Targeting CDK7 in oncology: The avenue forward

https://doi.org/10.1016/j.pharmthera.2022.108229Get rights and content

Abstract

Cyclin-dependent kinase (CDK) 7 is best characterized for the ability to regulate biological processes, including the cell cycle and gene transcription. Abnormal CDK7 activity is observed in various tumours and represents a driving force for tumourigenesis. Therefore, CDK7 may be an appealing target for cancer treatment. Whereas, the enthusiasm for CDK7-targeted therapeutic strategy is mitigated due to the widely possessed belief that this protein is essential for normal cells. Indeed, the fact confronts the consensus. This is the first review to introduce the role of CDK7 in pan-cancers via a combined analysis of comprehensive gene information and (pre)clinical research results. We also discuss the recent advances in protein structure and summarize the understanding of mechanisms underlying CDK7 function. These endeavours highlight the pivotal roles of CDK7 in tumours and may contribute to the development of effective CDK7 inhibitors within the strategy of structure-based drug discovery for cancer therapy.

Introduction

All living organisms on Earth must cope with constant changes in the extracellular and intracellular environment. The development of elegant cell cycle pathways and transcriptional programs allows cells to respond to stimuli by regulating cell division and gene expression, which are critical for cell proliferation and growth (Malumbres, 2014). Cyclin-dependent protein kinases (CDKs) are identified as core components of these fundamental processes, for which a Nobel Prize was awarded in 2001 (Whittaker, Mallinger, Workman, & Clarke, 2017). Uncontrolled proliferative ability and gene dysregulation are hallmarks of cancer (Zielinska & Katanaev, 2019). For two decades, targeting CDKs to block cell division and promote apoptosis has gained more attention in cancer treatment (Pack, Daigh, Chung, & Meyer, 2021; Parua & Fisher, 2020).

As a prominent member of the CDK family, CDK7 in combination with cyclin H and MAT1, serves as the CDK-activating kinase complex (CAK) to phosphorylate the cell cycle-associated CDKs and the carboxy-terminal repeat domain (CTD) of RNA polymerase II (Pol II) (Chen et al., 2021; Rengachari, Schilbach, Aibara, Dienemann, & Cramer, 2021). These two distinct roles have been elegantly portrayed and placed CDK7 under the spotlight of oncology. Amplification of CDK7 has been observed in several cancer types and is correlated with poor prognosis (Patel et al., 2016; Zeng et al., 2021). In last several years, significant efforts from the pharmaceutical industry and academia have ensured CDK7 of seat among the appealing targets for cancer therapy (Wei et al., 2021; Zhang et al., 2020; Zhang et al., 2020; Zhang et al., 2020). Specifically, a series of selective inhibitors of CDK7 have been developed with effective pro-apoptotic and anti-tumour activity, revealing that CDK7 affects the transcription of key cancer-dependent genes, thereby leading to abnormal activation of classical cancer pathways (Kalan et al., 2017; Zhou et al., 2019). Currently, two active phase I clinical trials involving CDK7 inhibitors (SY-5609 and CT7001) have been conducted in patients with breast cancer, pancreatic cancer, and advanced solid malignancies (including small cell lung cancer (SCLC) and ovarian cancer) (Marineau et al., 2022; Sava, Fan, Coombes, Buluwela, & Ali, 2020). To date, two studies have elucidated the structure of CDK7 and its relevant complex, CAK. In 2004, the first X-ray protein structure of human CDK7-ATP was outlined (Lolli, Lowe, Brown, & Johnson, 2004). CAK was structurally unrevealed until 2020 (Greber et al., 2020). The resolution of the cryo-electron microscopy (cryo-EM) structure of human CDK7-cyclin H-MAT1-ATPγS was determined at 2.8 Å. In the same year, cryo-EM structures of human CAK in complex with THZ1 and CT7001 were determined at 3.3 Å and 2.5 Å, respectively (Greber et al., 2020; Greber, Remis, Ali, & Nogales, 2021). These refined structures offer vital insights into CDK7 function and may provide an avenue for structure-based drug discovery.

This review highlights the recent advances in the structure and function of CDK7 and analyses CDK7 gene-related information in various cancer types as well as in classical tumour signalling pathways using bioinformatics tools. Moreover, it focuses on the new CDK7 blockers for cancer therapy and briefly discusses CDK7 blockers that can be developed in the future. This is the first review to display an overview painting of CDK7, which confirms the promising status of CDK7 as an anti-tumour target via a combined analysis of comprehensive gene information and (pre)clinical research results.

Section snippets

The CDK family

Based on the kinase domain sequence, the CDK family belongs to the CMGC group of kinases, along with glycogen synthase kinase-3β (Gsk3β), mitogen-activated protein kinases (MAPKs), CDK-like kinases, and members of the dual-specificity tyrosine-regulated kinase (DYRK) family (Sang et al., 2019). Evolutionary relationships between CDKs have been identified (Fig. 1), indicating that these CDKs can be divided into two subfamilies: cell cycle-associated CDKs (CDKs 1–6 and CDKs 14–18) and

Structural biology and mechanism related to CDK7

In the early 1990s, CDK7 was discovered as a component of a protein complex that phosphorylates and activates cell cycle–associated CDKs (Fisher, 2019). In parallel, in a seemingly unrelated field-gene transcription, researchers focused on protein purification within the human transcription factor TFIIH complex, which catalyzes RNA Pol II, and identified these proteins as a CDK7-Cyclin H-MAT1 trimer (CAK module). Although repeated observations confirmed that CDK7 impinged on the cell cycle and

CDK7 and cancer

Cancer is characterized by uncontrolled cell division and transcription. Therefore, understanding the underlying mechanisms is vital for effective cancer therapy. CDK7 emerges as an important regulatory enzyme that drives both cell cycle phase transitions and gene transcription to ensure an organized cell division and proliferation. Increasing evidence reveals that deregulated CDK7 is involved in the progression of various cancers (Huang et al., 2021; Patel et al., 2016; Yuan, Li, & Yu, 2022).

Rationale for targeting CDK7

In normal cells, cyclin activity is tightly controlled by specific transcription, protein degradation, and several CDK proteins (Sherr & Roberts, 2004). However, these mechanisms are generally dysregulated in most human cancers, resulting in anomalous cell cycle-related proteins (Ingham & Schwartz, 2017). Moreover, genetic lesions within the essential cell cycle machinery cause hyperactivation and contribute to the development of most tumour types. Notably, CDK7, a member of the CDKs, could

Conclusions

The journey of serine-threonine kinase inhibitors began in 2009 with everolimus (the approval of mTOR inhibitor) in the treatment of advanced kidney cancer (Colwell, 2016), followed by the approval of inhibitors targeting other serine-threonine kinases such as CDKs. The development and FDA approval of CDK4 and CDK6 inhibitors—Pfizer's palbociclib in 2015 for the treatment of HR+/HER2 breast cancer represents a significant milestone in cancer chemotherapy (Beaver et al., 2015). This has

Conflict of interest statement

The authors declare that there are no conflicts of interest.

Acknowledgments

This work was supported by the Natural Science Foundation of Shaanxi Province 2022JQ-815 and National Natural Science Foundation of China 31771119 and 31972902.

References (140)

  • R.P. Fisher et al.

    Alternative mechanisms of Cak assembly require an assembly factor or an activating kinase

    Cell

    (1995)
  • B.J. Greber et al.

    2.5 angstrom-resolution structure of human CDK-activating kinase bound to the clinical inhibitor ICEC0942

    Biophysical Journal

    (2021)
  • R. Jeselsohn et al.

    Allele-specific chromatin recruitment and therapeutic vulnerabilities of ESR1 activating mutations

    Cancer Cell

    (2018)
  • S. Kalan et al.

    Activation of the p53 transcriptional program sensitizes cancer cells to Cdk7 inhibitors

    Cell Reports

    (2017)
  • G. Lolli et al.

    The crystal structure of human CDK7 and its protein recognition properties

    Structure

    (2004)
  • S. Nagaraja et al.

    Transcriptional dependencies in diffuse intrinsic pontine glioma

    Cancer Cell

    (2017)
  • C.M. Olson et al.

    Development of a selective CDK7 covalent inhibitor reveals predominant cell-cycle phenotype

    Cell Chemical Biology

    (2019)
  • G. Petroni et al.

    Cancer immunotherapy with CDK7 inhibitors

    Trends in Cancer

    (2020)
  • R. Roskoski

    Cyclin-dependent protein serine/threonine kinase inhibitors as anticancer drugs

    Pharmacological Research

    (2019)
  • M.M. Schachter et al.

    A Cdk7-Cdk4 T-loop phosphorylation cascade promotes G1 progression

    Molecular Cell

    (2013)
  • C.J. Sherr et al.

    The RB and p53 pathways in cancer

    Cancer Cell

    (2002)
  • E.K. Ainscow et al.

    CT7001: An orally bioavailable CDK7 inhibitor is a potential therapy for breast, small-cell lung and haematological cancers

    Cancer Research

    (2018)
  • S. Ali et al.

    The development of a selective cyclin-dependent kinase inhibitor that shows antitumor activity

    Cancer Research

    (2009)
  • Y.M. Attia et al.

    Targeting CDK7 reverses tamoxifen resistance through regulating stemness in ER+ breast cancer

    Pharmacological Reports

    (2022)
  • Y.M. Attia et al.

    Blockade of CDK7 reverses endocrine therapy resistance in breast cancer

    International Journal of Molecular Sciences

    (2020)
  • J.A. Beaver et al.

    FDA approval: Palbociclib for the treatment of postmenopausal patients with Estrogen receptor-positive, HER2-negative metastatic breast cancer

    Clinical Cancer Research

    (2015)
  • D. Bhurta et al.

    Analyzing the scaffold diversity of cyclin-dependent kinase inhibitors and revisiting the clinical and preclinical pipeline

    Medicinal Research Reviews

    (2022)
  • X. Bisteau et al.

    CDK4 T172 phosphorylation is central in a CDK7-dependent bidirectional CDK4/CDK2 interplay mediated by p21 phosphorylation at the restriction point

    PLoS Genetics

    (2013)
  • F. Cayrol et al.

    THZ1 targeting CDK7 suppresses STAT transcriptional activity and sensitizes T-cell lymphomas to BCL2 inhibitors

    Nature Communications

    (2017)
  • F.X. Chen et al.

    Born to run: Control of transcription elongation by RNA polymerase II

    Nature Reviews Molecular Cell Biology

    (2018)
  • X.Z. Chen et al.

    Structures of the human mediator and mediator-bound preinitiation complex

    Science

    (2021)
  • Y.J. Choi et al.

    Efficacy of the novel CDK7 inhibitor QS1189 in mantle cell lymphoma

    Scientific Reports

    (2019)
  • J. Chou et al.

    Transcription-associated cyclin-dependent kinases as targets and biomarkers for cancer therapy

    Cancer Discovery

    (2020)
  • J. Colwell

    FDA approves drug combo for kidney cancer

    Cancer Discovery

    (2016)
  • P. Cramer

    Organization and regulation of gene transcription

    Nature

    (2019)
  • F.B. Dang et al.

    Ubiquitin signaling in cell cycle control and tumorigenesis

    Cell Death and Differentiation

    (2021)
  • B. Decaesteker et al.

    TBX2 is a neuroblastoma core regulatory circuitry component enhancing MYCN/FOXM1 reactivation of DREAM targets

    Nature Communications

    (2018)
  • G. Della Chiara et al.

    Epigenomic landscape of human colorectal cancer unveils an aberrant core of pan-cancer enhancers orchestrated by YAP/TAZ

    Nature Communications

    (2021)
  • I. Dogan Turacli et al.

    Flavopiridol's effects on metastasis in KRAS mutant lung adenocarcinoma cells

    Journal of Cellular Biochemistry

    (2019)
  • A.D. Durbin et al.

    Selective gene dependencies in MYCN-amplified neuroblastoma include the core transcriptional regulatory circuitry

    Nature Genetics

    (2018)
  • R.P. Fisher

    Cdk7: A kinase at the core of transcription and in the crosshairs of cancer drug discovery

    Transcription

    (2019)
  • M. Ganuza et al.

    Genetic inactivation of Cdk7 leads to cell cycle arrest and induces premature aging due to adult stem cell exhaustion

    EMBO Journal

    (2012)
  • Y. Gao et al.

    Synergistic anti-tumor effect of combining selective CDK7 and BRD4 inhibition in neuroblastoma

    Frontiers in Oncology

    (2022)
  • S. Goel et al.

    Targeting CDK4 and CDK6 in cancer

    Nature Reviews. Cancer

    (2022)
  • M.N. Gonzalez et al.

    Causes and consequences of RNA polymerase II stalling during transcript elongation

    Nature Reviews Molecular Cell Biology

    (2021)
  • E.K. Grant et al.

    A photoaffinity displacement assay and probes to study the cyclin-dependent kinase family

    Angewandte Chemie, International Edition

    (2019)
  • B.J. Greber et al.

    The cryo-electron microscopy structure of human transcription factor IIH

    Nature

    (2017)
  • B.J. Greber et al.

    The cryoelectron microscopy structure of the human CDK-activating kinase

    Proceedings of the National Academy of Sciences of the United States of America

    (2020)
  • B.J. Greber et al.

    The complete structure of the human TFIIH core complex

    Elife

    (2019)
  • S.A. Greenall et al.

    Cyclin-dependent kinase 7 is a therapeutic target in high-grade glioma

    Oncogenesis

    (2017)
  • Cited by (8)

    View all citing articles on Scopus
    View full text