A patent review of selective CDK9 inhibitors in treating cancer

ABSTRACT Introduction The dysregulation of CDK9 protein is greatly related to the proliferation and differentiation of various cancers due to its key role in the regulation of RNA transcription. Moreover, CDK9 inhibition can markedly downregulate the anti-apoptotic protein Mcl-1 which is essential for the survival of tumors. Thus, targeting CDK9 is considered to be a promising strategy for antitumor drug development, and the development of selective CDK9 inhibitors has gained increasing attention. Areas covered This review focuses on the development of selective CDK9 inhibitors reported in patent publications during the period 2020–2022, which were searched from SciFinder and Cortellis Drug Discovery Intelligence. Expert opinion Given that pan-CDK9 inhibitors may lead to serious side effects due to poor selectivity, the investigation of selective CDK9 inhibitors has attracted widespread attention. CDK9 inhibitors make some advance in treating solid tumors and possess the therapeutic potential in EGFR-mutant lung cancer. CDK9 inhibitors with short half-life and intravenous administration might result in transient target engagement and contribute to a better safety profile in vivo. However, more efforts are urgently needed to accelerate the development of CDK9 inhibitors, including the research on new binding modes between ligand and receptor or new protein binding sites.

[1]  Jung-Ae Kim,et al.  CDK9 inhibitors downregulate DKK1 expression to suppress the metastatic potential of HCC cells , 2023, Genes & Genomics.

[2]  Zhanxiang Wang,et al.  The CDK inhibitor AT7519 inhibits human glioblastoma cell growth by inducing apoptosis, pyroptosis and cell cycle arrest , 2023, Cell Death & Disease.

[3]  Oula El Atat,et al.  Molecular targeted therapy: A new avenue in glioblastoma treatment. , 2022, Oncology letters.

[4]  G. Curigliano,et al.  ‘Why is survival with triple negative breast cancer so low? insights and talking points from preclinical and clinical research’ , 2022, Expert opinion on investigational drugs.

[5]  Xiu-ming Lu,et al.  C-MYC Inhibited Ferroptosis and Promoted Immune Evasion in Ovarian Cancer Cells through NCOA4 Mediated Ferritin Autophagy , 2022, Cells.

[6]  S. Peña-Llopis,et al.  The non-apoptotic function of Caspase-8 in negatively regulating the CDK9-mediated Ser2 phosphorylation of RNA polymerase II in cervical cancer , 2022, Cellular and Molecular Life Sciences.

[7]  M. Loda,et al.  Castration-resistant prostate cancer cells are dependent on the high activity of CDK7. , 2022, Journal of cancer research and clinical oncology.

[8]  S. Knapp,et al.  Macrocyclization of Quinazoline-Based EGFR Inhibitors Leads to Exclusive Mutant Selectivity for EGFR L858R and Del19. , 2022, Journal of medicinal chemistry.

[9]  J. Byrd,et al.  VIP152 is a selective CDK9 inhibitor with pre-clinical in vitro and in vivo efficacy in chronic lymphocytic leukemia , 2022, Leukemia.

[10]  Tian Xie,et al.  Recent advances of novel fourth generation EGFR inhibitors in overcoming C797S mutation of lung cancer therapy. , 2022, European journal of medicinal chemistry.

[11]  S. Hehlgans,et al.  Prognostic Impact of Caspase-8, CDK9 and Phospho-CDK9 (Thr 186) Expression in Patients with Uterine Cervical Cancer Treated with Definitive Chemoradiation and Brachytherapy , 2022, Cancers.

[12]  W. Liu,et al.  SNS-032 attenuates liver fibrosis by anti-active hepatic stellate cells via inhibition of cyclin dependent kinase 9 , 2022, Frontiers in Pharmacology.

[13]  S. Gou,et al.  Design, synthesis and anticancer evaluation of selective 2,4-disubstituted pyrimidine CDK9 inhibitors. , 2022, European journal of medicinal chemistry.

[14]  W. Pao,et al.  Discovery of Novel Allosteric EGFR L858R Inhibitors for the Treatment of Non-Small-Cell Lung Cancer as a Single Agent or in Combination with Osimertinib. , 2022, Journal of medicinal chemistry.

[15]  Kaixian Chen,et al.  Discovery of Small-Molecule Degraders of the CDK9-Cyclin T1 Complex for Targeting Transcriptional Addiction in Prostate Cancer. , 2022, Journal of medicinal chemistry.

[16]  D. Taatjes,et al.  Chemical inhibitors of transcription-associated kinases. , 2022, Current opinion in chemical biology.

[17]  Yan Fan,et al.  CDK9 inhibitors in cancer research. , 2022, RSC Medicinal Chemistry.

[18]  Yadong Chen,et al.  Discovery of 2H-benzo[b][1,4]oxazin-3(4H)-one derivatives as potent and selective CDK9 inhibitors that enable transient target engagement for the treatment of hematologic malignancies. , 2022, European journal of medicinal chemistry.

[19]  A. Alimonti,et al.  Single-cell transcriptomics identifies Mcl-1 as a target for senolytic therapy in cancer , 2022, Nature Communications.

[20]  Yuetong Wang,et al.  Artesunate improves venetoclax plus cytarabine AML cell targeting by regulating the Noxa/Bim/Mcl-1/p-Chk1 axis , 2022, Cell death & disease.

[21]  P. Antosik,et al.  The Prognostic Role of CDK9 in Bladder Cancer , 2022, Cancers.

[22]  K. Bhalla,et al.  Efficacy of CDK9 inhibition in therapy of post-myeloproliferative neoplasm (MPN) secondary (s) AML cells , 2022, Blood cancer journal.

[23]  K. Garber The PROTAC gold rush , 2021, Nature biotechnology.

[24]  Fangbing Liu,et al.  Cotargeting of Bcl-2 and Mcl-1 Shows Promising Antileukemic Activity against AML Cells Including those with Acquired Cytarabine Resistance. , 2021, Experimental Hematology.

[25]  A. Ferguson,et al.  Discovery of a Series of 7-Azaindoles as Potent and Highly Selective CDK9 Inhibitors for Transient Target Engagement. , 2021, Journal of medicinal chemistry.

[26]  Qiulian Wu,et al.  Transcription Elongation Machinery Is a Druggable Dependency and Potentiates Immunotherapy in Glioblastoma Stem Cells. , 2021, Cancer discovery.

[27]  S. Bharate,et al.  Analyzing the scaffold diversity of cyclin‐dependent kinase inhibitors and revisiting the clinical and preclinical pipeline , 2021, Medicinal research reviews.

[28]  C. Leung,et al.  Inhibition of the CDK9–cyclin T1 protein–protein interaction as a new approach against triple-negative breast cancer , 2021, Acta pharmaceutica Sinica. B.

[29]  Yu-bo Zhou,et al.  Discovery of Potent and Selective CDK9 Degraders for Targeting Transcription Regulation in Triple-Negative Breast Cancer. , 2021, Journal of medicinal chemistry.

[30]  Hong Liu,et al.  Rational Design and Development of Novel CDK9 Inhibitors for the Treatment of Acute Myeloid Leukemia. , 2021, Journal of medicinal chemistry.

[31]  G. Gomatou,et al.  Cyclin-dependent kinase (CDK) inhibitors in solid tumors: a review of clinical trials , 2021, Clinical and Translational Oncology.

[32]  Q. Mo,et al.  Inhibitors Targeting CDK9 Show High Efficacy against Osimertinib and AMG510 Resistant Lung Adenocarcinoma Cells , 2021, Cancers.

[33]  H. Briem,et al.  Changing for the Better: Discovery of the Highly Potent and Selective CDK9 Inhibitor VIP152 Suitable for Once Weekly Intravenous Dosing for the Treatment of Cancer. , 2021, Journal of medicinal chemistry.

[34]  C. Leung,et al.  Discovery of a tetrahydroisoquinoline-based CDK9-cyclin T1 protein–protein interaction inhibitor as an anti-proliferative and anti-migration agent against triple-negative breast cancer cells , 2021, Genes & diseases.

[35]  M. Gilbert,et al.  Targeting CDK9 for the Treatment of Glioblastoma , 2021, Cancers.

[36]  Tariq Ahmad Masoodi,et al.  Cyclin-dependent kinase 9 (CDK9) predicts recurrence in Middle Eastern epithelial ovarian cancer , 2021, Journal of Ovarian Research.

[37]  Shudong Wang,et al.  CDK9: A Comprehensive Review of Its Biology, and Its Role as a Potential Target for Anti-Cancer Agents , 2021, Frontiers in Oncology.

[38]  K. Strebhardt,et al.  Targeting CDK9 for Anti-Cancer Therapeutics , 2021, Cancers.

[39]  S. Guhan,et al.  The molecular context of vulnerability for CDK9 suppression in triple wild-type melanoma. , 2021, Journal of Investigative Dermatology.

[40]  Tuersunayi Abudureheman,et al.  CDK9 Inhibitor Induces the Apoptosis of B-Cell Acute Lymphocytic Leukemia by Inhibiting c-Myc-Mediated Glycolytic Metabolism , 2021, Frontiers in Cell and Developmental Biology.

[41]  Fen Ma,et al.  Phosphorylation of the androgen receptor at Ser81 is co‐sustained by CDK1 and CDK9 and leads to AR‐mediated transactivation in prostate cancer , 2021, Molecular oncology.

[42]  F. Appelbaum,et al.  NCCN Guidelines Insights: Acute Myeloid Leukemia, Version 2.2021. , 2021, Journal of the National Comprehensive Cancer Network : JNCCN.

[43]  Aisha A. Alsfouk Small molecule inhibitors of cyclin-dependent kinase 9 for cancer therapy , 2021, Journal of enzyme inhibition and medicinal chemistry.

[44]  Jianbing Hou,et al.  CSN6 promotes melanoma proliferation and metastasis by controlling the UBR5-mediated ubiquitination and degradation of CDK9 , 2021, Cell death & disease.

[45]  A. Ferguson,et al.  Discovery of AZD4573, a Potent and Selective Inhibitor of CDK9 That Enables Short Duration of Target Engagement for the Treatment of Hematological Malignancies. , 2020, Journal of medicinal chemistry.

[46]  R. Montironi,et al.  Androgen Receptor Signaling Pathway in Prostate Cancer: From Genetics to Clinical Applications , 2020, Cells.

[47]  Y. Zhuang,et al.  Acetyl-bufalin shows potent efficacy against non-small-cell lung cancer by targeting the CDK9/STAT3 signalling pathway , 2020, British Journal of Cancer.

[48]  N. Lu,et al.  A novel kinase inhibitor, LZT-106, downregulates Mcl-1 and sensitizes colorectal cancer cells to BH3 mimetic ABT-199 by targeting CDK9 and GSK-3β signaling. , 2020, Cancer letters.

[49]  Charles Y. Lin,et al.  Modulating Androgen Receptor-Driven Transcription in Prostate Cancer with Selective CDK9 Inhibitors. , 2020, Cell chemical biology.

[50]  Zhiyu Li,et al.  Recent Developments in the Biology and Medicinal Chemistry of CDK9 Inhibitors: An Update. , 2020, Journal of medicinal chemistry.

[51]  Yanmin Zhang,et al.  Discovery of coumarin derivatives as potent and selective cyclin-dependent kinase 9 (CDK9) inhibitors with high antitumour activity. , 2020, European journal of medicinal chemistry.

[52]  Xiuling Zhi,et al.  CDK9 inhibitor CDKI-73 is synergetic lethal with PARP inhibitor olaparib in BRCA1 wide-type ovarian cancer. , 2020, American journal of cancer research.

[53]  S. Fawell,et al.  AZD4573 Is a Highly Selective CDK9 Inhibitor That Suppresses MCL-1 and Induces Apoptosis in Hematologic Cancer Cells , 2019, Clinical Cancer Research.

[54]  L. Selth,et al.  Novel and highly selective CDK9 inhibitors suppress proliferation of triple negative breast cancer (TNBC) cells in vitro , 2019, Oncology Abstracts.

[55]  H. Ding,et al.  Design of wogonin-inspired selective cyclin-dependent kinase 9 (CDK9) inhibitors with potent in vitro and in vivo antitumor activity. , 2019, European journal of medicinal chemistry.

[56]  L. Drew,et al.  Abstract 2500: Transient CDK9 inhibition with AZD4573 modulates Bfl-1 in preclinical lymphoma models , 2019, Molecular and Cellular Biology / Genetics.

[57]  X. Tian,et al.  Recent development of CDK inhibitors: An overview of CDK/inhibitor co-crystal structures. , 2019, European journal of medicinal chemistry.

[58]  Z. Duan,et al.  Cyclin-dependent kinase 9 (CDK9) is a novel prognostic marker and therapeutic target in osteosarcoma , 2018, EBioMedicine.

[59]  Lindsey A. Marsh,et al.  Antineoplastic effects of selective CDK9 inhibition with atuveciclib on cancer stem-like cells in triple-negative breast cancer , 2018, Oncotarget.

[60]  J. Espinosa,et al.  Therapeutic targeting of transcriptional cyclin-dependent kinases , 2018, Transcription.

[61]  C. Zahnow,et al.  Targeting CDK9 Reactivates Epigenetically Silenced Genes in Cancer , 2018, Cell.

[62]  I. D’Orso,et al.  CDK9: a signaling hub for transcriptional control , 2018, Transcription.

[63]  L. Drew,et al.  Abstract 310: AZD4573, a novel CDK9 inhibitor, rapidly induces cell death in hematological tumor models through depletion of Mcl1 , 2018, Molecular and Cellular Biology / Genetics.

[64]  H. Briem,et al.  Identification of Atuveciclib (BAY 1143572), the First Highly Selective, Clinical PTEFb/CDK9 Inhibitor for the Treatment of Cancer , 2017, ChemMedChem.

[65]  R. Young,et al.  Transcriptional Addiction in Cancer , 2017, Cell.

[66]  Sonali M. Smith,et al.  Feasibility of interim positron emission tomography (PET)-adapted therapy in HIV-positive patients with advanced Hodgkin lymphoma (HL): a sub-analysis of SWOG S0816 Phase 2 trial , 2017, Leukemia & lymphoma.

[67]  Y. Fernández-Marrero,et al.  Survival control of malignant lymphocytes by anti-apoptotic MCL-1 , 2016, Leukemia.

[68]  Daniel C. Factor,et al.  RBPJ maintains brain tumor-initiating cells through CDK9-mediated transcriptional elongation. , 2016, The Journal of clinical investigation.

[69]  Mu-Kuan Chen,et al.  Transcriptional regulation of Mcl-1 plays an important role of cellular protective effector of vincristine-triggered autophagy in oral cancer cells , 2015, Expert opinion on therapeutic targets.

[70]  Agnieszka K. Witkiewicz,et al.  The history and future of targeting cyclin-dependent kinases in cancer therapy , 2015, Nature Reviews Drug Discovery.

[71]  John G. Moffat,et al.  Phenotypic screening in cancer drug discovery — past, present and future , 2014, Nature Reviews Drug Discovery.

[72]  J. Butera Phenotypic screening as a strategic component of drug discovery programs targeting novel antiparasitic and antimycobacterial agents: an editorial. , 2013, Journal of medicinal chemistry.

[73]  J. Karn,et al.  Phosphorylation of CDK9 at Ser175 Enhances HIV Transcription and Is a Marker of Activated P-TEFb in CD4+ T Lymphocytes , 2013, PLoS pathogens.

[74]  W. Hahn,et al.  CDK9 regulates AR promoter selectivity and cell growth through serine 81 phosphorylation. , 2010, Molecular endocrinology.

[75]  Jitender Verma,et al.  3D-QSAR in drug design--a review. , 2010, Current topics in medicinal chemistry.

[76]  A. Giordano,et al.  Cell cycle kinases as therapeutic targets for cancer , 2009, Nature Reviews Drug Discovery.

[77]  Joshua M. Korn,et al.  Comprehensive genomic characterization defines human glioblastoma genes and core pathways , 2008, Nature.

[78]  Q. Zheng,et al.  CDK inhibitors in cancer therapy, an overview of recent development. , 2021, American journal of cancer research.

[79]  R. Roskoski Cyclin-dependent protein serine/threonine kinase inhibitors as anticancer drugs. , 2019, Pharmacological research.