1
|
Loganathan T, Priya Doss C G. A comprehensive high-throughput screening approach for discovering inhibitors targeting the menin-MLL1 interaction. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 143:69-95. [PMID: 39843145 DOI: 10.1016/bs.apcsb.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
The prognosis for mixed-lineage leukemia (MLL), particularly in young children, remains a significant health concern due to the limited therapeutic options available. MLL refers to KMT2A chromosomal translocations that produce MLL fusion proteins. The protein menin, which is essential for the malignant potential of these MLL fusion proteins, offers novel targets for acute leukemia treatment. This study reports the identification of potential new inhibitors of MLL-mediated leukemia targeting menin through the screening of two distinct drug libraries and existing inhibitors. The 3D structure of the protein was retrieved from the Protein Data Bank (ID: 8IG0). The drug libraries, sourced from public repositories such as the 'Epigenetic Drug Library' and 'The FDA-anticancer Drug Library,' yielded top candidates like Tozaseritib and Panobinostat, which exhibited the highest binding energy scores in the Glide virtual screening module. Additionally, 31 known menin-MLL1 inhibitors were identified through PDB screening and subsequently docked with the menin protein. The top three inhibitors (M-525, M-808, and MI-89) were selected for further analysis. Five menin-ligand complexes were validated using molecular dynamics analysis and Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA) calculations to verify the stability and binding mechanisms.These findings provide insights into the molecular mechanisms of these drugs and lay the groundwork for future clinical development aimed at improving outcomes for acute myeloid leukemia (AML) patients.
Collapse
Affiliation(s)
- Tamizhini Loganathan
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - George Priya Doss C
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India.
| |
Collapse
|
2
|
Liang J, Qiao G, Zhang Y, Yuan Y, Liu Z, Jiang Y, Zhang Y, Deng Z, Yu L, Lin H, Ma L, Luo Y, Zhou Y, Hu H, Liu X, Zhang J. Ailanthone targets the KMT2A-MEN1 complex to suppress lung metastasis of osteosarcoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156258. [PMID: 39579612 DOI: 10.1016/j.phymed.2024.156258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 09/04/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Lung metastasis is the leading cause of death in patients with osteosarcoma (OS), and new drugs are urgently needed. Epigenetic reprogramming is a recently proposed hallmark of malignancy; therefore, targeting epigenetic enzymes might provide a novel therapeutic strategy for OS lung metastasis. We recently reported that ailanthone (AIL), a natural product isolated from the Chinese medicinal plant Ailanthus altissima, inhibits OS cell growth and induces substantial metabolic changes; however, its direct targets remain unclear. PURPOSE To identify the direct targets of AIL in OS and to explore the effects of AIL on OS lung metastasis in vivo. STUDY DESIGN Direct target proteins of AIL and downstream signaling pathways were identified in Saos-2 and U-2OS OS cells. The in vivo effects of AIL on OS lung metastasis were investigated using a mouse model. METHODS A novel surface plasmon resonance-high-performance liquid chromatography-mass spectrometry (SPR-HPLC-MS) assay was used to determine direct targets of AIL in OS. A cellular thermal shift assay, molecular docking analysis, enzyme activity assay, qRT-PCR, western blotting, chromatin immunoprecipitation assay, and reverse tests were performed to confirm the target and downstream pathway of AIL. A tumor xenograft model was used to verify the efficacy and mechanisms in vivo. RESULTS Histone-lysine N-methyltransferase 2A (KMT2A) together with its scaffold protein menin (MEN1) were identified as direct target proteins of AIL in OS. AIL induced the autophagic degradation of the KMT2A-MEN1 complex. Moreover, AIL inhibited intracellular H3K4 methyltransferase activity and epigenetically inhibited the transcription of genes in the serine biosynthetic pathway (SSP). Furthermore, AIL suppressed OS lung metastasis and downregulated KMT2A, MEN1, and SSP in mouse models. CONCLUSION This work showed that AIL targets the KMT2A-MEN1 complex and inhibits SSP to suppress OS lung metastasis. Notably, AIL exhibits new mechanisms of action, distinct from those of existing anti-OS drugs. On the basis of these findings, we proposed a novel strategy to treat OS by targeting epigenetic enzymes and cancer metabolism.
Collapse
Affiliation(s)
- Jinrong Liang
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China; Department of Oncology, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Guanglei Qiao
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Yawen Zhang
- Department of Oncology, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Ying Yuan
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Zimei Liu
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Yue Jiang
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Yan Zhang
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Zhoufeng Deng
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Liping Yu
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Hongjian Lin
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Lijun Ma
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Yanli Luo
- Department of Pathology, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yan Zhou
- Department of Oncology, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Haiyan Hu
- Shanghai Clinical Research Ward (SCRW), Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Xin Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Jianjun Zhang
- Department of Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China.
| |
Collapse
|
3
|
Jamil A, Qureshi Z, El-amir ZM, Kupakuwana-Suk G, Akram H, Ahmad M, Huselton E. Targeting menin for precision therapy in high-risk acute myeloid leukemia. Leuk Res Rep 2024; 23:100495. [PMID: 39811412 PMCID: PMC11732178 DOI: 10.1016/j.lrr.2024.100495] [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: 11/04/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Objective This mini-review provides an overview of the current evidence for Revumenib, a first-in-class menin inhibitor, in treating AML with KMT2A rearrangements or NPM1 mutations. This therapy represents a promising advancement by selectively disrupting leukemogenic pathways. Summary The clinical promise of Revumenib in genetically defined AML highlights its potential role in shaping the future treatment landscape. This mini-review underscores the need for ongoing trials to define optimal dosing, safety protocols, and combination therapies, with the ultimate goal of establishing Revumenib as a standard of care for high-risk AML subsets.
Collapse
Affiliation(s)
- Abdur Jamil
- Department of Medicine, Samaritan Medical Centre Watertown, NY, USA
| | - Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, Connecticut, USA
| | | | - Gillian Kupakuwana-Suk
- Leukemia Fellowship Program at The Ottawa Hospital Division of Hematology Department of Medicine University of Ottawa, Ottawa, Canada
| | - Hamzah Akram
- Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Mohsin Ahmad
- CHRISTUS Southeast Texas - St. Elizabeth Hospital, Beaumont, Texas, USA
| | - Eric Huselton
- University of Rochester, Department of Hematology/Oncology, Rochester, NY, USA
| |
Collapse
|
4
|
Nadiminti KVG, Sahasrabudhe KD, Liu H. Menin inhibitors for the treatment of acute myeloid leukemia: challenges and opportunities ahead. J Hematol Oncol 2024; 17:113. [PMID: 39558390 PMCID: PMC11575055 DOI: 10.1186/s13045-024-01632-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024] Open
Abstract
The AML treatment landscape has significantly changed in recent years with the approval of targeted therapies in the front-line and relapsed/refractory settings, including inhibitors of FLT3 and IDH1/2 mutations. More importantly, approval of the combination of the BCl-2 inhibitor, venetoclax, and hypomethylating agents or low dose cytarabine provided unprecedented breakthrough for the frontline treatment of older, unfit AML patients. Even with all this exciting progress, more targeted therapies for AML treatment are needed. Recent development of menin inhibitors targeting AML with KMT2A rearrangements or NPM1 mutations could represent a promising new horizon of treatment for patients within these subsets of AML. Our current review will focus on a summary and updates of recent developments of menin inhibitors in the treatment of AML, on the challenges ahead arising from drug resistance, as well as on the opportunities of novel combinations with menin inhibitors.
Collapse
Affiliation(s)
- Kalyan V G Nadiminti
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI, 53705-2281, USA.
| | - Kieran D Sahasrabudhe
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI, 53705-2281, USA
| | - Hongtao Liu
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI, 53705-2281, USA
| |
Collapse
|
5
|
Al-nakhle HH, Yagoub HS, Alrehaili RY, Shaqroon OA, Khan MK, Alsharif GS. Elucidating the role of MLL1 nsSNPs: Structural and functional alterations and their contribution to leukemia development. PLoS One 2024; 19:e0304986. [PMID: 39405275 PMCID: PMC11478856 DOI: 10.1371/journal.pone.0304986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/21/2024] [Indexed: 10/19/2024] Open
Abstract
(1) BACKGROUND The Mixed lineage leukemia 1 (MLL1) gene, located on chromosome 11q23, plays a pivotal role in histone lysine-specific methylation and is consistently associated with various types of leukemia. Non-synonymous Single Nucleotide Polymorphisms (nsSNPs) have been tied to numerous diseases, including cancers, and have become valuable cancer biomarkers. There's a notable gap in studies probing the influence of SNPs on MLL1 protein structure, function, and subsequent modifications. (2) METHODS We utilized an array of bioinformatics tools, including PredictSNP, InterPro, ConSurf, I-Mutant2.0, MUpro, Musitedeep, Project HOPE, RegulomeDB, Mutpred2, and both CScape and CScape Somatic, to meticulously analyze the consequences of nsSNPs in the MLL1 gene. (3) RESULTS Out of 2,097 nsSNPs analyzed, 62 were determined to be significantly pathogenic by the PredictSNP tool, with ten crucial MLL1 functional domains identified using InterPro. Additionally, 50 of these nsSNPs had high conservation scores, hinting at potential effects on protein structure and function, while 32 were found to undermine MLL1 protein stability. Notably, four nsSNPs were deemed oncogenic, with two identified as cancer drivers. The nsSNP, D2724G, between the MLL1 protein's FY-rich domains, could disrupt proteolytic cleavage, altering gene expression patterns and potentially promoting cancer. (4) CONCLUSIONS Our research provides a comprehensive assessment of nsSNPs' impact in the MLL1 protein structure and function and consequently on leukemia development, suggesting potential avenues for personalized treatment, early detection, improved prognosis, and a deeper understanding of hematological malignancy genesis.
Collapse
Affiliation(s)
- Hakeemah H. Al-nakhle
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Monawarah, Saudi Arabia
| | - Hind S. Yagoub
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Monawarah, Saudi Arabia
- Faculty of Medical Laboratory Sciences, Omdurman Islamic University, Omdurman, Sudan
| | - Rahaf Y. Alrehaili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Monawarah, Saudi Arabia
| | - Ola A. Shaqroon
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Monawarah, Saudi Arabia
| | - Minna K. Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Monawarah, Saudi Arabia
| | - Ghaidaa S. Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Al-Madinah Al-Monawarah, Saudi Arabia
| |
Collapse
|
6
|
Ray J, Clegg B, Grembecka J, Cierpicki T. Drug-resistant menin variants retain high binding affinity and interactions with MLL1. J Biol Chem 2024; 300:107777. [PMID: 39276940 PMCID: PMC11490872 DOI: 10.1016/j.jbc.2024.107777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/15/2024] [Accepted: 09/05/2024] [Indexed: 09/17/2024] Open
Abstract
Menin is an essential oncogenic cofactor of MLL1 fusion proteins in acute leukemias and inhibitors of the menin-MLL1 interaction are under evaluation in clinical trials. Recent studies found emerging resistance to menin inhibitor treatment in patients with leukemia as a result of somatic mutations in menin. To understand how patient mutations in menin affect the interaction with MLL1, we performed systematic characterization of the binding affinity of these menin mutants (T349M, M327I, G331R and G331D) and the N-terminal fragment of MLL1. We also determined the crystal structures of menin patient mutants and their complexes with MLL1-derived peptides. We found that drug-resistant mutations in menin occur at a site adjacent to the MLL1 binding site, but they do not affect MLL1 binding to menin. On the contrary, our structural analysis shows that all these point mutations in menin generate steric clash with menin inhibitors. We also found that mutation G331D results in a very slow dissociation of MLL1 from menin and this mutant might be particularly difficult to inhibit with small molecule drugs. This work provides structural information to support the development of a new generation of small molecule inhibitors that overcome resistance caused by menin mutations.
Collapse
Affiliation(s)
- Joshua Ray
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Bradley Clegg
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA; Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA; Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan, USA.
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA; Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan, USA; Department of Biophysics, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
7
|
Kwon MC, Thuring JW, Querolle O, Dai X, Verhulst T, Pande V, Marien A, Goffin D, Wenge DV, Yue H, Cutler JA, Jin C, Perner F, Hogeling SM, Shaffer PL, Jacobs F, Vinken P, Cai W, Keersmaekers V, Eyassu F, Bhogal B, Verstraeten K, El Ashkar S, Perry JA, Jayaguru P, Barreyro L, Kuchnio A, Darville N, Krosky D, Urbanietz G, Verbist B, Edwards JP, Cowley GS, Kirkpatrick R, Steele R, Ferrante L, Guttke C, Daskalakis N, Pietsch EC, Wilson DM, Attar R, Elsayed Y, Fischer ES, Schuringa JJ, Armstrong SA, Packman K, Philippar U. Preclinical efficacy of the potent, selective menin-KMT2A inhibitor JNJ-75276617 (bleximenib) in KMT2A- and NPM1-altered leukemias. Blood 2024; 144:1206-1220. [PMID: 38905635 PMCID: PMC11419783 DOI: 10.1182/blood.2023022480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 06/23/2024] Open
Abstract
ABSTRACT The interaction between menin and histone-lysine N-methyltransferase 2A (KMT2A) is a critical dependency for KMT2A- or nucleophosmin 1 (NPM1)-altered leukemias and an emerging opportunity for therapeutic development. JNJ-75276617 (bleximenib) is a novel, orally bioavailable, potent, and selective protein-protein interaction inhibitor of the binding between menin and KMT2A. In KMT2A-rearranged (KMT2A-r) and NPM1-mutant (NPM1c) acute myeloid leukemia (AML) cells, JNJ-75276617 inhibited the association of the menin-KMT2A complex with chromatin at target gene promoters, resulting in reduced expression of several menin-KMT2A target genes, including MEIS1 and FLT3. JNJ-75276617 displayed potent antiproliferative activity across several AML and acute lymphoblastic leukemia (ALL) cell lines and patient samples harboring KMT2A or NPM1 alterations in vitro. In xenograft models of AML and ALL, JNJ-75276617 reduced leukemic burden and provided a significant dose-dependent survival benefit accompanied by expression changes of menin-KMT2A target genes. JNJ-75276617 demonstrated synergistic effects with gilteritinib in vitro in AML cells harboring KMT2A-r. JNJ-75276617 further exhibited synergistic effects with venetoclax and azacitidine in AML cells bearing KMT2A-r in vitro, and significantly increased survival in mice. Interestingly, JNJ-75276617 showed potent antiproliferative activity in cell lines engineered with recently discovered mutations (MEN1M327I or MEN1T349M) that developed in patients refractory to the menin-KMT2A inhibitor revumenib. A cocrystal structure of menin in complex with JNJ-75276617 indicates a unique binding mode distinct from other menin-KMT2A inhibitors, including revumenib. JNJ-75276617 is being clinically investigated for acute leukemias harboring KMT2A or NPM1 alterations, as a monotherapy for relapsed/refractory acute leukemia (NCT04811560), or in combination with AML-directed therapies (NCT05453903).
Collapse
MESH Headings
- Nucleophosmin
- Humans
- Animals
- Mice
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/antagonists & inhibitors
- Histone-Lysine N-Methyltransferase/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Nuclear Proteins/antagonists & inhibitors
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Xenograft Model Antitumor Assays
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/antagonists & inhibitors
- Proto-Oncogene Proteins/metabolism
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Mice, SCID
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
Collapse
Affiliation(s)
| | | | - Olivier Querolle
- Discovery Product Development and Supply, Janssen R&D, Beerse, Belgium
| | - Xuedong Dai
- Discovery Product Development and Supply, Janssen R&D, Shanghai, China
| | | | - Vineet Pande
- Discovery Product Development and Supply, Janssen R&D, Beerse, Belgium
| | - Ann Marien
- Discovery Oncology, Janssen R&D, Beerse, Belgium
| | - Dries Goffin
- Discovery Oncology, Janssen R&D, Beerse, Belgium
| | - Daniela V. Wenge
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children's Hospital, and Harvard Medical School, Boston, MA
| | - Hong Yue
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Jevon A. Cutler
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children's Hospital, and Harvard Medical School, Boston, MA
| | - Cyrus Jin
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Florian Perner
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children's Hospital, and Harvard Medical School, Boston, MA
| | - Shanna M. Hogeling
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Paul L. Shaffer
- Discovery Product Development and Supply, Janssen R&D, Spring House, PA
| | - Frank Jacobs
- Discovery Product Development and Supply, Janssen R&D, Beerse, Belgium
| | - Petra Vinken
- Discovery Product Development and Supply, Janssen R&D, Beerse, Belgium
| | - Wei Cai
- Discovery Product Development and Supply, Janssen R&D, Shanghai, China
| | | | | | - Balpreet Bhogal
- Discovery Product Development and Supply, Janssen R&D, Spring House, PA
| | | | | | - Jennifer A. Perry
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children's Hospital, and Harvard Medical School, Boston, MA
| | | | | | - Anna Kuchnio
- Discovery Oncology, Janssen R&D, Beerse, Belgium
| | - Nicolas Darville
- Discovery Product Development and Supply, Janssen R&D, Beerse, Belgium
| | - Daniel Krosky
- Discovery Product Development and Supply, Janssen R&D, Spring House, PA
| | - Gregor Urbanietz
- Discovery Product Development and Supply, Janssen R&D, Beerse, Belgium
| | | | - James P. Edwards
- Discovery Product Development and Supply, Janssen R&D, Spring House, PA
| | - Glenn S. Cowley
- Discovery Product Development and Supply, Janssen R&D, Spring House, PA
| | | | - Ruth Steele
- Discovery Product Development and Supply, Janssen R&D, Spring House, PA
| | | | | | | | | | - David M. Wilson
- Discovery Product Development and Supply, Janssen R&D, Beerse, Belgium
| | - Ricardo Attar
- Translational Research, Janssen R&D, Spring House, PA
| | | | - Eric S. Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Jan Jacob Schuringa
- Department of Experimental Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Scott A. Armstrong
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children's Hospital, and Harvard Medical School, Boston, MA
| | | | | |
Collapse
|
8
|
Hintzen JCJ, Mecinović J. Peptide-based inhibitors of epigenetic proteins. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 212:25-65. [PMID: 40122647 DOI: 10.1016/bs.pmbts.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Epigenetic drug discovery has become an integral part of medicinal chemistry in the past two decades. Targeting epigenetic proteins-enzymes that modify histone proteins and DNA (writers and erasers) and proteins that recognize such modifications (readers)-has been firmly established as a medicinal strategy for treatment of many human diseases, including cancer and neurological disorders. In this chapter, we systematically describe peptide-based inhibitors of structurally and functionally diverse classes of epigenetic proteins. We show that epigenetic writers, such as DNA methyltransferases, histone methyltransferases and histone acetyltransferases, can be efficiently inhibited by peptides possessing nonproteinogenic amino acids. Moreover, the activity of epigenetic erasers, including TET enzymes, histone demethylases, and histone deacetylases, can be selectively modulated by diverse linear and cyclic peptides. Furthermore, we discuss chromatin-binding epigenetic reader proteins that can be inhibited by histone-mimicking peptides. Overall, this chapter highlights that peptides provide an important molecular platform for epigenetic drug discovery programmes in academia and industry.
Collapse
Affiliation(s)
- Jordi C J Hintzen
- Department of Chemistry and Molecular Biology, Wallenberg Center for Molecular and Translational Medicine, University of Gothenburg, Göteborg, Sweden.
| | - Jasmin Mecinović
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
9
|
Zhuang K, Leng L, Su X, Wang S, Su Y, Chen Y, Yuan Z, Zi L, Li J, Xie W, Yan S, Xia Y, Wang H, Li H, Chen Z, Yuan T, Zhang J. Menin Deficiency Induces Autism-Like Behaviors by Regulating Foxg1 Transcription and Participates in Foxg1-Related Encephalopathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307953. [PMID: 38582517 PMCID: PMC11200012 DOI: 10.1002/advs.202307953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/18/2024] [Indexed: 04/08/2024]
Abstract
FOXG1 syndrome is a developmental encephalopathy caused by FOXG1 (Forkhead box G1) mutations, resulting in high phenotypic variability. However, the upstream transcriptional regulation of Foxg1 expression remains unclear. This report demonstrates that both deficiency and overexpression of Men1 (protein: menin, a pathogenic gene of MEN1 syndrome known as multiple endocrine neoplasia type 1) lead to autism-like behaviors, such as social defects, increased repetitive behaviors, and cognitive impairments. Multifaceted transcriptome analyses revealed that Foxg1 signaling is predominantly altered in Men1 deficiency mice, through its regulation of the Alpha Thalassemia/Mental Retardation Syndrome X-Linked (Atrx) factor. Atrx recruits menin to bind to the transcriptional start region of Foxg1 and mediates the regulation of Foxg1 expression by H3K4me3 (Trimethylation of histone H3 lysine 4) modification. The deficits observed in menin deficient mice are rescued by the over-expression of Foxg1, leading to normalized spine growth and restoration of hippocampal synaptic plasticity. These findings suggest that menin may have a putative role in the maintenance of Foxg1 expression, highlighting menin signaling as a potential therapeutic target for Foxg1-related encephalopathy.
Collapse
Affiliation(s)
- Kai Zhuang
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Lige Leng
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Xiao Su
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Shuzhong Wang
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Yuemin Su
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Yanbing Chen
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Ziqi Yuan
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Liu Zi
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Jieyin Li
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Wenting Xie
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Sihan Yan
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Yujun Xia
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Han Wang
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Huifang Li
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Zhenyi Chen
- Department of AnesthesiologyFirst Affiliated HospitalCollege of MedicineXiamen UniversityXiamenFujian361105China
| | - Tifei Yuan
- Shanghai Mental Health CenterShanghai Jiaotong University School of MedicineShanghai200030China
| | - Jie Zhang
- Institute of NeuroscienceCollege of MedicineXiamen UniversityXiamenFujian361105China
- Department of AnesthesiologyFirst Affiliated HospitalCollege of MedicineXiamen UniversityXiamenFujian361105China
- The Key Laboratory of Neural and Vascular BiologyMinistry of EducationCollege of Basic MedicineHebei Medical UniversityShijiazhuang050017China
| |
Collapse
|
10
|
Candoni A, Coppola G. A 2024 Update on Menin Inhibitors. A New Class of Target Agents against KMT2A-Rearranged and NPM1-Mutated Acute Myeloid Leukemia. Hematol Rep 2024; 16:244-254. [PMID: 38651453 PMCID: PMC11036224 DOI: 10.3390/hematolrep16020024] [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: 01/22/2024] [Revised: 04/01/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
Menin inhibitors are new and promising agents currently in clinical development that target the HOX/MEIS1 transcriptional program which is critical for leukemogenesis in histone-lysine N-methyltransferase 2A-rearranged (KMT2Ar) and in NPM1-mutated (NPM1mut) acute leukemias. The mechanism of action of this new class of agents is based on the disruption of the menin-KMT2A complex (consisting of chromatin remodeling proteins), leading to the differentiation and apoptosis of AML cells expressing KMT2A or with mutated NPM1. To date, this new class of drugs has been tested in phase I and II clinical trials, both alone and in combination with synergistic drugs showing promising results in terms of response rates and safety in heavily pre-treated acute leukemia patients. In this brief review, we summarize the key findings on menin inhibitors, focusing on the mechanism of action and preliminary clinical data on the treatment of acute myeloid leukemia with this promising new class of agents, particularly revumenib and ziftomenib.
Collapse
Affiliation(s)
- Anna Candoni
- Section of Haematology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41123 Modena, Italy;
| | | |
Collapse
|
11
|
Gao Y, Vakoc CR. Therapeutic index of targeting select chromatin complexes in human cancer patients. Curr Opin Genet Dev 2024; 85:102162. [PMID: 38401489 PMCID: PMC11072572 DOI: 10.1016/j.gde.2024.102162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/21/2024] [Accepted: 01/29/2024] [Indexed: 02/26/2024]
Abstract
Aberrant chromatin regulation can promote the initiation and progression of human cancer. An improved understanding of such mechanisms has resulted in the identification of cancers with an enhanced dependency on specific chromatin regulatory proteins relative to nonmalignant cell types. Hence, targeting of such complexes with small molecules has significant therapeutic potential in oncology. In recent years, several drugs have been developed and evaluated in human cancer patients, which can influence tumor biology by reprogramming of chromatin structure. In this review, we summarize several of the known mechanisms that endow cancer cells with a powerful dependency on chromatin regulation that exceeds the requirements for normal tissue homeostasis. We also summarize the remarkable small-molecule inhibitors that exploit chromatin regulator dependencies with a clear therapeutic benefit in human cancer patients.
Collapse
Affiliation(s)
- Yuan Gao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA. https://twitter.com/@yuangao_yg
| | | |
Collapse
|
12
|
Maher SP, Bakowski MA, Vantaux A, Flannery EL, Andolina C, Gupta M, Antonova-Koch Y, Argomaniz M, Cabrera-Mora M, Campo B, Chao AT, Chatterjee AK, Cheng WT, Chuenchob E, Cooper CA, Cottier K, Galinski MR, Harupa-Chung A, Ji H, Joseph SB, Lenz T, Lonardi S, Matheson J, Mikolajczak SA, Moeller T, Orban A, Padín-Irizarry V, Pan K, Péneau J, Prudhomme J, Roesch C, Ruberto AA, Sabnis SS, Saney CL, Sattabongkot J, Sereshki S, Suriyakan S, Ubalee R, Wang Y, Wasisakun P, Yin J, Popovici J, McNamara CW, Joyner CJ, Nosten F, Witkowski B, Le Roch KG, Kyle DE. A Drug Repurposing Approach Reveals Targetable Epigenetic Pathways in Plasmodium vivax Hypnozoites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.31.526483. [PMID: 36778461 PMCID: PMC9915689 DOI: 10.1101/2023.01.31.526483] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Radical cure of Plasmodium vivax malaria must include elimination of quiescent 'hypnozoite' forms in the liver; however, the only FDA-approved treatments are contraindicated in many vulnerable populations. To identify new drugs and drug targets for hypnozoites, we screened the Repurposing, Focused Rescue, and Accelerated Medchem (ReFRAME) library and a collection of epigenetic inhibitors against P. vivax liver stages. From both libraries, we identified inhibitors targeting epigenetics pathways as selectively active against P. vivax and P. cynomolgi hypnozoites. These include DNA methyltransferase (DNMT) inhibitors as well as several inhibitors targeting histone post-translational modifications. Immunofluorescence staining of Plasmodium liver forms showed strong nuclear 5-methylcystosine signal, indicating liver stage parasite DNA is methylated. Using bisulfite sequencing, we mapped genomic DNA methylation in sporozoites, revealing DNA methylation signals in most coding genes. We also demonstrated that methylation level in proximal promoter regions as well as in the first exon of the genes may affect, at least partially, gene expression in P. vivax. The importance of selective inhibitors targeting epigenetic features on hypnozoites was validated using MMV019721, an acetyl-CoA synthetase inhibitor that affects histone acetylation and was previously reported as active against P. falciparum blood stages. In summary, our data indicate that several epigenetic mechanisms are likely modulating hypnozoite formation or persistence and provide an avenue for the discovery and development of improved radical cure antimalarials.
Collapse
Affiliation(s)
- S. P. Maher
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
| | - M. A. Bakowski
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - A. Vantaux
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - E. L. Flannery
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | - C. Andolina
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit; Mae Sot, Tak, 63110, Thailand
| | - M. Gupta
- Department of Molecular, Cell, and Systems Biology, University of California; Riverside, CA, 92521, USA
| | - Y. Antonova-Koch
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - M. Argomaniz
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - M. Cabrera-Mora
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Emory National Primate Research Center, Emory University; Atlanta, GA, 30329, USA
| | - B. Campo
- Medicines for Malaria Venture (MMV); Geneva, 1215, Switzerland
| | - A. T. Chao
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | - A. K. Chatterjee
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - W. T. Cheng
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - E. Chuenchob
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | - C. A. Cooper
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
| | | | - M. R. Galinski
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Emory National Primate Research Center, Emory University; Atlanta, GA, 30329, USA
- Division of Infectious Diseases, Department of Medicine, Emory University; Atlanta, GA, 30329, USA
| | - A. Harupa-Chung
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | - H. Ji
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - S. B. Joseph
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - T. Lenz
- Department of Molecular, Cell, and Systems Biology, University of California; Riverside, CA, 92521, USA
| | - S. Lonardi
- Department of Computer Science and Engineering, University of California; Riverside, CA, 92521, USA
| | - J. Matheson
- Department of Microbiology and Immunology, University of Otago; Dunedin, 9016, New Zealand
| | - S. A. Mikolajczak
- Novartis Institute for Tropical Diseases, Novartis Institutes for Biomedical Research; Emeryville, CA, 94608, USA
| | | | - A. Orban
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - V. Padín-Irizarry
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
- School of Sciences, Clayton State University; Morrow, GA, 30260, USA
| | - K. Pan
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - J. Péneau
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - J. Prudhomme
- Department of Molecular, Cell, and Systems Biology, University of California; Riverside, CA, 92521, USA
| | - C. Roesch
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - A. A. Ruberto
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
| | - S. S. Sabnis
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - C. L. Saney
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
| | - J. Sattabongkot
- Mahidol Vivax Research Unit, Mahidol University; Bangkok, 10400, Thailand
| | - S. Sereshki
- Department of Computer Science and Engineering, University of California; Riverside, CA, 92521, USA
| | - S. Suriyakan
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit; Mae Sot, Tak, 63110, Thailand
| | - R. Ubalee
- Department of Entomology, Armed Forces Research Institute of Medical Sciences (AFRIMS); Bangkok, 10400, Thailand
| | - Y. Wang
- Department of Chemistry, University of California; Riverside, CA, 92521
- Environmental Toxicology Graduate Program, University of California; Riverside, CA, 92521, USA
| | - P. Wasisakun
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit; Mae Sot, Tak, 63110, Thailand
| | - J. Yin
- Environmental Toxicology Graduate Program, University of California; Riverside, CA, 92521, USA
| | - J. Popovici
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - C. W. McNamara
- Calibr, a division of The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - C. J. Joyner
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA, 30602, USA
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Emory National Primate Research Center, Emory University; Atlanta, GA, 30329, USA
| | - F. Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit; Mae Sot, Tak, 63110, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford; Oxford, OX3 7LG, UK
| | - B. Witkowski
- Malaria Molecular Epidemiology Unit, Institute Pasteur of Cambodia; Phnom Penh, 120 210, Cambodia
| | - K. G. Le Roch
- Department of Molecular, Cell, and Systems Biology, University of California; Riverside, CA, 92521, USA
| | - D. E. Kyle
- Center for Tropical & Emerging Global Disease, University of Georgia; Athens, GA, 30602, USA
| |
Collapse
|
13
|
Yoon H, Rutter JC, Li YD, Ebert BL. Induced protein degradation for therapeutics: past, present, and future. J Clin Invest 2024; 134:e175265. [PMID: 38165043 PMCID: PMC10760958 DOI: 10.1172/jci175265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Abstract
The concept of induced protein degradation by small molecules has emerged as a promising therapeutic strategy that is particularly effective in targeting proteins previously considered "undruggable." Thalidomide analogs, employed in the treatment of multiple myeloma, stand as prime examples. These compounds serve as molecular glues, redirecting the CRBN E3 ubiquitin ligase to degrade myeloma-dependency factors, IKZF1 and IKZF3. The clinical success of thalidomide analogs demonstrates the therapeutic potential of induced protein degradation. Beyond molecular glue degraders, several additional modalities to trigger protein degradation have been developed and are currently under clinical evaluation. These include heterobifunctional degraders, polymerization-induced degradation, ligand-dependent degradation of nuclear hormone receptors, disruption of protein interactions, and various other strategies. In this Review, we will provide a concise overview of various degradation modalities, their clinical applications, and potential future directions in the field of protein degradation.
Collapse
Affiliation(s)
- Hojong Yoon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Justine C. Rutter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yen-Der Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Benjamin L. Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Aryal S, Lu R. HOXA9 Regulome and Pharmacological Interventions in Leukemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:405-430. [PMID: 39017854 DOI: 10.1007/978-3-031-62731-6_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
HOXA9, an important transcription factor (TF) in hematopoiesis, is aberrantly expressed in numerous cases of both acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) and is a strong indicator of poor prognosis in patients. HOXA9 is a proto-oncogene which is both sufficient and necessary for leukemia transformation. HOXA9 expression in leukemia correlates with patient survival outcomes and response to therapy. Chromosomal transformations (such as NUP98-HOXA9), mutations, epigenetic dysregulation (e.g., MLL- MENIN -LEDGF complex or DOT1L/KMT4), transcription factors (such as USF1/USF2), and noncoding RNA (such as HOTTIP and HOTAIR) regulate HOXA9 mRNA and protein during leukemia. HOXA9 regulates survival, self-renewal, and progenitor cell cycle through several of its downstream target TFs including LMO2, antiapoptotic BCL2, SOX4, and receptor tyrosine kinase FLT3 and STAT5. This dynamic and multilayered HOXA9 regulome provides new therapeutic opportunities, including inhibitors targeting DOT1L/KMT4, MENIN, NPM1, and ENL proteins. Recent findings also suggest that HOXA9 maintains leukemia by actively repressing myeloid differentiation genes. This chapter summarizes the recent advances understanding biochemical mechanisms underlying HOXA9-mediated leukemogenesis, the clinical significance of its abnormal expression, and pharmacological approaches to treat HOXA9-driven leukemia.
Collapse
Affiliation(s)
- Sajesan Aryal
- Department of Medicine, Division of Hematology/Oncology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Rui Lu
- Department of Medicine, Division of Hematology/Oncology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA.
| |
Collapse
|
15
|
Wolffhardt TM, Ketzer F, Telese S, Wirth T, Ushmorov A. Dependency of B-Cell Acute Lymphoblastic Leukemia and Multiple Myeloma Cell Lines on MEN1 Extends beyond MEN1-KMT2A Interaction. Int J Mol Sci 2023; 24:16472. [PMID: 38003662 PMCID: PMC10670986 DOI: 10.3390/ijms242216472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Menin/MEN1 is a scaffold protein that participates in proliferation, regulation of gene transcription, DNA damage repair, and signal transduction. In hematological malignancies harboring the KMT2A/MLL1 (MLLr) chromosomal rearrangements, the interaction of the oncogenic fusion protein MLLr with MEN1 has been shown to be essential. MEN1 binders inhibiting the MEN1 and KMT2A interaction have been shown to be effective against MLLr AML and B-ALL in experimental models and clinical studies. We hypothesized that in addition to the MEN1-KMT2A interaction, alternative mechanisms might be instrumental in the MEN1 dependency of leukemia. We first mined and analyzed data from publicly available gene expression databases, finding that the dependency of B-ALL cell lines on MEN1 did not correlate with the presence of MLLr. Using shRNA-mediated knockdown, we found that all tested B-ALL cell lines were sensitive to MEN1 depletion, independent of the underlying driver mutations. Most multiple myeloma cell lines that did not harbor MLLr were also sensitive to the genetic depletion of MEN1. We conclude that the oncogenic role of MEN1 is not limited to the interaction with KMT2A. Our results suggest that targeted degradation of MEN1 or the development of binders that induce global changes in the MEN1 protein structure may be more efficient than the inhibition of individual MEN1 protein interactions.
Collapse
Affiliation(s)
- Tatjana Magdalena Wolffhardt
- Institute of Physiological Chemistry, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany; (T.M.W.); (S.T.)
| | - Franz Ketzer
- Center for Molecular and Cellular Oncology, Yale School of Medicine, New Haven, CT 06510, USA;
| | - Stefano Telese
- Institute of Physiological Chemistry, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany; (T.M.W.); (S.T.)
| | - Thomas Wirth
- Institute of Physiological Chemistry, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany; (T.M.W.); (S.T.)
| | - Alexey Ushmorov
- Institute of Physiological Chemistry, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany; (T.M.W.); (S.T.)
| |
Collapse
|
16
|
Wu D, Li Y, Zheng L, Xiao H, Ouyang L, Wang G, Sun Q. Small molecules targeting protein-protein interactions for cancer therapy. Acta Pharm Sin B 2023; 13:4060-4088. [PMID: 37799384 PMCID: PMC10547922 DOI: 10.1016/j.apsb.2023.05.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/28/2023] [Accepted: 05/22/2023] [Indexed: 10/07/2023] Open
Abstract
Protein-protein interactions (PPIs) are fundamental to many biological processes that play an important role in the occurrence and development of a variety of diseases. Targeting the interaction between tumour-related proteins with emerging small molecule drugs has become an attractive approach for treatment of human diseases, especially tumours. Encouragingly, selective PPI-based therapeutic agents have been rapidly advancing over the past decade, providing promising perspectives for novel therapies for patients with cancer. In this review we comprehensively clarify the discovery and development of small molecule modulators of PPIs from multiple aspects, focusing on PPIs in disease, drug design and discovery strategies, structure-activity relationships, inherent dilemmas, and future directions.
Collapse
Affiliation(s)
- Defa Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Yang Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Lang Zheng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Huan Xiao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Qiu Sun
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, Innovation Center of Nursing Research, West China Hospital, Sichuan University /West China School of Nursing, Sichuan University, Chengdu 610041, China
- West China Medical Publishers, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
17
|
Chin L, Wong CYG, Gill H. Targeting and Monitoring Acute Myeloid Leukaemia with Nucleophosmin-1 ( NPM1) Mutation. Int J Mol Sci 2023; 24:3161. [PMID: 36834572 PMCID: PMC9958584 DOI: 10.3390/ijms24043161] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023] Open
Abstract
Mutations in NPM1, also known as nucleophosmin-1, B23, NO38, or numatrin, are seen in approximately one-third of patients with acute myeloid leukaemia (AML). A plethora of treatment strategies have been studied to determine the best possible approach to curing NPM1-mutated AML. Here, we introduce the structure and function of NPM1 and describe the application of minimal residual disease (MRD) monitoring using molecular methods by means of quantitative polymerase chain reaction (qPCR), droplet digital PCR (ddPCR), next-generation sequencing (NGS), and cytometry by time of flight (CyTOF) to target NPM1-mutated AML. Current drugs, now regarded as the standard of care for AML, as well as potential drugs still under development, will also be explored. This review will focus on the role of targeting aberrant NPM1 pathways such as BCL-2 and SYK; as well as epigenetic regulators (RNA polymerase), DNA intercalators (topoisomerase II), menin inhibitors, and hypomethylating agents. Aside from medication, the effects of stress on AML presentation have been reported, and some possible mechanisms outlined. Moreover, targeted strategies will be briefly discussed, not only for the prevention of abnormal trafficking and localisation of cytoplasmic NPM1 but also for the elimination of mutant NPM1 proteins. Lastly, the advancement of immunotherapy such as targeting CD33, CD123, and PD-1 will be mentioned.
Collapse
Affiliation(s)
| | | | - Harinder Gill
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
18
|
Dissecting MENIN in bivalent gene regulation. Nat Cell Biol 2023; 25:209-210. [PMID: 36635502 DOI: 10.1038/s41556-022-01063-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
19
|
Discovery of cysteine-targeting covalent histone methyltransferase inhibitors. Eur J Med Chem 2023; 246:115028. [PMID: 36528996 DOI: 10.1016/j.ejmech.2022.115028] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/02/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022]
Abstract
Post-translational methylation of histone lysine or arginine residues by histone methyltransferases (HMTs) plays crucial roles in gene regulation and diverse physiological processes and is implicated in a plethora of human diseases, especially cancer. Therefore, histone methyltransferases have been increasingly recognized as potential therapeutic targets. Consequently, the discovery and development of histone methyltransferase inhibitors have been pursued with steadily increasing interest over the past decade. However, the disadvantages of limited clinical efficacy, moderate selectivity, and propensity for acquired resistance have hindered the development of HMTs inhibitors. Targeted covalent modification represents a proven strategy for kinase drug development and has gained increasing attention in HMTs drug discovery. In this review, we focus on the discovery, characterization, and biological applications of covalent inhibitors for HMTs with emphasis on advancements in the field. In addition, we identify the challenges and future directions in this fast-growing research area of drug discovery.
Collapse
|
20
|
Shiah JV, Johnson DE, Grandis JR. Transcription Factors and Cancer: Approaches to Targeting. Cancer J 2023; 29:38-46. [PMID: 36693157 PMCID: PMC9881838 DOI: 10.1097/ppo.0000000000000639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
ABSTRACT Cancer is defined by the presence of uncontrollable cell growth, whereby improper proliferative signaling has overcome regulation by cellular mechanisms. Transcription factors are uniquely situated at the helm of signaling, merging extracellular stimuli with intracellular responses. Therefore, this class of proteins plays a pivotal role in coordinating the correct gene expression levels for maintaining normal cellular functions. Dysregulation of transcription factor activity unsurprisingly drives tumorigenesis and oncogenic transformation. Although this imparts considerable therapeutic potential to targeting transcription factors, their lack of enzymatic activity renders intervention challenging and has contributed to a sense that transcription factors are "undruggable." Yet, enduring efforts to elucidate strategies for targeting transcription factors as well as a deeper understanding of their interactions with binding partners have led to advancements that are emerging to counter this narrative. Here, we highlight some of these approaches, focusing primarily on therapeutics that have advanced to the clinic.
Collapse
Affiliation(s)
- Jamie V Shiah
- From the Department Otolaryngology-Head and Neck Surgery, University of California at San Francisco, San Francisco, CA
| | | | | |
Collapse
|
21
|
Chemical biology and pharmacology of histone lysine methylation inhibitors. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194840. [PMID: 35753676 DOI: 10.1016/j.bbagrm.2022.194840] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 12/20/2022]
Abstract
Histone lysine methylation is a post-translational modification that plays a key role in the epigenetic regulation of a broad spectrum of biological processes. Moreover, the dysregulation of histone lysine methyltransferases (KMTs) has been implicated in the pathogenesis of several diseases particularly cancer. Due to their pathobiological importance, KMTs have garnered immense attention over the last decade as attractive therapeutic targets. These endeavors have culminated in tens of chemical probes that have been used to interrogate many aspects of histone lysine methylation. Besides, over a dozen inhibitors have been advanced to clinical trials, including the EZH2 inhibitor tazemetostat approved for the treatment of follicular lymphoma and advanced epithelioid sarcoma. In this Review, we highlight the chemical biology and pharmacology of KMT inhibitors and targeted protein degraders focusing on the clinical development of EZH1/2, DOT1L, Menin-MLL, and WDR5-MLL inhibitors. We also briefly discuss the pharmacologic targeting of other KMTs.
Collapse
|
22
|
Kim T, Jeong K, Kim E, Yoon K, Choi J, Park JH, Kim JH, Kim HS, Youn HD, Cho EJ. Menin Enhances Androgen Receptor-Independent Proliferation and Migration of Prostate Cancer Cells. Mol Cells 2022; 45:202-215. [PMID: 35014621 PMCID: PMC9001152 DOI: 10.14348/molcells.2021.0206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/19/2021] [Accepted: 12/07/2021] [Indexed: 11/27/2022] Open
Abstract
The androgen receptor (AR) is an important therapeutic target for treating prostate cancer (PCa). Moreover, there is an increasing need for understanding the AR-independent progression of tumor cells such as neuroendocrine prostate cancer (NEPC). Menin, which is encoded by multiple endocrine neoplasia type 1 (MEN1), serves as a direct link between AR and the mixed-lineage leukemia (MLL) complex in PCa development by activating AR target genes through histone H3 lysine 4 methylation. Although menin is a critical component of AR signaling, its tumorigenic role in AR-independent PCa cells remains unknown. Here, we compared the role of menin in AR-positive and AR-negative PCa cells via RNAi-mediated or pharmacological inhibition of menin. We demonstrated that menin was involved in tumor cell growth and metastasis in PCa cells with low or deficient levels of AR. The inhibition of menin significantly diminished the growth of PCa cells and induced apoptosis, regardless of the presence of AR. Additionally, transcriptome analysis showed that the expression of many metastasis-associated genes was perturbed by menin inhibition in AR-negative DU145 cells. Furthermore, wound-healing assay results showed that menin promoted cell migration in AR-independent cellular contexts. Overall, these findings suggest a critical function of menin in tumorigenesis and provide a rationale for drug development against menin toward targeting high-risk metastatic PCa, especially those independent of AR.
Collapse
Affiliation(s)
- Taewan Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Kwanyoung Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Eunji Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Kwanghyun Yoon
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Jinmi Choi
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Jae Hyeon Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Jae-Hwan Kim
- NineBiopharm, Co., Ltd., Cheongju 28161, Korea
- National Creative Research Center for Epigenome Reprogramming Network, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Hong-Duk Youn
- National Creative Research Center for Epigenome Reprogramming Network, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Eun-Jung Cho
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
23
|
Targeting matrix metallopeptidase 2 by hydroxyurea selectively kills acute myeloid mixed-lineage leukemia. Cell Death Dis 2022; 8:180. [PMID: 35396375 PMCID: PMC8993889 DOI: 10.1038/s41420-022-00989-4] [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: 12/16/2021] [Revised: 03/02/2022] [Accepted: 03/21/2022] [Indexed: 12/02/2022]
Abstract
Oncogene-induced tumorigenesis results in the variation of epigenetic modifications, and in addition to promoting cell immortalization, cancer cells undergo more intense cellular stress than normal cells and depend on other support genes for survival. Chromosomal translocations of mixed-lineage leukemia (MLL) induce aggressive leukemias with an inferior prognosis. Unfortunately, most MLL-rearranged (MLL-r) leukemias are resistant to conventional chemotherapies. Here, we showed that hydroxyurea (HU) could kill MLL-r acute myeloid leukemia (AML) cells through the necroptosis process. HU target these cells by matrix metallopeptidase 2 (MMP2) deficiency rather than subordinate ribonucleotide reductase regulatory subunit M2 (RRM2) inhibition, where MLL directly regulates MMP2 expression and is decreased in most MLL-r AMLs. Moreover, iron chelation of HU is also indispensable for inducing cell stress, and MMP2 is the support factor to protect cells from death. Our preliminary study indicates that MMP2 might play a role in the nonsense-mediated mRNA decay pathway that prevents activation of unfolding protein response under innocuous endoplasmic reticulum stress. Hence, these results reveal a possible strategy of HU application in MLL-r AML treatment and shed new light upon HU repurposing.
Collapse
|
24
|
Bai H, Zhang SQ, Lei H, Wang F, Ma M, Xin M. Menin-MLL protein-protein interaction inhibitors: a patent review (2014-2021). Expert Opin Ther Pat 2022; 32:507-522. [PMID: 35202550 DOI: 10.1080/13543776.2022.2045947] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Chromosomal translocations involving the mixed-lineage leukemia (MLL, KMT2A, MLL1) genes result in the production of MLL fusion proteins, which cause abnormal transcriptional regulation leading to acute leukemia (AL). Menin (MEN1) protein is essential for MLL to regulate the expression of related target genes. High affinity interactions between the amino terminus of MLL proteins and Menin proteins are required to mediate the oncogenic transformation of MLL fusion proteins. Therefore, inhibition of Menin and MLL interaction is a potential therapeutic strategy for MLL rearrangement (MLL-r) leukemia and can provide a new choice for treatment of other diseases. Therefore, researchers have made great efforts to explore small molecule Menin-MLL interaction inhibitors. AREAS COVERED This review is to provide an overview of the patented Menin-MLL protein protein interaction inhibitors from 2014 to 2021. EXPERT OPINION Menin-MLL interaction inhibitors have therapeutic potential in the treatment of acute leukemia, such as acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL). Nowadays, SNDX-5613 and KO-539 have been granted orphan drug designation by the FDA for treatment of refractory/relapsed leukemia and AML, respectively. In addition, they are undergoing clinical evaluation for other indications. It is clear that Menin-MLL interaction inhibitors have promising benefits in the clinical treatment of acute leukemia and other diseases.
Collapse
Affiliation(s)
- Huanrong Bai
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - San-Qi Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Hao Lei
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Fang Wang
- Hebei Chemical & Pharmaceutical College, Shijiazhuang 050000, P.R. China
| | - Mengyan Ma
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Minhang Xin
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
25
|
He L, Boulant S, Stanifer M, Guo C, Nießen A, Chen M, Felix K, Bergmann F, Strobel O, Schimmack S. The link between menin and pleiotrophin in the tumor biology of pancreatic neuroendocrine neoplasms. Cancer Sci 2022; 113:1575-1586. [PMID: 35179814 PMCID: PMC9128182 DOI: 10.1111/cas.15301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/26/2022] [Accepted: 01/30/2022] [Indexed: 11/29/2022] Open
Abstract
MEN1, which encodes menin protein, is the most frequently mutated gene in pancreatic neuroendocrine neoplasms (pNEN). Pleiotrophin (PTN) was reported being a downstream factor of menin and to promote metastasis in different tumor entities. In this study, the effect of menin and its link to PTN were assessed on features of pNEN cells and outcome of pNEN patients. The expression of menin and PTN in pNEN patient tissues were examined by qRT-PCR and western blot and compared to their metastasis status. Functional assays, including transwell migration/invasion and scratch wound healing assays, were performed on specifically designed CRISPR/Cas9-mediated MEN1-knockout (MEN1-KO) pNEN cell lines (BON1MEN1-KO and QGP1MEN1-KO ) to study the metastasis of pNEN. Among 30 menin negative pNEN patients, 21 revealed a strong protein expression of PTN. This combination was associated with metastasis and shorter disease-free survival. Accordingly, in BON1MEN1-KO and QGP1MEN1-KO cells, PTN protein expression was positively associated with enhanced cell migration and invasion, which could be reversed by PTN silencing. PTN is a predicting factor of metastatic behavior of menin-deficient-pNEN. In vitro, menin is able to both promote and suppress the metastasis of pNEN by regulating PTN expression depending on the tumoral origin of pNEN cells.
Collapse
Affiliation(s)
- Liping He
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, P.R. China
| | - Steeve Boulant
- Center for Integrative Infectious Disease Research, Heidelberg University, Heidelberg, Germany
| | - Megan Stanifer
- Center for Integrative Infectious Disease Research, Heidelberg University, Heidelberg, Germany
| | - Cuncai Guo
- Center for Integrative Infectious Disease Research, Heidelberg University, Heidelberg, Germany
| | - Anna Nießen
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Mingyi Chen
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Department of Breast Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310003, P.R. China
| | - Klaus Felix
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Frank Bergmann
- Institute of Pathology, Heidelberg University, Heidelberg, Germany
| | - Oliver Strobel
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Simon Schimmack
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
26
|
Lei H, Zhang SQ, Fan S, Bai HR, Zhao HY, Mao S, Xin M. Recent Progress of Small Molecule Menin-MLL Interaction Inhibitors as Therapeutic Agents for Acute Leukemia. J Med Chem 2021; 64:15519-15533. [PMID: 34726905 DOI: 10.1021/acs.jmedchem.1c00872] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mixed lineage leukemia (MLL) gene rearrangements are associated with acute leukemia. The protein menin is regarded as a critical oncogenic cofactor of the resulting MLL fusion proteins in acute leukemia. A direct interaction between menin and the MLL amino terminal sequences is necessary for MLL fusion protein-mediated leukemogenesis. Thus, inhibition of the interaction between menin and MLL has emerged as a novel therapeutic strategy. Recent improvements in structural biology and chemical reactivity have promoted the design and development of selective and potent menin-MLL interaction inhibitors. In this Perspective, different classes of menin-MLL interaction inhibitors are comprehensively summarized. Further research potential, challenges, and opportunities in the field are also discussed.
Collapse
Affiliation(s)
- Hao Lei
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - San-Qi Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Shu Fan
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Huan-Rong Bai
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Hong-Yi Zhao
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Shuai Mao
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Minhang Xin
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| |
Collapse
|
27
|
Cherif C, Nguyen DT, Paris C, Le TK, Sefiane T, Carbuccia N, Finetti P, Chaffanet M, Kaoutari AE, Vernerey J, Fazli L, Gleave M, Manai M, Barthélémy P, Birnbaum D, Bertucci F, Taïeb D, Rocchi P. Menin inhibition suppresses castration-resistant prostate cancer and enhances chemosensitivity. Oncogene 2021; 41:125-137. [PMID: 34711954 PMCID: PMC8724010 DOI: 10.1038/s41388-021-02039-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 09/09/2021] [Accepted: 09/27/2021] [Indexed: 11/09/2022]
Abstract
Disease progression and therapeutic resistance of prostate cancer (PC) are linked to multiple molecular events that promote survival and plasticity. We previously showed that heat shock protein 27 (HSP27) acted as a driver of castration-resistant phenotype (CRPC) and developed an oligonucleotides antisense (ASO) against HSP27 with evidence of anti-cancer activity in men with CRPC. Here, we show that the tumor suppressor Menin (MEN1) is highly regulated by HSP27. Menin is overexpressed in high-grade PC and CRPC. High MEN1 mRNA expression is associated with decreased biochemical relapse-free and overall survival. Silencing Menin with ASO technology inhibits CRPC cell proliferation, tumor growth, and restores chemotherapeutic sensitivity. ChIP-seq analysis revealed differential DNA binding sites of Menin in various prostatic cells, suggesting a switch from tumor suppressor to oncogenic functions in CRPC. These data support the evaluation of ASO against Menin for CRPC. ![]()
Collapse
Affiliation(s)
- Chaïma Cherif
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France.,Laboratory of Biochemistry and Molecular Biology, Science University of Tunis, 2092, El Manar, Tunis, Tunisia
| | - Dang Tan Nguyen
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Clément Paris
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Thi Khanh Le
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Thibaud Sefiane
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Nadine Carbuccia
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Pascal Finetti
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Max Chaffanet
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Abdessamad El Kaoutari
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Julien Vernerey
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - Ladan Fazli
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Martin Gleave
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Mohamed Manai
- Laboratory of Biochemistry and Molecular Biology, Science University of Tunis, 2092, El Manar, Tunis, Tunisia
| | - Philippe Barthélémy
- ARNA Laboratory, INSERM U1212, CNRS UMR 5320, University of Bordeaux, F-33076 Bordeaux, France
| | - Daniel Birnbaum
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - François Bertucci
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France
| | - David Taïeb
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France.,Biophysics and Nuclear Medicine Department, La Timone University Hospital, European Center for Research in Medical Imaging, Aix-Marseille University, F-13005 Marseille, France
| | - Palma Rocchi
- Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille, Inserm UMR 1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix-Marseille University, 27 Bd. Leï Roure, F-13009 Marseille, France.
| |
Collapse
|
28
|
Antisense Oligonucleotide-Based Therapeutic against Menin for Triple-Negative Breast Cancer Treatment. Biomedicines 2021; 9:biomedicines9070795. [PMID: 34356858 PMCID: PMC8301388 DOI: 10.3390/biomedicines9070795] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/20/2021] [Accepted: 06/30/2021] [Indexed: 01/01/2023] Open
Abstract
The tumor suppressor menin has dual functions, acting either as a tumor suppressor or as an oncogene/oncoprotein, depending on the oncological context. Triple-negative breast cancer (TNBC) is characterized by the lack of expression of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (ERBB2/HER2) and is often a basal-like breast cancer. TNBC is associated with a dismal prognosis and an insufficient response to chemotherapies. Previously, menin was shown to play a proliferative role in ER-positive breast cancer; however, the functions of menin in TNBC remain unknown. Here, we have demonstrated that menin is expressed in various TNBC subtypes with the strongest expression in the TNBC Hs 578T cells. The depletion of menin by an antisense oligonucleotide (ASO) inhibits cell proliferation, enhances apoptosis in Hs 578T cells, highlighting the oncogenic functions of menin in this TNBC model. ASO-based menin silencing also delays the tumor progression of TNBC xenografts. Analysis of the menin interactome suggests that menin could drive TNBC tumorigenesis through the regulation of MLL/KMT2A-driven transcriptional activity, mRNA 3′-end processing and apoptosis. The study provides a rationale behind the use of ASO-based therapy, targeting menin in monotherapy or in combination with chemo or PARP inhibitors for menin-positive TNBC treatments.
Collapse
|
29
|
Zhang M, Aguilar A, Xu S, Huang L, Chinnaswamy K, Sleger T, Wang B, Gross S, Nicolay BN, Ronseaux S, Harvey K, Wang Y, McEachern D, Kirchhoff PD, Liu Z, Stuckey J, Tron AE, Liu T, Wang S. Discovery of M-1121 as an Orally Active Covalent Inhibitor of Menin-MLL Interaction Capable of Achieving Complete and Long-Lasting Tumor Regression. J Med Chem 2021; 64:10333-10349. [PMID: 34196551 DOI: 10.1021/acs.jmedchem.1c00789] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Targeting the menin-MLL protein-protein interaction is being pursued as a new therapeutic strategy for the treatment of acute leukemia carrying MLL-rearrangements (MLLr leukemia). Herein, we report M-1121, a covalent and orally active inhibitor of the menin-MLL interaction capable of achieving complete and persistent tumor regression. M-1121 establishes covalent interactions with Cysteine 329 located in the MLL binding pocket of menin and potently inhibits growth of acute leukemia cell lines carrying MLL translocations with no activity in cell lines with wild-type MLL. Consistent with the mechanism of action, M-1121 drives dose-dependent down-regulation of HOXA9 and MEIS1 gene expression in the MLL-rearranged MV4;11 leukemia cell line. M-1121 is orally bioavailable and shows potent antitumor activity in vivo with tumor regressions observed at tolerated doses in the MV4;11 subcutaneous and disseminated models of MLL-rearranged leukemia. Together, our findings support development of an orally active covalent menin inhibitor as a new therapy for MLLr leukemia.
Collapse
Affiliation(s)
- Meng Zhang
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Angelo Aguilar
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Shilin Xu
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Liyue Huang
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | | | - Taryn Sleger
- Agios Pharmaceuticals, Inc., Cambridge, Massachusetts 02139, United States
| | - Bo Wang
- Agios Pharmaceuticals, Inc., Cambridge, Massachusetts 02139, United States
| | - Stefan Gross
- Agios Pharmaceuticals, Inc., Cambridge, Massachusetts 02139, United States
| | - Brandon N Nicolay
- Agios Pharmaceuticals, Inc., Cambridge, Massachusetts 02139, United States
| | - Sebastien Ronseaux
- Agios Pharmaceuticals, Inc., Cambridge, Massachusetts 02139, United States
| | - Kaitlin Harvey
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yu Wang
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Donna McEachern
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Paul D Kirchhoff
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Zhaomin Liu
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jeanne Stuckey
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Adriana E Tron
- Agios Pharmaceuticals, Inc., Cambridge, Massachusetts 02139, United States
| | - Tao Liu
- Agios Pharmaceuticals, Inc., Cambridge, Massachusetts 02139, United States
| | - Shaomeng Wang
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States.,Department of Medicinal Chemistry, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
30
|
Gao K, Shaabani S, Xu R, Zarganes-Tzitzikas T, Gao L, Ahmadianmoghaddam M, Groves MR, Dömling A. Nanoscale, automated, high throughput synthesis and screening for the accelerated discovery of protein modifiers. RSC Med Chem 2021; 12:809-818. [PMID: 34124680 PMCID: PMC8152715 DOI: 10.1039/d1md00087j] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/14/2021] [Indexed: 11/26/2022] Open
Abstract
Hit finding in early drug discovery is often based on high throughput screening (HTS) of existing and historical compound libraries, which can limit chemical diversity, is time-consuming, very costly, and environmentally not sustainable. On-the-fly compound synthesis and in situ screening in a highly miniaturized and automated format has the potential to greatly reduce the medicinal chemistry environmental footprint. Here, we used acoustic dispensing technology to synthesize a library in a 1536 well format based on the Groebcke-Blackburn-Bienaymé reaction (GBB-3CR) on a nanomole scale. The unpurified library was screened by differential scanning fluorimetry (DSF) and cross-validated using microscale thermophoresis (MST) against the oncogenic protein-protein interaction menin-MLL. Several GBB reaction products were found as μM menin binder, and the structural basis of the interactions with menin was elucidated by co-crystal structure analysis. Miniaturization and automation of the organic synthesis and screening process can lead to an acceleration in the early drug discovery process, which is an alternative to classical HTS and a step towards the paradigm of continuous manufacturing.
Collapse
Affiliation(s)
- Kai Gao
- Pharmacy Department, Drug Design group, University of Groningen A. Deusinglaan 1 9700 AD Groningen The Netherlands
| | - Shabnam Shaabani
- Pharmacy Department, Drug Design group, University of Groningen A. Deusinglaan 1 9700 AD Groningen The Netherlands
| | - Ruixue Xu
- Pharmacy Department, Drug Design group, University of Groningen A. Deusinglaan 1 9700 AD Groningen The Netherlands
| | - Tryfon Zarganes-Tzitzikas
- Pharmacy Department, Drug Design group, University of Groningen A. Deusinglaan 1 9700 AD Groningen The Netherlands
| | - Li Gao
- Pharmacy Department, Drug Design group, University of Groningen A. Deusinglaan 1 9700 AD Groningen The Netherlands
| | - Maryam Ahmadianmoghaddam
- Pharmacy Department, Drug Design group, University of Groningen A. Deusinglaan 1 9700 AD Groningen The Netherlands
| | - Matthew R Groves
- Pharmacy Department, Drug Design group, University of Groningen A. Deusinglaan 1 9700 AD Groningen The Netherlands
| | - Alexander Dömling
- Pharmacy Department, Drug Design group, University of Groningen A. Deusinglaan 1 9700 AD Groningen The Netherlands
| |
Collapse
|
31
|
Grieselhuber NR, Mims AS. Novel Targeted Therapeutics in Acute Myeloid Leukemia: an Embarrassment of Riches. Curr Hematol Malig Rep 2021; 16:192-206. [PMID: 33738705 DOI: 10.1007/s11899-021-00621-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW Acute myeloid leukemia (AML) is an aggressive malignancy of the bone marrow that has a poor prognosis with traditional cytotoxic chemotherapy, especially in elderly patients. In recent years, small molecule inhibitors targeting AML-associated IDH1, IDH2, and FLT3 mutations have been FDA approved. However, the majority of AML cases do not have a targetable mutation. A variety of novel agents targeting both previously untargetable mutations and general pathways in AML are currently being investigated. Herein, we review selected new targeted therapies currently in early-phase clinical investigation in AML. RECENT FINDINGS The DOT1L inhibitor pinometostat in KMT2A-rearranged AML, the menin inhibitors KO-539 and SYNDX-5613 in KMT2Ar and NPM1-mutated AML, and the mutant TP53 inhibitor APR-246 are examples of novel agents targeting specific mutations in AML. In addition, BET inhibitors, polo-like kinase inhibitors, and MDM2 inhibitors are promising new drug classes for AML which do not depend on the presence of a particular mutation. AML remains in incurable disease for many patients but advances in genomics, epigenetics, and drug discovery have led to the development of many potential novel therapeutic agents, many of which are being investigated in ongoing clinical trials. Additional studies will be necessary to determine how best to incorporate these novel agents into routine clinical treatment of AML.
Collapse
Affiliation(s)
- Nicole R Grieselhuber
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Alice S Mims
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
32
|
Zhang Y, Rong D, Li B, Wang Y. Targeting Epigenetic Regulators with Covalent Small-Molecule Inhibitors. J Med Chem 2021; 64:7900-7925. [PMID: 33599482 DOI: 10.1021/acs.jmedchem.0c02055] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epigenetic regulation of gene expression plays a critical role in various physiological processes, and epigenetic dysregulation is implicated in a number of diseases, prominently including cancer. Epigenetic regulators have been validated as potential therapeutic targets, and significant progress has been made in the discovery and development of epigenetic-based inhibitors. However, successful epigenetic drug discovery is still facing challenges, including moderate selectivity, limited efficacy, and acquired drug resistance. Inspired by the advantages of covalent small-molecule inhibitors, targeted covalent inhibition has attracted increasing interest in epigenetic drug discovery. In this review, we comprehensively summarize the structure-based design and characterization of covalent inhibitors targeting epigenetic writers, readers, and erasers and highlight their potential benefits in enhancing selectivity across the enzyme family and improving in vivo efficacy. We also discuss the challenges and opportunities of covalent small-molecule inhibitors and hope to shed light on future epigenetic drug discovery.
Collapse
Affiliation(s)
- Yi Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Deqin Rong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Bingbing Li
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yuanxiang Wang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
33
|
Abstract
PURPOSE OF THE REVIEW Infant leukemia is a rare, distinct subgroup of pediatric acute leukemias diagnosed in children under 1 year of age and characterized by unique, aggressive biology. Here, we review its clinical presentation, underlying molecular biology, current treatment strategies, and novel therapeutic approaches. RECENT FINDINGS Infant leukemias are associated with high-risk molecular features and high rates of chemotherapy resistance. International collaborative clinical trials have led to better understanding of the underlying molecular biology, refined risk-based stratification, and investigated the use of hematopoietic stem cell transplantation. However, intensification of chemotherapy has failed to improve outcomes, and current regimens are associated with significant treatment-related and long-term toxicities. Infants with leukemia remain a challenging group to treat. We must continue collaborative efforts to move beyond traditional cytotoxic chemotherapy, incorporate molecularly targeted strategies and immunotherapy, and increase access to clinical trials to improve outcomes for this high-risk group of patients.
Collapse
|
34
|
Nelakurti DD, Pappula AL, Rajasekaran S, Miles WO, Petreaca RC. Comprehensive Analysis of MEN1 Mutations and Their Role in Cancer. Cancers (Basel) 2020; 12:cancers12092616. [PMID: 32937789 PMCID: PMC7565326 DOI: 10.3390/cancers12092616] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/04/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Cancers are characterized by accumulation of genetic mutations in key cell cycle regulators that alter or disable the function of these genes. Such mutations can be inherited or arise spontaneously during the life of the individual. The MEN1 gene prevents uncontrolled cell division and it is considered a tumor suppressor. Inherited MEN1 mutations are associated with certain parathyroid and pancreatic syndromes while spontaneous mutations have been detected in cancer cells. We investigated whether inherited mutations appear in cancer cells which would suggest that patients with parathyroid and pancreatic syndromes have a predisposition to develop cancer. We find a weak correlation between the spectrum of inherited mutations and those appearing spontaneously. Thus, inherited MEN1 mutations may not be a good predictor of tumorigenesis. Abstract MENIN is a scaffold protein encoded by the MEN1 gene that functions in multiple biological processes, including cell proliferation, migration, gene expression, and DNA damage repair. MEN1 is a tumor suppressor gene, and mutations that disrupts MEN1 function are common to many tumor types. Mutations within MEN1 may also be inherited (germline). Many of these inherited mutations are associated with a number of pathogenic syndromes of the parathyroid and pancreas, and some also predispose patients to hyperplasia. In this study, we cataloged the reported germline mutations from the ClinVar database and compared them with the somatic mutations detected in cancers from the Catalogue of Somatic Mutations in Cancer (COSMIC) database. We then used statistical software to determine the probability of mutations being pathogenic or driver. Our data show that many confirmed germline mutations do not appear in tumor samples. Thus, most mutations that disable MEN1 function in tumors are somatic in nature. Furthermore, of the germline mutations that do appear in tumors, only a fraction has the potential to be pathogenic or driver mutations.
Collapse
Affiliation(s)
- Devi D. Nelakurti
- Biomedical Science Undergraduate Program, The Ohio State University Medical School, Columbus, OH 43210, USA;
| | - Amrit L. Pappula
- Computer Science and Engineering Undergraduate Program, The Ohio State University, Columbus, OH 43210, USA;
| | - Swetha Rajasekaran
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA;
| | - Wayne O. Miles
- Department of Cancer Biology and Genetics, The Ohio State University Medical School, Columbus, OH 43210, USA;
| | - Ruben C. Petreaca
- Department of Molecular Genetics, The Ohio State University, Marion, OH 43302, USA
- Correspondence:
| |
Collapse
|
35
|
Covalent and noncovalent constraints yield a figure eight-like conformation of a peptide inhibiting the menin-MLL interaction. Eur J Med Chem 2020; 207:112748. [PMID: 32882610 DOI: 10.1016/j.ejmech.2020.112748] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/04/2020] [Accepted: 08/08/2020] [Indexed: 11/21/2022]
Abstract
The interaction between menin and mixed lineage leukemia (MLL) was identified as an interesting target for treating some cancers including acute leukemia. On the basis of the known crystal structure of the MBM1-menin complex (MBM - menin binding motif), several cyclic peptides were designed. Elaboration of the effective cyclization strategy using a metathesis reaction allowed for a successfully large number of derivatives to be obtained. Subsequent optimization of the loop size, as well as N-terminal, central and C-terminal parts of the studied peptides resulted in structures exhibiting low nanomolar activities. A crystal structure of an inhibitor-menin complex revealed a compact conformation of the ligand molecule, which is stabilized not only by the introduction of a covalent linker but also three intramolecular hydrogen bonds. The inhibitor adopts a figure eight-like conformation, which perfectly fits the cleft of menin. We demonstrated that the development of compact, miniprotein-like structures is a highly effective approach for inhibition of protein-protein interactions.
Collapse
|
36
|
Linhares BM, Grembecka J, Cierpicki T. Targeting epigenetic protein-protein interactions with small-molecule inhibitors. Future Med Chem 2020; 12:1305-1326. [PMID: 32551894 PMCID: PMC7421387 DOI: 10.4155/fmc-2020-0082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 05/01/2020] [Indexed: 02/07/2023] Open
Abstract
Epigenetic protein-protein interactions (PPIs) play essential roles in regulating gene expression, and their dysregulations have been implicated in many diseases. These PPIs are comprised of reader domains recognizing post-translational modifications on histone proteins, and of scaffolding proteins that maintain integrities of epigenetic complexes. Targeting PPIs have become focuses for development of small-molecule inhibitors and anticancer therapeutics. Here we summarize efforts to develop small-molecule inhibitors targeting common epigenetic PPI domains. Potent small molecules have been reported for many domains, yet small domains that recognize methylated lysine side chains on histones are challenging in inhibitor development. We posit that the development of potent inhibitors for difficult-to-prosecute epigenetic PPIs may be achieved by interdisciplinary approaches and extensive explorations of chemical space.
Collapse
Affiliation(s)
- Brian M Linhares
- Biophysics Program, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular, Cellular & Developmental Biology, Yale University, New Haven, CT 06511, USA
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Tomasz Cierpicki
- Biophysics Program, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
37
|
Xu S, Aguilar A, Huang L, Xu T, Zheng K, McEachern D, Przybranowski S, Foster C, Zawacki K, Liu Z, Chinnaswamy K, Stuckey J, Wang S. Discovery of M-808 as a Highly Potent, Covalent, Small-Molecule Inhibitor of the Menin-MLL Interaction with Strong In Vivo Antitumor Activity. J Med Chem 2020; 63:4997-5010. [PMID: 32338903 PMCID: PMC7981784 DOI: 10.1021/acs.jmedchem.0c00547] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Targeting the menin-MLL protein-protein interaction is a new therapeutic strategy for the treatment of acute leukemia carrying MLL fusion (MLL leukemia). We describe herein the structure-based optimization of a class of covalent menin inhibitors, which led to the discovery of M-808 (16) as a highly potent and efficacious covalent menin inhibitor. M-808 effectively inhibits leukemia cell growth at low nanomolar concentrations and is capable of achieving partial tumor regression in an MV4;11 xenograft tumor model in mice at a well-tolerated dose schedule. Determination of the co-crystal structure of M-808 in complex with menin provides a structural basis for their high-affinity, covalent interactions. M-808 represents a promising, covalent menin inhibitor for further optimization and evaluation toward developing a new therapy for the treatment of MLL leukemia.
Collapse
Affiliation(s)
- Shilin Xu
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Angelo Aguilar
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Liyue Huang
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Tianfeng Xu
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ke Zheng
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Donna McEachern
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Sally Przybranowski
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Caroline Foster
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Kaitlin Zawacki
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Zhaomin Liu
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | | | - Jeanne Stuckey
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Shaomeng Wang
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States,Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States,Department of Medicinal Chemistry, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States,Corresponding Author Phone: 1-734-615-0362;
| |
Collapse
|
38
|
Chen J, Glasser CL. New and Emerging Targeted Therapies for Pediatric Acute Myeloid Leukemia (AML). CHILDREN (BASEL, SWITZERLAND) 2020; 7:E12. [PMID: 32050659 PMCID: PMC7072702 DOI: 10.3390/children7020012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 12/16/2022]
Abstract
The relapse rate for children with acute myeloid leukemia (AML) remains high despite advancements in risk classification, multi-agent chemotherapy intensification, stem cell transplantation, and supportive care guidelines. Prognosis for this subgroup of children with relapsed/refractory AML remains poor. It is well known that the ceiling of chemotherapy intensification has been reached, limited by acute and chronic toxicity, necessitating alternative treatment approaches. In the last several years, our improved understanding of disease biology and critical molecular pathways in AML has yielded a variety of new drugs to target these specific pathways. This review provides a summary of antibody drug conjugates (ADCs), small molecule inhibitors, and tyrosine kinase inhibitors with an emphasis on those that are currently under clinical evaluation or soon to open in early phase trials for children with relapsed/refractory AML.
Collapse
Affiliation(s)
- Jing Chen
- Division of Pediatric Hematology/Oncology, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Chana L. Glasser
- Division of Pediatric Hematology/Oncology, NYU Winthrop Hospital, Mineola, NY 11501, USA
| |
Collapse
|
39
|
Transcriptional addiction in mixed lineage leukemia: new avenues for target therapies. BLOOD SCIENCE 2019; 1:50-56. [PMID: 35402805 PMCID: PMC8975088 DOI: 10.1097/bs9.0000000000000011] [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: 06/11/2019] [Accepted: 07/02/2019] [Indexed: 11/25/2022] Open
Abstract
Mixed lineage leukemia (MLL) is an aggressive and refractory blood cancer that predominantly occurs in pediatric patients and is often associated with poor prognosis and dismal outcomes. Thus far, no effective target therapy for the treatment of MLL leukemia is available. MLL leukemia is caused by the rearrangement of MLL genes at 11q23, which generates various MLL chimeric proteins that promote leukemogenesis through transcriptional misregulation of MLL target genes. Biochemical studies on MLL chimeras have identified that the most common partners exist in the superelongation complex (SEC) and DOT1L complex, which activate or sustain MLL target gene expression through processive transcription elongation. The results of these studies indicate a transcription-related mechanism for MLL leukemogenesis and maintenance. In this study, we first review the history of MLL leukemia and its related clinical features. Then, we discuss the biological functions of MLL and MLL chimeras, significant cooperating events, and transcriptional addiction mechanisms in MLL leukemia with an emphasis on potential and rational therapy development. Collectively, we believe that targeting the transcriptional addiction mediated by SEC and the DOT1L complex will provide new avenues for target therapies in MLL leukemia and serve as a novel paradigm for targeting transcriptional addiction in other cancers.
Collapse
|
40
|
Cicaloni V, Trezza A, Pettini F, Spiga O. Applications of in Silico Methods for Design and Development of Drugs Targeting Protein-Protein Interactions. Curr Top Med Chem 2019; 19:534-554. [PMID: 30836920 DOI: 10.2174/1568026619666190304153901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 01/02/2019] [Accepted: 01/25/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Identification of Protein-Protein Interactions (PPIs) is a major challenge in modern molecular biology and biochemistry research, due to the unquestionable role of proteins in cells, biological process and pathological states. Over the past decade, the PPIs have evolved from being considered a highly challenging field of research to being investigated and examined as targets for pharmacological intervention. OBJECTIVE Comprehension of protein interactions is crucial to known how proteins come together to build signalling pathways, to carry out their functions, or to cause diseases, when deregulated. Multiplicity and great amount of PPIs structures offer a huge number of new and potential targets for the treatment of different diseases. METHODS Computational techniques are becoming predominant in PPIs studies for their effectiveness, flexibility, accuracy and cost. As a matter of fact, there are effective in silico approaches which are able to identify PPIs and PPI site. Such methods for computational target prediction have been developed through molecular descriptors and data-mining procedures. RESULTS In this review, we present different types of interactions between protein-protein and the application of in silico methods for design and development of drugs targeting PPIs. We described computational approaches for the identification of possible targets on protein surface and to detect of stimulator/ inhibitor molecules. CONCLUSION A deeper study of the most recent bioinformatics methodologies for PPIs studies is vital for a better understanding of protein complexes and for discover new potential PPI modulators in therapeutic intervention.
Collapse
Affiliation(s)
- Vittoria Cicaloni
- Department of Biotechnology, Chemistry and Pharmacy (Dept. of Excellence 2018-2022), University of Siena, via A. Moro 2, 53100 Siena, Italy.,Toscana Life Sciences Foundation, via Fiorentina 1, 53100 Siena, Italy
| | - Alfonso Trezza
- Department of Biotechnology, Chemistry and Pharmacy (Dept. of Excellence 2018-2022), University of Siena, via A. Moro 2, 53100 Siena, Italy
| | - Francesco Pettini
- Department of Biotechnology, Chemistry and Pharmacy (Dept. of Excellence 2018-2022), University of Siena, via A. Moro 2, 53100 Siena, Italy
| | - Ottavia Spiga
- Department of Biotechnology, Chemistry and Pharmacy (Dept. of Excellence 2018-2022), University of Siena, via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
41
|
Aguilar A, Zheng K, Xu T, Xu S, Huang L, Fernandez-Salas E, Liu L, Bernard D, Harvey KP, Foster C, McEachern D, Stuckey J, Chinnaswamy K, Delproposto J, Kampf JW, Wang S. Structure-Based Discovery of M-89 as a Highly Potent Inhibitor of the Menin-Mixed Lineage Leukemia (Menin-MLL) Protein-Protein Interaction. J Med Chem 2019; 62:6015-6034. [PMID: 31244110 DOI: 10.1021/acs.jmedchem.9b00021] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Inhibition of the menin-mixed lineage leukemia (MLL) protein-protein interaction is a promising new therapeutic strategy for the treatment of acute leukemia carrying MLL fusion (MLL leukemia). We describe herein our structure-based design, synthesis, and evaluation of a new class of small-molecule inhibitors of the menin-MLL interaction (hereafter called menin inhibitors). Our efforts have resulted in the discovery of highly potent menin inhibitors, as exemplified by compound 42 (M-89). M-89 binds to menin with a Kd value of 1.4 nM and effectively engages cellular menin protein at low nanomolar concentrations. M-89 inhibits cell growth in the MV4;11 and MOLM-13 leukemia cell lines carrying MLL fusion with IC50 values of 25 and 55 nM, respectively, and demonstrates >100-fold selectivity over the HL-60 leukemia cell line lacking MLL fusion. The determination of a co-crystal structure of M-89 in a complex with menin provides the structural basis for their high-affinity interaction. Further optimization of M-89 may lead to a new class of therapy for the treatment of MLL leukemia.
Collapse
|
42
|
Balasubramanian K, Gupta SP. Quantum Molecular Dynamics, Topological, Group Theoretical and Graph Theoretical Studies of Protein-Protein Interactions. Curr Top Med Chem 2019; 19:426-443. [PMID: 30836919 DOI: 10.2174/1568026619666190304152704] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 11/08/2018] [Accepted: 11/28/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Protein-protein interactions (PPIs) are becoming increasingly important as PPIs form the basis of multiple aggregation-related diseases such as cancer, Creutzfeldt-Jakob, and Alzheimer's diseases. This mini-review presents hybrid quantum molecular dynamics, quantum chemical, topological, group theoretical, graph theoretical, and docking studies of PPIs. We also show how these theoretical studies facilitate the discovery of some PPI inhibitors of therapeutic importance. OBJECTIVE The objective of this review is to present hybrid quantum molecular dynamics, quantum chemical, topological, group theoretical, graph theoretical, and docking studies of PPIs. We also show how these theoretical studies enable the discovery of some PPI inhibitors of therapeutic importance. METHODS This article presents a detailed survey of hybrid quantum dynamics that combines classical and quantum MD for PPIs. The article also surveys various developments pertinent to topological, graph theoretical, group theoretical and docking studies of PPIs and highlight how the methods facilitate the discovery of some PPI inhibitors of therapeutic importance. RESULTS It is shown that it is important to include higher-level quantum chemical computations for accurate computations of free energies and electrostatics of PPIs and Drugs with PPIs, and thus techniques that combine classical MD tools with quantum MD are preferred choices. Topological, graph theoretical and group theoretical techniques are shown to be important in studying large network of PPIs comprised of over 100,000 proteins where quantum chemical and other techniques are not feasible. Hence, multiple techniques are needed for PPIs. CONCLUSION Drug discovery and our understanding of complex PPIs require multifaceted techniques that involve several disciplines such as quantum chemistry, topology, graph theory, knot theory and group theory, thus demonstrating a compelling need for a multi-disciplinary approach to the problem.
Collapse
Affiliation(s)
- Krishnan Balasubramanian
- School of Molecular Sciences, Arizona State University, Tempe, Arizona, AZ 85287-1604, United States
| | - Satya P Gupta
- Department of Pharmaceutical Technology, Meerut Institute of Engineering Technology, Meerut-250002, India
| |
Collapse
|
43
|
Goldman SL, Hassan C, Khunte M, Soldatenko A, Jong Y, Afshinnekoo E, Mason CE. Epigenetic Modifications in Acute Myeloid Leukemia: Prognosis, Treatment, and Heterogeneity. Front Genet 2019; 10:133. [PMID: 30881380 PMCID: PMC6405641 DOI: 10.3389/fgene.2019.00133] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/08/2019] [Indexed: 01/09/2023] Open
Abstract
Leukemia, specifically acute myeloid leukemia (AML), is a common malignancy that can be differentiated into multiple subtypes based on leukemogenic history and etiology. Although genetic aberrations, particularly cytogenetic abnormalities and mutations in known oncogenes, play an integral role in AML development, epigenetic processes have been shown as a significant and sometimes independent dynamic in AML pathophysiology. Here, we summarize how tumors evolve and describe AML through an epigenetic lens, including discussions on recent discoveries that include prognostics from epialleles, changes in RNA function for hematopoietic stem cells and the epitranscriptome, and novel epigenetic treatment options. We further describe the limitations of treatment in the context of the high degree of heterogeneity that characterizes acute myeloid leukemia.
Collapse
Affiliation(s)
- Samantha L Goldman
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States.,The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States.,University of Maryland, College Park, MD, United States
| | - Ciaran Hassan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States.,The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States.,Yale College, New Haven, CT, United States
| | - Mihir Khunte
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States.,Yale College, New Haven, CT, United States
| | - Arielle Soldatenko
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States.,Yale College, New Haven, CT, United States
| | - Yunji Jong
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States.,Yale College, New Haven, CT, United States
| | - Ebrahim Afshinnekoo
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States.,The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States.,The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, United States
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States.,The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States.,The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, United States.,The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
44
|
Kim P, Zhou X. FusionGDB: fusion gene annotation DataBase. Nucleic Acids Res 2019; 47:D994-D1004. [PMID: 30407583 PMCID: PMC6323909 DOI: 10.1093/nar/gky1067] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/05/2018] [Accepted: 11/01/2018] [Indexed: 12/26/2022] Open
Abstract
Gene fusion is one of the hallmarks of cancer genome via chromosomal rearrangement initiated by DNA double-strand breakage. To date, many fusion genes (FGs) have been established as important biomarkers and therapeutic targets in multiple cancer types. To better understand the function of FGs in cancer types and to promote the discovery of clinically relevant FGs, we built FusionGDB (Fusion Gene annotation DataBase) available at https://ccsm.uth.edu/FusionGDB. We collected 48 117 FGs across pan-cancer from three representative fusion gene resources: the improved database of chimeric transcripts and RNA-seq data (ChiTaRS 3.1), an integrative resource for cancer-associated transcript fusions (TumorFusions), and The Cancer Genome Atlas (TCGA) fusions by Gao et al. For these ∼48K FGs, we performed functional annotations including gene assessment across pan-cancer fusion genes, open reading frame (ORF) assignment, and retention search of 39 protein features based on gene structures of multiple isoforms with different breakpoints. We also provided the fusion transcript and amino acid sequences according to multiple breakpoints and transcript isoforms. Our analyses identified 331, 303 and 667 in-frame FGs with retaining kinase, DNA-binding, and epigenetic factor domains, respectively, as well as 976 FGs lost protein-protein interaction. FusionGDB provides six categories of annotations: FusionGeneSummary, FusionProtFeature, FusionGeneSequence, FusionGenePPI, RelatedDrug and RelatedDisease.
Collapse
Affiliation(s)
- Pora Kim
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaobo Zhou
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
45
|
Fang J, Liu C, Wang Q, Lin P, Cheng F. In silico polypharmacology of natural products. Brief Bioinform 2018; 19:1153-1171. [PMID: 28460068 DOI: 10.1093/bib/bbx045] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Indexed: 01/03/2025] Open
Abstract
Natural products with polypharmacological profiles have demonstrated promise as novel therapeutics for various complex diseases, including cancer. Currently, many gaps exist in our knowledge of which compounds interact with which targets, and experimentally testing all possible interactions is infeasible. Recent advances and developments of systems pharmacology and computational (in silico) approaches provide powerful tools for exploring the polypharmacological profiles of natural products. In this review, we introduce recent progresses and advances of computational tools and systems pharmacology approaches for identifying drug targets of natural products by focusing on the development of targeted cancer therapy. We survey the polypharmacological and systems immunology profiles of five representative natural products that are being considered as cancer therapies. We summarize various chemoinformatics, bioinformatics and systems biology resources for reconstructing drug-target networks of natural products. We then review currently available computational approaches and tools for prediction of drug-target interactions by focusing on five domains: target-based, ligand-based, chemogenomics-based, network-based and omics-based systems biology approaches. In addition, we describe a practical example of the application of systems pharmacology approaches by integrating the polypharmacology of natural products and large-scale cancer genomics data for the development of precision oncology under the systems biology framework. Finally, we highlight the promise of cancer immunotherapies and combination therapies that target tumor ecosystems (e.g. clones or 'selfish' sub-clones) via exploiting the immunological and inflammatory 'side' effects of natural products in the cancer post-genomics era.
Collapse
Affiliation(s)
- Jiansong Fang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuang Liu
- Alibaba Research Center for Complexity Sciences at the Hangzhou Normal University, Hangzhou, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ping Lin
- National Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Feixiong Cheng
- Department of Biomedical Informatics, Vanderbilt University Medical Center in Nashville (United States)
| |
Collapse
|
46
|
Abdel-Magid AF. Potential Treatment of Acute Leukemia with Inhibitors of Menin/MLL Interaction. ACS Med Chem Lett 2018; 9:868-869. [PMID: 30258531 DOI: 10.1021/acsmedchemlett.8b00342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Indexed: 11/28/2022] Open
Affiliation(s)
- Ahmed F. Abdel-Magid
- Therachem Research Medilab, LLC., 100 Jade Park, Chelsea, Alabama 35043, United States
| |
Collapse
|
47
|
Chen QB, Gao J, Zou GA, Xin XL, Aisa HA. Piperidine Alkaloids with Diverse Skeletons from Anacyclus pyrethrum. JOURNAL OF NATURAL PRODUCTS 2018; 81:1474-1482. [PMID: 29775308 DOI: 10.1021/acs.jnatprod.8b00239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Fifteen new piperidine derivatives, pyracyclumines A-J (1-10), including five pairs of enantiomers, (+)-1/(-)-1 to (+)-5/(-)-5, together with three known compounds, agrocybenine (11), 4,6,6-trimethyl-5,6-dihydro-2(1 H)-pyridone (12), and 3,5,5-trimethyl-1,5-dihydro-2 H-pyrrol-2-one (13), were isolated from the roots of Anacyclus pyrethrum. Pyracyclumines A, B, and H (1, 2, and 8) possess a novel 6/5/6/6 dimeric piperidine skeleton, a unique 6/5/6 dimeric piperidine skeleton, and a 1,4,6-triazaindan skeleton, respectively. Pyracyclumine C (3) is based on a rare cyclopentane-piperidine framework. The structures of the isolated compounds were established by analysis of their NMR and HRESIMS data. The racemic pyracyclumines A-E (1-5) were further separated by chiral HPLC to give the enantiomers (+)-1/(-)-1 to (+)-5/(-)-5, for which the absolute configurations were determined by comparison of their experimental and calculated ECD spectra. The plausible biogenetic pathways of these piperidine alkaloids were proposed starting from the basic units of compounds 12 and 13. All of the isolated compounds were tested for their inhibitory effects on menin-mixed lineage leukemia 1 protein-protein interaction.
Collapse
Affiliation(s)
- Qi-Bin Chen
- Key Laboratory of Plant Resources and Chemistry in Arid Zone and State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry , Chinese Academy of Sciences , Urumqi 830011 , People's Republic of China
| | - Jie Gao
- Key Laboratory of Plant Resources and Chemistry in Arid Zone and State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry , Chinese Academy of Sciences , Urumqi 830011 , People's Republic of China
| | - Guo-An Zou
- Key Laboratory of Plant Resources and Chemistry in Arid Zone and State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry , Chinese Academy of Sciences , Urumqi 830011 , People's Republic of China
| | - Xue-Lei Xin
- Key Laboratory of Plant Resources and Chemistry in Arid Zone and State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry , Chinese Academy of Sciences , Urumqi 830011 , People's Republic of China
| | - Haji Akber Aisa
- Key Laboratory of Plant Resources and Chemistry in Arid Zone and State Key Laboratory Basis of Xinjiang Indigenous Medicinal Plants Resource Utilization, Xinjiang Technical Institute of Physics and Chemistry , Chinese Academy of Sciences , Urumqi 830011 , People's Republic of China
| |
Collapse
|
48
|
Borkin D, Klossowski S, Pollock J, Miao H, Linhares BM, Kempinska K, Jin Z, Purohit T, Wen B, He M, Sun D, Cierpicki T, Grembecka J. Complexity of Blocking Bivalent Protein-Protein Interactions: Development of a Highly Potent Inhibitor of the Menin-Mixed-Lineage Leukemia Interaction. J Med Chem 2018; 61:4832-4850. [PMID: 29738674 PMCID: PMC7029623 DOI: 10.1021/acs.jmedchem.8b00071] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The protein-protein interaction between menin and mixed-lineage leukemia 1 (MLL1) plays an important role in development of acute leukemia with translocations of the MLL1 gene and in solid tumors. Here, we report the development of a new generation of menin-MLL1 inhibitors identified by structure-based optimization of the thienopyrimidine class of compounds. This work resulted in compound 28 (MI-1481), which showed very potent inhibition of the menin-MLL1 interaction (IC50 = 3.6 nM), representing the most potent reversible menin-MLL1 inhibitor reported to date. The crystal structure of the menin-28 complex revealed a hydrogen bond with Glu366 and hydrophobic interactions, which contributed to strong inhibitory activity of 28. Compound 28 also demonstrates pronounced activity in MLL leukemia cells and in vivo in MLL leukemia models. Thus, 28 is a valuable menin-MLL1 inhibitor that can be used for potential therapeutic applications and in further studies regarding the role of menin in cancer.
Collapse
Affiliation(s)
- Dmitry Borkin
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Szymon Klossowski
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jonathan Pollock
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hongzhi Miao
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Brian M. Linhares
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Zhuang Jin
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Trupta Purohit
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Bo Wen
- College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Miao He
- College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Duxin Sun
- College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA,Corresponding author; Jolanta Grembecka, PhD, Associate Professor, Department of Pathology, University of Michigan, 1150 West Medical Center Dr, MSRB I, Room 4510D, Ann Arbor, MI, 48108, , Tel. 734-615-9319
| |
Collapse
|
49
|
Malouf C, Ottersbach K. Molecular processes involved in B cell acute lymphoblastic leukaemia. Cell Mol Life Sci 2018; 75:417-446. [PMID: 28819864 PMCID: PMC5765206 DOI: 10.1007/s00018-017-2620-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 08/01/2017] [Accepted: 08/04/2017] [Indexed: 12/19/2022]
Abstract
B cell leukaemia is one of the most frequent malignancies in the paediatric population, but also affects a significant proportion of adults in developed countries. The majority of infant and paediatric cases initiate the process of leukaemogenesis during foetal development (in utero) through the formation of a chromosomal translocation or the acquisition/deletion of genetic material (hyperdiploidy or hypodiploidy, respectively). This first genetic insult is the major determinant for the prognosis and therapeutic outcome of patients. B cell leukaemia in adults displays similar molecular features as its paediatric counterpart. However, since this disease is highly represented in the infant and paediatric population, this review will focus on this demographic group and summarise the biological, clinical and epidemiological knowledge on B cell acute lymphoblastic leukaemia of four well characterised subtypes: t(4;11) MLL-AF4, t(12;21) ETV6-RUNX1, t(1;19) E2A-PBX1 and t(9;22) BCR-ABL1.
Collapse
Affiliation(s)
- Camille Malouf
- MRC Centre for Regenerative Medicine, The University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Katrin Ottersbach
- MRC Centre for Regenerative Medicine, The University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
50
|
Castelli G, Pelosi E, Testa U. Targeting histone methyltransferase and demethylase in acute myeloid leukemia therapy. Onco Targets Ther 2017; 11:131-155. [PMID: 29343972 PMCID: PMC5749389 DOI: 10.2147/ott.s145971] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clonal disorder of myeloid progenitors characterized by the acquisition of chromosomal abnormalities, somatic mutations, and epigenetic changes that determine a consistent degree of biological and clinical heterogeneity. Advances in genomic technologies have increasingly shown the complexity and heterogeneity of genetic and epigenetic alterations in AML. Among the genetic alterations occurring in AML, frequent are the genetic alterations at the level of various genes involved in the epigenetic control of the DNA methylome and histone methylome. In fact, genes involved in DNA demethylation (such as DNMT3A, TET2, IDH1, and IDH2) or histone methylation and demethylation (EZH2, MLL, DOT1L) are frequently mutated in primary and secondary AML. Furthermore, some histone demethylases, such as LSD1, are frequently overexpressed in AML. These observations have strongly supported a major role of dysregulated epigenetic regulatory processes in leukemia onset and development. This conclusion was further supported by the observation that mutations in genes encoding epigenetic modifiers, such as DMT3A, ASXL1, TET2, IDH1, and IDH2, are usually acquired early and are present in the founding leukemic clone. These observations have contributed to development of the idea that targeting epigenetic abnormalities could represent a potentially promising strategy for the development of innovative treatments of AML. In this review, we analyze those proteins and their inhibitors that have already reached the first stages of clinical trials in AML, namely the histone methyltransferase DOT1L, the demethylase LSD1, and the MLL-interacting protein menin.
Collapse
Affiliation(s)
- Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| | - Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|