Elsevier

DNA Repair

Volume 107, November 2021, 103203
DNA Repair

Review Article
Targeting Wee1 kinase as a therapeutic approach in Hematological Malignancies

https://doi.org/10.1016/j.dnarep.2021.103203Get rights and content

Abstract

Hematologic malignancies include various diseases that develop from hematopoietic stem cells of bone marrow or lymphatic organs. Currently, conventional DNA-damage-based chemotherapy drugs are approved as standard therapeutic regimens for these malignancies. Although many improvements have been made, patients with relapsed or refractory hematological malignancies have a poor prognosis. Therefore, novel and practical therapeutic approaches are required for the treatment of these diseases. Interestingly several studies have shown that targeting Wee1 kinase in the Hematological malignancies, including AML, ALL, CML, CLL, DLBCL, BL, MCL, etc., can be an effective therapeutic strategy. It plays an essential role in regulating the cell cycle process by abrogating the G2–M cell-cycle checkpoint, which provides time for DNA damage repair before mitotic entry. Consistently, Wee1 overexpression is observed in various Hematological malignancies. Also, in healthy normal cells, repairing DNA damages occurs due to G1-S checkpoint function; however, in the cancer cells, which have an impaired G1–S checkpoint, the damaged DNA repair process depends on the G2–M checkpoint function. Thus, Wee1 inhibition could be a promising target in the presence of DNA damage in order to potentiate multiple therapeutic drugs. This review summarized the potentials and challenges of Wee1 inhibition combined with other therapies as a novel effective therapeutic strategy in Hematological malignancies.

Introduction

Hematological malignancies include various malignant transformations originating from the blood, bone marrow, and lymphatic system. Three major types of these malignancies are leukemia, lymphoma, and multiple myeloma [1]. Current treatment of these diseases includes chemotherapy, radiotherapy, stem cell transplantation, and immunotherapy. However, due to the low specificity and high toxicity, the utility of chemotherapeutic drugs for Hematological malignancies is limited [2,3]. Thereby, there is a striking necessity to improve the efficacy and activity of conventional chemotherapeutic drugs.

Studies have shown that the cell cycle checkpoints play a critical role in malignant cells ' fate in exposure to anticancer treatments [4]. Furthermore, a genome-wide functional screen among the proteins in Dependency Map (DepMap) portal (www.depmap.org) and Oncomine database (www.Oncomine.org) demonstrates Wee1 kinase overexpression in the malignant cells compared to the normal cells. According to data from the DepMap site, liquid tumor cell lines are more sensitive to blockade of the Wee1 than solid tumor cell lines. Among hematopoietic cancer cell lines, the LOUCY cell line (related to ALL) is the most sensitive and the MLMA cell line (related to hairy cell leukemia) is the most resistant cell line to blockade of the Wee1. Also, among the solid tumor cell lines, CPCN cell line (related to SCLC disease) is the most sensitive and PCL15A cell line (related to head and neck cancer) is the most resistant cell line. Wee1, as a critical G2-M and G1-S checkpoints kinase, regulates cellular fate decisions consisting of progression, arrest, or apoptosis induction in the cell cycle process [5,6]. In response to the presence of DNA damage, Wee1-inhibitory phosphorylation of cyclin-dependent kinase 1 (CDK1) results in delay in the cell cycle progression [7], which provides time for DNA-damage repair, thus enable malignant cells to complete the cell cycle process by achieving sufficient genomic integrity [8]. In this regard, it has been demonstrated that genetic or pharmacologic inhibition of Wee1, together with anticancer treatments, inhibits more proliferation and induces apoptosis in malignant cell lines and primary patient samples, which validated the Wee1 kinase targeting potential as a therapeutic approach in Hematological malignancies [9]. This efficacy enhancement is due to abrogated cell cycle arrest that enforces mitosis entry in the cells harboring DNA damages and induces apoptosis in mitotic catastrophe process [[10], [11], [12]]. Thus, there is a rationale for targeting Wee1 to enhance the efficacy of conventional treatments that act by DNA damage induction [13].

The potent WEE1 inhibitor AZD1775 has advanced to clinical trials in combination with DNA-damaging agents such as carboplatin, cisplatin, docetaxel, 5-fluorouracil, gemcitabine, irinotecan, paclitaxel, pemetrexed, temozolomide, topotecan, or irradiation in various cancer types [[14], [15], [16], [17]]. Current clinical data in many cancer types with different genetic abnormalities support a combination of AZD1775 with DNA damaging agents. In addition, it has been demonstrated that AZD1775 can enhance the antitumor efficacy of anticancer therapeutics. This diverse effectiveness demonstrates that the action of AZD1775 may be dependent on a variety of cellular mechanisms [18].

In recent years, several studies devoted to improving these patients' therapy have led to the development of novel combined Wee1 inhibition and conventional treatments [9,[19], [20], [21], [22], [23]] (Table 1, Table 2, Table 3). This review describes current studies on Wee1 inhibition as a targeted therapy combined with other available treatments and limitations and potential advantages of these combination approaches to treat various types of Hematological malignancies, which hopefully will translate soon into a benefit to clinical practice in oncology.

Section snippets

Biology and function of Wee1

Historically Wee1 tyrosine kinase was first discovered in Schizosaccharomyces pombe yeast in 1978. The genes that had mutated in the yeast due to high temperatures, and cause yeast's reduced size through cell division, were called Wee1 [24]. This gene was then found in human cells, which was essential for the survival of mammalian embryos [25]. Several checkpoints, including the G1-S, S phase, and G2-M checkpoints, are in the cell cycle (Fig. 1). The prominent and well-known role of Wee1 kinase

Mitotic catastrophe

As mentioned above, Wee1 as a key regulator of G2-M transition, activates the G2-M checkpoint in the presence of substantial DNA damages [12]. Wee1 kinase, by induction of G2-M cell cycle arrest, allows cancer cells to accumulate mutations without regulatory mechanisms such as apoptosis [43]. Cancer cells with substantial DNA damages, by the initiation of premature mitosis will, die in apoptotic pathways called mitotic catastrophe, but Wee1 kinase activation prevents DNA damaged cells from

Replication stress and DNA damage responses

Specific molecular mechanisms are responsible for fixing most transient stalled replication fork cases, such as DNA damage response. However, sometimes persistent stalled replication fork activates specific cellular responses such as replication stress to ensure proper DNA replication before mitosis initiation [48].

In brief, Wee1, as a part of cell cycle checkpoints in response to DNA damages inhibits cell cycle progression and induces DNA repair process activation, thus playing a critical role

Wee1 inhibitors for cancer therapy

AZD1775 (previously known as MK1775; Merck KGaA, now Adavosertib; AstraZeneca) is a specific Wee1 kinase inhibitor. However, it has been shown that AZD1775 have a binding affinity to plk1 kinase and jak2, 3, and some other kinases [[67], [68], [69]]. Numerous studies have revealed the antitumor effects of AZD1775, either alone or in combination with other treatments [[70], [71], [72], [73], [74], [75]]. Based on studies that evaluate the effects of AZD1775 as a single agent against cancer

Wee1 inhibition combined with cytarabine

In acute myeloid leukemia (AML) treatment, Ara-C (cytarabine) is utilized both as a single agent and in combination with other drugs [89,90]. Since single Ara-C utility in refractory or relapsed AML has insufficient efficacy, it is necessary to find therapeutics to promote Ara-C efficiency. Further studies have shown G2-M checkpoint activation upon Ara-C treatment in AML cells [91,92]. Moreover, Wee1-inhibitory phosphorylation of CDK1 in this checkpoint results in cell cycle progression arrest.

Combined Wee1 inhibition and Plk1 blockers

Plk1 (polo-like kinase 1) is the early trigger of G2-M transition through activation of CDC25C that dephosphorylates and activates the Cdk2. In CML cancer cells, Plk1 inhibition results in G2/M cell cycle arrest followed by cell death. In the treatment of CML, both Danusertib and Volasertib (Plk1 inhibitors) effectively inhibited cell growth by forcing cells to stop the cell cycle and inducing apoptosis. Furthermore, Plk1 inhibition increased the G2/M and decreased the G0/G1 cells' ratio in a

Combined Wee1 inhibition with doxorubicin

Interestingly, Wee1 kinase has an essential role in the G2-M checkpoint (which ensures chromatin integrity and cell cycle progression) [25] and plays an essential role in histone synthesis regulation and consequently spindle formation [128]. In this regard, Wee1 prevents histone synthesis by inhibitory phosphorylation of histone H2B to maintain an appropriate balance in the histone/DNA ratio before mitosis. Thus, inhibition of Wee1 directly induces S-phase transition to mitosis without

Wee1 inhibition in CLL

To our knowledge, there is no study evaluating Wee1 inhibition combined with other therapeutic agents. However, several studies demonstrated that Wee1 as G2 checkpoint Kinase has high expression levels in primary CLL patient-derived samples. Regarding the critical role of Wee1 in the DDR pathway, Wee1 inhibition can be considered a therapeutic target in CLL [156]. Consistently, Wee1 mRNA overexpression has been reported in CLL patients compared to healthy control groups. It has been

Combined Wee1 and CHK1 inhibition in DLBCL

Diffuse large B-cell lymphoma (DLBCL) is the most common type of Non-Hodgkin Lymphoma with an invasive prognosis. MYC oncogene mutations are common in DLBCL patients, representing a more aggressive phenotype and poor prognosis [159]. Furthermore, high MYC protein expression in DLBCLs is also associated with a poor prognosis. Therefore, MYC inhibition is a rational target in the treatment of DLBCL; however, the inability for effective targeting limited its utility. It has been demonstrated that

Combined Wee1 and Plk1 inhibition in SM

Advanced systemic mastocytosis (SM) patients exhibit a weak response to conventional drugs and poor prognosis. Tyrosine kinase inhibitor myostosterone has recently been approved for mastocytosis; however, some patients demonstrate resistance or relapse after treatment. Plk1 inhibition (by volasertib and danusertib) in the SM leukemia cell lines and primary neoplastic MCs obtained from SM patients indicates that Plk1 phosphorylation results in inhibited growth induced apoptotic cell death in

Concluding remarks

Many anticancer therapeutic drugs have been developed to inhibit various cellular and/or molecular pathways. The association between Wee1 and hematologic malignancies is well established. Inhibitors of Wee1 (such as AZD1775) were shown to induce apoptosis in malignant cells selectively and have been extensively investigated as a single agent and combined with other drugs in several malignancies. AZD1775 was an essential inhibitor in preclinical studies in which silencing of the Wee1 gene by

Declaration of Competing Interest

The authors report no declarations of interest.

Acknowledgment

We would like to thank the Tabriz University of Medical Sciences for financial support of this study (grant number: 64476).

References (184)

  • L.I. Toledo et al.

    Targeting ATR and Chk1 kinases for cancer treatment: a new model for new (and old) drugs

    Mol. Oncol.

    (2011)
  • A.N. Blackford et al.

    ATM, ATR, and DNA-PK: the trinity at the heart of the DNA damage response

    Mol. Cell

    (2017)
  • A. Ciccia et al.

    The DNA damage response: making it safe to play with knives

    Mol. Cell

    (2010)
  • R. Buisson et al.

    Distinct but concerted roles of ATR, DNA-PK, and Chk1 in countering replication stress during S phase

    Mol. Cell

    (2015)
  • V. D’Angiolella et al.

    Cyclin F-mediated degradation of ribonucleotide reductase M2 controls genome integrity and DNA repair

    Cell

    (2012)
  • K.C. Cuneo et al.

    Wee1 kinase inhibitor AZD1775 radiosensitizes hepatocellular carcinoma regardless of TP53 mutational status through induction of replication stress

    Int. J. Radiat. Oncol. Biol. Phys.

    (2016)
  • S.X. Pfister et al.

    Inhibiting WEE1 selectively kills histone H3K36me3-deficient cancers by dNTP starvation

    Cancer Cell

    (2015)
  • G. Wang et al.

    Synergistic antitumor interactions between MK-1775 and panobinostat in preclinical models of pancreatic cancer

    Cancer Lett.

    (2015)
  • R.A. Mesa et al.

    Heat shock protein 90 inhibition sensitizes acute myelogenous leukemia cells to cytarabine

    Blood

    (2005)
  • R. Tibes et al.

    RNAi screening of the kinome with cytarabine in leukemias

    Blood

    (2012)
  • D.E. Banker et al.

    Cell cycle perturbations in acute myeloid leukemia samples following in vitro exposures to therapeutic agents

    Leuk. Res.

    (1998)
  • P. Bose et al.

    Histone deacetylase inhibitor (HDACI) mechanisms of action: emerging insights

    Pharmacol. Ther.

    (2014)
  • S.E. Mir et al.

    In silico analysis of kinase expression identifies WEE1 as a gatekeeper against mitotic catastrophe in glioblastoma

    Cancer Cell

    (2010)
  • G. Visani et al.

    Nanomedicine strategies for hematological malignancies: what is next?

    Nanomedicine

    (2014)
  • G. Ghalamfarsa et al.

    The role of natural killer T cells in B cell malignancies

    Tumor Biol.

    (2013)
  • A.K. Burnett et al.

    Attempts to optimize induction and consolidation treatment in acute myeloid leukemia: results of the MRC AML12 trial

    J. Clin. Oncol.

    (2010)
  • C.C. Chen et al.

    CHK1 inhibition as a strategy for targeting Fanconi Anemia (FA) DNA repair pathway deficient tumors

    Mol. Cancer

    (2009)
  • S.H. Cho et al.

    Chk1 is essential for tumor cell viability following activation of the replication checkpoint

    Cell Cycle

    (2005)
  • H. Niida et al.

    Specific role of Chk1 phosphorylations in cell survival and checkpoint activation

    Mol. Cell. Biol.

    (2007)
  • K.D. Davies et al.

    Chk1 inhibition and Wee1 inhibition combine synergistically to impede cellular proliferation

    Cancer Biol. Ther.

    (2011)
  • C.C. Porter 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)
  • H. Hirai et al.

    MK-1775, a small molecule Wee1 inhibitor, enhances anti-tumor efficacy of various DNA-damaging agents, including 5-fluorouracil

    Cancer Biol. Ther.

    (2010)
  • H. Hirai et al.

    Small-molecule inhibition of Wee1 kinase by MK-1775 selectively sensitizes p53-deficient tumor cells to DNA-damaging agents

    Mol. Cancer Ther.

    (2009)
  • P.C.D.W. Hamer et al.

    WEE1 kinase targeting combined with DNA-damaging cancer therapy catalyzes mitotic catastrophe

    Clin. Cancer Res.

    (2011)
  • A. Stathis et al.

    Targeting Wee1-like protein kinase to treat cancer

    Drug News Perspect.

    (2010)
  • S. Leijen et al.

    A phase I pharmacological and pharmacodynamic study of MK-1775, a Wee1 tyrosine kinase inhibitor, in monotherapy and combination with gemcitabine, cisplatin, or carboplatin in patients with advanced solid tumors

    J. Clin. Oncol.

    (2010)
  • J. Schellens et al.

    A phase I and pharmacological study of MK-1775, a Wee1 tyrosine kinase inhibitor, in both monotherapy and in combination with gemcitabine, cisplatin, or carboplatin in patients with advanced solid tumors

    J. Clin. Oncol.

    (2009)
  • J. Schellens et al.

    Update on a phase I pharmacologic and pharmacodynamic study of MK-1775, a Wee1 tyrosine kinase inhibitor, in monotherapy and combination with gemcitabine, cisplatin, or carboplatin in patients with advanced solid tumors

    J. Clin. Oncol.

    (2011)
  • S. Leijen et al.

    Phase II study of WEE1 inhibitor AZD1775 plus carboplatin in patients with TP53-mutated ovarian cancer refractory or resistant to first-line therapy within 3 months

    J. Clin. Oncol.

    (2016)
  • T.B. Garcia 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)
  • M. Cozzi et al.

    Antitumor activity of new pyrazolo [3, 4-d] pyrimidine SRC kinase inhibitors in Burkitt lymphoma cell lines and its enhancement by WEE1 inhibition

    Cell Cycle

    (2012)
  • A. Ghelli Luserna Di Rorà et al.

    Synergism through WEE1 and CHK1 inhibition in acute lymphoblastic leukemia

    Cancers

    (2019)
  • S. Rana et al.

    Deregulated expression of circadian clock and clock-controlled cell cycle genes in chronic lymphocytic leukemia

    Mol. Biol. Rep.

    (2014)
  • L. Zhou et al.

    A regimen combining the Wee1 inhibitor AZD1775 with HDAC inhibitors targets human acute myeloid leukemia cells harboring various genetic mutations

    Leukemia

    (2015)
  • P. Thuriaux et al.

    Mutants altered in the control co-ordinating cell division with cell growth in the fission yeast Schizosaccharomyces pombe

    Mol. Gen. Genet.

    (1978)
  • C.H. McGowan et al.

    Cell cycle regulation of human WEE1

    EMBO J.

    (1995)
  • K. Do et al.

    Wee1 kinase as a target for cancer therapy

    Cell Cycle

    (2013)
  • M.B. Kastan et al.

    Cell-cycle checkpoints and cancer

    Nature

    (2004)
  • M. Malumbres et al.

    Cell cycle, CDKs and cancer: a changing paradigm

    Nat. Rev. Cancer

    (2009)
  • E. Aleem et al.

    Mouse models of cell cycle regulators: new paradigms

    Cell Cycle Regulation

    (2006)
  • Cited by (0)

    View full text