151
|
Rohban MH, Fuller AM, Tan C, Goldstein JT, Syangtan D, Gutnick A, DeVine A, Nijsure MP, Rigby M, Sacher JR, Corsello SM, Peppler GB, Bogaczynska M, Boghossian A, Ciotti GE, Hands AT, Mekareeya A, Doan M, Gale JP, Derynck R, Turbyville T, Boerckel JD, Singh S, Kiessling LL, Schwarz TL, Varelas X, Wagner FF, Kafri R, Eisinger-Mathason TSK, Carpenter AE. Virtual screening for small-molecule pathway regulators by image-profile matching. Cell Syst 2022; 13:724-736.e9. [PMID: 36057257 PMCID: PMC9509476 DOI: 10.1016/j.cels.2022.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/14/2022] [Accepted: 08/09/2022] [Indexed: 02/08/2023]
Abstract
Identifying the chemical regulators of biological pathways is a time-consuming bottleneck in developing therapeutics and research compounds. Typically, thousands to millions of candidate small molecules are tested in target-based biochemical screens or phenotypic cell-based screens, both expensive experiments customized to each disease. Here, our uncustomized, virtual, profile-based screening approach instead identifies compounds that match to pathways based on the phenotypic information in public cell image data, created using the Cell Painting assay. Our straightforward correlation-based computational strategy retrospectively uncovered the expected, known small-molecule regulators for 32% of positive-control gene queries. In prospective, discovery mode, we efficiently identified new compounds related to three query genes and validated them in subsequent gene-relevant assays, including compounds that phenocopy or pheno-oppose YAP1 overexpression and kill a Yap1-dependent sarcoma cell line. This image-profile-based approach could replace many customized labor- and resource-intensive screens and accelerate the discovery of biologically and therapeutically useful compounds.
Collapse
Affiliation(s)
- Mohammad H Rohban
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ashley M Fuller
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ceryl Tan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Department of Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Deepsing Syangtan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Amos Gutnick
- FM Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Ann DeVine
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Madhura P Nijsure
- Departments of Orthopaedic Surgery & Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Megan Rigby
- Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Joshua R Sacher
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Steven M Corsello
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Grace B Peppler
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, USA
| | - Marta Bogaczynska
- Departments of Cell/Tissue Biology and Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew Boghossian
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gabrielle E Ciotti
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison T Hands
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aroonroj Mekareeya
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Minh Doan
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jennifer P Gale
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rik Derynck
- Departments of Cell/Tissue Biology and Anatomy, University of California, San Francisco, San Francisco, CA, USA
| | - Thomas Turbyville
- Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Joel D Boerckel
- Departments of Orthopaedic Surgery & Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Shantanu Singh
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Laura L Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Thomas L Schwarz
- FM Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Xaralabos Varelas
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Florence F Wagner
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ran Kafri
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Department of Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - T S Karin Eisinger-Mathason
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Anne E Carpenter
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
152
|
Zhang T, Wang Y, Xie M, Ji X, Luo X, Chen X, Zhang B, Liu D, Feng Y, Sun M, Huang W, Xia L. HGF-mediated elevation of ETV1 facilitates hepatocellular carcinoma metastasis through upregulating PTK2 and c-MET. J Exp Clin Cancer Res 2022; 41:275. [PMID: 36109787 PMCID: PMC9479266 DOI: 10.1186/s13046-022-02475-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/24/2022] [Indexed: 12/14/2022] Open
Abstract
Background Metastasis is a major determinant of death in patients with hepatocellular carcinoma (HCC). Dissecting key molecular mediators that promote this malignant feature may help yield novel therapeutic insights. Here, we investigated the role of E-twenty-six transformation-specific variant 1 (ETV1), a member of the E-twenty-six transformation-specific (ETS) family, in HCC metastasis. Methods The clinical significance of ETV1 and its target genes in two independent cohorts of HCC patients who underwent curative resection were assessed by Kaplan–Meier analysis and Multivariate Cox proportional hazards model. Luciferase reporter assay and chromatin immunoprecipitation assay were used to detect the transcriptional regulation of target gene promoters by ETV1. The effect of ETV1 on invasiveness and metastasis of HCC were detected by transwell assays and the orthotopically metastatic model. Results ETV1 expression was frequently elevated in human HCC specimens. Increased ETV1 expression was associated with the malignant biological characteristics and poor prognosis of HCC patients. ETV1 facilitated invasion and metastasis of HCC cells in vitro and in vivo. Mechanistically, ETV1 promoted HCC metastasis via upregulating metastasis-related genes, including protein tyrosine kinase 2 (PTK2) and MET. Down-regulated the expression of PTK2 or tyrosine protein kinase Met (c-MET) decreased ETV1-mediated HCC metastasis. Hepatocyte growth factor (HGF) upregulated ETV1 expression through activating c-MET-ERK1/2-ELK1 pathway. Notably, in two independent cohorts, patients with positive coexpression of ETV1/PTK2 or ETV1/c-MET had worse prognosis. Furthermore, the combination of PTK2 inhibitor defactinib and c-MET inhibitor capmatinib significantly suppressed HCC metastasis induced by ETV1. Conclusion This study uncovers functional and prognostic roles for ETV1 in HCC and exposes a positive feedback loop of HGF-ERK1/2-ETV1-c-MET. Targeting this pathway may provide a potential therapeutic intervention for ETV1-overexpressing HCC. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02475-2.
Collapse
|
153
|
Kooijman JJ, van Riel WE, Dylus J, Prinsen MBW, Grobben Y, de Bitter TJJ, van Doornmalen AM, Melis JJTM, Uitdehaag JCM, Narumi Y, Kawase Y, de Roos JADM, Willemsen-Seegers N, Zaman GJR. Comparative kinase and cancer cell panel profiling of kinase inhibitors approved for clinical use from 2018 to 2020. Front Oncol 2022; 12:953013. [PMID: 36185300 PMCID: PMC9516332 DOI: 10.3389/fonc.2022.953013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/26/2022] [Indexed: 11/23/2022] Open
Abstract
During the last two decades, kinase inhibitors have become the major drug class for targeted cancer therapy. Although the number of approved kinase inhibitors increases rapidly, comprehensive in vitro profiling and comparison of inhibitor activities is often lacking in the public domain. Here we report the extensive profiling and comparison of 21 kinase inhibitors approved by the FDA for oncology indications since June 2018 and 13 previously approved comparators on panels of 255 biochemical kinase assays and 134 cancer cell line viability assays. Comparison of the cellular inhibition profiles of the EGFR inhibitors gefitinib, dacomitinib, and osimertinib identified the uncommon EGFR p.G719S mutation as a common response marker for EGFR inhibitors. Additionally, the FGFR inhibitors erdafitinib, infigratinib, and pemigatinib potently inhibited the viability of cell lines which harbored oncogenic alterations in FGFR1-3, irrespective of the specific clinical indications of the FGFR inhibitors. These results underscore the utility of in vitro kinase inhibitor profiling in cells for identifying new potential stratification markers for patient selection. Furthermore, comparison of the in vitro inhibition profiles of the RET inhibitors pralsetinib and selpercatinib revealed they had very similar biochemical and cellular selectivity. As an exception, an NTRK3 fusion-positive cell line was potently inhibited by pralsetinib but not by selpercatinib, which could be explained by the targeting of TRK kinases in biochemical assays by pralsetinib but not selpercatinib. This illustrates that unexpected differences in cellular activities between inhibitors that act through the same primary target can be explained by subtle differences in biochemical targeting. Lastly, FLT3-mutant cell lines were responsive to both FLT3 inhibitors gilteritinib and midostaurin, and the PI3K inhibitor duvelisib. Biochemical profiling revealed that the FLT3 and PI3K inhibitors targeted distinct kinases, indicating that unique dependencies can be identified by combined biochemical and cellular profiling of kinase inhibitors. This study provides the first large scale kinase assay or cell panel profiling study for newly approved kinase inhibitors, and shows that comprehensive in vitro profiling of kinase inhibitors can provide rationales for therapy selection and indication expansion of approved kinase inhibitors.
Collapse
|
154
|
Shebl B, Ng D, Lalazar G, Rosemore C, Finkelstein TM, Migler RD, Zheng G, Zhang P, Jiang CS, Qureshi A, Vaughan R, Yarchoan M, de Jong YP, Rice CM, Coffino P, Ortiz MV, Zhou D, Simon SM. Targeting BCL-XL in fibrolamellar hepatocellular carcinoma. JCI Insight 2022; 7:e161820. [PMID: 36073545 PMCID: PMC9536265 DOI: 10.1172/jci.insight.161820] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
Abstract
Fibrolamellar hepatocellular carcinoma (FLC) is a rare and often lethal liver cancer with no proven effective systemic therapy. Inhibition of the antiapoptotic protein BCL-XL was found to synergize with a variety of systemic therapies in vitro using cells dissociated from patient-derived xenografts (PDX) of FLC or cells dissociated directly from surgical patient resections. As BCL-XL is physiologically expressed in platelets, prior efforts to leverage this vulnerability in other cancers have been hampered by severe thrombocytopenia. To overcome this toxicity, we treated FLC models with DT2216, a proteolysis targeting chimera (PROTAC) that directs BCL-XL for degradation via the von Hippel-Lindau (VHL) E3 ligase, which is minimally expressed in platelets. The combination of irinotecan and DT2216 in vitro on cells directly acquired from patients or in vivo using several xenografts derived from patients with FLC demonstrated remarkable synergy and at clinically achievable doses not associated with significant thrombocytopenia.
Collapse
Affiliation(s)
- Bassem Shebl
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
| | - Denise Ng
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
| | - Gadi Lalazar
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
| | - Carly Rosemore
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Tova M. Finkelstein
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
| | | | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Peiyi Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Caroline S. Jiang
- Hospital Biostatistics, The Rockefeller University, New York, New York, USA
| | - Adam Qureshi
- Hospital Biostatistics, The Rockefeller University, New York, New York, USA
| | - Roger Vaughan
- Hospital Biostatistics, The Rockefeller University, New York, New York, USA
| | - Mark Yarchoan
- The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ype P. de Jong
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, USA
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, New York, USA
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, USA
| | - Philip Coffino
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
| | - Michael V. Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Daohong Zhou
- Department of Biochemistry and Structural Biology and Center for Innovative Drug Discovery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Sanford M. Simon
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, New York, USA
| |
Collapse
|
155
|
Le Roux Ö, Pershing NLK, Kaltenbrun E, Newman NJ, Everitt JI, Baldelli E, Pierobon M, Petricoin EF, Counter CM. Genetically manipulating endogenous Kras levels and oncogenic mutations in vivo influences tissue patterning of murine tumorigenesis. eLife 2022; 11:e75715. [PMID: 36069770 PMCID: PMC9451540 DOI: 10.7554/elife.75715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 08/02/2022] [Indexed: 12/04/2022] Open
Abstract
Despite multiple possible oncogenic mutations in the proto-oncogene KRAS, unique subsets of these mutations are detected in different cancer types. As KRAS mutations occur early, if not being the initiating event, these mutational biases are ostensibly a product of how normal cells respond to the encoded oncoprotein. Oncogenic mutations can impact not only the level of active oncoprotein, but also engagement with proteins. To attempt to separate these two effects, we generated four novel Cre-inducible (LSL) Kras alleles in mice with the biochemically distinct G12D or Q61R mutations and encoded by native (nat) rare or common (com) codons to produce low or high protein levels. While there were similarities, each allele also induced a distinct transcriptional response shortly after activation in vivo. At one end of the spectrum, activating the KrasLSL-natG12D allele induced transcriptional hallmarks suggestive of an expansion of multipotent cells, while at the other end, activating the KrasLSL-comQ61R allele led to hallmarks of hyperproliferation and oncogenic stress. Evidence suggests that these changes may be a product of signaling differences due to increased protein expression as well as the specific mutation. To determine the impact of these distinct responses on RAS mutational patterning in vivo, all four alleles were globally activated, revealing that hematolymphopoietic lesions were permissive to the level of active oncoprotein, squamous tumors were permissive to the G12D mutant, while carcinomas were permissive to both these features. We suggest that different KRAS mutations impart unique signaling properties that are preferentially capable of inducing tumor initiation in a distinct cell-specific manner.
Collapse
Affiliation(s)
- Özgün Le Roux
- Department of Pharmacology & Cancer Biology, Duke University Medical CenterDurhamUnited States
| | - Nicole LK Pershing
- Department of Pharmacology & Cancer Biology, Duke University Medical CenterDurhamUnited States
| | - Erin Kaltenbrun
- Department of Pharmacology & Cancer Biology, Duke University Medical CenterDurhamUnited States
| | - Nicole J Newman
- Department of Pharmacology & Cancer Biology, Duke University Medical CenterDurhamUnited States
| | - Jeffrey I Everitt
- Department of Pathology, Duke University Medical CenterDurhamUnited States
| | - Elisa Baldelli
- Center for Applied Proteomics and Molecular Medicine, School of Systems Biology, George Mason UniversityManassasUnited States
| | - Mariaelena Pierobon
- Center for Applied Proteomics and Molecular Medicine, School of Systems Biology, George Mason UniversityManassasUnited States
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, School of Systems Biology, George Mason UniversityManassasUnited States
| | - Christopher M Counter
- Department of Pharmacology & Cancer Biology, Duke University Medical CenterDurhamUnited States
| |
Collapse
|
156
|
Mah KM, Wu W, Al-Ali H, Sun Y, Han Q, Ding Y, Muñoz M, Xu XM, Lemmon VP, Bixby JL. Compounds co-targeting kinases in axon regulatory pathways promote regeneration and behavioral recovery after spinal cord injury in mice. Exp Neurol 2022; 355:114117. [PMID: 35588791 PMCID: PMC9443329 DOI: 10.1016/j.expneurol.2022.114117] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 12/21/2022]
Abstract
Recovery from spinal cord injury (SCI) and other central nervous system (CNS) trauma is hampered by limits on axonal regeneration in the CNS. Regeneration is restricted by the lack of neuron-intrinsic regenerative capacity and by the repressive microenvironment confronting damaged axons. To address this challenge, we have developed a therapeutic strategy that co-targets kinases involved in both extrinsic and intrinsic regulatory pathways. Prior work identified a kinase inhibitor (RO48) with advantageous polypharmacology (co-inhibition of targets including ROCK2 and S6K1), which promoted CNS axon growth in vitro and corticospinal tract (CST) sprouting in a mouse pyramidotomy model. We now show that RO48 promotes neurite growth from sensory neurons and a variety of CNS neurons in vitro, and promotes CST sprouting and/or regeneration in multiple mouse models of spinal cord injury. Notably, these in vivo effects of RO48 were seen in several independent experimental series performed in distinct laboratories at different times. Finally, in a cervical dorsal hemisection model, RO48 not only promoted growth of CST axons beyond the lesion, but also improved behavioral recovery in the rotarod, gridwalk, and pellet retrieval tasks. Our results provide strong evidence for RO48 as an effective compound to promote axon growth and regeneration. Further, they point to strategies for increasing robustness of interventions in pre-clinical models.
Collapse
Affiliation(s)
- Kar Men Mah
- The Miami Project to Cure Paralysis, Dept of Neurological Surgery, University of Miami, Miami, FL, USA
| | - Wei Wu
- Department of Neurological Surgery, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hassan Al-Ali
- The Miami Project to Cure Paralysis, Dept of Neurological Surgery, University of Miami, Miami, FL, USA; Peggy and Harold Katz Family Drug Discovery Center, Dept of Medicine, University of Miami, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Yan Sun
- Department of Neurological Surgery, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Qi Han
- Department of Neurological Surgery, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ying Ding
- Department of Neurological Surgery, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Melissa Muñoz
- The Miami Project to Cure Paralysis, Dept of Neurological Surgery, University of Miami, Miami, FL, USA
| | - Xiao-Ming Xu
- Department of Neurological Surgery, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Vance P Lemmon
- The Miami Project to Cure Paralysis, Dept of Neurological Surgery, University of Miami, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA; Institute for Data Science and Computing, University of Miami, Miami, FL, USA.
| | - John L Bixby
- The Miami Project to Cure Paralysis, Dept of Neurological Surgery, University of Miami, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA; Dept of Molecular and Cellular Pharmacology, University of Miami, Miami, FL, USA.
| |
Collapse
|
157
|
Neuditschko B, King AP, Huang Z, Janker L, Bileck A, Borutzki Y, Marker SC, Gerner C, Wilson JJ, Meier-Menches SM. An Anticancer Rhenium Tricarbonyl Targets Fe‐S Cluster Biogenesis in Ovarian Cancer Cells. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202209136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Benjamin Neuditschko
- University of Vienna: Universitat Wien Department of Analytical Chemistry AUSTRIA
| | - A. Paden King
- Cornell University Department of Chemistry and Chemical Biology UNITED STATES
| | - Zhouyang Huang
- Cornell University Department of Chemistry and Chemical Biology UNITED STATES
| | - Lukas Janker
- University of Vienna Faculty of Chemistry: Universitat Wien Fakultat fur Chemie Department of Analytical Chemistry AUSTRIA
| | - Andrea Bileck
- University of Vienna: Universitat Wien Department of Analytical Chemistry AUSTRIA
| | - Yasmin Borutzki
- University of Vienna: Universitat Wien Institute of Inorganic Chemistry AUSTRIA
| | - Sierra C. Marker
- Cornell University Department of Chemistry and Chemical Biology UNITED STATES
| | - Christopher Gerner
- University of Vienna: Universitat Wien Department of Analytical Chemistry AUSTRIA
| | - Justin J. Wilson
- Cornell University Department of Chemistry and Chemical Biology UNITED STATES
| | - Samuel M. Meier-Menches
- University of Vienna: Universitat Wien Department of Analytical Chemistry Waehringer Str. 38 1090 Vienna AUSTRIA
| |
Collapse
|
158
|
Network Pharmacology of Adaptogens in the Assessment of Their Pleiotropic Therapeutic Activity. Pharmaceuticals (Basel) 2022; 15:ph15091051. [PMID: 36145272 PMCID: PMC9504187 DOI: 10.3390/ph15091051] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/11/2022] [Accepted: 08/19/2022] [Indexed: 02/07/2023] Open
Abstract
The reductionist concept, based on the ligand–receptor interaction, is not a suitable model for adaptogens, and herbal preparations affect multiple physiological functions, revealing polyvalent pharmacological activities, and are traditionally used in many conditions. This review, for the first time, provides a rationale for the pleiotropic therapeutic efficacy of adaptogens based on evidence from recent gene expression studies in target cells and where the network pharmacology and systems biology approaches were applied. The specific molecular targets and adaptive stress response signaling mechanisms involved in nonspecific modes of action of adaptogens are identified.
Collapse
|
159
|
Hu J, Jarusiewicz J, Du G, Nishiguchi G, Yoshimura S, Panetta JC, Li Z, Min J, Yang L, Chepyala D, Actis M, Reyes N, Smart B, Pui CH, Teachey DT, Rankovic Z, Yang JJ. Preclinical evaluation of proteolytic targeting of LCK as a therapeutic approach in T cell acute lymphoblastic leukemia. Sci Transl Med 2022; 14:eabo5228. [PMID: 36001679 PMCID: PMC9730446 DOI: 10.1126/scitranslmed.abo5228] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy, and there is an unmet need for targeted therapies, especially for patients with relapsed disease. We have recently identified pre-T cell receptor and lymphocyte-specific protein tyrosine kinase (LCK) signaling as a common therapeutic vulnerability in T-ALL. LCK inhibitor dasatinib showed efficacy against T-ALL in preclinical studies and in patients with T-ALL; however, this is transient in most cases. Leveraging the proteolysis targeting chimera (PROTAC) approach, we developed a series of LCK degraders using dasatinib as an LCK ligand and phenyl-glutarimide as a cereblon-directing moiety. Our lead compound SJ11646 exhibited marked efficiency in cereblon-mediated LCK degradation in T-ALL cells. Relative to dasatinib, SJ11646 showed up to three orders of magnitude higher cytotoxicity in LCK-activated T-ALL cell lines and primary leukemia samples in vitro, with drastically prolonged suppression of LCK signaling. In vivo pharmacokinetic and pharmacodynamic profiling indicated a 630% increase in the duration of LCK suppression by SJ11646 over dasatinib in patient-derived xenograft models of T-ALL, which translated into its extended leukemia-free survival over dasatinib in vivo. Last, SJ11646 retained a high binding affinity to 51 human kinases, particularly ABL1, KIT, and DDR1, all of which are known drug targets in other cancers. Together, our dasatinib-based phenyl-glutarimide PROTACs are promising therapeutic agents in T-ALL and valuable tools for developing degradation-based therapeutics for other cancers.
Collapse
Affiliation(s)
- Jianzhong Hu
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, 38105, USA
| | - Jamie Jarusiewicz
- Department of Chemical Biology & Therapeutics, St. Jude Children’s Research Hospital; Memphis, TN, 38105, USA
| | - Guoqing Du
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, 38105, USA
| | - Gisele Nishiguchi
- Department of Chemical Biology & Therapeutics, St. Jude Children’s Research Hospital; Memphis, TN, 38105, USA
| | - Satoshi Yoshimura
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, 38105, USA
| | - John C. Panetta
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, 38105, USA
| | - Zhenhua Li
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, 38105, USA
| | - Jaeki Min
- Department of Chemical Biology & Therapeutics, St. Jude Children’s Research Hospital; Memphis, TN, 38105, USA
| | - Lei Yang
- Department of Chemical Biology & Therapeutics, St. Jude Children’s Research Hospital; Memphis, TN, 38105, USA
| | - Divyabharathi Chepyala
- Department of Chemical Biology & Therapeutics, St. Jude Children’s Research Hospital; Memphis, TN, 38105, USA
| | - Marisa Actis
- Department of Chemical Biology & Therapeutics, St. Jude Children’s Research Hospital; Memphis, TN, 38105, USA
| | - Noemi Reyes
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, 38105, USA
| | - Brandon Smart
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, 38105, USA
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, 38105, USA
| | - David T. Teachey
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Zoran Rankovic
- Department of Chemical Biology & Therapeutics, St. Jude Children’s Research Hospital; Memphis, TN, 38105, USA,,Correspondence to: Jun J. Yang Ph.D., Member, Department of Pharmacy and Pharmaceutical Sciences, Department of Oncology, ; Zoran Rankovic Ph.D., Director, CBT Chemistry Centers, Department of Chemical Biology & Therapeutics, ; St. Jude Children’s Research Hospital, 262 Danny Thomas Pl., Memphis, TN 38105
| | - Jun J. Yang
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN, 38105, USA,,Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, 38105, USA,,Correspondence to: Jun J. Yang Ph.D., Member, Department of Pharmacy and Pharmaceutical Sciences, Department of Oncology, ; Zoran Rankovic Ph.D., Director, CBT Chemistry Centers, Department of Chemical Biology & Therapeutics, ; St. Jude Children’s Research Hospital, 262 Danny Thomas Pl., Memphis, TN 38105
| |
Collapse
|
160
|
Inferring tumor-specific cancer dependencies through integrating ex vivo drug response assays and drug-protein profiling. PLoS Comput Biol 2022; 18:e1010438. [PMID: 35994503 PMCID: PMC9436053 DOI: 10.1371/journal.pcbi.1010438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 09/01/2022] [Accepted: 07/26/2022] [Indexed: 11/22/2022] Open
Abstract
The development of cancer therapies may be improved by the discovery of tumor-specific molecular dependencies. The requisite tools include genetic and chemical perturbations, each with its strengths and limitations. Chemical perturbations can be readily applied to primary cancer samples at large scale, but mechanistic understanding of hits and further pharmaceutical development is often complicated by the fact that a chemical compound has affinities to multiple proteins. To computationally infer specific molecular dependencies of individual cancers from their ex vivo drug sensitivity profiles, we developed a mathematical model that deconvolutes these data using measurements of protein-drug affinity profiles. Through integrating a drug-kinase profiling dataset and several drug response datasets, our method, DepInfeR, correctly identified known protein kinase dependencies, including the EGFR dependence of HER2+ breast cancer cell lines, the FLT3 dependence of acute myeloid leukemia (AML) with FLT3-ITD mutations and the differential dependencies on the B-cell receptor pathway in the two major subtypes of chronic lymphocytic leukemia (CLL). Furthermore, our method uncovered new subgroup-specific dependencies, including a previously unreported dependence of high-risk CLL on Checkpoint kinase 1 (CHEK1). The method also produced a detailed map of the kinase dependencies in a heterogeneous set of 117 CLL samples. The ability to deconvolute polypharmacological phenotypes into underlying causal molecular dependencies should increase the utility of high-throughput drug response assays for functional precision oncology. As survival and proliferation of cancer cells depend on molecular aberrations that can be highly specific to cancer types and individual tumors, identifying such dependence is pivotal to designing individualized tumor therapy. Chemical perturbations, through screening of bioactive compounds using primary cancer cells, provide an important tool for identifying tumor-specific dependencies. However, many chemical compounds bind multiple proteins, which complicates interpreting screening results and pinpointing the phenotype-causing target. To overcome this challenge and increase the utility of drug screening approaches for functional precision medicine, we developed a computational framework, DepInfeR, to identify tumor-specific dependencies on druggable proteins through integrating two sources of information: drug sensitivity assays and drug-protein affinity profiling. Our approach correctly identifies known kinase dependencies, which validates our approach. Furthermore, by integrating a newly generated drug screening dataset on primary tumor samples, we discovered a previously unreported survival dependence on Checkpoint kinase 1 (CHEK1) by a molecular subgroup of chronic lymphocytic leukemia samples, highlighting the clinical potential of our method.
Collapse
|
161
|
Development of actionable targets of multi-kinase inhibitors (AToMI) screening platform to dissect kinase targets of staurosporines in glioblastoma cells. Sci Rep 2022; 12:13796. [PMID: 35963891 PMCID: PMC9376105 DOI: 10.1038/s41598-022-18118-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/05/2022] [Indexed: 11/08/2022] Open
Abstract
Therapeutic resistance to kinase inhibitors constitutes a major unresolved clinical challenge in cancer and especially in glioblastoma. Multi-kinase inhibitors may be used for simultaneous targeting of multiple target kinases and thereby potentially overcome kinase inhibitor resistance. However, in most cases the identification of the target kinases mediating therapeutic effects of multi-kinase inhibitors has been challenging. To tackle this important problem, we developed an actionable targets of multi-kinase inhibitors (AToMI) strategy and used it for characterization of glioblastoma target kinases of staurosporine derivatives displaying synergy with protein phosphatase 2A (PP2A) reactivation. AToMI consists of interchangeable modules combining drug-kinase interaction assay, siRNA high-throughput screening, bioinformatics analysis, and validation screening with more selective target kinase inhibitors. As a result, AToMI analysis revealed AKT and mitochondrial pyruvate dehydrogenase kinase PDK1 and PDK4 as kinase targets of staurosporine derivatives UCN-01, CEP-701, and K252a that synergized with PP2A activation across heterogeneous glioblastoma cells. Based on these proof-of-principle results, we propose that the application and further development of AToMI for clinically applicable multi-kinase inhibitors could provide significant benefits in overcoming the challenge of lack of knowledge of the target specificity of multi-kinase inhibitors.
Collapse
|
162
|
Emdal KB, Palacio-Escat N, Wigerup C, Eguchi A, Nilsson H, Bekker-Jensen DB, Rönnstrand L, Kazi JU, Puissant A, Itzykson R, Saez-Rodriguez J, Masson K, Blume-Jensen P, Olsen JV. Phosphoproteomics of primary AML patient samples reveals rationale for AKT combination therapy and p53 context to overcome selinexor resistance. Cell Rep 2022; 40:111177. [PMID: 35947955 PMCID: PMC9380259 DOI: 10.1016/j.celrep.2022.111177] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 05/18/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease with variable patient responses to therapy. Selinexor, an inhibitor of nuclear export, has shown promising clinical activity for AML. To identify the molecular context for monotherapy sensitivity as well as rational drug combinations, we profile selinexor signaling responses using phosphoproteomics in primary AML patient samples and cell lines. Functional phosphosite scoring reveals that p53 function is required for selinexor sensitivity consistent with enhanced efficacy of selinexor in combination with the MDM2 inhibitor nutlin-3a. Moreover, combining selinexor with the AKT inhibitor MK-2206 overcomes dysregulated AKT-FOXO3 signaling in resistant cells, resulting in synergistic anti-proliferative effects. Using high-throughput spatial proteomics to profile subcellular compartments, we measure global proteome and phospho-proteome dynamics, providing direct evidence of nuclear translocation of FOXO3 upon combination treatment. Our data demonstrate the potential of phosphoproteomics and functional phosphorylation site scoring to successfully pinpoint key targetable signaling hubs for rational drug combinations. Phosphoproteomics with functional scoring uncovers context for selinexor sensitivity Functional p53 correlates with selinexor sensitivity, which is enhanced by nutlin-3a Dysregulated AKT-FOXO3 drives selinexor resistance, which is overcome with MK-2206 Spatial proteomics reveals selinexor-induced nucleocytoplasmic protein shuttling
Collapse
Affiliation(s)
- Kristina B Emdal
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolàs Palacio-Escat
- Heidelberg University, Faculty of Medicine and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant-Zentrum, Heidelberg, Germany; Heidelberg University, Faculty of Biosciences, Heidelberg, Germany; RWTH Aachen University, Faculty of Medicine, Joint Research Centre for Computational Biomedicine, Aachen, Germany
| | | | - Akihiro Eguchi
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Dorte B Bekker-Jensen
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Rönnstrand
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Julhash U Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | | | | | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant-Zentrum, Heidelberg, Germany; RWTH Aachen University, Faculty of Medicine, Joint Research Centre for Computational Biomedicine, Aachen, Germany.
| | | | | | - Jesper V Olsen
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
163
|
A Systems Biology Approach to Investigate Kinase Signal Transduction Networks That Are Involved in Triple Negative Breast Cancer Resistance to Cisplatin. J Pers Med 2022; 12:jpm12081277. [PMID: 36013226 PMCID: PMC9409860 DOI: 10.3390/jpm12081277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 11/24/2022] Open
Abstract
Triple negative breast cancer (TNBC) remains a therapeutic challenge due to the lack of targetable genetic alterations and the frequent development of resistance to the standard cisplatin-based chemotherapies. Here, we have taken a systems biology approach to investigate kinase signal transduction networks that are involved in TNBC resistance to cisplatin. Treating a panel of cisplatin-sensitive and cisplatin-resistant TNBC cell lines with a panel of kinase inhibitors allowed us to reconstruct two kinase signalling networks that characterise sensitive and resistant cells. The analysis of these networks suggested that the activation of the PI3K/AKT signalling pathway is critical for cisplatin resistance. Experimental validation of the computational model predictions confirmed that TNBC cell lines with activated PI3K/AKT signalling are sensitive to combinations of cisplatin and PI3K/AKT pathway inhibitors. Thus, our results reveal a new therapeutic approach that is based on identifying targeted therapies that synergise with conventional chemotherapies.
Collapse
|
164
|
The synthesis of PROTAC molecule and new target KAT6A identification of CDK9 inhibitor iCDK9. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.107741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
165
|
Karim M, Saul S, Ghita L, Sahoo MK, Ye C, Bhalla N, Lo CW, Jin J, Park JG, Martinez-Gualda B, East MP, Johnson GL, Pinsky BA, Martinez-Sobrido L, Asquith CRM, Narayanan A, De Jonghe S, Einav S. Numb-associated kinases are required for SARS-CoV-2 infection and are cellular targets for antiviral strategies. Antiviral Res 2022; 204:105367. [PMID: 35738348 PMCID: PMC9212491 DOI: 10.1016/j.antiviral.2022.105367] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/08/2022] [Accepted: 06/15/2022] [Indexed: 12/02/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to pose serious threats to global health. We previously reported that AAK1, BIKE and GAK, members of the Numb-associated kinase family, control intracellular trafficking of multiple RNA viruses during viral entry and assembly/egress. Here, using both genetic and pharmacological approaches, we probe the functional relevance of NAKs for SARS-CoV-2 infection. siRNA-mediated depletion of AAK1, BIKE, GAK, and STK16, the fourth member of the NAK family, suppressed SARS-CoV-2 infection in human lung epithelial cells. Both known and novel small molecules with potent AAK1/BIKE, GAK or STK16 activity suppressed SARS-CoV-2 infection. Moreover, combination treatment with the approved anti-cancer drugs, sunitinib and erlotinib, with potent anti-AAK1/BIKE and GAK activity, respectively, demonstrated synergistic effect against SARS-CoV-2 infection in vitro. Time-of-addition experiments revealed that pharmacological inhibition of AAK1 and BIKE suppressed viral entry as well as late stages of the SARS-CoV-2 life cycle. Lastly, suppression of NAKs expression by siRNAs inhibited entry of both wild type and SARS-CoV-2 pseudovirus. These findings provide insight into the roles of NAKs in SARS-CoV-2 infection and establish a proof-of-principle that pharmacological inhibition of NAKs can be potentially used as a host-targeted approach to treat SARS-CoV-2 with potential implications to other coronaviruses.
Collapse
Affiliation(s)
- Marwah Karim
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, CA, USA
| | - Sirle Saul
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, CA, USA
| | - Luca Ghita
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, CA, USA
| | - Malaya Kumar Sahoo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Nishank Bhalla
- National Center for Biodefence and Infectious Disease, Biomedical Research Laboratory, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Chieh-Wen Lo
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, CA, USA
| | - Jing Jin
- Vitalant Research Institute, San Francisco, CA, USA
| | - Jun-Gyu Park
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Belén Martinez-Gualda
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Michael Patrick East
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Gary L Johnson
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Benjamin A Pinsky
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Christopher R M Asquith
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, 70211, Finland
| | - Aarthi Narayanan
- National Center for Biodefence and Infectious Disease, Biomedical Research Laboratory, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Steven De Jonghe
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Shirit Einav
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, CA, USA; Department of Microbiology and Immunology, Stanford University, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA.
| |
Collapse
|
166
|
Optimization of the 4-anilinoquin(az)oline scaffold as epidermal growth factor receptor (EGFR) inhibitors for chordoma utilizing a toxicology profiling assay platform. Sci Rep 2022; 12:12820. [PMID: 35896603 PMCID: PMC9329436 DOI: 10.1038/s41598-022-15552-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022] Open
Abstract
The 4-anilinoquin(az)oline is a well-known kinase inhibitor scaffold incorporated in clinical inhibitors including gefitinib, erlotinib, afatinib, and lapatinib, all of which have previously demonstrated activity against chordoma cell lines in vitro. We screened a focused array of compounds based on the 4-anilinoquin(az)oline scaffold against both U-CH1 and the epidermal growth factor receptor (EGFR) inhibitor resistant U-CH2. To prioritize the hit compounds for further development, we screened the compound set in a multiparameter cell health toxicity assay. The de-risked compounds were then screened against a wider panel of patient derived cell lines and demonstrated low micromolar efficacy in cells. We also investigated the properties that gave rise to the toxophore markers, including the structural and electronic features, while optimizing for EGFR in-cell target engagement. These de-risked leads present a potential new therapeutic avenue for treatment of chordomas and new chemical tools and probe compound 45 (UNC-CA359) to interrogate EGFR mediated disease phenotypes.
Collapse
|
167
|
Bosdriesz E, Fernandes Neto JM, Sieber A, Bernards R, Blüthgen N, Wessels LF. Identifying mutant-specific multi-drug combinations using Comparative Network Reconstruction. iScience 2022; 25:104760. [PMID: 35992065 PMCID: PMC9385552 DOI: 10.1016/j.isci.2022.104760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/30/2022] [Accepted: 07/11/2022] [Indexed: 10/28/2022] Open
|
168
|
Makki Almansour N. Computational exploration of maternal embryonic leucine zipper kinase (MELK) as a cancer drug target. Saudi J Biol Sci 2022; 29:103335. [PMID: 35769060 PMCID: PMC9235044 DOI: 10.1016/j.sjbs.2022.103335] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/20/2022] [Accepted: 05/29/2022] [Indexed: 11/30/2022] Open
Abstract
Maternal embryonic leucine zipper kinase (MELK) is of vital importance due to its significant role in cancer development and its association with poor prognosis in different cancers. Here, we employed several computer aided drug design approaches to shortlist potential binding molecules of MELK. For virtual screening, asinex oncology library (containing 6334 drugs) and comprehensive marine natural products database (containing approximately 32,000 drugs) were used. The study identified two drug molecules: Top-2 and Top-3 as high affinity binding MELK molecules compared to the control co-crystalized Top-1 inhibitor. Both the shortlisted compounds and the control showed high stable binding free energy and high GOLD score. The compounds and control also reported stable dynamics with root mean square deviations (RMSD) value ∼ 2 Å in 500 ns. Similarly, the MELK active site residues were observed in good stability with the compounds. Further, it was noticed the compounds/control formed multiple hydrogen bonds with the MELK active pocket residues which is the main reason of high intermolecular stability. Atomic level binding free energies determined van der Waals and electrostatic energies to play vital role in stable complex formation. From drug likeness and pharmacokinetics perspective, the compounds are ideal molecules for further investigation. Overall, the results are promising and might be tested in in vivo and in vitro studies against MELK.
Collapse
Affiliation(s)
- Nahlah Makki Almansour
- Department of Biology, College of Science, University of Hafr Al Batin, Hafr Al Batin 31991, Saudi Arabia
| |
Collapse
|
169
|
Heilig CE, Laßmann A, Mughal SS, Mock A, Pirmann S, Teleanu V, Renner M, Andresen C, Köhler BC, Aybey B, Bauer S, Siveke JT, Hamacher R, Folprecht G, Richter S, Schröck E, Brandts CH, Ahrens M, Hohenberger P, Egerer G, Kindler T, Boerries M, Illert AL, von Bubnoff N, Apostolidis L, Jost PJ, Westphalen CB, Weichert W, Keilholz U, Klauschen F, Beck K, Winter U, Richter D, Möhrmann L, Bitzer M, Schulze-Osthoff K, Brors B, Mechtersheimer G, Kreutzfeldt S, Heining C, Lipka DB, Stenzinger A, Schlenk RF, Horak P, Glimm H, Hübschmann D, Fröhling S. Gene expression-based prediction of pazopanib efficacy in sarcoma. Eur J Cancer 2022; 172:107-118. [PMID: 35763870 DOI: 10.1016/j.ejca.2022.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/28/2022] [Accepted: 05/12/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND The multi-receptor tyrosine kinase inhibitor pazopanib is approved for the treatment of advanced soft-tissue sarcoma and has also shown activity in other sarcoma subtypes. However, its clinical efficacy is highly variable, and no reliable predictors exist to select patients who are likely to benefit from this drug. PATIENTS AND METHODS We analysed the molecular profiles and clinical outcomes of patients with pazopanib-treated sarcoma enrolled in a prospective observational study by the German Cancer Consortium, DKTK MASTER, that employs whole-genome/exome sequencing and transcriptome sequencing to inform the care of young adults with advanced cancer across histology and patients with rare cancers. RESULTS Among 109 patients with available whole-genome/exome sequencing data, there was no correlation between clinical parameters, specific genetic alterations or mutational signatures and clinical outcome. In contrast, the analysis of a subcohort of 62 patients who underwent molecular analysis before pazopanib treatment and had transcriptome sequencing data available showed that mRNA levels of NTRK3 (hazard ratio [HR] = 0.53, p = 0.021), IGF1R (HR = 1.82, p = 0.027) and KDR (HR = 0.50, p = 0.011) were independently associated with progression-free survival (PFS). Based on the expression of these multi-receptor tyrosine kinase genes, i.e. the features NTRK3-high, IGF1R-low and KDR-high, we developed a pazopanib efficacy predictor that stratified patients into three groups with significantly different PFS (p < 0.0001). Application of the pazopanib efficacy predictor to an independent cohort of patients with pazopanib-treated sarcoma from DKTK MASTER (n = 43) confirmed its potential to separate patient groups with significantly different PFS (p = 0.02), whereas no such association was observed in patients with sarcoma from DKTK MASTER (n = 97) or The Cancer Genome Atlas sarcoma cohort (n = 256) who were not treated with pazopanib. CONCLUSION A score based on the combined expression of NTRK3, IGF1R and KDR allows the identification of patients with sarcoma and with good, intermediate and poor outcome following pazopanib therapy and warrants prospective investigation as a predictive tool to optimise the use of this drug in the clinic.
Collapse
Affiliation(s)
- Christoph E Heilig
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany. https://twitter.com/ChrisHeiligMD
| | - Andreas Laßmann
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sadaf S Mughal
- Division of Applied Bioinformatics, DKFZ, Heidelberg, Germany
| | - Andreas Mock
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany. https://twitter.com/am0ck
| | - Sebastian Pirmann
- Computational Oncology Group, Molecular Precision Oncology Program, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Veronica Teleanu
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marcus Renner
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Carolin Andresen
- Pattern Recognition and Digital Medicine Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Heidelberg, Germany
| | - Bruno C Köhler
- Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany. https://twitter.com/koehlerlab
| | - Bogac Aybey
- Division of Applied Bioinformatics, DKFZ, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Sebastian Bauer
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; DKTK, Essen, Germany. https://twitter.com/seppobauer
| | - Jens T Siveke
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; DKTK, Essen, Germany; Division of Solid Tumor Translational Oncology, DKTK, Essen, and DKFZ, Heidelberg, Germany; Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Rainer Hamacher
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; DKTK, Essen, Germany
| | - Gunnar Folprecht
- Department of Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Stephan Richter
- Department of Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Evelin Schröck
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany; Center for Personalized Oncology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany; DKTK, Dresden, Germany
| | - Christian H Brandts
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, Frankfurt, Germany; Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany; Frankfurt Cancer Institute, Frankfurt, Germany; DKTK, Frankfurt, Germany
| | - Marit Ahrens
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Peter Hohenberger
- Department of Surgery, Mannheim University Medical Center, Heidelberg University, Mannheim, Germany; Sarcoma Unit, Interdisciplinary Tumor Center Mannheim, Mannheim University Medical Center, Heidelberg University, Mannheim, Germany
| | - Gerlinde Egerer
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Kindler
- UCT Mainz, Johannes Gutenberg University Mainz, Mainz, Germany; Department of Hematology, Medical Oncology and Pneumology, University Medical Center, Mainz, Germany; DKTK, Mainz, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, University of Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Comprehensive Cancer Center Freiburg, University of Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; DKTK, Freiburg, Germany
| | - Anna L Illert
- Comprehensive Cancer Center Freiburg, University of Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; DKTK, Freiburg, Germany; Department of Internal Medicine I, University of Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nikolas von Bubnoff
- Department of Internal Medicine I, University of Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - Leonidas Apostolidis
- Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany
| | - Philipp J Jost
- Department of Hematology and Oncology, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany; Division of Clinical Oncology, Department of Medicine, Medical University of Graz, Graz, Austria; DKTK, Munich, Germany
| | - C Benedikt Westphalen
- DKTK, Munich, Germany; Comprehensive Cancer Center, University Hospital, Ludwig Maximilians University Munich, Munich, Germany; Department of Medicine III, University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Wilko Weichert
- DKTK, Munich, Germany; Institute of Pathology, Technical University Munich, Munich, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany; DKTK, Berlin, Germany
| | - Frederick Klauschen
- DKTK, Berlin, Germany; Institute of Pathology, Charité - Universitätsmedizin Berlin, And Berlin Institute of Health, Berlin, Germany; Institute of Pathology, Ludwig Maximilians University Munich, Munich, Germany
| | - Katja Beck
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Ulrike Winter
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Daniela Richter
- Center for Personalized Oncology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany; DKTK, Dresden, Germany; Department of Translational Medical Oncology, NCT Dresden and DKFZ, Dresden, Germany
| | - Lino Möhrmann
- Center for Personalized Oncology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany; DKTK, Dresden, Germany; Department of Translational Medical Oncology, NCT Dresden and DKFZ, Dresden, Germany
| | - Michael Bitzer
- Department of Internal Medicine I, University Hospital, Eberhard-Karls University, Tübingen, Germany; DKTK, Tübingen, Germany
| | - Klaus Schulze-Osthoff
- DKTK, Tübingen, Germany; Department of Molecular Medicine, Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany
| | - Benedikt Brors
- German Cancer Consortium (DKTK), Heidelberg, Germany; Division of Applied Bioinformatics, DKFZ, Heidelberg, Germany
| | | | - Simon Kreutzfeldt
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Christoph Heining
- Center for Personalized Oncology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany; DKTK, Dresden, Germany; Department of Translational Medical Oncology, NCT Dresden and DKFZ, Dresden, Germany. https://twitter.com/ChrisHeining
| | - Daniel B Lipka
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany. https://twitter.com/dblipka1
| | - Albrecht Stenzinger
- German Cancer Consortium (DKTK), Heidelberg, Germany; Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Richard F Schlenk
- German Cancer Consortium (DKTK), Heidelberg, Germany; Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany; Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany; NCT Trial Center, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Peter Horak
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany. https://twitter.com/PeterHorak_MD
| | - Hanno Glimm
- Center for Personalized Oncology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany; DKTK, Dresden, Germany; Department of Translational Medical Oncology, NCT Dresden and DKFZ, Dresden, Germany
| | - Daniel Hübschmann
- German Cancer Consortium (DKTK), Heidelberg, Germany; Computational Oncology Group, Molecular Precision Oncology Program, NCT Heidelberg and DKFZ, Heidelberg, Germany; Pattern Recognition and Digital Medicine Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Heidelberg, Germany
| | - Stefan Fröhling
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
170
|
Zhang XR, Ouyang YC, Meng TG, Zhang HY, Yue W, Yan FZ, Xue Y, Schatten H, Wang ZB, Sun QY. OTSSP167 leads to follicular dysplasia and negatively affects oocyte quality in mice. Toxicology 2022; 476:153243. [PMID: 35760214 DOI: 10.1016/j.tox.2022.153243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/11/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022]
Abstract
OTSSP167 is an anti-tumor drug significantly inhibiting tumor growth in xenotransplantation studies using mouse breast, lung, prostate, and pancreatic cancer cell lines. Its phase I clinical trial has been completed, indicating its great potential for future treatment of solid tumors. However, its drug-related adverse effects on reproductive systems have not yet been reported. In this study, we evaluated the effects of OTSSP167 on reproduction of female mice by determining oocyte quality and follicular development. We selected four-week-old female ICR mice for a 21-day intraperitoneal injection of OTSSP167 at a dose of 5mg/kg/d. We found that OTSSP167 could block the meiotic process of oocytes, leading to a decrease in oocyte maturation and ovulated oocyte numbers, as well as a decrease in the quality of oocytes. The results showed that OTSSP167 treatment caused disordered spindle assembly, decreased mitochondria membrane potential, and increased accumulation of reactive oxygen species in oocytes. Further investigation showed that OTSSP167 induced DNA double-strand breaks, as indicated by increased levels of γH2AX in oocytes of primordial follicles and granulosa cells of growing follicles, which induced follicular atresia and decreased the numbers of follicles at various growing stages. Our study suggests that OTSSP167 treatment may have serious effects on the ovary and consequences for female cancer patients, providing strong evidence for the necessity of protecting female fertility in clinical OTSSP167 trials.
Collapse
Affiliation(s)
- Xin-Ran Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China; College of Life Science, University of Chinese Academy of Science, 100101, Beijing
| | - Ying-Chun Ouyang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Tie-Gang Meng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Hong-Yong Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Wei Yue
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China; College of Life Science, University of Chinese Academy of Science, 100101, Beijing
| | - Feng-Ze Yan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China; College of Life Science, University of Chinese Academy of Science, 100101, Beijing
| | - Yue Xue
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, USA
| | - Zhen-Bo Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China; College of Life Science, University of Chinese Academy of Science, 100101, Beijing
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
| |
Collapse
|
171
|
Omar AM, Khayat MT, Ahmed F, Muhammad YA, Malebari AM, Ibrahim SM, Khan MI, Shah DK, Childers WE, El-Araby ME. SAR Probing of KX2-391 Provided Analogues With Juxtaposed Activity Profile Against Major Oncogenic Kinases. Front Oncol 2022; 12:879457. [PMID: 35669422 PMCID: PMC9166630 DOI: 10.3389/fonc.2022.879457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Tirbanibulin (KX2-391, KX-01), a dual non-ATP (substrate site) Src kinase and tubulin-polymerization inhibitor, demonstrated a universal anti-cancer activity for variety of cancer types. The notion that KX2-391 is a highly selective Src kinase inhibitor have been challenged by recent reports on the activities of this drug against FLT3-ITD mutations in some leukemic cell lines. Therefore, we hypothesized that analogues of KX2-391 may inhibit oncogenic kinases other than Src. A set of 4-aroylaminophenyl-N-benzylacetamides were synthesized and found to be more active against leukemia cell lines compared to solid tumor cell lines. N-(4-(2-(benzylamino)-2-oxoethyl)phenyl)-4-chlorobenzamide (4e) exhibited activities at IC50 0.96 µM, 1.62 µM, 1.90 µM and 4.23 µM against NB4, HL60, MV4-11 and K562 leukemia cell lines, respectively. We found that underlying mechanisms of 4e did not include tubulin polymerization or Src inhibition. Such results interestingly suggested that scaffold-hopping of KX2-391 may change the two main underlying cytotoxic mechanisms (Src and tubulin). Kinase profiling using two methods revealed that 4e significantly reduces the activities of some other potent oncogenic kinases like the MAPK member ERK1/2 (>99%) and it also greatly upregulates the pro-apoptotic c-Jun kinase (84%). This research also underscores the importance of thorough investigation of total kinase activities as part of the structure-activity relationship studies.
Collapse
Affiliation(s)
- Abdelsattar M Omar
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, King Abdulaziz University, Jeddah, Saudi Arabia.,Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah, Saudi Arabia.,Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Al-Azhar University, Cairo, Egypt
| | - Maan T Khayat
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Farid Ahmed
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Yosra A Muhammad
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Azizah M Malebari
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sara M Ibrahim
- Faculty of Science, Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad I Khan
- Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah, Saudi Arabia.,Faculty of Science, Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Dhaval K Shah
- School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Wayne E Childers
- Moulder Center for Drug Discovery Research, School of Pharmacy, Department of Pharmaceutical Sciences, Temple University, Philadelphia, PA, United States
| | - Moustafa E El-Araby
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, King Abdulaziz University, Jeddah, Saudi Arabia.,Centre for Artificial Intelligence in Precision Medicines, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
172
|
Guardado Rivas MO, Stuart SD, Thach D, Dahan M, Shorr R, Zachar Z, Bingham PM. Evidence for a novel, effective approach to targeting carcinoma catabolism exploiting the first-in-class, anti-cancer mitochondrial drug, CPI-613. PLoS One 2022; 17:e0269620. [PMID: 35675354 PMCID: PMC9176802 DOI: 10.1371/journal.pone.0269620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/24/2022] [Indexed: 11/19/2022] Open
Abstract
Clinical targeting of the altered metabolism of tumor cells has long been considered an attractive hypothetical approach. However, this strategy has yet to perform well clinically. Metabolic redundancy is among the limitations on effectiveness of many approaches, engendering intrinsic single-agent resistance or efficient evolution of such resistance. We describe new studies of the multi-target, tumor-preferential inhibition of the mitochondrial tricarboxylic acid (TCA) cycle by the first-in-class drug CPI-613® (devimistat). By suppressing the TCA hub, indispensable to many metabolic pathways, CPI-613 substantially reduces the effective redundancy of tumor catabolism. This TCA cycle suppression also engenders an apparently homeostatic accelerated, inefficient consumption of nutrient stores in carcinoma cells, eroding some sources of drug resistance. Nonetheless, sufficiently abundant, cell line-specific lipid stores in carcinoma cells are among remaining sources of CPI-613 resistance in vitro and during the in vivo pharmacological drug pulse. Specifically, the fatty acid beta-oxidation step delivers electrons directly to the mitochondrial electron transport system (ETC), by-passing the TCA cycle CPI-613 target and producing drug resistance. Strikingly, tested carcinoma cell lines configure much of this fatty acid flow to initially traverse the peroxisome enroute to additional mitochondrial beta-oxidation. This feature facilitates targeting as clinically practical agents disrupting this flow are available. Two such agents significantly sensitize an otherwise fully CPI-613-resistant carcinoma xenograft in vivo. These and related results are strong empirical support for a potentially general class of strategies for enhanced clinical targeting of carcinoma catabolism.
Collapse
Affiliation(s)
- Moises O. Guardado Rivas
- Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, United States of America
- Graduate Program in Genetics, Stony Brook University, Stony Brook, NY, United States of America
- Rafael Pharmaceuticals, Cranbury, NJ, United States of America
| | - Shawn D. Stuart
- Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, United States of America
- Rafael Pharmaceuticals, Cranbury, NJ, United States of America
| | - Daniel Thach
- Rafael Pharmaceuticals, Cranbury, NJ, United States of America
| | - Michael Dahan
- Rafael Pharmaceuticals, Cranbury, NJ, United States of America
| | - Robert Shorr
- Rafael Pharmaceuticals, Cranbury, NJ, United States of America
| | - Zuzana Zachar
- Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, United States of America
| | - Paul M. Bingham
- Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, United States of America
- * E-mail:
| |
Collapse
|
173
|
Catalytic activity in vitro of the human protein kinase ASK1 mutants: Experimental and molecular simulation study. Comput Biol Chem 2022; 99:107712. [PMID: 35689994 DOI: 10.1016/j.compbiolchem.2022.107712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 11/21/2022]
Abstract
Kinases have become an important class of targets for drug discovery since the milestone approval of imatinib in 2001. Although a great success has been achieved for targeting kinases with over 70 inhibitors approved by the FDA, it is inevitable that drug resistance would emerge during treatment. Thus, assessment of the kinase mutations is an essential issue for the development of the next generation inhibitors. Apoptosis signal-regulating kinase 1 (ASK1) is a crucial regulator of classical mitogen-activated protein kinase cascade that is being explored under several clinical trials as a promising target. Herein, we investigate the catalytic activity in vitro of ASK1 by constructing two mutants: M754T and H729L, from gatekeeper and αC-helix, respectively. Compared to wild type, the mutation of M754T and H729L results in a roughly 3-fold and 2-fold decrease in binding affinity experimentally. In addition, their binding modes with substrate are theoretically predicted and compared by molecular dynamics. Trajectory analyses of simulations indicate that the decrease of binding affinity should be attributed to the loss of H-bond interaction with gatekeeper methionine. Unexpectedly, the conformation of αC-helix in H729L mutant did not alter significantly during the simulations, although the putatively important H-bond with H729 is lost. These simulations showed the regulatory role of H729 in αC-helix is maintained by leucine residue through the interaction with non-polar residues around H729 site.
Collapse
|
174
|
Betge J, Rindtorff N, Sauer J, Rauscher B, Dingert C, Gaitantzi H, Herweck F, Srour-Mhanna K, Miersch T, Valentini E, Boonekamp KE, Hauber V, Gutting T, Frank L, Belle S, Gaiser T, Buchholz I, Jesenofsky R, Härtel N, Zhan T, Fischer B, Breitkopf-Heinlein K, Burgermeister E, Ebert MP, Boutros M. The drug-induced phenotypic landscape of colorectal cancer organoids. Nat Commun 2022; 13:3135. [PMID: 35668108 PMCID: PMC9170716 DOI: 10.1038/s41467-022-30722-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 05/12/2022] [Indexed: 12/14/2022] Open
Abstract
Patient-derived organoids resemble the biology of tissues and tumors, enabling ex vivo modeling of human diseases. They have heterogeneous morphologies with unclear biological causes and relationship to treatment response. Here, we use high-throughput, image-based profiling to quantify phenotypes of over 5 million individual colorectal cancer organoids after treatment with >500 small molecules. Integration of data using multi-omics modeling identifies axes of morphological variation across organoids: Organoid size is linked to IGF1 receptor signaling, and cystic vs. solid organoid architecture is associated with LGR5 + stemness. Treatment-induced organoid morphology reflects organoid viability, drug mechanism of action, and is biologically interpretable. Inhibition of MEK leads to cystic reorganization of organoids and increases expression of LGR5, while inhibition of mTOR induces IGF1 receptor signaling. In conclusion, we identify shared axes of variation for colorectal cancer organoid morphology, their underlying biological mechanisms, and pharmacological interventions with the ability to move organoids along them. The heterogeneity underlying cancer organoid phenotypes is not yet well understood. Here, the authors develop an imaging analysis assay for high throughput phenotypic screening of colorectal organoids that allows to define specific morphological changes that occur following different drug treatments.
Collapse
Affiliation(s)
- Johannes Betge
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Heidelberg, Germany.,Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany.,German Cancer Research Center (DKFZ), Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, Heidelberg, Germany.,DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.,Mannheim Cancer Center, Mannheim, Germany
| | - Niklas Rindtorff
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Heidelberg, Germany
| | - Jan Sauer
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Heidelberg, Germany.,German Cancer Research Center (DKFZ), Computational Genome Biology Group, Heidelberg, Germany
| | - Benedikt Rauscher
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Heidelberg, Germany
| | - Clara Dingert
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Heidelberg, Germany
| | - Haristi Gaitantzi
- Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany.,Mannheim Cancer Center, Mannheim, Germany
| | - Frank Herweck
- Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany.,Mannheim Cancer Center, Mannheim, Germany
| | - Kauthar Srour-Mhanna
- Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany.,German Cancer Research Center (DKFZ), Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, Heidelberg, Germany.,DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
| | - Thilo Miersch
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Heidelberg, Germany
| | - Erica Valentini
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Heidelberg, Germany
| | - Kim E Boonekamp
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Heidelberg, Germany
| | - Veronika Hauber
- Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany.,Mannheim Cancer Center, Mannheim, Germany
| | - Tobias Gutting
- Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany.,Mannheim Cancer Center, Mannheim, Germany.,Department of Internal Medicine IV, Heidelberg University, Heidelberg, Germany
| | - Larissa Frank
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Heidelberg, Germany
| | - Sebastian Belle
- Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany.,Mannheim Cancer Center, Mannheim, Germany
| | - Timo Gaiser
- Mannheim Cancer Center, Mannheim, Germany.,Heidelberg University, Institute of Pathology, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany
| | - Inga Buchholz
- Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany.,Mannheim Cancer Center, Mannheim, Germany
| | - Ralf Jesenofsky
- Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany.,Mannheim Cancer Center, Mannheim, Germany
| | - Nicolai Härtel
- Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany.,Mannheim Cancer Center, Mannheim, Germany
| | - Tianzuo Zhan
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Heidelberg, Germany.,Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany.,Mannheim Cancer Center, Mannheim, Germany
| | - Bernd Fischer
- German Cancer Research Center (DKFZ), Computational Genome Biology Group, Heidelberg, Germany
| | - Katja Breitkopf-Heinlein
- Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany.,Mannheim Cancer Center, Mannheim, Germany
| | - Elke Burgermeister
- Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany.,Mannheim Cancer Center, Mannheim, Germany
| | - Matthias P Ebert
- Heidelberg University, Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Mannheim, Germany. .,DKFZ-Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany. .,Mannheim Cancer Center, Mannheim, Germany.
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, and Heidelberg University, Medical Faculty Mannheim, Department of Cell and Molecular Biology, Heidelberg, Germany. .,German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
175
|
Schipper LJ, Zeverijn LJ, Garnett MJ, Voest EE. Can Drug Repurposing Accelerate Precision Oncology? Cancer Discov 2022; 12:1634-1641. [PMID: 35642948 DOI: 10.1158/2159-8290.cd-21-0612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 01/28/2022] [Accepted: 04/11/2022] [Indexed: 11/16/2022]
Abstract
Ongoing new insights in the field of cancer diagnostics, genomic profiling, and cancer behavior have raised the demand for novel, personalized cancer treatments. As the development of new cancer drugs is a challenging, costly, and time-consuming endeavor, drug repurposing is regarded as an attractive alternative to potentially accelerate this. In this review, we describe strategies for drug repurposing of anticancer agents, translation of preclinical findings in novel trial designs, and associated challenges. Furthermore, we provide suggestions to further utilize the potential of drug repurposing within precision oncology, with a focus on combinatorial approaches. SIGNIFICANCE Oncologic drug development is a timely and costly endeavor, with only few compounds progressing to meaningful therapy options. Although repurposing of existing agents for novel, oncologic indications provides an opportunity to accelerate this process, it is not without challenges.
Collapse
Affiliation(s)
- Luuk J Schipper
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands.,Oncode Institute, Utrecht, the Netherlands
| | - Laurien J Zeverijn
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands.,Oncode Institute, Utrecht, the Netherlands
| | | | - Emile E Voest
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, the Netherlands.,Oncode Institute, Utrecht, the Netherlands
| |
Collapse
|
176
|
Montaño JL, Wang BJ, Volk RF, Warrington SE, Garda VG, Hofmann KL, Chen LC, Zaro BW. Improved Electrophile Design for Exquisite Covalent Molecule Selectivity. ACS Chem Biol 2022; 17:1440-1449. [PMID: 35587148 DOI: 10.1021/acschembio.1c00980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Covalent inhibitors are viable therapeutics. However, off-target reactivity challenges the field. Chemists have attempted to solve this issue by varying the reactivity attributes of electrophilic warheads. Here, we report the development of an approach to increase the selectivity of covalent molecules that is independent of warhead reactivity features and can be used in concert with existing methods. Using the scaffold of the Bruton's tyrosine kinase (BTK) inhibitor Ibrutinib for our proof-of-concept, we reasoned that increasing the steric bulk of fumarate-based electrophiles on Ibrutinib should improve selectivity via the steric exclusion of off-targets but retain rates of cysteine reactivity comparable to that of an acrylamide. Using chemical proteomic techniques, we demonstrate that elaboration of the electrophile to a tert-butyl (t-Bu) fumarate ester decreases time-dependent off-target reactivity and abolishes time-independent off-target reactivity. While an alkyne-bearing probe analogue of Ibrutinib has 247 protein targets, our t-Bu fumarate probe analogue has only 7. Of these 7 targets, BTK is the only time-independent target. The t-Bu inhibitor itself is also more selective for BTK, reducing off-targets by 70%. We investigated the consequences of treatment with Ibrutinib and our t-Bu analogue and discovered that only 8 proteins are downregulated in response to treatment with the t-Bu analogue compared to 107 with Ibrutinib. Of these 8 proteins, 7 are also downregulated by Ibrutinib and a majority of these targets are associated with BTK biology. Taken together, these findings reveal an opportunity to increase cysteine-reactive covalent inhibitor selectivity through electrophilic structure optimization.
Collapse
Affiliation(s)
- José L. Montaño
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, California 94158, United States
| | - Brian J. Wang
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, California 94158, United States
| | - Regan F. Volk
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, California 94158, United States
| | - Sara E. Warrington
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, California 94158, United States
| | - Virginia G. Garda
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, California 94158, United States
| | - Katherine L. Hofmann
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, California 94158, United States
| | - Leo C. Chen
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, California 94158, United States
| | - Balyn W. Zaro
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, California 94158, United States
| |
Collapse
|
177
|
Wu X, Xu Y, Liang Q, Yang X, Huang J, Wang J, Zhang H, Shi J. Recent Advances in Dual PI3K/mTOR Inhibitors for Tumour Treatment. Front Pharmacol 2022; 13:875372. [PMID: 35614940 PMCID: PMC9124774 DOI: 10.3389/fphar.2022.875372] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/14/2022] [Indexed: 12/31/2022] Open
Abstract
The PI3K-Akt-mTOR pathway is a viable target for cancer treatment and can be used to treat various malignant tumours, including follicular lymphoma and breast cancer. Both enzymes, PI3K and mTOR, are critical in this pathway. Hence, in recent years, an array of inhibitors targeting these two targets have been studied, showing dual PI3K/mTOR inhibition compared with single targeting small molecule inhibitors. Inhibitors not only inhibit cell proliferation but also promote cell apoptosis. These inhibitors show high potency and little drug resistance even at low doses, suggesting that PI3K/mTOR inhibitors are promising cancer drugs. Herein, we summarised the recent research of PI3K/mTOR dual inhibitors—for example, structure-activity relationship, pharmacokinetics, and clinical practice, and briefly commented on them. Clinical Trial Registration:https://clinicaltrials.gov.
Collapse
Affiliation(s)
- Xianbo Wu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Yihua Xu
- School of Basic Medical Science, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qi Liang
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Xinwei Yang
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Jianli Huang
- First Clinical College of Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jie Wang
- First Clinical College of Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hong Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
178
|
A Comprehensive Overview of Globally Approved JAK Inhibitors. Pharmaceutics 2022; 14:pharmaceutics14051001. [PMID: 35631587 PMCID: PMC9146299 DOI: 10.3390/pharmaceutics14051001] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/22/2022] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
Janus kinase (JAK) is a family of cytoplasmic non-receptor tyrosine kinases that includes four members, namely JAK1, JAK2, JAK3, and TYK2. The JAKs transduce cytokine signaling through the JAK-STAT pathway, which regulates the transcription of several genes involved in inflammatory, immune, and cancer conditions. Targeting the JAK family kinases with small-molecule inhibitors has proved to be effective in the treatment of different types of diseases. In the current review, eleven of the JAK inhibitors that received approval for clinical use have been discussed. These drugs are abrocitinib, baricitinib, delgocitinib, fedratinib, filgotinib, oclacitinib, pacritinib, peficitinib, ruxolitinib, tofacitinib, and upadacitinib. The aim of the current review was to provide an integrated overview of the chemical and pharmacological data of the globally approved JAK inhibitors. The synthetic routes of the eleven drugs were described. In addition, their inhibitory activities against different kinases and their pharmacological uses have also been explained. Moreover, their crystal structures with different kinases were summarized, with a primary focus on their binding modes and interactions. The proposed metabolic pathways and metabolites of these drugs were also illustrated. To sum up, the data in the current review could help in the design of new JAK inhibitors with potential therapeutic benefits in inflammatory and autoimmune diseases.
Collapse
|
179
|
Beusch CM, Sabatier P, Zubarev RA. Ion-Based Proteome-Integrated Solubility Alteration Assays for Systemwide Profiling of Protein-Molecule Interactions. Anal Chem 2022; 94:7066-7074. [PMID: 35506705 PMCID: PMC9118197 DOI: 10.1021/acs.analchem.2c00391] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
![]()
Unbiased drug target
engagement deconvolution and mechanism of
action elucidation are major challenges in drug development. Modification-free
target engagement methods, such as thermal proteome profiling, have
gained increasing popularity in the last several years. However, these
methods have limitations, and, in any case, new orthogonal approaches
are needed. Here, we present a novel isothermal method for comprehensive
characterization of protein solubility alterations using the effect
on protein solubility of cations and anions in the Hofmeister series.
We combine the ion-based protein precipitation approach with Proteome-Integrated
Solubility Alteration (PISA) analysis and use this I-PISA assay to
delineate the targets of several anticancer drugs both in cell lysates
and intact cells. Finally, we demonstrate that I-PISA can detect solubility
changes in minute amounts of sample, opening chemical proteomics applications
to small and rare biological material.
Collapse
Affiliation(s)
- Christian M Beusch
- Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17177, Sweden
| | - Pierre Sabatier
- Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17177, Sweden
| | - Roman A Zubarev
- Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17177, Sweden.,Department of Pharmacological & Technological Chemistry, I.M. Sechenov First Moscow State Medical University, Moscow 119146, Russia.,The National Medical Research Centre for Endocrinology, Moscow 115478, Russia
| |
Collapse
|
180
|
Sunderhaus A, Imran R, Enoh E, Adedeji A, Obafemi T, Abdel Aziz MH. Comparative expression of soluble, active human kinases in specialized bacterial strains. PLoS One 2022; 17:e0267226. [PMID: 35439268 PMCID: PMC9017934 DOI: 10.1371/journal.pone.0267226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/05/2022] [Indexed: 11/19/2022] Open
Abstract
Kinases act as molecular switches for cellular functions and are involved in multiple human pathogeneses, most notably cancer. There is a continuous need for soluble and active kinases for in-vitro drug discovery and structural biology purposes. Kinases remain challenging to express using Escherichia coli, the most widely utilized host for heterologous expression. In this work, four bacterial strains, BL21 (DE3), BL21 (DE3) pLysS, Rosetta, and Arctic Express, were chosen for parallel expression trials along with BL21 (DE3) complemented with folding chaperones DnaJ/K and GroEL/ES to compare their performance in producing soluble and active human kinases. Three representative diverse kinases were studied, Epidermal Growth Factor Receptor kinase domain, Aurora Kinase A kinase domain, and Mitogen-activated protein Kinase Kinase. The genes encoding the kinases were subcloned into pET15b bacterial plasmid and transformed into the bacterial strains. Soluble kinase expression was tested using different IPTG concentrations (1–0.05 mM) at varying temperatures (37°C– 10°C) and induction times (3–24 hours). The optimum conditions for each kinase in all strains were then used for 1L large scale cultures from which each kinase was purified to compare yield, purity, oligomerization status, and activity. Although using specialized strains achieved improvements in yield and/or activity for the three kinases, none of the tested strains was universally superior, highlighting the individuality in kinase expression.
Collapse
Affiliation(s)
- Allison Sunderhaus
- Department of Pharmaceutical Sciences and Health Outcomes, Fisch College of Pharmacy, The University of Texas at Tyler, Tyler, Texas, United States of America
| | - Ramsha Imran
- Department of Pharmaceutical Sciences and Health Outcomes, Fisch College of Pharmacy, The University of Texas at Tyler, Tyler, Texas, United States of America
| | - Elanzou Enoh
- Department of Pharmaceutical Sciences and Health Outcomes, Fisch College of Pharmacy, The University of Texas at Tyler, Tyler, Texas, United States of America
| | - Adesola Adedeji
- Department of Pharmaceutical Sciences and Health Outcomes, Fisch College of Pharmacy, The University of Texas at Tyler, Tyler, Texas, United States of America
| | - Taiye Obafemi
- Department of Pharmaceutical Sciences and Health Outcomes, Fisch College of Pharmacy, The University of Texas at Tyler, Tyler, Texas, United States of America
| | - May H. Abdel Aziz
- Department of Pharmaceutical Sciences and Health Outcomes, Fisch College of Pharmacy, The University of Texas at Tyler, Tyler, Texas, United States of America
- * E-mail:
| |
Collapse
|
181
|
Abstract
Proteins are the molecular effectors of the information encoded in the genome. Proteomics aims at understanding the molecular functions of proteins in their biological context. In contrast to transcriptomics and genomics, the study of proteomes provides deeper insight into the dynamic regulatory layers encoded at the protein level, such as posttranslational modifications, subcellular localization, cell signaling, and protein-protein interactions. Currently, mass spectrometry (MS)-based proteomics is the technology of choice for studying proteomes at a system-wide scale, contributing to clinical biomarker discovery and fundamental molecular biology. MS technologies are continuously being developed to fulfill the requirements of speed, resolution, and quantitative accuracy, enabling the acquisition of comprehensive proteomes. In this review, we present how MS technology and acquisition methods have evolved to meet the requirements of cutting-edge proteomics research, which is describing the human proteome and its dynamic posttranslational modifications with unprecedented depth. Finally, we provide a perspective on studying proteomes at single-cell resolution. Expected final online publication date for the Annual Review of Genomics and Human Genetics, Volume 23 is October 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Ana Martinez-Val
- Novo Nordisk Foundation Center for Protein Research, Proteomics Program, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Ulises H Guzmán
- Novo Nordisk Foundation Center for Protein Research, Proteomics Program, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, Proteomics Program, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| |
Collapse
|
182
|
van Linde ME, Labots M, Brahm CG, Hovinga KE, De Witt Hamer PC, Honeywell RJ, de Goeij-de Haas R, Henneman AA, Knol JC, Peters GJ, Dekker H, Piersma SR, Pham TV, Vandertop WP, Jiménez CR, Verheul HM. Tumor Drug Concentration and Phosphoproteomic Profiles After Two Weeks of Treatment With Sunitinib in Patients with Newly Diagnosed Glioblastoma. Clin Cancer Res 2022; 28:1595-1602. [PMID: 35165100 PMCID: PMC9365363 DOI: 10.1158/1078-0432.ccr-21-1933] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/14/2021] [Accepted: 02/09/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE Tyrosine kinase inhibitors (TKI) have poor efficacy in patients with glioblastoma (GBM). Here, we studied whether this is predominantly due to restricted blood-brain barrier penetration or more to biological characteristics of GBM. PATIENTS AND METHODS Tumor drug concentrations of the TKI sunitinib after 2 weeks of preoperative treatment was determined in 5 patients with GBM and compared with its in vitro inhibitory concentration (IC50) in GBM cell lines. In addition, phosphotyrosine (pTyr)-directed mass spectrometry (MS)-based proteomics was performed to evaluate sunitinib-treated versus control GBM tumors. RESULTS The median tumor sunitinib concentration of 1.9 μmol/L (range 1.0-3.4) was 10-fold higher than in concurrent plasma, but three times lower than sunitinib IC50s in GBM cell lines (median 5.4 μmol/L, 3.0-8.5; P = 0.01). pTyr-phosphoproteomic profiles of tumor samples from 4 sunitinib-treated versus 7 control patients revealed 108 significantly up- and 23 downregulated (P < 0.05) phosphopeptides for sunitinib treatment, resulting in an EGFR-centered signaling network. Outlier analysis of kinase activities as a potential strategy to identify drug targets in individual tumors identified nine kinases, including MAPK10 and INSR/IGF1R. CONCLUSIONS Achieved tumor sunitinib concentrations in patients with GBM are higher than in plasma, but lower than reported for other tumor types and insufficient to significantly inhibit tumor cell growth in vitro. Therefore, alternative TKI dosing to increase intratumoral sunitinib concentrations might improve clinical benefit for patients with GBM. In parallel, a complex profile of kinase activity in GBM was found, supporting the potential of (phospho)proteomic analysis for the identification of targets for (combination) treatment.
Collapse
Affiliation(s)
- Myra E. van Linde
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Mariette Labots
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Cyrillo G. Brahm
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Koos E. Hovinga
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Philip C. De Witt Hamer
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Richard J. Honeywell
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Pharmacy, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Richard de Goeij-de Haas
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Alex A. Henneman
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Jaco C. Knol
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Godefridus J. Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | - Henk Dekker
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Sander R. Piersma
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Thang V. Pham
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - William P. Vandertop
- Department of Neurosurgery, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Connie R. Jiménez
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Henk M.W. Verheul
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Medical Oncology, Radboud UMC, Nijmegen, the Netherlands
| |
Collapse
|
183
|
Differentiating Inhibitors of Closely Related Protein Kinases with Single- or Multi-Target Activity via Explainable Machine Learning and Feature Analysis. Biomolecules 2022; 12:biom12040557. [PMID: 35454147 PMCID: PMC9032434 DOI: 10.3390/biom12040557] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 01/01/2023] Open
Abstract
Protein kinases are major drug targets. Most kinase inhibitors are directed against the adenosine triphosphate (ATP) cofactor binding site, which is largely conserved across the human kinome. Hence, such kinase inhibitors are often thought to be promiscuous. However, experimental evidence and activity data for publicly available kinase inhibitors indicate that this is not generally the case. We have investigated whether inhibitors of closely related human kinases with single- or multi-kinase activity can be differentiated on the basis of chemical structure. Therefore, a test system consisting of two distinct kinase triplets has been devised for which inhibitors with reported triple-kinase activities and corresponding single-kinase activities were assembled. Machine learning models derived on the basis of chemical structure distinguished between these multi- and single-kinase inhibitors with high accuracy. A model-independent explanatory approach was applied to identify structural features determining accurate predictions. For both kinase triplets, the analysis revealed decisive features contained in multi-kinase inhibitors. These features were found to be absent in corresponding single-kinase inhibitors, thus providing a rationale for successful machine learning. Mapping of features determining accurate predictions revealed that they formed coherent and chemically meaningful substructures that were characteristic of multi-kinase inhibitors compared with single-kinase inhibitors.
Collapse
|
184
|
Lee PY, Yeoh Y, Low TY. A recent update on small‐molecule kinase inhibitors for targeted cancer therapy and their therapeutic insights from mass spectrometry‐based proteomic analysis. FEBS J 2022. [DOI: 10.1111/febs.16442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/21/2022] [Accepted: 03/18/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Pey Yee Lee
- UKM Medical Molecular Biology Institute (UMBI) Universiti Kebangsaan Malaysia Kuala Lumpur Malaysia
| | - Yeelon Yeoh
- UKM Medical Molecular Biology Institute (UMBI) Universiti Kebangsaan Malaysia Kuala Lumpur Malaysia
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI) Universiti Kebangsaan Malaysia Kuala Lumpur Malaysia
| |
Collapse
|
185
|
The emerging role of mass spectrometry-based proteomics in drug discovery. Nat Rev Drug Discov 2022; 21:637-654. [PMID: 35351998 DOI: 10.1038/s41573-022-00409-3] [Citation(s) in RCA: 136] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2022] [Indexed: 12/14/2022]
Abstract
Proteins are the main targets of most drugs; however, system-wide methods to monitor protein activity and function are still underused in drug discovery. Novel biochemical approaches, in combination with recent developments in mass spectrometry-based proteomics instrumentation and data analysis pipelines, have now enabled the dissection of disease phenotypes and their modulation by bioactive molecules at unprecedented resolution and dimensionality. In this Review, we describe proteomics and chemoproteomics approaches for target identification and validation, as well as for identification of safety hazards. We discuss innovative strategies in early-stage drug discovery in which proteomics approaches generate unique insights, such as targeted protein degradation and the use of reactive fragments, and provide guidance for experimental strategies crucial for success.
Collapse
|
186
|
Abstract
With several marketed drugs, allosteric inhibition of kinases has translated to pharmacological effects and clinical benefits comparable to those from orthosteric inhibition. However, despite much effort over more than 20 years, the number of kinase targets associated with FDA-approved allosteric drugs is limited, suggesting the challenges in identifying and validating allosteric inhibitors. Here we review the principles of allosteric inhibition, summarize the discovery of allosteric MEK1/2 and BCR-ABL1 inhibitors, and discuss the approaches to screening and demonstrating the functional activity of allosteric pocket ligands.
Collapse
Affiliation(s)
- Yue Pan
- Relay Therapeutics, 399 Binney Street, Cambridge, Massachusetts 02139, United States
| | - Mary M Mader
- Relay Therapeutics, 399 Binney Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
187
|
Malchow S, Korepanova A, Panchal SC, McClure RA, Longenecker KL, Qiu W, Zhao H, Cheng M, Guo J, Klinge KL, Trusk P, Pratt SD, Li T, Kurnick MD, Duan L, Shoemaker AR, Gopalakrishnan SM, Warder SE, Shotwell JB, Lai A, Sun C, Osuma AT, Pappano WN. The HPK1 Inhibitor A-745 Verifies the Potential of Modulating T Cell Kinase Signaling for Immunotherapy. ACS Chem Biol 2022; 17:556-566. [PMID: 35188729 DOI: 10.1021/acschembio.1c00819] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hematopoietic progenitor kinase 1 (HPK1) is an MAP4K family member within the Ste20-like serine/threonine branch of the kinome. HPK1 expression is limited to hematopoietic cells and has a predominant role as a negative regulator of T cell function. Because of the central/dominant role in negatively regulating T cell function, HPK1 has long been in the center of interest as a potential pharmacological target for immune therapy. The development of a small molecule HPK1 inhibitor remains challenging because of the need for high specificity relative to other kinases, including additional MAP4K family members, that are required for efficient immune cell activation. Here, we report the identification of the selective and potent HPK1 chemical probe, A-745. In unbiased cellular kinase-binding assays, A-745 demonstrates an excellent cellular selectivity binding profile within pharmacologically relevant concentrations. This HPK1 selectivity translates to an in vitro immune cell activation phenotype reminiscent of Hpk1-deficient and Hpk1-kinase-dead T cells, including augmented proliferation and cytokine production. The results from this work give a path forward for further developmental efforts to generate additional selective and potent small molecule HPK1 inhibitors with the pharmacological properties for immunotherapy.
Collapse
Affiliation(s)
- Sven Malchow
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Alla Korepanova
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Sanjay C. Panchal
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Ryan A. McClure
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | | | - Wei Qiu
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Hongyu Zhao
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Min Cheng
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Jun Guo
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Kelly L. Klinge
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Patricia Trusk
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Steven D. Pratt
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Tao Li
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Matthew D. Kurnick
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Lishu Duan
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Alex R. Shoemaker
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | | | - Scott E. Warder
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - J. Brad Shotwell
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Albert Lai
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Chaohong Sun
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - Augustine T. Osuma
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| | - William N. Pappano
- AbbVie Inc., 1 North Waukegan Rd., North Chicago, Illinois 60064, United States
| |
Collapse
|
188
|
Costantini S, Meloni A, Spasiano A, Cinque P, Ricchi P. The unexpected impact of cabozantinib on red blood cells consumption in patients with transfusion-dependent thalassemia. Ann Hematol 2022; 101:1621-1623. [PMID: 35246730 DOI: 10.1007/s00277-022-04810-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 02/28/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Silvia Costantini
- U.O.S.D. Malattie Rare del Globulo Rosso, Azienda Ospedaliera Di Rilievo Nazionale "A. Cardarelli", Via A.Cardarelli 9, 80131, Naples, Italy
| | - Antonella Meloni
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, Pisa, Italy
| | - Anna Spasiano
- U.O.S.D. Malattie Rare del Globulo Rosso, Azienda Ospedaliera Di Rilievo Nazionale "A. Cardarelli", Via A.Cardarelli 9, 80131, Naples, Italy
| | - Patrizia Cinque
- U.O.S.D. Malattie Rare del Globulo Rosso, Azienda Ospedaliera Di Rilievo Nazionale "A. Cardarelli", Via A.Cardarelli 9, 80131, Naples, Italy
| | - Paolo Ricchi
- U.O.S.D. Malattie Rare del Globulo Rosso, Azienda Ospedaliera Di Rilievo Nazionale "A. Cardarelli", Via A.Cardarelli 9, 80131, Naples, Italy.
| |
Collapse
|
189
|
Montinaro A, Areso Zubiaur I, Saggau J, Kretz AL, Ferreira RMM, Hassan O, Kitzig E, Müller I, El-Bahrawy MA, von Karstedt S, Kulms D, Liccardi G, Lemke J, Walczak H. Potent pro-apoptotic combination therapy is highly effective in a broad range of cancers. Cell Death Differ 2022; 29:492-503. [PMID: 34535764 PMCID: PMC8901660 DOI: 10.1038/s41418-021-00869-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/14/2022] Open
Abstract
Primary or acquired therapy resistance is a major obstacle to the effective treatment of cancer. Resistance to apoptosis has long been thought to contribute to therapy resistance. We show here that recombinant TRAIL and CDK9 inhibition cooperate in killing cells derived from a broad range of cancers, importantly without inducing detectable adverse events. Remarkably, the combination of TRAIL with CDK9 inhibition was also highly effective on cancers resistant to both, standard-of-care chemotherapy and various targeted therapeutic approaches. Dynamic BH3 profiling revealed that, mechanistically, combining TRAIL with CDK9 inhibition induced a drastic increase in the mitochondrial priming of cancer cells. Intriguingly, this increase occurred irrespective of whether the cancer cells were sensitive or resistant to chemo- or targeted therapy. We conclude that this pro-apoptotic combination therapy has the potential to serve as a highly effective new treatment option for a variety of different cancers. Notably, this includes cancers that are resistant to currently available treatment modalities.
Collapse
Affiliation(s)
- Antonella Montinaro
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Itziar Areso Zubiaur
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Julia Saggau
- CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany
- Center for Biochemistry, Medical Faculty, Joseph-Stelzmann-Str. 52, University of Cologne, 50931, Cologne, Germany
| | - Anna-Laura Kretz
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Rute M M Ferreira
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Omar Hassan
- CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany
- Center for Biochemistry, Medical Faculty, Joseph-Stelzmann-Str. 52, University of Cologne, 50931, Cologne, Germany
| | - Ella Kitzig
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Ines Müller
- Experimental Dermatology, Department of Dermatology, Technical University Dresden, Dresden, Germany
| | - Mona A El-Bahrawy
- Department of Histopathology, Imperial College London, London, W12 0NN, UK
| | - Silvia von Karstedt
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
- CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany
- Department of Translational Genomics, Center of Integrated Oncology Cologne-Bonn, Medical Faculty, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne, Medical Faculty, University Hospital of Cologne, 50931, Cologne, Germany
| | - Dagmar Kulms
- Experimental Dermatology, Department of Dermatology, Technical University Dresden, Dresden, Germany
| | - Gianmaria Liccardi
- Center for Biochemistry, Medical Faculty, Joseph-Stelzmann-Str. 52, University of Cologne, 50931, Cologne, Germany
| | - Johannes Lemke
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Henning Walczak
- Centre for Cell Death, Cancer, and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK.
- CECAD Cluster of Excellence, University of Cologne, 50931, Cologne, Germany.
- Center for Biochemistry, Medical Faculty, Joseph-Stelzmann-Str. 52, University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
190
|
Selective multi-kinase inhibition sensitizes mesenchymal pancreatic cancer to immune checkpoint blockade by remodeling the tumor microenvironment. NATURE CANCER 2022; 3:318-336. [PMID: 35122074 PMCID: PMC7612546 DOI: 10.1038/s43018-021-00326-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 12/13/2021] [Indexed: 12/14/2022]
Abstract
KRAS-mutant pancreatic ductal adenocarcinoma (PDAC) is highly immunosuppressive and resistant to targeted and immunotherapies. Among the different PDAC subtypes, basal-like mesenchymal PDAC, which is driven by allelic imbalance, increased gene dosage and subsequent high expression levels of oncogenic KRAS, shows the most aggressive phenotype and strongest therapy resistance. In the present study, we performed a systematic high-throughput combination drug screen and identified a synergistic interaction between the MEK inhibitor trametinib and the multi-kinase inhibitor nintedanib, which targets KRAS-directed oncogenic signaling in mesenchymal PDAC. This combination treatment induces cell-cycle arrest and cell death, and initiates a context-dependent remodeling of the immunosuppressive cancer cell secretome. Using a combination of single-cell RNA-sequencing, CRISPR screens and immunophenotyping, we show that this combination therapy promotes intratumor infiltration of cytotoxic and effector T cells, which sensitizes mesenchymal PDAC to PD-L1 immune checkpoint inhibition. Overall, our results open new avenues to target this aggressive and therapy-refractory mesenchymal PDAC subtype.
Collapse
|
191
|
Kelly RL, Huehls AM, Venkatachalam A, Huntoon CJ, Machida YJ, Karnitz LM. Intra-S phase checkpoint kinase Chk1 dissociates replication proteins Treslin and TopBP1 through multiple mechanisms during replication stress. J Biol Chem 2022; 298:101777. [PMID: 35231445 PMCID: PMC8965152 DOI: 10.1016/j.jbc.2022.101777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/17/2022] [Accepted: 02/19/2022] [Indexed: 11/17/2022] Open
Abstract
Replication stress impedes DNA polymerase progression causing activation of the ataxia telangiectasia and Rad3-related signaling pathway, which promotes the intra-S phase checkpoint activity through phosphorylation of checkpoint kinase 1 (Chk1). Chk1 suppresses replication origin firing, in part, by disrupting the interaction between the preinitiation complex components Treslin and TopBP1, an interaction that is mediated by TopBP1 BRCT domain-binding to two cyclin-dependent kinase (CDK) phosphorylation sites, T968 and S1000, in Treslin. Two nonexclusive models for how Chk1 regulates the Treslin–TopBP1 interaction have been proposed in the literature: in one model, these proteins dissociate due to a Chk1-induced decrease in CDK activity that reduces phosphorylation of the Treslin sites that bind TopBP1 and in the second model, Chk1 directly phosphorylates Treslin, resulting in dissociation of TopBP1. However, these models have not been formally examined. We show here that Treslin T968 phosphorylation was decreased in a Chk1-dependent manner, while Treslin S1000 phosphorylation was unchanged, demonstrating that T968 and S1000 are differentially regulated. However, CDK2-mediated phosphorylation alone did not fully account for Chk1 regulation of the Treslin–TopBP1 interaction. We also identified additional Chk1 phosphorylation sites on Treslin that contributed to disruption of the Treslin–TopBP1 interaction, including S1114. Finally, we showed that both of the proposed mechanisms regulate origin firing in cancer cell line models undergoing replication stress, with the relative roles of each mechanism varying among cell lines. This study demonstrates that Chk1 regulates Treslin through multiple mechanisms to promote efficient dissociation of Treslin and TopBP1 and furthers our understanding of Treslin regulation during the intra-S phase checkpoint.
Collapse
Affiliation(s)
- Rebecca L Kelly
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Amelia M Huehls
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Annapoorna Venkatachalam
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Yuichi J Machida
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA; Division of Oncology Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Larry M Karnitz
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA; Division of Oncology Research, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
192
|
Bagheri S, Rahban M, Bostanian F, Esmaeilzadeh F, Bagherabadi A, Zolghadri S, Stanek A. Targeting Protein Kinases and Epigenetic Control as Combinatorial Therapy Options for Advanced Prostate Cancer Treatment. Pharmaceutics 2022; 14:515. [PMID: 35335890 PMCID: PMC8949110 DOI: 10.3390/pharmaceutics14030515] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 02/15/2022] [Accepted: 02/21/2022] [Indexed: 02/02/2023] Open
Abstract
Prostate cancer (PC), the fifth leading cause of cancer-related mortality worldwide, is known as metastatic bone cancer when it spreads to the bone. Although there is still no effective treatment for advanced/metastatic PC, awareness of the molecular events that contribute to PC progression has opened up opportunities and raised hopes for the development of new treatment strategies. Androgen deprivation and androgen-receptor-targeting therapies are two gold standard treatments for metastatic PC. However, acquired resistance to these treatments is a crucial challenge. Due to the role of protein kinases (PKs) in the growth, proliferation, and metastases of prostatic tumors, combinatorial therapy by PK inhibitors may help pave the way for metastatic PC treatment. Additionally, PC is known to have epigenetic involvement. Thus, understanding epigenetic pathways can help adopt another combinatorial treatment strategy. In this study, we reviewed the PKs that promote PC to advanced stages. We also summarized some PK inhibitors that may be used to treat advanced PC and we discussed the importance of epigenetic control in this cancer. We hope the information presented in this article will contribute to finding an effective treatment for the management of advanced PC.
Collapse
Affiliation(s)
- Soghra Bagheri
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran;
| | - Mahdie Rahban
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614335, Iran; (M.R.); (F.B.)
| | - Fatemeh Bostanian
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614335, Iran; (M.R.); (F.B.)
| | - Fatemeh Esmaeilzadeh
- Department of Biology, Jahrom Branch, Islamic Azad University, Jahrom 7414785318, Iran;
| | - Arash Bagherabadi
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil 5619911367, Iran;
| | - Samaneh Zolghadri
- Department of Biology, Jahrom Branch, Islamic Azad University, Jahrom 7414785318, Iran;
| | - Agata Stanek
- Department of Internal Medicine, Angiology and Physical Medicine, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Batorego 15 St, 41-902 Bytom, Poland
| |
Collapse
|
193
|
Cordo’ V, Meijer MT, Hagelaar R, de Goeij-de Haas RR, Poort VM, Henneman AA, Piersma SR, Pham TV, Oshima K, Ferrando AA, Zaman GJR, Jimenez CR, Meijerink JPP. Phosphoproteomic profiling of T cell acute lymphoblastic leukemia reveals targetable kinases and combination treatment strategies. Nat Commun 2022; 13:1048. [PMID: 35217681 PMCID: PMC8881579 DOI: 10.1038/s41467-022-28682-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/26/2022] [Indexed: 01/05/2023] Open
Abstract
Protein kinase inhibitors are amongst the most successful cancer treatments, but targetable kinases activated by genomic abnormalities are rare in T cell acute lymphoblastic leukemia. Nevertheless, kinases can be activated in the absence of genetic defects. Thus, phosphoproteomics can provide information on pathway activation and signaling networks that offer opportunities for targeted therapy. Here, we describe a mass spectrometry-based global phosphoproteomic profiling of 11 T cell acute lymphoblastic leukemia cell lines to identify targetable kinases. We report a comprehensive dataset consisting of 21,000 phosphosites on 4,896 phosphoproteins, including 217 kinases. We identify active Src-family kinases signaling as well as active cyclin-dependent kinases. We validate putative targets for therapy ex vivo and identify potential combination treatments, such as the inhibition of the INSR/IGF-1R axis to increase the sensitivity to dasatinib treatment. Ex vivo validation of selected drug combinations using patient-derived xenografts provides a proof-of-concept for phosphoproteomics-guided design of personalized treatments.
Collapse
Affiliation(s)
- Valentina Cordo’
- grid.487647.ePrincess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Mariska T. Meijer
- grid.487647.ePrincess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Rico Hagelaar
- grid.487647.ePrincess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Richard R. de Goeij-de Haas
- grid.12380.380000 0004 1754 9227OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Vera M. Poort
- grid.487647.ePrincess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Alex A. Henneman
- grid.12380.380000 0004 1754 9227OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Sander R. Piersma
- grid.12380.380000 0004 1754 9227OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Thang V. Pham
- grid.12380.380000 0004 1754 9227OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Koichi Oshima
- grid.239585.00000 0001 2285 2675Institute for Cancer Genetics, Columbia University Medical Center, New York, NY USA
| | - Adolfo A. Ferrando
- grid.239585.00000 0001 2285 2675Institute for Cancer Genetics, Columbia University Medical Center, New York, NY USA
| | | | - Connie R. Jimenez
- grid.12380.380000 0004 1754 9227OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Centers, VU University, Amsterdam, The Netherlands
| | - Jules P. P. Meijerink
- grid.487647.ePrincess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands ,Present Address: Acerta Pharma (member of the AstraZeneca group), Oss, The Netherlands
| |
Collapse
|
194
|
Owens AE, Iannotti MJ, Sanchez TW, Voss T, Kapoor A, Hall MD, Marugan JJ, Michael S, Southall N, Henderson MJ. High-Throughput Cellular Thermal Shift Assay Using Acoustic Transfer of Protein Lysates. ACS Chem Biol 2022; 17:322-330. [PMID: 35119255 PMCID: PMC10026039 DOI: 10.1021/acschembio.1c00760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cellular thermal shift assay (CETSA) is a valuable method to confirm target engagement within a complex cellular environment, by detecting changes in a protein's thermal stability upon ligand binding. The classical CETSA method measures changes in the thermal stability of endogenous proteins using immunoblotting, which is low-throughput and laborious. Reverse-phase protein arrays (RPPAs) have been demonstrated as a detection modality for CETSA; however, the reported procedure requires manual processing steps that limit throughput and preclude screening applications. We developed a high-throughput CETSA using an acoustic RPPA (HT-CETSA-aRPPA) protocol that is compatible with 96- and 384-well microplates from start-to-finish, using low speed centrifugation to remove thermally destabilized proteins. The utility of HT-CETSA-aRPPA for guiding structure-activity relationship studies was demonstrated for inhibitors of lactate dehydrogenase A. Additionally, a collection of kinase inhibitors was screened to identify compounds that engage MEK1, a clinically relevant kinase target.
Collapse
Affiliation(s)
- Ashley E. Owens
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, USA
| | - Michael J. Iannotti
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, USA
| | - Tino W. Sanchez
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, USA
| | - Ty Voss
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, USA
| | - Abhijeet Kapoor
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, USA
| | - Matthew D. Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, USA
| | - Juan J. Marugan
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, USA
| | - Sam Michael
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, USA
| | - Noel Southall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, USA
| | - Mark J. Henderson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, 20850, USA
- Corresponding Author: Mark Henderson;
| |
Collapse
|
195
|
Beetham H, Griffith BG, Murina O, Loftus AE, Parry DA, Temps C, Culley J, Muir M, Unciti-Broceta A, Sims AH, Byron A, Brunton VG. Loss of Integrin-Linked Kinase Sensitizes Breast Cancer to SRC Inhibitors. Cancer Res 2022; 82:632-647. [PMID: 34921014 PMCID: PMC9621571 DOI: 10.1158/0008-5472.can-21-0373] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 09/02/2021] [Accepted: 12/13/2021] [Indexed: 01/07/2023]
Abstract
SRC is a nonreceptor tyrosine kinase with key roles in breast cancer development and progression. Despite this, SRC tyrosine kinase inhibitors have so far failed to live up to their promise in clinical trials, with poor overall response rates. We aimed to identify possible synergistic gene-drug interactions to discover new rational combination therapies for SRC inhibitors. An unbiased genome-wide CRISPR-Cas9 knockout screen in a model of triple-negative breast cancer revealed that loss of integrin-linked kinase (ILK) and its binding partners α-Parvin and PINCH-1 sensitizes cells to bosutinib, a clinically approved SRC/ABL kinase inhibitor. Sensitivity to bosutinib did not correlate with ABL dependency; instead, bosutinib likely induces these effects by acting as a SRC tyrosine kinase inhibitor. Furthermore, in vitro and in vivo models showed that loss of ILK enhanced sensitivity to eCF506, a novel and highly selective inhibitor of SRC with a unique mode of action. Whole-genome RNA sequencing following bosutinib treatment in ILK knockout cells identified broad changes in the expression of genes regulating cell adhesion and cell-extracellular matrix. Increased sensitivity to SRC inhibition in ILK knockout cells was associated with defective adhesion, resulting in reduced cell number as well as increased G1 arrest and apoptosis. These findings support the potential of ILK loss as an exploitable therapeutic vulnerability in breast cancer, enhancing the effectiveness of clinical SRC inhibitors. SIGNIFICANCE A CRISPR-Cas9 screen reveals that loss of integrin-linked kinase synergizes with SRC inhibition, providing a new opportunity for enhancing the clinical effectiveness of SRC inhibitors in breast cancer.
Collapse
Affiliation(s)
- Henry Beetham
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, United Kingdom
| | - Billie G.C. Griffith
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, United Kingdom
| | - Olga Murina
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, United Kingdom
| | - Alexander E.P. Loftus
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, United Kingdom
| | - David A. Parry
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, United Kingdom
| | - Carolin Temps
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, United Kingdom
| | - Jayne Culley
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, United Kingdom
| | - Morwenna Muir
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, United Kingdom
| | - Asier Unciti-Broceta
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, United Kingdom
| | - Andrew H. Sims
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, United Kingdom
| | - Adam Byron
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, United Kingdom
| | - Valerie G. Brunton
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, United Kingdom
| |
Collapse
|
196
|
Sasaki K, Yamauchi T, Semba Y, Nogami J, Imanaga H, Terasaki T, Nakao F, Akahane K, Inukai T, Verhoeyen E, Akashi K, Maeda T. Genome-wide CRISPR-Cas9 screen identifies rationally designed combination therapies for CRLF2-rearranged Ph-like ALL. Blood 2022; 139:748-760. [PMID: 34587248 PMCID: PMC9632759 DOI: 10.1182/blood.2021012976] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/14/2021] [Indexed: 02/05/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) harboring the IgH-CRLF2 rearrangement (IgH-CRLF2-r) exhibits poor clinical outcomes and is the most common subtype of Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL). While multiple chemotherapeutic regimens, including ruxolitinib monotherapy and/or its combination with chemotherapy, are being tested, their efficacy is reportedly limited. To identify molecules/pathways relevant for IgH-CRLF2-r ALL pathogenesis, we performed genome-wide CRISPR-Cas9 dropout screens in the presence or absence of ruxolitinib using 2 IgH-CRLF2-r ALL lines that differ in RAS mutational status. To do so, we employed a baboon envelope pseudotyped lentiviral vector system, which enabled, for the first time, highly efficient transduction of human B cells. While single-guide RNAs (sgRNAs) targeting CRLF2, IL7RA, or JAK1/2 significantly affected cell fitness in both lines, those targeting STAT5A, STAT5B, or STAT3 did not, suggesting that STAT signaling is largely dispensable for IgH-CRLF2-r ALL cell survival. We show that regulators of RAS signaling are critical for cell fitness and ruxolitinib sensitivity and that CRKL depletion enhances ruxolitinib sensitivity in RAS wild-type (WT) cells. Gilteritinib, a pan-tyrosine kinase inhibitor that blocks CRKL phosphorylation, effectively killed RAS WT IgH-CRLF2-r ALL cells in vitro and in vivo, either alone or combined with ruxolitinib. We further show that combining gilteritinib with trametinib, a MEK1/2 inhibitor, is an effective means to target IgH-CRLF2-r ALL cells regardless of RAS mutational status. Our study delineates molecules/pathways relevant for CRLF2-r ALL pathogenesis and could suggest rationally designed combination therapies appropriate for disease subtypes.
Collapse
Affiliation(s)
- Kensuke Sasaki
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Takuji Yamauchi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Yuichiro Semba
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Jumpei Nogami
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Hiroshi Imanaga
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Tatsuya Terasaki
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Fumihiko Nakao
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Koshi Akahane
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Takeshi Inukai
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Els Verhoeyen
- CIRI-International Center for Infectiology Research, INSERM, Unité 1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
- Université Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France; and
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Takahiro Maeda
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
- Department of Pediatrics, School of Medicine, University of Yamanashi, Yamanashi, Japan
- CIRI-International Center for Infectiology Research, INSERM, Unité 1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, Unité Mixte de Recherche (UMR) 5308, Ecole Normale Supérieure de Lyon, Université Lyon, Lyon, France
- Université Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France; and
- Division of Precision Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
197
|
Targeting a splicing-mediated drug resistance mechanism in prostate cancer by inhibiting transcriptional regulation by PKCβ1. Oncogene 2022; 41:1536-1549. [PMID: 35087237 PMCID: PMC8913362 DOI: 10.1038/s41388-022-02179-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/14/2021] [Accepted: 01/05/2022] [Indexed: 11/08/2022]
Abstract
The androgen receptor (AR) is a central driver of aggressive prostate cancer. After initial treatment with androgen receptor signaling inhibitors (ARSi), reactivation of AR signaling leads to resistance. Alternative splicing of AR mRNA yields the AR-V7 splice variant, which is currently an undruggable mechanism of ARSi resistance: AR-V7 lacks a ligand binding domain, where hormones and anti-androgen antagonists act, but still activates AR signaling. We reveal PKCβ as a druggable regulator of transcription and splicing at the AR genomic locus. We identify a clinical PKCβ inhibitor in combination with an FDA-approved anti-androgen as an approach for repressing AR genomic locus expression, including expression of AR-V7, while antagonizing full-length AR. PKCβ inhibition reduces total AR gene expression, thus reducing AR-V7 protein levels and sensitizing prostate cancer cells to current anti-androgen therapies. We demonstrate that this combination may be a viable therapeutic strategy for AR-V7-positive prostate cancer.
Collapse
|
198
|
Ruan C, Ning W, Liu Z, Zhang X, Fang Z, Li Y, Dang Y, Xue Y, Ye M. Precipitate-Supported Thermal Proteome Profiling Coupled with Deep Learning for Comprehensive Screening of Drug Target Proteins. ACS Chem Biol 2022; 17:252-262. [PMID: 34989232 DOI: 10.1021/acschembio.1c00936] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Although thermal proteome profiling (TPP) acts as a popular modification-free approach for drug target deconvolution, some key problems are still limiting screening sensitivity. In the prevailing TPP workflow, only the soluble fractions are analyzed after thermal treatment, while the precipitate fractions that also contain abundant information of drug-induced stability shifts are discarded; the sigmoid melting curve fitting strategy used for data processing suffers from discriminations for a part of human proteome with multiple transitions. In this study, a precipitate-supported TPP (PSTPP) assay was presented for unbiased and comprehensive analysis of protein-drug interactions at the proteome level. In PSTPP, only these temperatures where significant precipitation is observed were applied to induce protein denaturation and the complementary information contained in both supernatant fractions and precipitate fractions was used to improve the screening specificity and sensitivity. In addition, a novel image recognition algorithm based on deep learning was developed to recognize the target proteins, which circumvented the problems that exist in the sigmoid curve fitting strategy. PSTPP assay was validated by identifying the known targets of methotrexate, raltitrexed, and SNS-032 with good performance. Using a promiscuous kinase inhibitor, staurosporine, we delineated 99 kinase targets with a specificity up to 83% in K562 cell lysates, which represented a significant improvement over the existing thermal shift methods. Furthermore, the PSTPP strategy was successfully applied to analyze the binding targets of rapamycin, identifying the well-known targets, FKBP1A, as well as revealing a few other potential targets.
Collapse
Affiliation(s)
- Chengfei Ruan
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wanshan Ning
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, Hubei 430074, China
| | - Zhen Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China
| | - Xiaolei Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China
| | - Zheng Fang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanan Li
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China
| | - Yongjun Dang
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yu Xue
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology (HUST), Wuhan, Hubei 430074, China
| | - Mingliang Ye
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China
| |
Collapse
|
199
|
Rácz A, Palkó R, Csányi D, Riedl Z, Bajusz D, Keserű GM. Consensus Virtual Screening Identified [1,2,4]Triazolo[1,5-b]isoquinolines As MELK Inhibitor Chemotypes. ChemMedChem 2022; 17:e202100569. [PMID: 34632716 PMCID: PMC9298037 DOI: 10.1002/cmdc.202100569] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/06/2021] [Indexed: 11/09/2022]
Abstract
Maternal Embryonic Leucine-zipper Kinase (MELK) is a current oncotarget involved in a diverse range of human cancers, with the usage of MELK inhibitors being explored clinically. Here, we aimed to discover new MELK inhibitor chemotypes from our in-house compound library with a consensus-based virtual screening workflow, employing three screening concepts. After careful retrospective validation, prospective screening and in vitro enzyme inhibition testing revealed a series of [1,2,4]triazolo[1,5-b]isoquinolines as a new structural class of MELK inhibitors, with the lead compound of the series exhibiting a sub-micromolar inhibitory activity. The structure-activity relationship of the series was explored by testing further analogs based on a structure-guided selection process. Importantly, the present work marks the first disclosure of the synthesis and bioactivity of this class of compounds.
Collapse
Affiliation(s)
- Anita Rácz
- Plasma Chemistry Research GroupResearch Centre for Natural SciencesMagyar tudósok krt. 21117BudapestHungary
- Medicinal Chemistry Research GroupResearch Centre for Natural SciencesMagyar tudósok krt. 21117BudapestHungary
| | - Roberta Palkó
- Medicinal Chemistry Research GroupResearch Centre for Natural SciencesMagyar tudósok krt. 21117BudapestHungary
- Present affiliation: Organocatalysis Research GroupResearch Centre for Natural SciencesMagyar tudósok krt. 21117BudapestHungary
| | - Dorottya Csányi
- Medicinal Chemistry Research GroupResearch Centre for Natural SciencesMagyar tudósok krt. 21117BudapestHungary
| | - Zsuzsanna Riedl
- Medicinal Chemistry Research GroupResearch Centre for Natural SciencesMagyar tudósok krt. 21117BudapestHungary
| | - Dávid Bajusz
- Medicinal Chemistry Research GroupResearch Centre for Natural SciencesMagyar tudósok krt. 21117BudapestHungary
| | - György M. Keserű
- Medicinal Chemistry Research GroupResearch Centre for Natural SciencesMagyar tudósok krt. 21117BudapestHungary
| |
Collapse
|
200
|
Douglass EF, Allaway RJ, Szalai B, Wang W, Tian T, Fernández-Torras A, Realubit R, Karan C, Zheng S, Pessia A, Tanoli Z, Jafari M, Wan F, Li S, Xiong Y, Duran-Frigola M, Bertoni M, Badia-i-Mompel P, Mateo L, Guitart-Pla O, Chung V, Tang J, Zeng J, Aloy P, Saez-Rodriguez J, Guinney J, Gerhard DS, Califano A. A community challenge for a pancancer drug mechanism of action inference from perturbational profile data. Cell Rep Med 2022; 3:100492. [PMID: 35106508 PMCID: PMC8784774 DOI: 10.1016/j.xcrm.2021.100492] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 08/08/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022]
Abstract
The Columbia Cancer Target Discovery and Development (CTD2) Center is developing PANACEA, a resource comprising dose-responses and RNA sequencing (RNA-seq) profiles of 25 cell lines perturbed with ∼400 clinical oncology drugs, to study a tumor-specific drug mechanism of action. Here, this resource serves as the basis for a DREAM Challenge assessing the accuracy and sensitivity of computational algorithms for de novo drug polypharmacology predictions. Dose-response and perturbational profiles for 32 kinase inhibitors are provided to 21 teams who are blind to the identity of the compounds. The teams are asked to predict high-affinity binding targets of each compound among ∼1,300 targets cataloged in DrugBank. The best performing methods leverage gene expression profile similarity analysis as well as deep-learning methodologies trained on individual datasets. This study lays the foundation for future integrative analyses of pharmacogenomic data, reconciliation of polypharmacology effects in different tumor contexts, and insights into network-based assessments of drug mechanisms of action. Drug-perturbed RNA sequencing data can be used to identify drug targets Technology-based drug-target definitions often subsume literature definitions Literature and screening datasets provide complementary information on drug mechanisms
Collapse
Affiliation(s)
- Eugene F. Douglass
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave., New York, NY 10032, USA
- Pharmaceutical and Biomedical Sciences, University of Georgia, 250 W. Green Street, Athens, GA 30602, USA
| | - Robert J. Allaway
- Computational Oncology Group, Sage Bionetworks, 2901 Third Ave., Ste 330, Seattle, WA 98121, USA
| | - Bence Szalai
- Semmelweis University, Faculty of Medicine, Department of Physiology, Budapest, Hungary
| | - Wenyu Wang
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tingzhong Tian
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing 100084, China
| | - Adrià Fernández-Torras
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Ron Realubit
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave., New York, NY 10032, USA
| | - Charles Karan
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave., New York, NY 10032, USA
| | - Shuyu Zheng
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Alberto Pessia
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ziaurrehman Tanoli
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mohieddin Jafari
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Fangping Wan
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing 100084, China
| | - Shuya Li
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing 100084, China
| | - Yuanpeng Xiong
- Department of Computer Science and Technology, Tsinghua University, Beijing 100084, China
| | - Miquel Duran-Frigola
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Martino Bertoni
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Pau Badia-i-Mompel
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Lídia Mateo
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Oriol Guitart-Pla
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| | - Verena Chung
- Computational Oncology Group, Sage Bionetworks, 2901 Third Ave., Ste 330, Seattle, WA 98121, USA
| | | | - Jing Tang
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jianyang Zeng
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing 100084, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing 100084, China
| | - Patrick Aloy
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Justin Guinney
- Computational Oncology Group, Sage Bionetworks, 2901 Third Ave., Ste 330, Seattle, WA 98121, USA
| | - Daniela S. Gerhard
- Office of Cancer Genomics, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Andrea Califano
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave., New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave., New York, NY 10032, USA
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY 10032, USA
- Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, 701 W 168th Street, New York, NY 10032, USA
- Department of Biomedical Informatics, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY 10032, USA
- Corresponding author
| |
Collapse
|