colon cancer SW620 cell apoptosis via the tubulin polymerization-independent activation of the nuclear factor-κB/IκB kinase signal pathway. Mol Cancer Ther. 2007;6(10):2786–97.CrossRef Seung HL, Seung MS, Dong JS, et al. Epothilones induce human colon cancer SW620 cell apoptosis via the tubulin polymerization-independent activation of the nuclear factor-κB/IκB kinase signal pathway. Mol Cancer Ther. 2007;6(10):2786–97.CrossRef
91.
Zurück zum Zitat Zhang R, Zhu L, Zhang L, et al. PTEN enhances G2/M arrest in etoposide-treated MCF-7 cells through activation of the ATM pathway. Oncol Rep. 2016;35(5):2707–14.PubMedCrossRef Zhang R, Zhu L, Zhang L, et al. PTEN enhances G2/M arrest in etoposide-treated MCF-7 cells through activation of the ATM pathway. Oncol Rep. 2016;35(5):2707–14.PubMedCrossRef
92.
Zurück zum Zitat Pitts TM, Simmons DM, Bagby SM, et al. Wee1 inhibition enhances the anti-tumor effects of capecitabine in preclinical models of triple-negative breast cancer. Cancers. 2020;12(3):719.PubMedCentralCrossRef Pitts TM, Simmons DM, Bagby SM, et al. Wee1 inhibition enhances the anti-tumor effects of capecitabine in preclinical models of triple-negative breast cancer. Cancers. 2020;12(3):719.PubMedCentralCrossRef
93.
Zurück zum Zitat Rajeshkumar NV, De Oliveira E, Ottenhof N, et al. MK-1775, a potent Wee1 inhibitor, synergizes with gemcitabine to achieve tumor regressions, selectively in p53-deficient pancreatic cancer xenografts. Clin Cancer Res. 2011;17(9):2799–806.PubMedPubMedCentralCrossRef Rajeshkumar NV, De Oliveira E, Ottenhof N, et al. MK-1775, a potent Wee1 inhibitor, synergizes with gemcitabine to achieve tumor regressions, selectively in p53-deficient pancreatic cancer xenografts. Clin Cancer Res. 2011;17(9):2799–806.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Koh SB, Wallez Y, Dunlop CR, et al. Mechanistic distinctions between CHK1 and WEE1 inhibition guide the scheduling of triple therapy with gemcitabine. Cancer Res. 2018;78(11):3054–66.PubMedPubMedCentralCrossRef Koh SB, Wallez Y, Dunlop CR, et al. Mechanistic distinctions between CHK1 and WEE1 inhibition guide the scheduling of triple therapy with gemcitabine. Cancer Res. 2018;78(11):3054–66.PubMedPubMedCentralCrossRef
95.
96.
Zurück zum Zitat Aarts M, Sharpe R, Garcia-Murillas I, et al. Forced mitotic entry of S-phase cells as a therapeutic strategy induced by inhibition of WEE1. Cancer Discov. 2012;2(6):524–39.PubMedCrossRef Aarts M, Sharpe R, Garcia-Murillas I, et al. Forced mitotic entry of S-phase cells as a therapeutic strategy induced by inhibition of WEE1. Cancer Discov. 2012;2(6):524–39.PubMedCrossRef
97.
Zurück zum Zitat Morgan MA, Onono FO, Spielmann HP, et al. Modulation of anthracycline-induced cytotoxicity by targeting the prenylated proteome in myeloid leukemia cells. J Mol Med. 2012;90(2):149–61.PubMedCrossRef Morgan MA, Onono FO, Spielmann HP, et al. Modulation of anthracycline-induced cytotoxicity by targeting the prenylated proteome in myeloid leukemia cells. J Mol Med. 2012;90(2):149–61.PubMedCrossRef
98.
Zurück zum Zitat Subhash VV, Tan SH, Yeo MS, et al. ATM expression predicts veliparib and irinotecan sensitivity in gastric cancer by mediating p53-independent regulation of cell cycle and apoptosis. Mol Cancer Ther. 2016;15(12):3087–96.PubMedCrossRef Subhash VV, Tan SH, Yeo MS, et al. ATM expression predicts veliparib and irinotecan sensitivity in gastric cancer by mediating p53-independent regulation of cell cycle and apoptosis. Mol Cancer Ther. 2016;15(12):3087–96.PubMedCrossRef
99.
Zurück zum Zitat Yin Y, Shen Q, Tao R, et al. Wee1 inhibition can suppress tumor proliferation and sensitize p53 mutant colonic cancer cells to the anticancer effect of irinotecan. Mol Med Rep. 2018;17(2):3344–9.PubMed Yin Y, Shen Q, Tao R, et al. Wee1 inhibition can suppress tumor proliferation and sensitize p53 mutant colonic cancer cells to the anticancer effect of irinotecan. Mol Med Rep. 2018;17(2):3344–9.PubMed
100.
Zurück zum Zitat Jan YH, Heck DE, Laskin DL, Laskin JD. Sulfur mustard analog mechlorethamine (Bis(2-chloroethyl)methylamine) modulates cell cycle progression via the DNA damage response in human lung epithelial A549 cells. Chem Res Toxicol. 2019;32(6):1123–33.PubMedPubMedCentralCrossRef Jan YH, Heck DE, Laskin DL, Laskin JD. Sulfur mustard analog mechlorethamine (Bis(2-chloroethyl)methylamine) modulates cell cycle progression via the DNA damage response in human lung epithelial A549 cells. Chem Res Toxicol. 2019;32(6):1123–33.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Mahbub A, Le Maitre C, Haywood-Small S, et al. Dietary polyphenols influence antimetabolite agents: Methotrexate, 6-mercaptopurine and 5-fluorouracil in leukemia cell lines. Oncotarget. 2017;8(62):104877–93.PubMedPubMedCentralCrossRef Mahbub A, Le Maitre C, Haywood-Small S, et al. Dietary polyphenols influence antimetabolite agents: Methotrexate, 6-mercaptopurine and 5-fluorouracil in leukemia cell lines. Oncotarget. 2017;8(62):104877–93.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Costantini DL, Villani DF, Vallis KA, Reilly RM. Methotrexate, paclitaxel, and doxorubicin radiosensitize HER2-amplified human breast cancer cells to the auger electron-emitting radiotherapeutic agent 111In-NLS-trastuzumab. J Nucl Med. 2010;51(3):477–83.PubMedCrossRef Costantini DL, Villani DF, Vallis KA, Reilly RM. Methotrexate, paclitaxel, and doxorubicin radiosensitize HER2-amplified human breast cancer cells to the auger electron-emitting radiotherapeutic agent 111In-NLS-trastuzumab. J Nucl Med. 2010;51(3):477–83.PubMedCrossRef
103.
Zurück zum Zitat Di Rorà AGL, Bocconcelli M, Ferrari A, et al. Synergism through WEE1 and CHK1 inhibition in acute lymphoblastic leukemia. Cancers (Basel). 2019;11(11):1654.CrossRef Di Rorà AGL, Bocconcelli M, Ferrari A, et al. Synergism through WEE1 and CHK1 inhibition in acute lymphoblastic leukemia. Cancers (Basel). 2019;11(11):1654.CrossRef
104.
Zurück zum Zitat Guerriero E, Sorice A, Capone F, et al. Vitamin C effect on mitoxantrone-induced cytotoxicity in human breast cancer cell lines. PLoS One. 2014;9(12):e115287.PubMedPubMedCentralCrossRef Guerriero E, Sorice A, Capone F, et al. Vitamin C effect on mitoxantrone-induced cytotoxicity in human breast cancer cell lines. PLoS One. 2014;9(12):e115287.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Voland C, Bord A, Péleraux A, et al. Repression of cell cycle-related proteins by oxaliplatin but not cisplatin in human colon cancer cells. Mol Cancer Ther. 2006;5(9):2149–57.PubMedCrossRef Voland C, Bord A, Péleraux A, et al. Repression of cell cycle-related proteins by oxaliplatin but not cisplatin in human colon cancer cells. Mol Cancer Ther. 2006;5(9):2149–57.PubMedCrossRef
106.
Zurück zum Zitat Lal S, Zarei M, Chand SN, et al. WEE1 inhibition in pancreatic cancer cells is dependent on DNA repair status in a context dependent manner. Sci Rep. 2016;6:33323.PubMedPubMedCentralCrossRef Lal S, Zarei M, Chand SN, et al. WEE1 inhibition in pancreatic cancer cells is dependent on DNA repair status in a context dependent manner. Sci Rep. 2016;6:33323.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Lewis CW, Jin Z, Macdonald D, et al. Prolonged mitotic arrest induced by Wee1 inhibition sensitizes breast cancer cells to paclitaxel. Oncotarget. 2017;8(43):73705–22.PubMedPubMedCentralCrossRef Lewis CW, Jin Z, Macdonald D, et al. Prolonged mitotic arrest induced by Wee1 inhibition sensitizes breast cancer cells to paclitaxel. Oncotarget. 2017;8(43):73705–22.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Chen KC, Yang TY, Wu CC, et al. Pemetrexed induces S-phase arrest and apoptosis via a deregulated activation of Akt signaling pathway. PLoS One. 2014;9(5):e97888.PubMedPubMedCentralCrossRef Chen KC, Yang TY, Wu CC, et al. Pemetrexed induces S-phase arrest and apoptosis via a deregulated activation of Akt signaling pathway. PLoS One. 2014;9(5):e97888.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Hirai H, Arai T, Okada M, et al. MK-1775, a small molecule Wee1 inhibitor, enhances antitumor efficacy of various DNA-damaging agents, including 5-fluorouracil. Cancer Biol Ther. 2010;9(7):514–22.PubMedCrossRef Hirai H, Arai T, Okada M, et al. MK-1775, a small molecule Wee1 inhibitor, enhances antitumor efficacy of various DNA-damaging agents, including 5-fluorouracil. Cancer Biol Ther. 2010;9(7):514–22.PubMedCrossRef
111.
Zurück zum Zitat Maier P, Hartmann L, Wenz F, Herskind C. Cellular pathways in response to ionizing radiation and their targetability for tumor radiosensitization. Int J Mol Sci. 2016;17(1):102.PubMedCentralCrossRef Maier P, Hartmann L, Wenz F, Herskind C. Cellular pathways in response to ionizing radiation and their targetability for tumor radiosensitization. Int J Mol Sci. 2016;17(1):102.PubMedCentralCrossRef
112.
Zurück zum Zitat Li J, Chen W, Zhang P, Li N. Topoisomerase II trapping agent teniposide induces apoptosis and G2/M or S phase arrest of oral squamous cell carcinoma. World J Surg Oncol. 2006;4:41.PubMedPubMedCentralCrossRef Li J, Chen W, Zhang P, Li N. Topoisomerase II trapping agent teniposide induces apoptosis and G2/M or S phase arrest of oral squamous cell carcinoma. World J Surg Oncol. 2006;4:41.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Wotring LL, Roti Roti JL. Thioguanine-induced S and G2 blocks and their significance to the mechanism of cytotoxicity. Cancer Res. 1980;40(5):1458–62.PubMed Wotring LL, Roti Roti JL. Thioguanine-induced S and G2 blocks and their significance to the mechanism of cytotoxicity. Cancer Res. 1980;40(5):1458–62.PubMed
114.
Zurück zum Zitat Nguyen D, Zajac-Kaye M, Rubinstein L, et al. Poly(ADP-ribose) polymerase inhibition enhances p53-dependent and -independent DNA damage responses induced by DNA damaging agent. Cell Cycle. 2011;10(23):4074–82.PubMedPubMedCentralCrossRef Nguyen D, Zajac-Kaye M, Rubinstein L, et al. Poly(ADP-ribose) polymerase inhibition enhances p53-dependent and -independent DNA damage responses induced by DNA damaging agent. Cell Cycle. 2011;10(23):4074–82.PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Shumway SD, Kubica JL, Guertin AD, et al. Abstract 2969: a Wee1 kinase inhibitor, MK-1775, sensitizes cervical carcinoma cell lines to cisplatin and topotecan. Cancer Res. 2011;71(8 Supplement):2969. Shumway SD, Kubica JL, Guertin AD, et al. Abstract 2969: a Wee1 kinase inhibitor, MK-1775, sensitizes cervical carcinoma cell lines to cisplatin and topotecan. Cancer Res. 2011;71(8 Supplement):2969.
116.
Zurück zum Zitat Brandl MB, Pasquier E, Li F, et al. Computational analysis of image-based drug profiling predicts synergistic drug combinations: applications in triple-negative breast cancer. Mol Oncol. 2014;8(8):1548–60.PubMedPubMedCentralCrossRef Brandl MB, Pasquier E, Li F, et al. Computational analysis of image-based drug profiling predicts synergistic drug combinations: applications in triple-negative breast cancer. Mol Oncol. 2014;8(8):1548–60.PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Tu Y, Cheng S, Zhang S, et al. Vincristine induces cell cycle arrest and apoptosis in SH-SY5Y human neuroblastoma cells. Int J Mol Med. 2013;31(1):113–9.PubMedCrossRef Tu Y, Cheng S, Zhang S, et al. Vincristine induces cell cycle arrest and apoptosis in SH-SY5Y human neuroblastoma cells. Int J Mol Med. 2013;31(1):113–9.PubMedCrossRef
118.
Zurück zum Zitat Visconti R, Della Monica R, Palazzo L, et al. The Fcp1-Wee1-Cdk1 axis affects spindle assembly checkpoint robustness and sensitivity to antimicrotubule cancer drugs. Cell Death Differ. 2015;22(9):1551–60.PubMedPubMedCentralCrossRef Visconti R, Della Monica R, Palazzo L, et al. The Fcp1-Wee1-Cdk1 axis affects spindle assembly checkpoint robustness and sensitivity to antimicrotubule cancer drugs. Cell Death Differ. 2015;22(9):1551–60.PubMedPubMedCentralCrossRef
119.
Zurück zum Zitat Zhu JY, Cuellar RA, Berndt N, et al. Structural basis of Wee kinases functionality and inactivation by diverse small molecule inhibitors. J Med Chem. 2017;60(18):7863–75.PubMedPubMedCentralCrossRef Zhu JY, Cuellar RA, Berndt N, et al. Structural basis of Wee kinases functionality and inactivation by diverse small molecule inhibitors. J Med Chem. 2017;60(18):7863–75.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat Restelli V, Chilà R, Lupi M, et al. Characterization of a mantle cell lymphoma cell line resistant to the Chk1 inhibitor PF-00477736. Oncotarget. 2015;6(35):37229–40.PubMedPubMedCentralCrossRef Restelli V, Chilà R, Lupi M, et al. Characterization of a mantle cell lymphoma cell line resistant to the Chk1 inhibitor PF-00477736. Oncotarget. 2015;6(35):37229–40.PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Qi W, Xie C, Li C, et al. CHK1 plays a critical role in the anti-leukemic activity of the wee1 inhibitor MK-1775 in acute myeloid leukemia cells. J Hematol Oncol. 2014;7(1):53.PubMedPubMedCentralCrossRef Qi W, Xie C, Li C, et al. CHK1 plays a critical role in the anti-leukemic activity of the wee1 inhibitor MK-1775 in acute myeloid leukemia cells. J Hematol Oncol. 2014;7(1):53.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Young LA, O’Connor LO, de Renty C, et al. Differential activity of ATR and Wee1 inhibitors in a highly sensitive subpopulation of DLBCL linked to replication stress. Cancer Res. 2019;79(14):3762–75.PubMedCrossRef Young LA, O’Connor LO, de Renty C, et al. Differential activity of ATR and Wee1 inhibitors in a highly sensitive subpopulation of DLBCL linked to replication stress. Cancer Res. 2019;79(14):3762–75.PubMedCrossRef
123.
Zurück zum Zitat Bridges KA, Hirai H, Buser CA, et al. MK-1775, a novel wee1 kinase inhibitor, radiosensitizes p53-defective human tumor cells. Clin Cancer Res. 2011;17(17):5638–48.PubMedPubMedCentralCrossRef Bridges KA, Hirai H, Buser CA, et al. MK-1775, a novel wee1 kinase inhibitor, radiosensitizes p53-defective human tumor cells. Clin Cancer Res. 2011;17(17):5638–48.PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Ma H, Takahashi A, Sejimo Y, et al. Targeting of carbon ion-induced G2 checkpoint activation in lung cancer cells using Wee-1 inhibitor MK-1775. Radiat Res. 2016;185(2):e52.CrossRef Ma H, Takahashi A, Sejimo Y, et al. Targeting of carbon ion-induced G2 checkpoint activation in lung cancer cells using Wee-1 inhibitor MK-1775. Radiat Res. 2016;185(2):e52.CrossRef
125.
Zurück zum Zitat Lindenblatt D, Terraneo N, Pellegrini G, et al. Combination of lutetium-177 labelled anti-L1CAM antibody chCE7 with the clinically relevant protein kinase inhibitor MK1775: a novel combination against human ovarian carcinoma. BMC Cancer. 2018;18(1):922.PubMedPubMedCentralCrossRef Lindenblatt D, Terraneo N, Pellegrini G, et al. Combination of lutetium-177 labelled anti-L1CAM antibody chCE7 with the clinically relevant protein kinase inhibitor MK1775: a novel combination against human ovarian carcinoma. BMC Cancer. 2018;18(1):922.PubMedPubMedCentralCrossRef
126.
Zurück zum Zitat Parsels LA, Karnak D, Parsels JD, et al. PARP1 Trapping and DNA replication stress enhance radiosensitization with combined WEE1 and PARP inhibitors. Mol Cancer Res. 2018;16(2):222–32.PubMedCrossRef Parsels LA, Karnak D, Parsels JD, et al. PARP1 Trapping and DNA replication stress enhance radiosensitization with combined WEE1 and PARP inhibitors. Mol Cancer Res. 2018;16(2):222–32.PubMedCrossRef
127.
Zurück zum Zitat Caldwell JT, Edwards H, Buck SA, et al. Targeting the wee1 kinase for treatment of pediatric Down syndrome acute myeloid leukemia. Pediatr Blood Cancer. 2014;61(10):1767–73.PubMedPubMedCentralCrossRef Caldwell JT, Edwards H, Buck SA, et al. Targeting the wee1 kinase for treatment of pediatric Down syndrome acute myeloid leukemia. Pediatr Blood Cancer. 2014;61(10):1767–73.PubMedPubMedCentralCrossRef
129.
Zurück zum Zitat Tibes R, Ferreira Coutinho D, Tuen MT, et al. DNA damage repair interference By WEE1 inhibition with AZD1775 overcomes combined azacitidine and Venetoclax resistance in acute myeloid leukmeia (AML). Blood. 2019;134(Supplement_1):2559.CrossRef Tibes R, Ferreira Coutinho D, Tuen MT, et al. DNA damage repair interference By WEE1 inhibition with AZD1775 overcomes combined azacitidine and Venetoclax resistance in acute myeloid leukmeia (AML). Blood. 2019;134(Supplement_1):2559.CrossRef
130.
Zurück zum Zitat Qi W, Zhang W, Edwards H, et al. Synergistic anti-leukemic interactions between panobinostat and MK-1775 in acute myeloid leukemia ex vivo. Cancer Biol Ther. 2015;16(12):1784–93.PubMedPubMedCentralCrossRef Qi W, Zhang W, Edwards H, et al. Synergistic anti-leukemic interactions between panobinostat and MK-1775 in acute myeloid leukemia ex vivo. Cancer Biol Ther. 2015;16(12):1784–93.PubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Tanaka N, Patel AA, Tang L, et al. Replication stress leading to apoptosis within the S-phase contributes to synergism between vorinostat and AZD1775 in HNSCC harboring high-risk TP53 mutation. Clin Cancer Res. 2017;23(21):6541–54.PubMedPubMedCentralCrossRef Tanaka N, Patel AA, Tang L, et al. Replication stress leading to apoptosis within the S-phase contributes to synergism between vorinostat and AZD1775 in HNSCC harboring high-risk TP53 mutation. Clin Cancer Res. 2017;23(21):6541–54.PubMedPubMedCentralCrossRef
132.
Zurück zum Zitat Qi W, Xu X, Wang M, et al. Inhibition of Wee1 sensitizes AML cells to ATR inhibitor VE-822-induced DNA damage and apoptosis. Biochem Pharmacol. 2019;164:273–82.PubMedCrossRef Qi W, Xu X, Wang M, et al. Inhibition of Wee1 sensitizes AML cells to ATR inhibitor VE-822-induced DNA damage and apoptosis. Biochem Pharmacol. 2019;164:273–82.PubMedCrossRef
133.
Zurück zum Zitat Restelli V, Lupi M, Chila R, et al. DNA damage response inhibitor combinations exert synergistic antitumor activity in aggressive B-cell lymphomas. Mol Cancer Ther. 2019;18(7):1255–64.PubMedCrossRef Restelli V, Lupi M, Chila R, et al. DNA damage response inhibitor combinations exert synergistic antitumor activity in aggressive B-cell lymphomas. Mol Cancer Ther. 2019;18(7):1255–64.PubMedCrossRef
134.
Zurück zum Zitat Bukhari AB, Lewis CW, Pearce JJ, et al. Inhibiting Wee1 and ATR kinases produces tumor-selective synthetic lethality and suppresses metastasis. J Clin Invest. 2019;129(3):1329–44.PubMedPubMedCentralCrossRef Bukhari AB, Lewis CW, Pearce JJ, et al. Inhibiting Wee1 and ATR kinases produces tumor-selective synthetic lethality and suppresses metastasis. J Clin Invest. 2019;129(3):1329–44.PubMedPubMedCentralCrossRef
135.
Zurück zum Zitat Jin J, Fang H, Yang F, et al. Combined inhibition of ATR and WEE1 as a novel therapeutic strategy in triple-negative breast cancer. Neoplasia (United States). 2018;20(5):478–88.CrossRef Jin J, Fang H, Yang F, et al. Combined inhibition of ATR and WEE1 as a novel therapeutic strategy in triple-negative breast cancer. Neoplasia (United States). 2018;20(5):478–88.CrossRef
136.
Zurück zum Zitat Weisberg E, Nonami A, Chen Z, et al. Identification of Wee1 as a novel therapeutic target for mutant RAS-driven acute leukemia and other malignancies. Leukemia. 2014;29(1):27–37.PubMedPubMedCentralCrossRef Weisberg E, Nonami A, Chen Z, et al. Identification of Wee1 as a novel therapeutic target for mutant RAS-driven acute leukemia and other malignancies. Leukemia. 2014;29(1):27–37.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Li F, Guo E, Huang J, et al. mTOR inhibition overcomes primary and acquired resistance to Wee1 inhibition by augmenting replication stress in epithelial ovarian cancers. Am J Cancer Res. 2020;10(3):908–24.PubMedPubMedCentral Li F, Guo E, Huang J, et al. mTOR inhibition overcomes primary and acquired resistance to Wee1 inhibition by augmenting replication stress in epithelial ovarian cancers. Am J Cancer Res. 2020;10(3):908–24.PubMedPubMedCentral
138.
Zurück zum Zitat Sen T, Tong P, Diao L, et al. Targeting AXL and mTOR pathway overcomes primary and acquired resistance to WEE1 inhibition in small-cell lung cancer. Clin Cancer Res. 2017;23(20):6239–54.PubMedPubMedCentralCrossRef Sen T, Tong P, Diao L, et al. Targeting AXL and mTOR pathway overcomes primary and acquired resistance to WEE1 inhibition in small-cell lung cancer. Clin Cancer Res. 2017;23(20):6239–54.PubMedPubMedCentralCrossRef
139.
140.
Zurück zum Zitat Chila R, Basana A, Lupi M, et al. Combined inhibition of Chk1 and Wee1 as a new therapeutic strategy for mantle cell lymphoma. Oncotarget. 2015;6(5):3394–408.PubMedCrossRef Chila R, Basana A, Lupi M, et al. Combined inhibition of Chk1 and Wee1 as a new therapeutic strategy for mantle cell lymphoma. Oncotarget. 2015;6(5):3394–408.PubMedCrossRef
141.
Zurück zum Zitat Restelli V, Vagni M, Arribas AJ, et al. Inhibition of CHK1 and WEE1 as a new therapeutic approach in diffuse large B cell lymphomas with MYC deregulation. Br J Haematol. 2018;181(1):129–33.PubMedCrossRef Restelli V, Vagni M, Arribas AJ, et al. Inhibition of CHK1 and WEE1 as a new therapeutic approach in diffuse large B cell lymphomas with MYC deregulation. Br J Haematol. 2018;181(1):129–33.PubMedCrossRef
142.
Zurück zum Zitat Chaudhuri L, Vincelette ND, Koh BD, et al. CHK1 and WEE1 inhibition combine synergistically to enhance therapeutic efficacy in acute myeloid leukemia ex vivo. Haematologica. 2014;99(4):688–96.PubMedPubMedCentralCrossRef Chaudhuri L, Vincelette ND, Koh BD, et al. CHK1 and WEE1 inhibition combine synergistically to enhance therapeutic efficacy in acute myeloid leukemia ex vivo. Haematologica. 2014;99(4):688–96.PubMedPubMedCentralCrossRef
143.
Zurück zum Zitat De Jong MRW, Langendonk M, Reitsma B, et al. WEE1 inhibition enhances anti-apoptotic dependency as a result of premature mitotic entry and DNA damage. Cancers (Basel). 2019;11(11):1743.CrossRef De Jong MRW, Langendonk M, Reitsma B, et al. WEE1 inhibition enhances anti-apoptotic dependency as a result of premature mitotic entry and DNA damage. Cancers (Basel). 2019;11(11):1743.CrossRef
144.
Zurück zum Zitat Fang Y, McGrail DJ, Sun C, et al. Sequential therapy with PARP and WEE1 inhibitors minimizes toxicity while maintaining efficacy. Cancer Cell. 2019;35(6):851–867.e7.PubMedPubMedCentralCrossRef Fang Y, McGrail DJ, Sun C, et al. Sequential therapy with PARP and WEE1 inhibitors minimizes toxicity while maintaining efficacy. Cancer Cell. 2019;35(6):851–867.e7.PubMedPubMedCentralCrossRef
145.
Zurück zum Zitat Lallo A, Frese KK, Morrow CJ, et al. The combination of the PARP inhibitor olaparib and the WEE1 Inhibitor AZD1775 as a new therapeutic option for small cell lung cancer. Clin Cancer Res. 2018;24(20):5153–64.PubMedCrossRef Lallo A, Frese KK, Morrow CJ, et al. The combination of the PARP inhibitor olaparib and the WEE1 Inhibitor AZD1775 as a new therapeutic option for small cell lung cancer. Clin Cancer Res. 2018;24(20):5153–64.PubMedCrossRef
146.
Zurück zum Zitat Garcia TB, Snedeker JC, Baturin D, et al. A small-molecule inhibitor of WEE1, AZD1775, synergizes with olaparib by impairing homologous recombination and enhancing DNA damage and apoptosis in acute leukemia. Mol Cancer Ther. 2017;16(10):2058–68.PubMedPubMedCentralCrossRef Garcia TB, Snedeker JC, Baturin D, et al. A small-molecule inhibitor of WEE1, AZD1775, synergizes with olaparib by impairing homologous recombination and enhancing DNA damage and apoptosis in acute leukemia. Mol Cancer Ther. 2017;16(10):2058–68.PubMedPubMedCentralCrossRef
147.
Zurück zum Zitat Lee JW, Parameswaran J, Sandoval-Schaefer T, et al. Combined aurora kinase A (AURKA) and WEE1 inhibition demonstrates synergistic antitumor effect in squamous cell carcinoma of the head and neck. Clin Cancer Res. 2019;25(11):3430–42.PubMedPubMedCentralCrossRef Lee JW, Parameswaran J, Sandoval-Schaefer T, et al. Combined aurora kinase A (AURKA) and WEE1 inhibition demonstrates synergistic antitumor effect in squamous cell carcinoma of the head and neck. Clin Cancer Res. 2019;25(11):3430–42.PubMedPubMedCentralCrossRef
148.
Zurück zum Zitat Chen G, Zhang B, Xu H, et al. Suppression of Sirt1 sensitizes lung cancer cells to WEE1 inhibitor MK-1775-induced DNA damage and apoptosis. Oncogene. 2017;36(50):6863–72.PubMedCrossRef Chen G, Zhang B, Xu H, et al. Suppression of Sirt1 sensitizes lung cancer cells to WEE1 inhibitor MK-1775-induced DNA damage and apoptosis. Oncogene. 2017;36(50):6863–72.PubMedCrossRef
149.
Zurück zum Zitat Francis AM, Alexander A, Liu Y, et al. CDK4/6 inhibitors sensitize Rb-positive sarcoma cells to Wee1 kinase inhibition through reversible cell-cycle arrest. Mol Cancer Ther. 2017;16(9):1751–64.PubMedPubMedCentralCrossRef Francis AM, Alexander A, Liu Y, et al. CDK4/6 inhibitors sensitize Rb-positive sarcoma cells to Wee1 kinase inhibition through reversible cell-cycle arrest. Mol Cancer Ther. 2017;16(9):1751–64.PubMedPubMedCentralCrossRef
150.
Zurück zum Zitat Takashima Y, Kikuchi E, Kikuchi J, et al. Bromodomain and extraterminal domain inhibition synergizes with WEE1-inhibitor AZD1775 effect by impairing nonhomologous end joining and enhancing DNA damage in nonsmall cell lung cancer. Int J Cancer. 2020;146(4):1114–24.PubMedCrossRef Takashima Y, Kikuchi E, Kikuchi J, et al. Bromodomain and extraterminal domain inhibition synergizes with WEE1-inhibitor AZD1775 effect by impairing nonhomologous end joining and enhancing DNA damage in nonsmall cell lung cancer. Int J Cancer. 2020;146(4):1114–24.PubMedCrossRef
151.
Zurück zum Zitat Panek RL, Lu GH, Klutchko SR, et al. In vitro pharmacological characterization of PD 166285, a new nanomolar potent and broadly active protein tyrosine kinase inhibitor. J Pharmacol Exp Ther. 1997;283(3):1433–44.PubMed Panek RL, Lu GH, Klutchko SR, et al. In vitro pharmacological characterization of PD 166285, a new nanomolar potent and broadly active protein tyrosine kinase inhibitor. J Pharmacol Exp Ther. 1997;283(3):1433–44.PubMed
152.
Zurück zum Zitat Duan L, Perez RE, Hansen M, et al. Increasing cisplatin sensitivity by scheduledependent inhibition of AKT and Chk1. Cancer Biol Ther. 2014;15(12):1600–12.PubMedPubMedCentralCrossRef Duan L, Perez RE, Hansen M, et al. Increasing cisplatin sensitivity by scheduledependent inhibition of AKT and Chk1. Cancer Biol Ther. 2014;15(12):1600–12.PubMedPubMedCentralCrossRef
153.
Zurück zum Zitat Blackwood E, Epler J, Yen I, et al. Combination drug scheduling defines a “window of opportunity” for chemopotentiation of gemcitabine by an orally bioavailable, selective ChK1 inhibitor, GNE-900. Mol Cancer Ther. 2013;12(10):1968–80.PubMedCrossRef Blackwood E, Epler J, Yen I, et al. Combination drug scheduling defines a “window of opportunity” for chemopotentiation of gemcitabine by an orally bioavailable, selective ChK1 inhibitor, GNE-900. Mol Cancer Ther. 2013;12(10):1968–80.PubMedCrossRef
154.
Zurück zum Zitat Levinson NM, Boxer SG. Structural and spectroscopic analysis of the kinase inhibitor bosutinib and an isomer of bosutinib binding to the Abl tyrosine kinase domain. PLoS One. 2012;7(4):e29828.PubMedPubMedCentralCrossRef Levinson NM, Boxer SG. Structural and spectroscopic analysis of the kinase inhibitor bosutinib and an isomer of bosutinib binding to the Abl tyrosine kinase domain. PLoS One. 2012;7(4):e29828.PubMedPubMedCentralCrossRef
155.
Zurück zum Zitat Beeharry N, Banina E, Hittle J, et al. Re-purposing clinical kinase inhibitors to enhance chemosensitivity by overriding checkpoints. Cell Cycle. 2014;13(14):2172–91.PubMedPubMedCentralCrossRef Beeharry N, Banina E, Hittle J, et al. Re-purposing clinical kinase inhibitors to enhance chemosensitivity by overriding checkpoints. Cell Cycle. 2014;13(14):2172–91.PubMedPubMedCentralCrossRef
156.
Zurück zum Zitat Mendez E, Rodriguez CP, Kao MC, et al. A phase I clinical trial of AZD1775 in combination with neoadjuvant weekly docetaxel and cisplatin before definitive therapy in head and neck squamous cell carcinoma. Clin Cancer Res. 2018;24(12):2740–8.PubMedPubMedCentralCrossRef Mendez E, Rodriguez CP, Kao MC, et al. A phase I clinical trial of AZD1775 in combination with neoadjuvant weekly docetaxel and cisplatin before definitive therapy in head and neck squamous cell carcinoma. Clin Cancer Res. 2018;24(12):2740–8.PubMedPubMedCentralCrossRef
157.
Zurück zum Zitat Do K, Wilsker D, Ji J, et al. Phase I study of single-agent AZD1775 (MK-1775), a wee1 kinase inhibitor, in patients with refractory solid tumors. J Clin Oncol. 2015;33(30):3409–15.PubMedPubMedCentralCrossRef Do K, Wilsker D, Ji J, et al. Phase I study of single-agent AZD1775 (MK-1775), a wee1 kinase inhibitor, in patients with refractory solid tumors. J Clin Oncol. 2015;33(30):3409–15.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Leijen S, Van Geel RMJM, Pavlick AC, et al. Phase I study evaluating WEE1 inhibitor AZD1775 as monotherapy and in combination with gemcitabine, cisplatin, or carboplatin in patients with advanced solid tumors. J Clin Oncol. 2016;34(36):4371–80.PubMedCrossRefPubMedCentral Leijen S, Van Geel RMJM, Pavlick AC, et al. Phase I study evaluating WEE1 inhibitor AZD1775 as monotherapy and in combination with gemcitabine, cisplatin, or carboplatin in patients with advanced solid tumors. J Clin Oncol. 2016;34(36):4371–80.PubMedCrossRefPubMedCentral
159.
Zurück zum Zitat Leijen S, Van Geel RMJM, Sonke GS, et al. Phase II study of WEE1 inhibitor AZD1775 plus carboplatin in patientswith tp53-mutated ovarian cancer refractory or resistant to first-line therapy within 3 months. J Clin Oncol. 2016;34(36):4354–61.PubMedCrossRef Leijen S, Van Geel RMJM, Sonke GS, et al. Phase II study of WEE1 inhibitor AZD1775 plus carboplatin in patientswith tp53-mutated ovarian cancer refractory or resistant to first-line therapy within 3 months. J Clin Oncol. 2016;34(36):4354–61.PubMedCrossRef
160.
Zurück zum Zitat Moore KN, Chambers SK, Hamilton EP, et al. Adavosertib with chemotherapy (CT) in patients (pts) with platinum-resistant ovarian cancer (PPROC): an open label, four-arm, phase II study. J Clin Oncol. 2019;5(_suppl):5513.CrossRef Moore KN, Chambers SK, Hamilton EP, et al. Adavosertib with chemotherapy (CT) in patients (pts) with platinum-resistant ovarian cancer (PPROC): an open label, four-arm, phase II study. J Clin Oncol. 2019;5(_suppl):5513.CrossRef
161.
Zurück zum Zitat Yap TA, Plummer R, Azad NS, Helleday T. The DNA damaging revolution: PARP inhibitors and beyond. Am Soc Clin Oncol Educ B. 2019;39:185–95.CrossRef Yap TA, Plummer R, Azad NS, Helleday T. The DNA damaging revolution: PARP inhibitors and beyond. Am Soc Clin Oncol Educ B. 2019;39:185–95.CrossRef
162.
Zurück zum Zitat Forment JV, O’Connor MJ. Targeting the replication stress response in cancer. Pharmacol Ther. 2018;188:155–67.PubMedCrossRef Forment JV, O’Connor MJ. Targeting the replication stress response in cancer. Pharmacol Ther. 2018;188:155–67.PubMedCrossRef
163.
Zurück zum Zitat Fu S, Wang Y, Keyomarsi K, Meric-Bernstein F. Strategic development of AZD1775, a Wee1 kinase inhibitor, for cancer therapy. Expert Opin Investig Drugs. 2018;27(9):741–51.PubMedCrossRef Fu S, Wang Y, Keyomarsi K, Meric-Bernstein F. Strategic development of AZD1775, a Wee1 kinase inhibitor, for cancer therapy. Expert Opin Investig Drugs. 2018;27(9):741–51.PubMedCrossRef
164.
Zurück zum Zitat Qiu Z, Oleinick NL, Zhang J. ATR/CHK1 inhibitors and cancer therapy. Radiother Oncol. 2018;126(3):450–64.PubMedCrossRef Qiu Z, Oleinick NL, Zhang J. ATR/CHK1 inhibitors and cancer therapy. Radiother Oncol. 2018;126(3):450–64.PubMedCrossRef
165.
Zurück zum Zitat Ghelli Luserna Di Rora A, Iacobucci I, Martinelli G. The cell cycle checkpoint inhibitors in the treatment of leukemias. J Hematol Oncol. 2017;10(1):77.PubMedPubMedCentralCrossRef Ghelli Luserna Di Rora A, Iacobucci I, Martinelli G. The cell cycle checkpoint inhibitors in the treatment of leukemias. J Hematol Oncol. 2017;10(1):77.PubMedPubMedCentralCrossRef
166.
Zurück zum Zitat Lewis CW, Bukhari AB, Xiao EJ, et al. Upregulation of MyT1 promotes acquired resistance of cancer cells to WEE1 inhibition. Cancer Res. 2019;79(23):5971–85.PubMedCrossRef Lewis CW, Bukhari AB, Xiao EJ, et al. Upregulation of MyT1 promotes acquired resistance of cancer cells to WEE1 inhibition. Cancer Res. 2019;79(23):5971–85.PubMedCrossRef
167.
Zurück zum Zitat Garcia TB, Uluisik RC, van Linden AA, et al. Increased HDAC activity and c-MYC expression mediate acquired resistance to WEE1 inhibition in acute leukemia. Front Oncol. 2020;10:296.PubMedPubMedCentralCrossRef Garcia TB, Uluisik RC, van Linden AA, et al. Increased HDAC activity and c-MYC expression mediate acquired resistance to WEE1 inhibition in acute leukemia. Front Oncol. 2020;10:296.PubMedPubMedCentralCrossRef
Metadaten
Titel
A WEE1 family business: regulation of mitosis, cancer progression, and therapeutic target
verfasst von
Andrea Ghelli Luserna di Rorà
Claudio Cerchione
Giovanni Martinelli
Giorgia Simonetti
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Journal of Hematology & Oncology / Ausgabe 1/2020
Elektronische ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-020-00959-2

Weitere Artikel der Ausgabe 1/2020

Journal of Hematology & Oncology 1/2020 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.