1
|
Zattoni J, Vottero P, Carena G, Uliveto C, Pozzati G, Morabito B, Gitari E, Tuszynski J, Aminpour M. A comprehensive primer and review of PROTACs and their In Silico design. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 264:108687. [PMID: 40058081 DOI: 10.1016/j.cmpb.2025.108687] [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: 10/29/2024] [Revised: 01/28/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025]
Abstract
The cutting-edge technique of Proteolysis Targeting Chimeras, or PROTACs, has gained significant attention as a viable approach for specific protein degradation. This innovative technology has vast potential in fields such as cancer therapy and drug development. The development of effective and specific therapies for a range of diseases is within reach with PROTACs, which can target previously "undruggable" proteins while circumventing the off-target effects of conventional small molecule inhibitors. This manuscript aims to discuss the application of in silico techniques to the design of these groundbreaking molecules and develop PROTAC complexes, in order to identify potential PROTAC candidates with favorable drug-like properties. Additionally, this manuscript reviews the strengths and weaknesses of these methods to demonstrate their utility and highlights the challenges and future prospects of in silico PROTAC design. The present review provides a valuable and beginner-friendly resource for researchers and drug developers interested in using in silico methods for PROTAC design, specifically ternary structure prediction.
Collapse
Affiliation(s)
- Jacopo Zattoni
- Department of Biomedical Engineering, University of Alberta, Edmonton, T6G 1Z2, Canada
| | - Paola Vottero
- Department of Biomedical Engineering, University of Alberta, Edmonton, T6G 1Z2, Canada
| | - Gea Carena
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Chiara Uliveto
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Giulia Pozzati
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Benedetta Morabito
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Ebenezea Gitari
- Department of Biochemistry, University of Alberta, Edmonton, T6G 1Z2, Canada
| | - Jack Tuszynski
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; Department of Physics, University of Alberta, 11335 Saskatchewan Dr NW, Edmonton, T6G 2M9, Canada
| | - Maral Aminpour
- Department of Biomedical Engineering, University of Alberta, Edmonton, T6G 1Z2, Canada.
| |
Collapse
|
2
|
Zhang Y, Liu H, Zhen W, Jiang T, Cui J. Advancement of drugs conjugated with GalNAc in the targeted delivery to hepatocytes based on asialoglycoprotein receptor. Carbohydr Res 2025; 552:109426. [PMID: 40068307 DOI: 10.1016/j.carres.2025.109426] [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: 12/03/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 04/22/2025]
Abstract
The asialoglycoprotein receptor (ASGPR) is specifically expressed in hepatocytes. Sugar molecules, such as asialoglycoprotein, galactose, galactosamine, and N-acetyl galactosamine (GalNAc), have a high affinity for ASGPR. This review summarizes the structure of ASGPR, the distribution of this molecule in different cells, and the factors influencing the binding of GalNAc to ASGPR. We introduce the application of GalNAc in targeted delivery into hepatocytes by forming conjugated compounds with RNAs and small molecules, and the standard methods for synthesizing GalNAc are also briefly presented. This is to provide an overview of the current research on GalNAc and to shed light on the design of the new GalNAc.
Collapse
Affiliation(s)
- Yafang Zhang
- Baoding Key Laboratory for Precision Diagnosis and Treatment of Infectious Diseases in Children, Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, 071000, Hebei Province, China
| | - Hongliang Liu
- Pharmaron Beijing Co., Ltd. (China), Beijing, 100176, China
| | - Weina Zhen
- Baoding Key Laboratory for Precision Diagnosis and Treatment of Infectious Diseases in Children, Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, 071000, Hebei Province, China
| | - Tingting Jiang
- Baoding Key Laboratory for Precision Diagnosis and Treatment of Infectious Diseases in Children, Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, 071000, Hebei Province, China
| | - Jingxuan Cui
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China.
| |
Collapse
|
3
|
M S, Joga R, Gandhi K, Yerram S, Raghuvanshi RS, Srivastava S. Exploring the clinical trials, regulatory insights, and challenges of PROTACs in oncology. Semin Oncol 2025; 52:152339. [PMID: 40253775 DOI: 10.1016/j.seminoncol.2025.152339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 04/22/2025]
Abstract
While various targeted therapies exist for cancer, resistance mechanisms remain a significant challenge. Recent advancements in cancer treatment have led to the emergence of proteolysis-targeting chimeras (PROTACs), a promising technology utilizing hetero-bifunctional molecules to target and degrade proteins implicated in cancer progression through the ubiquitin-proteasome system (UPS). PROTACs offer a novel approach, with recent studies and clinical trials demonstrating promising outcomes in degrading endogenous proteins linked to cancer. This work explores classification, regulatory approvals, and ongoing clinical trials of PROTAC technology in cancer management. It emphasizes the importance of regulatory compliance to expedite approvals from relevant authorities. It also highlights challenges and opportunities associated with their implementation. Despite these preliminary efforts, PROTACs show immense potential in effectively addressing cancer. Their ability to target specific proteins for degradation represents a significant advancement in cancer therapeutics, offering new hope for improved outcomes in patient care.
Collapse
Affiliation(s)
- Sowndharya M
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Ramesh Joga
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Kajal Gandhi
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Sravani Yerram
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Rajeev Singh Raghuvanshi
- Central Drug Standard Control Organization (CDSCO), Ministry of Health & Family Welfare, Government of India, New Delhi, India
| | - Saurabh Srivastava
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India; Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India.
| |
Collapse
|
4
|
Zhong Y, Kim H, Qian C, Xie L, Chen X, Xiong Y, Hu J, Chen M, Guccione E, Shen Y, Jin J. Discovery of a Potent and Selective Protein Arginine Methyltransferase 5 (PRMT5) PROTAC Degrader. J Med Chem 2025. [PMID: 40232306 DOI: 10.1021/acs.jmedchem.5c00198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Protein arginine methyltransferase 5 (PRMT5) plays crucial roles in the regulation of various biological processes through the mono- and symmetric dimethylation of protein substrates. PRMT5 is overexpressed in various human cancers and its overexpression is associated with poor prognosis. We previously reported the first-in-class PRMT5 degrader, MS4322, which is also the only von Hippel-Lindau (VHL)-recruiting PRMT5 degrader to date. Here, we performed structure-activity relationship (SAR) studies exploring various linkers and ligands of VHL and PRMT5, which resulted in the best-in-class PRMT5 degrader, MS115 (compound 10). Compound 10 potently and selectively degraded PRMT5 and its coactivator, MEP50, in concentration-, time-, and ubiquitin-proteasome system-dependent manners. It displayed much improved PRMT5/MEP50 degradation potency over MS4322, which translated to better antiproliferative effect in both breast and prostate cancer cells. Overall, we discovered a highly potent and selective PRMT5/MEP50 complex degrader, which is an invaluable chemical biology tool and a potential cancer therapeutic.
Collapse
Affiliation(s)
- Yue Zhong
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Huensuk Kim
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Chao Qian
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Ling Xie
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Xian Chen
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Yan Xiong
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jacqueline Hu
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Matthew Chen
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Ernesto Guccione
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Yudao Shen
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
5
|
Bhole RP, Labhade S, Gurav SS. Conquering PROTAC molecular design and drugability. Bioanalysis 2025; 17:455-470. [PMID: 40114295 DOI: 10.1080/17576180.2025.2481021] [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: 12/17/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
PROTACs are reshaping drug discovery by enabling targeted protein degradation, overcoming the limitations of traditional inhibitors, and addressing previously "undruggable" proteins. The present perspective explores advancements in PROTAC molecular design, focusing on ligand discovery, E3 ligase recruitment, and ternary complex optimization. Integrating AI-driven modeling, FBDD, and SBDD accelerates PROTAC development. In contrast, emerging innovations, such as PHOTACs, hypoxia-responsive systems, and Ab-PROTACs, enhance precision and reduce systemic toxicity. Clinical successes, including ARV-110 for castration-resistant prostate cancer and ARV-471 for breast cancer, exemplify their ability to overcome resistance and provide durable effects. PROTACs are expanding into neurodegenerative diseases and rare conditions, highlighting their versatility. By addressing challenges in pharmacokinetics, safety, and scalability, PROTACs are poised to revolutionize precision medicine. This article presents a forward-looking perspective on conquering the molecular design and drugability of PROTACs, paving the path for transformative therapies.
Collapse
Affiliation(s)
- Ritesh P Bhole
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India
- Dr. D. Y. Patil Vidyapeeth, Dr. D. Y. Patil Dental College Hospital and Research Centre, Pimpri, Pune, India
| | - Sonali Labhade
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India
| | - Shilendra S Gurav
- Department of Pharmacognosy, Goa College of Pharmacy, Goa University, Panaji, India
| |
Collapse
|
6
|
Katerji M, Bergman KL, Lindberg E, Rubin MR, Funk AL, Woodroofe CC, Nyswaner K, Karpińska K, Serwa R, Marusiak A, Swenson RE, Brognard JF. Discovery of potent and selective PROTACs for the protein kinase LZK for the treatment of head and neck cancer. J Biol Chem 2025:108452. [PMID: 40157536 DOI: 10.1016/j.jbc.2025.108452] [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: 01/28/2025] [Revised: 02/28/2025] [Accepted: 03/12/2025] [Indexed: 04/01/2025] Open
Abstract
Leucine zipper-bearing kinase (LZK) is overexpressed in 20% of head and neck squamous cell carcinoma (HNSCC) cases and has emerged as a promising therapeutic target in this cancer subtype. LZK promotes HNSCC survival and proliferation by stabilizing c-MYC and GOF-p53 in kinase-dependent and -independent manners, respectively. Herein, we developed a new series of LZK degraders utilizing proteolysis-targeting chimera (PROTAC) technology by modulating the linker region or LZK warhead of LZK-targeting PROTAC-21A, previously developed by our lab. Among the 27 PROTACs synthesized and tested, PROTAC 17 was found to be the most potent, degrading LZK at 250 nM and suppressing HNSCC viability at 500 nM. In summary our lead PROTAC effectively targeted LZK for proteasomal degradation and inhibited oncogenic activity in HNSCC cell lines with amplified LZK.
Collapse
Affiliation(s)
- Meghri Katerji
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD, 21702, USA
| | - Knickole L Bergman
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD, 21702, USA
| | - Eric Lindberg
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, 20850, USA
| | - Maxine R Rubin
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD, 21702, USA
| | - Amy L Funk
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD, 21702, USA
| | - Carolyn C Woodroofe
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, 20850, USA
| | - Katherine Nyswaner
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD, 21702, USA
| | - Kamila Karpińska
- Laboratory of Molecular OncoSignalling, IMol Polish Academy of Sciences, Warsaw, Poland
| | - Remigiusz Serwa
- Proteomic Core Facility, IMol Polish Academy of Sciences, Warsaw, Poland
| | - Anna Marusiak
- Laboratory of Molecular OncoSignalling, IMol Polish Academy of Sciences, Warsaw, Poland
| | - Rolf E Swenson
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, 20850, USA.
| | - John F Brognard
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD, 21702, USA.
| |
Collapse
|
7
|
Dutta R, Devarajan A, Talluri A, Das R, Thayumanavan S. Dual-Action-Only PROTACs. J Am Chem Soc 2025; 147:9074-9078. [PMID: 40063962 DOI: 10.1021/jacs.5c00131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Proteolysis targeting chimera (PROTAC)-based degraders are highly potent pseudocatalytic drugs, but on-target off-site homing could yield undesirable consequences. We report here a generalizable AND-logic gated PROTAC, where the concurrent presence of two different disease-relevant endogenous stimuli liberates an active protein degrader. We design Dual-Action-Only PROTAC (DAO-PROTAC) molecules that are dormant and can only be activated in the presence of both hypoxia and cathepsin-L to degrade the protein of interest (POI). We also show that the dormancy of DAO-PROTACs translates to considerable mitigation of cytotoxicity, demonstrating the potential advantages over the corresponding free PROTAC and single-stimulus triggerable pro-PROTACs.
Collapse
Affiliation(s)
- Ranit Dutta
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Anirudh Devarajan
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Amelia Talluri
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Ritam Das
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| |
Collapse
|
8
|
Tregnago C, Benetton M, Ries RE, Peplinski JH, Alonzo TA, Stirewalt D, Othus M, Duployez N, Sonneveld E, Abrahamsson J, Fogelstrand L, von Neuhoff N, Hasle H, Reinhardt D, Meshinchi S, Locatelli F, Pigazzi M. Influence of Nucleophosmin ( NPM1) Genotypes on Outcome of Patients With AML: An AIEOP-BFM and COG-SWOG Intergroup Collaboration. J Clin Oncol 2025; 43:972-984. [PMID: 39621969 DOI: 10.1200/jco-24-01715] [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: 08/05/2024] [Revised: 09/25/2024] [Accepted: 10/03/2024] [Indexed: 03/01/2025] Open
Abstract
PURPOSE Several genomic subsets of NPM1 mutations with varying sequences (type A, B, D, etc) have been identified. Despite molecular heterogeneity, NPM1 mutations cumulatively portend a more favorable outcome, but biology and prognostic implications of different genomic subsets have not been extensively studied. In this multicentric study, we investigated the impact of NPM1 genotypes on patient's outcomes and interrogated the underlying biology of the different subtypes. MATERIALS AND METHODS Of more than 4,000 patients enrolled in multiple pediatric cooperative (AIEOP, BFM, ELAM02, NOPHO, DCOG, and COG trials), or adult (SWOG) trials, 348 pediatric and 75 adult AML patients with known NPM1 genotype and available outcome were selected for this study. Diverse NPM1 variants were correlated with the probabilities of overall survival (OS) and event-free survival. Nuclear localization and translational efficiency of the NPM1 variants was studied. RESULTS Evaluation of clinical outcome on the basis of NPM1 genotypes showed that patients with type A, B, and other rare variants had similarly favorable outcomes, whereas those with type D had a significantly worse outcome (OS of 63% for type D v 86% for type non-D, P = .005). Multivariate analysis confirmed type D as an independent prognostic factor associated with inferior OS (hazard ratio, 3; P = .005). In vitro, we demonstrated that in type D versus type A synonymous variants, codon optimality plays major roles in determining gene expression levels, and translation efficiency, which resulted in a more expressed NPM1-D mRNA and protein, mediating peculiar mitochondrial gene expression. CONCLUSION The evaluation of specific NPM1 genotypes identified AML patients with type D mutations being significantly associated with inferior outcomes, suggesting a reclassification of D cases to higher-risk groups.
Collapse
Affiliation(s)
- Claudia Tregnago
- Department of Women's and Children's Health, Onco-hematology Lab and Clinic, University of Padova, Padova, Italy
| | - Maddalena Benetton
- Department of Women's and Children's Health, Onco-hematology Lab and Clinic, University of Padova, Padova, Italy
| | - Rhonda E Ries
- Translational Sciences and Therapeutics, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jack H Peplinski
- Translational Sciences and Therapeutics, Fred Hutchinson Cancer Center, Seattle, WA
| | | | - Derek Stirewalt
- Translational Sciences and Therapeutics, Fred Hutchinson Cancer Center, Seattle, WA
| | - Megan Othus
- SWOG Statistics and Data Management Center, Fred Hutchinson Cancer Center, Seattle, WA
| | - Nicolas Duployez
- Laboratory of Hematology, Lille University Hospital, Lille, France
| | - Edwin Sonneveld
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Jonas Abrahamsson
- Institution for Clinical Sciences, Department of Pediatrics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Linda Fogelstrand
- Department of Laboratory Medicine, Sahlgrenska Academy University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Nils von Neuhoff
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Henrik Hasle
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Dirk Reinhardt
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Essen, Germany
| | - Soheil Meshinchi
- Translational Sciences and Therapeutics, Fred Hutchinson Cancer Center, Seattle, WA
| | - Franco Locatelli
- Department of Pediatric Hematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Catholic University of the Sacred Heart, Rome, Italy
| | - Martina Pigazzi
- Department of Women's and Children's Health, Onco-hematology Lab and Clinic, University of Padova, Padova, Italy
- Foundation Istituto Ricerca Pediatrica (IRP), Padova, Italy
| |
Collapse
|
9
|
Bhatia N, Kumar S, Goyal LD, Thareja S. Optimizing selective estrogen receptor degraders for anticancer drug development. Future Med Chem 2025; 17:637-640. [PMID: 40008677 PMCID: PMC11938951 DOI: 10.1080/17568919.2025.2467615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Affiliation(s)
- Neha Bhatia
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences Central University of Punjab, Bathinda, India
| | - Shashank Kumar
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, India
| | - Lajya Devi Goyal
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, Bathinda, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences Central University of Punjab, Bathinda, India
| |
Collapse
|
10
|
Miletić N, Weckesser J, Mosler T, Rathore R, Hoffmann ME, Gehrtz P, Schlesiger S, Hartung IV, Berner N, Wilhelm S, Müller J, Adhikari B, Němec V, Sivashanmugam SA, Elson L, Holzmann H, Schwalm MP, Hoffmann L, Abdul Azeez KR, Müller S, Kuster B, Wolf E, Đikić I, Knapp S. Workflow for E3 Ligase Ligand Validation for PROTAC Development. ACS Chem Biol 2025; 20:507-521. [PMID: 39932098 PMCID: PMC11851430 DOI: 10.1021/acschembio.4c00812] [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: 12/01/2024] [Revised: 01/20/2025] [Accepted: 02/03/2025] [Indexed: 02/22/2025]
Abstract
Proteolysis targeting chimeras (PROTACs) have gained considerable attention as a new modality in drug discovery. The development of PROTACs has been mainly focused on using CRBN (Cereblon) and VHL (Von Hippel-Lindau ligase) E3 ligase ligands. However, the considerable size of the human E3 ligase family, newly developed E3 ligase ligands, and the favorable druggability of some E3 ligase families hold the promise that novel degraders with unique pharmacological properties will be designed in the future using this large E3 ligase space. Here, we developed a workflow aiming to improve and streamline the evaluation of E3 ligase ligand efficiency for PROTAC development and the assessment of the corresponding "degradable" target space using broad-spectrum kinase inhibitors and the well-established VHL ligand VH032 as a validation system. Our study revealed VH032 linker attachment points that are highly efficient for kinase degradation as well as some of the pitfalls when using protein degradation as a readout. For instance, cytotoxicity was identified as a major mechanism leading to PROTAC- and VHL-independent kinase degradation. The combination of E3 ligase ligand negative controls, competition by kinase parent compounds, and neddylation and proteasome inhibitors was essential to distinguish between VHL-dependent and -independent kinase degradation events. We share here the findings and limitations of our study and hope that this study will provide guidance for future evaluations of new E3 ligase ligand systems for degrader development.
Collapse
Affiliation(s)
- Nebojša Miletić
- Institute
of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium (SGC), Buchmann Institute
for Life Sciences, Max-von-Laue-Str.
15, 60438 Frankfurt
am Main, Germany
| | - Janik Weckesser
- Institute
of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium (SGC), Buchmann Institute
for Life Sciences, Max-von-Laue-Str.
15, 60438 Frankfurt
am Main, Germany
| | - Thorsten Mosler
- Institute
of Biochemistry II, School of Medicine, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - Rajeshwari Rathore
- Institute
of Biochemistry II, School of Medicine, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - Marina E. Hoffmann
- Institute
of Biochemistry II, School of Medicine, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - Paul Gehrtz
- Medicinal
Chemistry, Global Research & Development, Merck Healthcare KGaA, 64293 Darmstadt, Germany
| | - Sarah Schlesiger
- Medicinal
Chemistry, Global Research & Development, Merck Healthcare KGaA, 64293 Darmstadt, Germany
| | - Ingo V. Hartung
- Medicinal
Chemistry, Global Research & Development, Merck Healthcare KGaA, 64293 Darmstadt, Germany
| | - Nicola Berner
- Chair
of Proteomics and Bioanalytics, Technical
University of Munich, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany
- German Cancer
Consortium (DKTK), partner site Munich, a partnership between DKFZ
and University Center Technical University of Munich, Frankfurt am Main 60590, Germany
| | - Stephanie Wilhelm
- Chair
of Proteomics and Bioanalytics, Technical
University of Munich, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany
| | - Juliane Müller
- Institute
of Biochemistry, University of Kiel, Rudolf-Höber-Str. 1, 24118 Kiel, Germany
| | - Bikash Adhikari
- Institute
of Biochemistry, University of Kiel, Rudolf-Höber-Str. 1, 24118 Kiel, Germany
| | - Václav Němec
- Institute
of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium (SGC), Buchmann Institute
for Life Sciences, Max-von-Laue-Str.
15, 60438 Frankfurt
am Main, Germany
| | - Saran Aswathaman Sivashanmugam
- Institute
of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium (SGC), Buchmann Institute
for Life Sciences, Max-von-Laue-Str.
15, 60438 Frankfurt
am Main, Germany
| | - Lewis Elson
- Institute
of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium (SGC), Buchmann Institute
for Life Sciences, Max-von-Laue-Str.
15, 60438 Frankfurt
am Main, Germany
| | - Hanna Holzmann
- Institute
of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium (SGC), Buchmann Institute
for Life Sciences, Max-von-Laue-Str.
15, 60438 Frankfurt
am Main, Germany
| | - Martin P. Schwalm
- Institute
of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium (SGC), Buchmann Institute
for Life Sciences, Max-von-Laue-Str.
15, 60438 Frankfurt
am Main, Germany
| | - Lasse Hoffmann
- Institute
of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium (SGC), Buchmann Institute
for Life Sciences, Max-von-Laue-Str.
15, 60438 Frankfurt
am Main, Germany
| | - Kamal Rayees Abdul Azeez
- Institute
of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium (SGC), Buchmann Institute
for Life Sciences, Max-von-Laue-Str.
15, 60438 Frankfurt
am Main, Germany
| | - Susanne Müller
- Institute
of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium (SGC), Buchmann Institute
for Life Sciences, Max-von-Laue-Str.
15, 60438 Frankfurt
am Main, Germany
| | - Bernhard Kuster
- Chair
of Proteomics and Bioanalytics, Technical
University of Munich, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany
- German Cancer
Consortium (DKTK), partner site Munich, a partnership between DKFZ
and University Center Technical University of Munich, Frankfurt am Main 60590, Germany
| | - Elmar Wolf
- Institute
of Biochemistry, University of Kiel, Rudolf-Höber-Str. 1, 24118 Kiel, Germany
| | - Ivan Đikić
- Institute
of Biochemistry II, School of Medicine, Goethe University Frankfurt, Frankfurt am Main 60590, Germany
| | - Stefan Knapp
- Institute
of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
- Structural
Genomics Consortium (SGC), Buchmann Institute
for Life Sciences, Max-von-Laue-Str.
15, 60438 Frankfurt
am Main, Germany
- German
Cancer Consortium (DKTK) site Frankfurt/Mainz, Frankfurt am Main 60590, Germany
| |
Collapse
|
11
|
Zhao XZ, Wang W, Mahmud MRA, Agama K, Pommier Y, Burke TR. Application of a bivalent "click" approach to target tyrosyl-DNA phosphodiesterase 1 (TDP1). RSC Med Chem 2025:d4md00824c. [PMID: 39990162 PMCID: PMC11843577 DOI: 10.1039/d4md00824c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/26/2025] [Indexed: 02/25/2025] Open
Abstract
Although inhibiting the DNA repair enzyme tyrosyl-DNA phosphodiesterase 1 (TDP1) synergizes with topoisomerase type I (TOP1) inhibitors in anticancer therapy, development of TDP1 inhibitors has been highly challenging. This may be due to the open and shallow nature of the TDP1 catalytic site and the necessity of competing with a large and highly extended substrate. The toolbox available to chemical biologists for studying TDP1 could be significantly enhanced by introducing the ability to selectively eliminate TDP1 using protein degraders. Our current work starts from phenyl imidazopyridine-based TDP1 inhibitors previously developed from small molecule microarrays (SMMs). Using crystal structures of lead inhibitors bound to TDP1, we designed and synthesized a series of bivalent proteolysis-targeting chimeras (PROTACs). The focus of our current work is to explore synthetic approaches that permit installation of E3 ligase-targeting functionality, while retaining the TDP1 binding. We employed copper-catalyzed azide-alkyne cycloaddition (CuAAC) "click" reactions to assemble PROTAC constituents with 1,2,3-triazole-containing linkers. With the addition of the relatively large parts of the linkers and E3-targeting moieties, we retained the ability to inhibit TDP1. The successful development of TDP1-directed PROTACS would yield a new therapeutic class that could potentially enhance the efficacy and selectivity of TOP1 inhibitors including those used as payloads in antibody drug conjugates (ADCs).
Collapse
Affiliation(s)
- Xue Zhi Zhao
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute Frederick MD USA
| | - Wenjie Wang
- Developmental Therapeutics Branch & Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute Bethesda MD USA
| | - Md Rasel Al Mahmud
- Developmental Therapeutics Branch & Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute Bethesda MD USA
| | - Keli Agama
- Developmental Therapeutics Branch & Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute Bethesda MD USA
| | - Yves Pommier
- Developmental Therapeutics Branch & Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute Bethesda MD USA
| | - Terrence R Burke
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute Frederick MD USA
| |
Collapse
|
12
|
Newman JA, Gavard AE, Imprachim N, Aitkenhead H, Sheppard HE, Te Poele R, Clarke PA, Hossain MA, Temme L, Oh HJ, Wells CI, Davis-Gilbert ZW, Workman P, Gileadi O, Drewry DH. Structural insights into human brachyury DNA recognition and discovery of progressible binders for cancer therapy. Nat Commun 2025; 16:1596. [PMID: 39952925 PMCID: PMC11828899 DOI: 10.1038/s41467-025-56213-1] [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: 05/09/2024] [Accepted: 01/10/2025] [Indexed: 02/17/2025] Open
Abstract
Brachyury is a transcription factor that plays an essential role in tumour growth of the rare bone cancer chordoma and is implicated in other solid tumours. Brachyury is minimally expressed in healthy tissues, making it a potential therapeutic target. Unfortunately, as a ligandless transcription factor, brachyury has historically been considered undruggable. To investigate direct targeting of brachyury by small molecules, we determine the structure of human brachyury both alone and in complex with DNA. The structures provide insights into DNA binding and the context of the chordoma associated G177D variant. We use crystallographic fragment screening to identify hotspots on numerous pockets on the brachyury surface. Finally, we perform follow-up chemistry on fragment hits and describe the progression of a thiazole chemical series into binders with low µM potency. Thus we show that brachyury is ligandable and provide an example of how crystallographic fragment screening may be used to target protein classes that are difficult to address using other approaches.
Collapse
Affiliation(s)
- Joseph A Newman
- Centre for Medicines Discovery, University of Oxford, Oxford, UK.
| | - Angeline E Gavard
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
- Exscientia, Oxford, UK
| | - Nergis Imprachim
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Hazel Aitkenhead
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
- Diamond Light Source Ltd, Didcot, UK
| | - Hadley E Sheppard
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, UK
- Sano Genetics Ltd, Cambridge, UK
| | - Robert Te Poele
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, UK
| | - Paul A Clarke
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, UK
| | - Mohammad Anwar Hossain
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Louisa Temme
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Institute of Pharmacy, University of Hamburg, Hamburg, Germany
| | - Hans J Oh
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Carrow I Wells
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- GlaxoSmithKline, Collegeville, PA, USA
| | - Zachary W Davis-Gilbert
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Paul Workman
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, UK.
| | - Opher Gileadi
- Centre for Medicines Discovery, University of Oxford, Oxford, UK
- SGC Karolinska, Centre for Molecular Medicine, Stockholm, Sweden
| | - David H Drewry
- SGC-UNC, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
13
|
Bonomini A, Mercorelli B, Loregian A. Antiviral strategies against influenza virus: an update on approved and innovative therapeutic approaches. Cell Mol Life Sci 2025; 82:75. [PMID: 39945883 PMCID: PMC11825441 DOI: 10.1007/s00018-025-05611-1] [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: 10/01/2024] [Revised: 01/27/2025] [Accepted: 02/02/2025] [Indexed: 02/16/2025]
Abstract
Influenza viruses still represent a great concern for Public Health by causing yearly seasonal epidemics and occasionally worldwide pandemics. Moreover, spillover events at the animal-human interface are becoming more frequent nowadays, also involving animal species not previously found as reservoirs. To restrict the effects of influenza virus epidemics, especially in at-risk population, and to prepare a drug arsenal for possible future pandemics, researchers worldwide have been working on the development of antiviral strategies since the 80's of the last century. One of the main obstacles is the considerable genomic variability of influenza viruses, which constantly poses the issues of drug-resistance emergence and immune evasion. This review summarizes the approved therapeutics for clinical management of influenza, promising new anti-flu compounds and monoclonal antibodies currently undergoing clinical evaluation, and molecules with efficacy against influenza virus in preclinical studies. Moreover, we discuss some innovative anti-influenza therapeutic approaches such as combination therapies and targeted protein degradation. Given the limited number of drugs approved for influenza treatment, there is a still strong need for novel potent anti-influenza drugs endowed with a high barrier to drug resistance and broad-spectrum activity against influenza viruses of animal origin that may be responsible of future large outbreaks and pandemics.
Collapse
Affiliation(s)
- Anna Bonomini
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, Italy.
- Microbiology and Virology Unit, Padua University Hospital, Padua, Italy.
| |
Collapse
|
14
|
Sun C, Liu S, Lau JW, Yang H, Chen Y, Xing B. Enzyme-Activated Orthogonal Proteolysis Chimeras for Tumor Microenvironment-Responsive Immunomodulation. Angew Chem Int Ed Engl 2025:e202423057. [PMID: 39932237 DOI: 10.1002/anie.202423057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Indexed: 02/20/2025]
Abstract
Precise modulation of dynamic and complex tumor microenvironment (TME) to disrupt tumorigenesis and reshape intratumoral immune infiltration has emerged as promising approaches for enhanced cancer therapy. Among recent innovations, proteolysis-targeting chimeras (PROTACs) represent a burgeoning chemical knockdown technology capable of degrading oncogenic protein homeostasis and inducing dynamic alternations within carcinoma settings, offering potential for antitumor manipulation. However, achieving selectivity in PROTACs that respond to disease environmental stimulation and precisely perturb on-target proteins remains challenging. The multi-step synthesis and limited permeability, attributed to high-molecular-weight and heterobifunctional structures, further hinder their in vivo efficacy. Herein, we present a unique TME-responsive enzyme-activated clickable PROTACs, which features a short peptide-tagged pomalidomide derivative to undergo tumor-specific cleavage by cathepsin protease to induce orthogonal crosslinking of the exposed cysteine with 2-cyanobenzothiazole-labeled epigenetic protein-ligand JQ1, facilitating in situ degrader formation within tumor regions only. Systematic protein profiling and proteomic analysis revealed that such TME-specific clickable-PROTACs not only selectively eliminate epigenetic proteins without tedious pre-synthesis to bridge disparate small-molecule bi-warhead fragments, but also demonstrated superior tumor penetration compared to conventional high-molecular-weight PROTACs. Importantly, these clickable-PROTACs efficiently downregulated immune checkpoint programmed death-ligand 1 (PD-L1) both in vitro and in vivo, remodeling TME for enhanced therapeutics, especially in anti-tumoral immunomodulation.
Collapse
Affiliation(s)
- Caixia Sun
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Songhan Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Jun Wei Lau
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| | - Hanyu Yang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Yun Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Bengang Xing
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hung Hom, Kowloon, Hong Kong SAR, China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| |
Collapse
|
15
|
Bhole RP, Kute P, Gurav SS. PROTACs in the treatment of viral diseases. Future Med Chem 2025; 17:267-269. [PMID: 39814466 PMCID: PMC11792865 DOI: 10.1080/17568919.2025.2453418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025] Open
Affiliation(s)
- Ritesh P. Bhole
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India
- Dr. D. Y. Patil Vidyapeeth, Dr. D. Y. Patil Dental College Hospital and Research Centre, Pimpri, India
| | - Payal Kute
- SSR College of Pharmacy, Silvassa, India
| | - Shilendra S. Gurav
- Department of Pharmacognosy, Goa College of Pharmacy, Goa University, Panaji, India
| |
Collapse
|
16
|
Mahajan A, Panzade G, Bhuniya T, Das P, Bhattacharjee B, Das S, Chowdhury A, Chakraborty K, Guha S, Samant A, Dey A, Ghosh S. Revolutionizing lung cancer treatment: Introducing PROTAC therapy as a novel paradigm in targeted therapeutics. Curr Probl Cancer 2025; 54:101172. [PMID: 39731828 DOI: 10.1016/j.currproblcancer.2024.101172] [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: 06/12/2024] [Revised: 09/23/2024] [Accepted: 12/18/2024] [Indexed: 12/30/2024]
Abstract
This comprehensive review explores the transformative potential of PROTAC (Proteolysis-Targeting Chimeras) therapy as a groundbreaking approach in the landscape of lung cancer treatment. The introduction provides a succinct overview of current challenges in lung cancer treatment, emphasizing the significance of targeted therapies. Focusing on PROTAC therapy, the article elucidates its mechanism of action, comparing it with traditional targeted therapies and highlighting the key components and design principles of PROTAC molecules. In the context of lung cancer, the review meticulously summarizes preclinical evidence, emphasizing efficacy and specificity gleaned from studies evaluating PROTAC therapy. It delves into the implications of this preclinical data, discussing potential advantages over existing targeted therapies. An update on ongoing clinical trials involving PROTAC therapy for lung cancer offers a snapshot of the current progress, with a summary of key outcomes and advancements in early-phase trials. The mechanistic insights into PROTAC therapy's impact on lung cancer cells are explored, alongside a discussion on potential biomarkers for patient stratification and response prediction. The influence of tumor heterogeneity on PROTAC therapy outcomes is also addressed. Safety and tolerability assessments, encompassing preclinical and clinical studies, are comprehensively evaluated, including a comparative analysis with traditional targeted therapies and strategies to mitigate side effects. Looking forward, the article discusses the future perspectives of PROTAC therapy in lung cancer treatment and addresses ongoing challenges, providing a nuanced exploration of potential combination therapies and synergistic approaches. In conclusion, the review summarizes key findings and insights, underscoring the tremendous potential of PROTAC therapy as a promising and innovative avenue in pursuing more effective lung cancer treatments.
Collapse
Affiliation(s)
- Atharva Mahajan
- Advance Centre for Treatment, Research and Education in Cancer (ACTREC), Navi Mumbai, Mumbai, Maharashtra, India
| | - Gauri Panzade
- Advance Centre for Treatment, Research and Education in Cancer (ACTREC), Navi Mumbai, Mumbai, Maharashtra, India
| | - Tiyasa Bhuniya
- Department of Biotechnology, National Institute of Technology Durgapur, West Bengal, India
| | - Purbasha Das
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| | | | - Sagnik Das
- Department of Microbiology, St Xavier's College (autonomous) Kolkata, West Bengal, India
| | - Ankita Chowdhury
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Delhi, India
| | - Kashmira Chakraborty
- Department of Chemistry and Chemical Biology, Indian Institute of Technology Dhanbad, Jharkhand, India
| | - Sudeepta Guha
- Department of Chemistry and Chemical Biology, Indian Institute of Technology Dhanbad, Jharkhand, India
| | - Anushka Samant
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Orissa, India
| | - Anuvab Dey
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.
| | - Subhrojyoti Ghosh
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India.
| |
Collapse
|
17
|
Ibrahim S, Umer Khan M, Khurram I, Rehman R, Rauf A, Ahmad Z, Aljohani ASM, Al Abdulmonem W, Quradha MM. Navigating PROTACs in Cancer Therapy: Advancements, Challenges, and Future Horizons. Food Sci Nutr 2025; 13:e70011. [PMID: 39898116 PMCID: PMC11786021 DOI: 10.1002/fsn3.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
Proteolysis Targeting Chimeras (PROTACs) have revolutionized cancer therapy by offering a selective and innovative approach to degrade key oncogenic proteins associated with various malignancies. These hybrid molecules exploit the ubiquitin-proteasome system, facilitating the degradation of target proteins through an event-driven mechanism, thereby overcoming drug resistance and enhancing selectivity. With diverse targets including androgen receptors, BTK, estrogen receptors, BET proteins, and BRAF, PROTACs offer a versatile strategy for personalized cancer treatment. Advantages of PROTACs over traditional small molecule inhibitors include their ability to operate at lower concentrations, catalyzing the degradation of multiple proteins of interest with reduced cytotoxicity. Notably, PROTACs address challenges associated with traditionally "undruggable" targets, expanding the therapeutic landscape of cancer therapy. Ongoing preclinical and clinical studies highlight the transformative potential of PROTACs, with promising results in prostate, breast, lung, melanoma, and colorectal cancers. Despite their potential, challenges persist in optimizing physicochemical properties and enhancing bioavailability. Further research is needed to refine PROTAC design and address complexities in molecule development. Nevertheless, the development of oral androgen receptor PROTACs represents a significant milestone, demonstrating the feasibility and efficacy of this innovative therapeutic approach. This review provides a comprehensive overview of PROTACs in cancer therapy, emphasizing their mechanism of action, advantages, and challenges. As PROTAC research progresses, continued exploration in both preclinical and clinical settings will be crucial to unlocking their full therapeutic potential and shaping the future of personalized cancer treatment.
Collapse
Affiliation(s)
- Saooda Ibrahim
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
- Centre for Applied Molecular BiologyUniversity of the PunjabLahorePakistan
| | - Muhammad Umer Khan
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| | - Iqra Khurram
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
- Centre for Applied Molecular BiologyUniversity of the PunjabLahorePakistan
| | - Raima Rehman
- Institute of Molecular Biology and BiotechnologyThe University of LahoreLahorePakistan
| | - Abdur Rauf
- Department of ChemistryUniversity of SwabiSwabiKhyber PakhtunkhwaPakistan
| | - Zubair Ahmad
- Department of ChemistryUniversity of SwabiSwabiKhyber PakhtunkhwaPakistan
| | - Abdullah S. M. Aljohani
- Department of Medical BiosciencesCollege of Veterinary Medicine, Qassim UniversityBuraydahSaudi Arabia
| | - Waleed Al Abdulmonem
- Department of PathologyCollege of Medicine, Qassim UniversityBuraydahSaudi Arabia
| | | |
Collapse
|
18
|
Fu MJ, Jin H, Wang SP, Shen L, Liu HM, Liu Y, Zheng YC, Dai XJ. Unleashing the Power of Covalent Drugs for Protein Degradation. Med Res Rev 2025. [PMID: 39834319 DOI: 10.1002/med.22101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/28/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Targeted protein degradation (TPD) has emerged as a significant therapeutic approach for a variety of diseases, including cancer. Advances in TPD techniques, such as molecular glue (MG) and lysosome-dependent strategies, have shown substantial progress since the inception of the first PROTAC in 2001. The PROTAC methodology represents the forefront of TPD technology, with ongoing evaluation in more than 20 clinical trials for the treatment of diverse medical conditions. Two prominent PROTACs, ARV-471 and ARV-110, are currently undergoing phase III and II clinical trials, respectively. Traditional PROTACs are encountering obstacles such as limited binding affinity and a restricted range of E3 ligase ligands for facilitating the protein of interest (POI) degradation. Covalent medicines offer the potential to enhance PROTAC efficacy by enabling the targeting of previously considered "undruggable" shallow binding sites. Strategic alterations allow PROTAC to establish covalent connections with particular target proteins, including Kirsten rat sarcoma viral oncogene homolog (KRAS), Bruton's tyrosine kinase (BTK), epidermal growth factor receptor (EGFR), as well as E3 ligases such as DDB1 and CUL4 associated factor 16 (DCAF16) and Kelch-like ECH-associated protein 1 (Keap1). The concept of covalent degradation has also been utilized in various new forms of degraders, including covalent molecule glue (MG), in-cell click-formed proteolysis targeting chimera (CLIPTAC), HaloPROTAC, lysosome-targeting chimera (LYTAC) and GlueTAC. This review focuses on recent advancements in covalent degraders beyond covalent PROTACs and examines obstacles and future directions pertinent to this field.
Collapse
Affiliation(s)
- Meng-Jie Fu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Hang Jin
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shao-Peng Wang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Liang Shen
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Hong-Min Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Liu
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi-Chao Zheng
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xing-Jie Dai
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardio-Cerebrovascular Drug, China Meheco Topfond Pharmaceutical Company, Zhumadian, Henan, China
| |
Collapse
|
19
|
London N. Covalent Proximity Inducers. Chem Rev 2025; 125:326-368. [PMID: 39692621 PMCID: PMC11719315 DOI: 10.1021/acs.chemrev.4c00570] [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: 07/31/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
Molecules that are able to induce proximity between two proteins are finding ever increasing applications in chemical biology and drug discovery. The ability to introduce an electrophile and make such proximity inducers covalent can offer improved properties such as selectivity, potency, duration of action, and reduced molecular size. This concept has been heavily explored in the context of targeted degradation in particular for bivalent molecules, but recently, additional applications are reported in other contexts, as well as for monovalent molecular glues. This is a comprehensive review of reported covalent proximity inducers, aiming to identify common trends and current gaps in their discovery and application.
Collapse
Affiliation(s)
- Nir London
- Department
of Chemical and Structural Biology, The
Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
20
|
Wozniak M, Czyz M. Exploring oncogenic roles and clinical significance of EZH2: focus on non-canonical activities. Ther Adv Med Oncol 2025; 17:17588359241306026. [PMID: 39776536 PMCID: PMC11705335 DOI: 10.1177/17588359241306026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
The enhancer of zeste homolog 2 (EZH2) is a catalytic component of Polycomb repressive complex 2 (PRC2) mediating the methylation of histone 3 lysine 27 (H3K27me3) and hence the epigenetic repression of target genes, known as canonical function. Growing evidence indicates that EZH2 has non-canonical roles that are exerted as PRC2-dependent and PRC2-independent methylation of non-histone proteins, and methyltransferase-independent interactions of EZH2 with various proteins contributing to gene expression regulation and alterations in the protein stability. EZH2 is frequently mutated and/or its expression is deregulated in various cancer types. The cancer sensitivity to inhibitors of EZH2 enzymatic activity and state-of-the-art approaches to deplete EZH2 with chemical degraders are discussed. This review also presents the clinical trials in various phases that evaluate the use of EZH2 inhibitors, both as monotherapy and in combination with other agents for the treatment of patients with diverse types of cancers.
Collapse
Affiliation(s)
- Michal Wozniak
- Department of Molecular Biology of Cancer, Medical University of Lodz, Lodz, Poland
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, Mazowiecka 6/8, Lodz 92-215, Poland
| |
Collapse
|
21
|
Chen J, Wu M, Mo J, Hong J, Wang W, Jin Y, Mao X, Liao X, Li K, Yu X, Chen S, Zeng S, Huang W, Xu H, Wu J, Cao J, Zhou Y, Ying M, Zhu C, He Q, Zhang B, Lin N, Dong X, Che J. Auto-RapTAC: A Versatile and Sustainable Platform for the Automated Rapid Synthesis and Evaluation of PROTAC. J Med Chem 2025. [PMID: 39754574 DOI: 10.1021/acs.jmedchem.4c02438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
The tedious synthesis and limited throughput biological evaluation remain a great challenge for discovering new proteolysis targeting chimera (PROTAC). To rapidly identify potential PROTAC lead compounds, we report a platform named Auto-RapTAC. Based on the modular characteristic of the PROTAC molecule, a streamlined workflow that integrates lab automation with "click chemistry" joint building-block libraries was constructed. This facilitates the autonomous generation of a variety of PROTACs, each with distinct linkers and E3 ligase ligands, all stored in biocompatible solutions. The ready-for-screening (R4S) approach, when paired with fluorescence-based assays, enables the efficient assessment of the PROTAC degradation activity in a high-throughput manner. To further test the capability of the platform, we identify six new PROTACs that target CDK2, CDK12, and BCL6 within a mere 8-day time frame for each target. In all, this platform could find broad application not only in discovering new PROTACs but also in the rapid development of novel heterobifunctional modalities.
Collapse
Affiliation(s)
- Jiexuan Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingfei Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jun Mo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ju Hong
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuheng Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinfei Mao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xueyan Liao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kailin Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaoli Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Sikang Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shenxin Zeng
- Center of Safety Evaluation and Research, School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Wenhai Huang
- Center of Safety Evaluation and Research, School of Pharmacy, Hangzhou Medical College, Hangzhou 310013, China
| | - Hongxia Xu
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jian Wu
- College of Computer Science and Technology, Zhejiang University, Hangzhou 310058, China
| | - Ji Cao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yubo Zhou
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Meidan Ying
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chengliang Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Zhang
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310024, China
| | - Nengming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310024, China
| | - Xiaowu Dong
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jinxin Che
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
22
|
Tan M, Li X, Cheng L, Long X, Cao G, Yu S, Ran H, Feng H, Wang H. Augmenting Protein Degradation Capacity of PROTAC through Energy Metabolism Regulation and Targeted Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2412837. [PMID: 39491551 DOI: 10.1002/adma.202412837] [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: 08/28/2024] [Indexed: 11/05/2024]
Abstract
The ubiquitin-proteasome system (UPS) is responsible for degrading over 70-80% of cellular proteins. Consequently, proteolysis-targeting chimeras (PROTACs) are developed to induce the ubiquitination and subsequent degradation of proteins of interest (POIs) by the UPS. To amplify the therapeutic efficacy of PROTACs, energy metabolism regulation is first harnessed to boost UPS function in tumor cells. Proteomic and ubiquitinome analyzes reveal that total ubiquitinated proteins and proteasome activity are significantly increased in 143B and MDA-MB-231 tumor cells following fasting-mimicking diet (FMD) treatment. As a result, the degradation efficiency of PROTACs targeting focal adhesion kinase (FAK-P) or bromodomain-containing protein 4 (BRD4-P) is significantly enhanced in FMD-treated 143B and MDA-MB-231 tumor cells. Then, silica-coated iron oxide nanoparticles are developed modified with tumor cell membranes for targeted delivery of PROTACs. Magnetic resonance imaging (MRI) and fluorescence imaging confirm that nanocarriers significantly improve the delivery efficiency of PROTACs in FMD-treated 143B or MDA-MB-231 tumors. In vivo studies demonstrate that the antitumor efficacy of FAK-P and BRD4-P is greatly augmented when combined with targeted delivery and FMD treatment. Overall, this study presents a strategy to enhance the efficacy of PROTACs in cancer therapy.
Collapse
Affiliation(s)
- Mixiao Tan
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Key Laboratory of Molecular Biology for Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Xiaoyang Li
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Long Cheng
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Xianli Long
- Key Laboratory of Molecular Biology for Infectious Diseases, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Guoliang Cao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Shengji Yu
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Helin Feng
- Department of Orthopedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hai Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
23
|
Wu D, Sun Q, Tang H, Xiao H, Luo J, Ouyang L, Sun Q. Acquired resistance to tyrosine kinase targeted therapy: mechanism and tackling strategies. Drug Resist Updat 2025; 78:101176. [PMID: 39642660 DOI: 10.1016/j.drup.2024.101176] [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: 09/17/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/09/2024]
Abstract
Over the past two decades, tyrosine kinase inhibitors (TKIs) have rapidly emerged as pivotal targeted agents, offering promising therapeutic prospects for patients. However, as the cornerstone of targeted therapies, an increasing number of TKIs have been found to develop acquired resistance during treatment, making the challenge of overcoming this resistance a primary focus of current research. This review comprehensively examines the evolution of TKIs from multiple perspectives, with particular emphasis on the mechanisms underlying acquired resistance, innovative drug design strategies, inherent challenges, and future directions.
Collapse
Affiliation(s)
- Defa Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Qian Sun
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China; West China Medical Publishers, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haolin Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Huan Xiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Jiaxiang Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China.
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China; West China Medical Publishers, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
24
|
Wang K, Qi Z, Guo J, Shen G, Ni X, Jiang S, Zhang K, Wang T, Zhang X. Discovery of small molecules for autophagy-lysosome degradation of immune checkpoint proteins. Eur J Med Chem 2024; 280:116958. [PMID: 39437574 DOI: 10.1016/j.ejmech.2024.116958] [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: 08/08/2024] [Revised: 09/20/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
Targeted protein degradation (TPD) technologies, particularly proteolysis targeting chimeras (PROTACs), have emerged as a promising branch of targeted therapy. Current ubiquitin-proteasome-dependent TPD technologies are limited to targeting intracellular proteins. Although the blockade of immune checkpoints has achieved great clinical success, most immune checkpoints are transmembrane proteins, which are difficult to be ubiquitinated and degraded by PROTACs. Herein, we developed a novel discovery strategy of bifunctional small molecules, which could mediate autophagy-lysosome degradation of immune checkpoints. F-1 was demonstrated to activate the autophagy-lysosome system, and conjugation of F-1 with inhibitors targeting programmed cell death-ligand 1 (PD-L1) or V-domain Ig suppressor of T-cell activation (VISTA) generated a new class of small molecules that effectively induce the degradation of PD-L1 or VISTA in tumor cells. The most promising PD-L1 degrader B3 significantly induced PD-L1 degradation in RKO cells through the autophagy-lysosome system and exhibited good tumor-inhibiting effects in vivo. Our work could expand the development of degraders targeting immune checkpoints and provide a promising discovery strategy for future autophagy-lysosome targeting degradation technology.
Collapse
Affiliation(s)
- Kaizhen Wang
- Department of Biomedical Engineering and Diagnostic Pharmacy, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhihao Qi
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Jiazheng Guo
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Guoqing Shen
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiang Ni
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Sheng Jiang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Kuojun Zhang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Tianyu Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xiangyu Zhang
- Department of Biomedical Engineering and Diagnostic Pharmacy, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
25
|
Cassim A, Dun MD, Gallego-Ortega D, Valdes-Mora F. EZHIP's role in diffuse midline glioma: echoes of oncohistones? Trends Cancer 2024; 10:1095-1105. [PMID: 39343635 DOI: 10.1016/j.trecan.2024.09.002] [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: 04/27/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024]
Abstract
The enhancer of zeste inhibitory protein (EZHIP) is typically expressed during germ cell development and has been classified as a cancer-testis antigen (CTA) in various cancers. In 2020, 4% of diffuse midline gliomas (DMGs) were shown to aberrantly express EZHIP, mirroring the DMG hallmark histone H3 K27M (H3K27M) oncohistone mutation. Similar to H3K27M, EZHIP is a negative regulator of polycomb repressive complex 2 (PRC2), leading to global epigenomic remodeling. In this opinion, we explore the similarities and disparities between H3K27M- and EZHIP-DMGs with a focus on their shared functional hallmark of PRC2 inhibition, their genetic and epigenomic landscapes, plausible differences in the cell of origin, and therapeutic avenues. Upcoming research on EZHIP will help better understand its role in gliomagenesis and DMG therapy.
Collapse
Affiliation(s)
- Afraah Cassim
- Cancer Epigenetic Biology and Therapeutics Laboratory, Children's Cancer Institute, Lowy Cancer Centre, Kensington, New South Wales, Australia; School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, New South Wales, Australia
| | - Matthew D Dun
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine, and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia; Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia; Paediatric Stream, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine, and Wellbeing, Callaghan, New South Wales, Australia
| | - David Gallego-Ortega
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, New South Wales, Australia; School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales Sydney, New South Wales, Australia; Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Fatima Valdes-Mora
- Cancer Epigenetic Biology and Therapeutics Laboratory, Children's Cancer Institute, Lowy Cancer Centre, Kensington, New South Wales, Australia; School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, New South Wales, Australia; School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales Sydney, New South Wales, Australia; Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
| |
Collapse
|
26
|
Raouf YS, Moreno-Yruela C. Slow-Binding and Covalent HDAC Inhibition: A New Paradigm? JACS AU 2024; 4:4148-4161. [PMID: 39610753 PMCID: PMC11600154 DOI: 10.1021/jacsau.4c00828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024]
Abstract
The dysregulated post-translational modification of proteins is an established hallmark of human disease. Through Zn2+-dependent hydrolysis of acyl-lysine modifications, histone deacetylases (HDACs) are key regulators of disease-implicated signaling pathways and tractable drug targets in the clinic. Early targeting of this family of 11 enzymes (HDAC1-11) afforded a first generation of broadly acting inhibitors with medicinal applications in oncology, specifically in cutaneous and peripheral T-cell lymphomas and in multiple myeloma. However, first-generation HDAC inhibitors are often associated with weak-to-modest patient benefits, dose-limited efficacies, pharmacokinetic liabilities, and recurring clinical toxicities. Alternative inhibitor design to target single enzymes and avoid toxic Zn2+-binding moieties have not overcome these limitations. Instead, recent literature has seen a shift toward noncanonical mechanistic approaches focused on slow-binding and covalent inhibition. Such compounds hold the potential of improving the pharmacokinetic and pharmacodynamic profiles of HDAC inhibitors through the extension of the drug-target residence time. This perspective aims to capture this emerging paradigm and discuss its potential to improve the preclinical/clinical outlook of HDAC inhibitors in the coming years.
Collapse
Affiliation(s)
- Yasir S. Raouf
- Department
of Chemistry, United Arab Emirates University, P.O. Box No. 15551 Al Ain, UAE
| | - Carlos Moreno-Yruela
- Laboratory
of Chemistry and Biophysics of Macromolecules (LCBM), Institute of
Chemical Sciences and Engineering (ISIC), School of Basic Sciences, École Polytechnique Fédérale
de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
27
|
Yim J, Park J, Kim G, Lee HH, Chung JS, Jo A, Koh M, Park J. Conditional PROTAC: Recent Strategies for Modulating Targeted Protein Degradation. ChemMedChem 2024; 19:e202400326. [PMID: 38993102 PMCID: PMC11581424 DOI: 10.1002/cmdc.202400326] [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: 04/30/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) have emerged as a promising technology for inducing targeted protein degradation by leveraging the intrinsic ubiquitin-proteasome system (UPS). While the potential druggability of PROTACs toward undruggable proteins has accelerated their rapid development and the wide-range of applications across diverse disease contexts, off-tissue effects and side-effects of PROTACs have recently received attentions to improve their efficacy. To address these issues, spatial or temporal target protein degradation by PROTACs has been spotlighted. In this review, we explore chemical strategies for modulating protein degradation in a cell type-specific (spatio-) and time-specific (temporal-) manner, thereby offering insights for expanding PROTAC applications to overcome the current limitations of target protein degradation strategy.
Collapse
Affiliation(s)
- Junhyeong Yim
- Department of ChemistryKangwon National UniversityChuncheon24341Republic of Korea
- Multidimensional Genomics Research CenterKangwon National UniversityChuncheon24341Republic of Korea
| | - Junyoung Park
- Department of ChemistryKangwon National UniversityChuncheon24341Republic of Korea
- Institute for Molecular Science and Fusion TechnologyKangwon National UniversityChuncheon24341Republic of Korea
| | - Gabin Kim
- Department of Chemistry and ChemistryInstitute for Functional MaterialsPusan National UniversityBusan46241Republic of Korea
| | - Hyung Ho Lee
- Department of UrologyUrological Cancer CenterResearch Institute and Hospital of National Cancer CenterGoyang10408Republic of Korea
| | - Jin Soo Chung
- Department of UrologyUrological Cancer CenterResearch Institute and Hospital of National Cancer CenterGoyang10408Republic of Korea
| | - Ala Jo
- Center for NanomedicineInstitute for Basic ScienceSeoul03722Republic of Korea
| | - Minseob Koh
- Department of Chemistry and ChemistryInstitute for Functional MaterialsPusan National UniversityBusan46241Republic of Korea
| | - Jongmin Park
- Department of ChemistryKangwon National UniversityChuncheon24341Republic of Korea
- Multidimensional Genomics Research CenterKangwon National UniversityChuncheon24341Republic of Korea
- Institute for Molecular Science and Fusion TechnologyKangwon National UniversityChuncheon24341Republic of Korea
| |
Collapse
|
28
|
Zhao M, Ma W, Liang J, Xie Y, Wei T, Zhang M, Qin J, Lao L, Tian R, Wu H, Cheng J, Li M, Liu Y, Hong L, Li G. Design, Synthesis, and Activity Evaluation of BRD4 PROTAC Based on Alkenyl Oxindole-DCAF11 Pair. J Med Chem 2024; 67:19428-19447. [PMID: 39475482 DOI: 10.1021/acs.jmedchem.4c01767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Proteolytic targeting chimera (PROTAC) represent an advanced strategy for targeting undruggable proteins, and the molecular warheads targeting E3 ligases play a crucial role. Recently, we explored an alkenyl oxindole warhead targeting the E3 ligase DCAF11 and sought to validate its potential. In this study, we synthesized a range of BRD4 PROTACs (8a-8o, 14a-14f, 22a-22m) with modified alkenyl oxindole warheads and developed a high-throughput screening system based on high-content imaging. We identified L134 (22a) as a potent BRD4 degrader, achieving BRD4 degradation (Dmax > 98%, DC50 = 7.36 nM) and demonstrating antitumor activity. Mechanically, BRD4 degradation by L134 was mediated through the ubiquitin-proteasome system in a DCAF11-dependent manner. Therefore, this study provides a rapid screening method for effective PROTACs and highlights the PROTAC L134 based on alkenyl oxindole-DCAF11 pair as a promising candidate for treating BRD4-driven cancers.
Collapse
Affiliation(s)
- Man Zhao
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Wenjing Ma
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Jinyi Liang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Yubao Xie
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Tianzi Wei
- Key University Laboratory of Metabolism and Health of Guangdong, Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ming Zhang
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Jiajie Qin
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Lingyin Lao
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Ruilin Tian
- Key University Laboratory of Metabolism and Health of Guangdong, Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Haiqiang Wu
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Jin Cheng
- The Affiliated Wuxi Center for Disease Control and Prevention, Wuxi Center for Disease Control and Prevention, Nanjing Medical University, Wuxi 214023, China
| | - Min Li
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yuyang Liu
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Liang Hong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Guofeng Li
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
29
|
Zhang Y, Wei S, Chen Z, Xu R, Li S, You L, Wu R, Zhang Y, Liao J, Xu X, Song E, Luo M. LncRNA FAISL Inhibits Calpain 2-Mediated Proteolysis of FAK to Promote Progression and Metastasis of Triple Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407493. [PMID: 39287113 PMCID: PMC11558121 DOI: 10.1002/advs.202407493] [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: 07/04/2024] [Revised: 08/22/2024] [Indexed: 09/19/2024]
Abstract
Triple negative breast cancer (TNBC) is the most aggressive subtype in breast tumors. When re-analyzing TCGA breast cancer dataset, we found cell adhesion molecules are highly enriched in differentially expressed genes in TNBC samples, among which Focal Adhesion Kinase (FAK) is most significantly associated with poor survival of TNBC patients. FAK is precisely modulated in the focal adhesion dynamics. To investigate whether lncRNAs regulate FAK signaling, we performed RNA immunoprecipitation sequencing and found FAISL (FAK Interacting and Stabilizing LncRNA) abundantly enriched in FAK-interacting lncRNAs and frequently overexpressed in TCGA TNBC tissues. FAISL promotes TNBC cell adhesion, cytoskeleton spreading, proliferation, and anchor-independent survival. FAISL doesn't affect FAK mRNA but positively regulates FAK protein level by blocking Calpain 2-mediated proteolysis. FAISL interacts with the C-terminus domain of FAK, whereby masks the binding site of Calpain 2 and prevents FAK cleavage. High level of FAISL correlates with FAK expression in tumor tissues and poor prognosis of TNBC patients. A siRNA delivery system targeting FAISL using reduction-responsive nanoparticles effectively inhibits tumor growth and metastasis in TNBC mouse models. Together, these findings uncover a lncRNA-mediated mechanism of regulating FAK proteolysis in the TNBC progression, and highlight the potential of targeting lncRNA FAISL for TNBC treatment.
Collapse
Affiliation(s)
- Yunmei Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
- Breast Tumor CenterSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
| | - Shiyu Wei
- Department of ImmunologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Zhengjie Chen
- Breast Tumor CenterSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
| | - Rui Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
| | - Shu‐Rong Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
| | - Lili You
- Department of EndocrinologySun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhou510120China
| | - Ruiyue Wu
- The First Clinical Medical CollegeLanzhou UniversityLanzhou730000China
| | - Yin Zhang
- Department of Cellular and Molecular Diagnostics CenterSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
| | - Jian‐You Liao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
- Breast Tumor CenterSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
| | - Man‐Li Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationGuangdong‐Hong Kong Joint Laboratory for RNA MedicineSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
- Medical Research CenterSun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhou510120China
| |
Collapse
|
30
|
Goyal A, Murkute SL, Bhowmik S, Prasad CP, Mohapatra P. Belling the "cat": Wnt/β-catenin signaling and its significance in future cancer therapies. Biochim Biophys Acta Rev Cancer 2024; 1879:189195. [PMID: 39413855 DOI: 10.1016/j.bbcan.2024.189195] [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: 05/30/2024] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024]
Abstract
The WNT/β-catenin is among one of the most extensively studied cellular signaling pathways involved in the initiation and progression of several deadly cancers. It is now understood that the WNT/β-catenin signaling, during tumor progression operates in a very complex fashion beyond the earlier assumed simple WNT 'On' or 'Off' mode as it recruits numerous WNT ligands, receptors, transcriptional factors and also cross-talks with other signaling molecules including the noncanonical WNT regulators. WNT/β-catenin signaling molecules are often mutated in different cancers which makes them very challenging to inhibit and sometimes ranks them among the undruggable targets. Furthermore, due to the evolutionary conservation of this pathway, inhibiting WNT/β-catenin has caused significant toxicity in normal cells. These challenges are reflected in clinical trial data, where the use of WNT/β-catenin inhibitors as standalone treatments remains limited. In this review, we have highlighted the crucial functional associations of diverse WNT/β-catenin signaling regulators with cancer progression and the phenotypic switching of tumor cells. Next, we have shed light on the roles of WNT/β-catenin signaling in drug resistance, clonal evolution, tumor heterogeneity, and immune evasion. The present review also focuses on various classes of routine and novel WNT/β-catenin therapeutic regimes while addressing the challenges associated with targeting the regulators of this complex pathway. In the light of multiple case studies on WNT/β-catenin inhibitors, we also highlighted the challenges and opportunities for future clinical trial strategies involving these treatments. Additionally, we have proposed strategies for future WNT/β-catenin-based drug discovery trials, emphasizing the potential of combination therapies and AI/ML-driven prediction approaches. Overall, here we showcased the opportunities, possibilities, and potentialities of WNT/β-catenin signaling modulatory therapeutic regimes as promising precision cancer medicines for the future.
Collapse
Affiliation(s)
- Akansha Goyal
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Satyajit Laxman Murkute
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Sujoy Bhowmik
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India
| | - Chandra Prakash Prasad
- Department of Medical Oncology Lab, DR BRA-IRCH, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Purusottam Mohapatra
- Department of Biotechnology, NIPER Guwahati, Sila Katamur, Changsari, 781101 Kamrup, Assam, India.
| |
Collapse
|
31
|
Wang Y, He Y, You Q, Wang L. Design of bifunctional molecules to accelerate post-translational modifications: achievements and challenges. Drug Discov Today 2024; 29:104194. [PMID: 39343161 DOI: 10.1016/j.drudis.2024.104194] [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: 06/03/2024] [Revised: 09/12/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Post-translational modifications (PTMs) of proteins are crucial for regulating biological processes and their dysregulation is linked to various diseases, highlighting PTM regulation as a significant target for drug development. Traditional drug targets often interact with multiple proteins, resulting in lower selectivity and inevitable adverse effects, which limits their clinical applicability. Recent advancements in bifunctional molecules, such as proteolysis-targeting chimeras (PROTACs), have shown promise in targeting PTMs precisely. However, regulatory mechanisms for many of the >600 known PTMs remain underexplored. This review examines current progress and challenges in designing bifunctional molecules for PTM regulation, focusing on effector selection and ligand design strategies, aiming to propel the utilization and advancement of bifunctional molecules to the forefront of PTM research.
Collapse
Affiliation(s)
- Yuxuan Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yanyi He
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Lei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
32
|
Dong Y, Ma T, Xu T, Feng Z, Li Y, Song L, Yao X, Ashby CR, Hao GF. Characteristic roadmap of linker governs the rational design of PROTACs. Acta Pharm Sin B 2024; 14:4266-4295. [PMID: 39525578 PMCID: PMC11544172 DOI: 10.1016/j.apsb.2024.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/11/2024] [Accepted: 04/02/2024] [Indexed: 11/16/2024] Open
Abstract
Proteolysis targeting chimera (PROTAC) technology represents a groundbreaking development in drug discovery, leveraging the ubiquitin‒proteasome system to specifically degrade proteins responsible for the disease. PROTAC is characterized by its unique heterobifunctional structure, which comprises two functional domains connected by a linker. The linker plays a pivotal role in determining PROTAC's biodegradative efficacy. Advanced and rationally designed functional linkers for PROTAC are under development. Nonetheless, the correlation between linker characteristics and PROTAC efficacy remains under-investigated. Consequently, this study will present a multidisciplinary analysis of PROTAC linkers and their impact on efficacy, thereby guiding the rational design of linkers. We will primarily discuss the structural types and characteristics of PROTAC linkers, and the optimization strategies used for their rational design. Furthermore, we will discuss how factors like linker length, group type, flexibility, and linkage site affect the biodegradation efficiency of PROTACs. We believe that this work will contribute towards the advancement of rational linker design in the PROTAC research area.
Collapse
Affiliation(s)
- Yawen Dong
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Tingting Ma
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Ting Xu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Zhangyan Feng
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Yonggui Li
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Lingling Song
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Xiaojun Yao
- Faculty of Applied Sciences, Macau Polytechnic University, Macau 999078, China
| | - Charles R. Ashby
- Department of Pharmaceutical Sciences, St. John's University, New York, NY 11439, USA
| | - Ge-Fei Hao
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals, Guizhou University, Guiyang 550025, China
| |
Collapse
|
33
|
Chen H, Gridnev A, Schlamowitz N, Hu W, Dey K, Zheng G, Misra JR. Targeted degradation of specific TEAD paralogs by small molecule degraders. Heliyon 2024; 10:e37829. [PMID: 39328531 PMCID: PMC11425103 DOI: 10.1016/j.heliyon.2024.e37829] [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: 03/20/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
The transcription factors, TEAD1-4 together with their co-activator YAP/TAZ function as key downstream effectors of the Hippo pathway. Hyperactivation of TEAD-YAP/TAZ activity is observed in many human cancers. TEAD1-4 possess distinct physiological and pathological functions, with conserved sequences and structures. Targeting specific isoforms within TEAD1-4 can serve as valuable chemical probes for investigating TEAD-related functions in both development and diseases. We report the TEAD-targeting proteolysis targeting chimera (PROTAC), HC278, which achieves effective and specific targeting of TEAD1 and TEAD3 at low nanomolar doses while weakly degrading TEAD2 and TEAD4 at higher doses. Proteomic analysis of >6000 proteins confirmed their highly selective TEAD1 and TEAD3 degradation. Consistently, HC278 can suppress the proliferation of YAP-dependent NCI-H226 mesothelioma cells. Mechanistic exploration revealed that both CRBN and proteasome systems are involved in the TEAD degradation induced by HC278. Moreover, RNA-seq and Gene Set Enrichment Analysis (GSEA) revealed that the YAP signature genes such as CTGF, CYR61, and ANKRD1 are significantly downregulated by HC278 treatment. Overall, HC278 serves as a valuable chemical tool for unraveling the intricate biological roles of TEAD1 and TEAD3 and holds the potential as a lead compound for developing targeted therapy for TEAD1/3-driven pathologies.
Collapse
Affiliation(s)
- Hui Chen
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Artem Gridnev
- Department of Pharmacological Sciences, Stony Brook University School of Medicine, Stony Brook, 11794, USA
- Graduate School of Biomedical Sciences, Oregon Health & Sciences University, Portland, OR, USA
| | - Netanya Schlamowitz
- Department of Pharmacological Sciences, Stony Brook University School of Medicine, Stony Brook, 11794, USA
- Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wanyi Hu
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Kuntala Dey
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Jyoti R. Misra
- Department of Pharmacological Sciences, Stony Brook University School of Medicine, Stony Brook, 11794, USA
| |
Collapse
|
34
|
Yan H, Zang R, Cui T, Liu Y, Zhang B, Zhao L, Li H, Zhou J, Wang H, Zeng Q, Xu L, Zhou Y, Pei X, Xi J, Yue W. PROTAC-mediated vimentin degradation promotes terminal erythroid differentiation of pluripotent stem cells. Stem Cell Res Ther 2024; 15:310. [PMID: 39294765 PMCID: PMC11412063 DOI: 10.1186/s13287-024-03910-1] [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: 06/10/2024] [Accepted: 08/28/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND Human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs), can undergo erythroid differentiation, offering a potentially invaluable resource for generating large quantities of erythroid cells. However, the majority of erythrocytes derived from hPSCs fail to enucleate compared with those derived from cord blood progenitors, with an unknown molecular basis for this difference. The expression of vimentin (VIM) is retained in erythroid cells differentiated from hPSCs but is absent in mature erythrocytes. Further exploration is required to ascertain whether VIM plays a critical role in enucleation and to elucidate the underlying mechanisms. METHODS In this study, we established a hESC line with reversible vimentin degradation (dTAG-VIM-H9) using the proteolysis-targeting chimera (PROTAC) platform. Various time-course studies, including erythropoiesis from CD34+ human umbilical cord blood and three-dimensional (3D) organoid culture from hESCs, morphological analysis, quantitative real-time PCR (qRT-PCR), western blotting, flow cytometry, karyotyping, cytospin, Benzidine-Giemsa staining, immunofluorescence assay, and high-speed cell imaging analysis, were conducted to examine and compare the characteristics of hESCs and those with vimentin degradation, as well as their differentiated erythroid cells. RESULTS Vimentin expression diminished during normal erythropoiesis in CD34+ cord blood cells, whereas it persisted in erythroid cells differentiated from hESC. Depletion of vimentin using the degradation tag (dTAG) system promotes erythroid enucleation in dTAG-VIM-H9 cells. Nuclear polarization of erythroblasts is elevated by elimination of vimentin. CONCLUSIONS VIM disappear during the normal maturation of erythroid cells, whereas they are retained in erythroid cells differentiated from hPSCs. We found that retention of vimentin during erythropoiesis impairs erythroid enucleation from hPSCs. Using the PROTAC platform, we validated that vimentin degradation by dTAG accelerates the enucleation rate in dTAG-VIM-H9 cells by enhancing nuclear polarization.
Collapse
Affiliation(s)
- Hao Yan
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Ruge Zang
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China.
| | - Tiantian Cui
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Yiming Liu
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Biao Zhang
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Lingpin Zhao
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Hongyu Li
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Juannian Zhou
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Haiyang Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Quan Zeng
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Lei Xu
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Yuqi Zhou
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Xuetao Pei
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Jiafei Xi
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China.
| | - Wen Yue
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China.
| |
Collapse
|
35
|
Chou CL, Lin CT, Kao CT, Lin CC. A Novel Rational PROTACs Design and Validation via AI-Driven Drug Design Approach. ACS OMEGA 2024; 9:38371-38384. [PMID: 39310161 PMCID: PMC11411543 DOI: 10.1021/acsomega.3c10183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 07/31/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024]
Abstract
The rational design of novel drug candidates presents a formidable challenge in modern drug discovery. Proteolysis-targeting chimeras (PROTACs) drug design is particularly demanding due to their limited crystal structure availability and design of a viable small molecule to bridge the protein of interest (POI) and ubiquitin-protein ligase (E3). An integrated approach that combines superimposition techniques and deep neural networks is demonstrated in this study to leverage the power of deep learning and structural biology to generate structurally diverse molecules with enhanced binding affinities. The superimposition technique ensures the congruence of initial and new protein-ligand pairs, which are evaluated via subsequent comprehensive screening using the root-mean-square deviation (RMSD), binding free energy (BFE), and buried solvent-accessible surface area (SASA). The final candidates are subjected to the incorporation of molecular dynamics (MD) and free energy perturbation (FEP) simulations to provide a quantitative evaluation of relative binding energies, reinforcing the efficacy and reliability of the generated molecules. The outcomes of the generated novel PROTACs molecules exhibit comparable structural attributes while demonstrating superior binding affinities within the binding pockets when contrasted with those of the established cocrystal ternary complexes. To enhance the generalizability of the workflow, we chose the ternary structure of the cellular inhibitor of apoptosis protein 1 (cIAP1) and Bruton's Tyrosine Kinase (BTK) for validating the chemical properties generated from the processes. The new linker molecules additionally showed superior affinity from the simulations. In summary, this methodology serves as an effective workflow to align computational predictions with current limitations, thereby introducing a novel paradigm in AI-driven drug design.
Collapse
Affiliation(s)
| | - Chieh-Te Lin
- Department
of Biomedical Engineering, University of
California Davis, Davis, California 95616, United States
| | | | | |
Collapse
|
36
|
Tejwani V, Carroll T, Macartney T, Bandau S, Alabert C, Saredi G, Toth R, Rouse J. PROTAC-mediated conditional degradation of the WRN helicase as a potential strategy for selective killing of cancer cells with microsatellite instability. Sci Rep 2024; 14:20824. [PMID: 39242638 PMCID: PMC11379953 DOI: 10.1038/s41598-024-71160-5] [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: 06/05/2024] [Accepted: 08/26/2024] [Indexed: 09/09/2024] Open
Abstract
Multiple studies have demonstrated that cancer cells with microsatellite instability (MSI) are intolerant to loss of the Werner syndrome helicase (WRN), whereas microsatellite-stable (MSS) cancer cells are not. Therefore, WRN represents a promising new synthetic lethal target for developing drugs to treat cancers with MSI. Given the uncertainty of how effective inhibitors of WRN activity will prove in clinical trials, and the likelihood of tumours developing resistance to WRN inhibitors, alternative strategies for impeding WRN function are needed. Proteolysis-targeting chimeras (PROTACs) are heterobifunctional small molecules that target specific proteins for degradation. Here, we engineered the WRN locus so that the gene product is fused to a bromodomain (Bd)-tag, enabling conditional WRN degradation with the AGB-1 PROTAC specific for the Bd-tag. Our data revealed that WRN degradation is highly toxic in MSI but not MSS cell lines. In MSI cells, WRN degradation caused G2/M arrest, chromosome breakage and ATM kinase activation. We also describe a multi-colour cell-based platform for facile testing of selective toxicity in MSI versus MSS cell lines. Together, our data show that a degrader approach is a potentially powerful way of targeting WRN in MSI cancers and paves the way for the development of WRN-specific PROTAC compounds.
Collapse
Affiliation(s)
- Vikram Tejwani
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, DD1 5EH, UK
| | - Thomas Carroll
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, DD1 5EH, UK
| | - Thomas Macartney
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, DD1 5EH, UK
| | - Susanne Bandau
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, Wellcome Trust Biocentre, University of Dundee, Dundee, DD1 5EH, UK
| | - Constance Alabert
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, Wellcome Trust Biocentre, University of Dundee, Dundee, DD1 5EH, UK
| | - Giulia Saredi
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, DD1 5EH, UK
| | - Rachel Toth
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, DD1 5EH, UK
| | - John Rouse
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, DD1 5EH, UK.
| |
Collapse
|
37
|
Huang Y, Liu W, Zhao C, Shi X, Zhao Q, Jia J, Wang A. Targeting cyclin-dependent kinases: From pocket specificity to drug selectivity. Eur J Med Chem 2024; 275:116547. [PMID: 38852339 DOI: 10.1016/j.ejmech.2024.116547] [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: 03/01/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
The development of selective modulators of cyclin-dependent kinases (CDKs), a kinase family with numerous members and functional variations, is a significant preclinical challenge. Recent advancements in crystallography have revealed subtle differences in the highly conserved CDK pockets. Exploiting these differences has proven to be an effective strategy for achieving excellent drug selectivity. While previous reports briefly discussed the structural features that lead to selectivity in individual CDK members, attaining inhibitor selectivity requires consideration of not only the specific structures of the target CDK but also the features of off-target members. In this review, we summarize the structure-activity relationships (SARs) that influence selectivity in CDK drug development and analyze the pocket features that lead to selectivity using molecular-protein binding models. In addition, in recent years, novel CDK modulators have been developed, providing more avenues for achieving selectivity. These cases were also included. We hope that these efforts will assist in the development of novel CDK drugs.
Collapse
Affiliation(s)
- Yaoguang Huang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Wenwu Liu
- School of Pharmaceutical Sciences, Tsinghua University, Haidian Dist., Beijing, 100084, People's Republic of China
| | - Changhao Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China
| | - Xiaoyu Shi
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | - Qingchun Zhao
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China; Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang, 110840, People's Republic of China.
| | - Jingming Jia
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China.
| | - Anhua Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China.
| |
Collapse
|
38
|
Lee S, Kim HR, Woo Y, Kim J, Kim HW, Park JY, Suh B, Choi Y, Ahn J, Ryu JH, Roe JS, Song J, Lee SH. UBX-390: A Novel Androgen Receptor Degrader for Therapeutic Intervention in Prostate Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400398. [PMID: 38958553 PMCID: PMC11434238 DOI: 10.1002/advs.202400398] [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: 01/10/2024] [Revised: 06/18/2024] [Indexed: 07/04/2024]
Abstract
The androgen receptor (AR) is an attractive target for treating prostate cancer, considering its role in the development and progression of localized and metastatic prostate cancer. The high global mortality burden of prostate cancer, despite medical treatments such as androgen deprivation or AR antagonist therapy, highlights the need to explore alternative strategies. One strategy involves the use of heterobifunctional degraders, also known as proteolysis-targeting chimeras, which are novel small-molecule therapeutics that inhibit amplified or mutated targets. Here, the study reports a novel cereblon-based AR degrader, UBX-390, and demonstrates its superior activity over established AR degraders, such as ARV-110 or ARCC-4, in prostate cancer cells under short- and long-term treatment conditions. UBX-390 suppresses chromatin binding and gene expression of AR and demonstrates substantial efficacy in the degradation of AR mutants in patients with treatment-resistant prostate cancer. UBX-390 is presented as an optimized AR degrader with remarkable potential for treating castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Soohyun Lee
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hwa-Ryeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yaejin Woo
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Jiyoung Kim
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Han Wool Kim
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Ji Youn Park
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Beomseon Suh
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Yuri Choi
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Jungmin Ahn
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Je Ho Ryu
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| | - Jae-Seok Roe
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Song Hee Lee
- Ubix Therapeutics, Seoul, 05836, Republic of Korea
| |
Collapse
|
39
|
Xu Q, Feng H, Li Z, Shao X. Acetyl-CoA Carboxylase Proteolysis-Targeting Chimeras: Conceptual Design and Application as Insecticides. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:18809-18815. [PMID: 39145990 DOI: 10.1021/acs.jafc.4c02793] [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: 08/16/2024]
Abstract
Novel approaches for pest control are essential to ensure a sufficient food supply for the growing global population. The development of new insecticides must meet rigorous regulatory requirements for safety and address the resistance issues of existing insecticides. Proteolysis-targeting chimeras (PROTACs), originally developed for human diseases, show promise in agriculture. They offer innovative insecticides tailored to overcome resistance, opening avenues for agricultural applications. In this study, we developed small-molecule degraders by incorporating pomalidomide as an E3 ligand. These degraders were linked to a ligand (spirotetratmat enol) targeting the ACC protein through a flexible chain, aiming to achieve the efficient control of insects. Compounds 9a-9d were designed, synthesized, and evaluated for biological activities and mechanisms. Among them, 9b exhibited superior potency against Aphis craccivora (LC50 = 107.8 μg mL-1) compared to others and effectively degraded ACC proteins through the ubiquitin-proteasome system. These findings highlight the potential of utilizing PROTAC-based approaches in the development of insecticides for efficient pest control.
Collapse
Affiliation(s)
- Qi Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Hao Feng
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zhong Li
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Xusheng Shao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
40
|
Choi HI, Choi J, Kim JW, Lee YH, Cho KH, Koo TS. Stability Evaluation and Pharmacokinetic Profiling of Vepdegestrant in Rodents Using Liquid Chromatography-Tandem Mass Spectrometry. Molecules 2024; 29:4048. [PMID: 39274896 PMCID: PMC11396746 DOI: 10.3390/molecules29174048] [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: 07/08/2024] [Revised: 08/12/2024] [Accepted: 08/20/2024] [Indexed: 09/16/2024] Open
Abstract
Vepdegestrant (formerly ARV-471), a novel proteolysis-targeting chimera (PROTAC), targets estrogen receptor alpha (ERα) for degradation, offering a promising option to treat advanced ER-positive breast cancer. We developed and validated a sensitive and rapid liquid chromatography-tandem mass spectrometry method to quantify vepdegestrant in rodent plasma using bavdegalutamide (formerly ARV-110) as an internal standard. Plasma samples were prepared with protein precipitation using acetonitrile and analyzed using reverse-phase C18 columns and a mobile phase of 10 mM ammonium formate in distilled water and acetonitrile. The method demonstrated linearity from 1 to 1000 ng/mL in mouse and rat plasma, meeting all validation criteria, and successfully applied to in vivo and in vitro studies. Pharmacokinetic analysis revealed low-to-moderate clearance (313.3, 1053 mL/h/kg) and oral bioavailability (17.91, 24.12%) of vepdegestrant in mice and rats, respectively. It was unstable in buffer solutions across pH 2-10 and in phosphate-buffered saline (pH 7.4), likely due to adsorption, but remained stable in mouse and rat plasma at varying temperatures. In liver microsomes, vepdegestrant exhibited moderate stability in rats but was stable in mice, dogs, and humans. These findings enhance the understanding of pharmacokinetic properties of vepdegestrant supporting further development of PROTAC drugs.
Collapse
Affiliation(s)
- Hae-In Choi
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jinyoung Choi
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jin Woo Kim
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Yoon Ha Lee
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Kwan Hyung Cho
- College of Pharmacy and Inje Institute of Pharmaceutical Sciences and Research, Inje University, Gimhae 50834, Republic of Korea
| | - Tae-Sung Koo
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| |
Collapse
|
41
|
Zhang J, Chen X, Chen C, Li F, Song X, Liu C, Liao K, Su MY, Tan CSH, Fang L, Rao H. Distinct Amino Acid-Based PROTACs Target Oncogenic Kinases for Degradation in Non-Small Cell Lung Cancer (NSCLC). J Med Chem 2024; 67:13666-13680. [PMID: 39114932 DOI: 10.1021/acs.jmedchem.4c00208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Proteolysis-targeting chimeras (PROTACs) selectively eliminate detrimental proteins by exploiting the ubiquitin-proteasome system (UPS), representing a promising therapeutic strategy against various diseases. Effective adaptations of degradation signal sequences and E3 ligases for PROTACs remain limited. Here, we employed three amino acids─Gly, Pro, and Lys─as the ligand to recruit the corresponding E3 ligases: CRL2ZYG11B/ZER1, GID4, and UBRs, to degrade EML4-ALK and mutant EGFR, two oncogenic drivers in NSCLC. We found that the extent of EML4-ALK and EGFR reduction can be easily fine-tuned by using different degradation signals. These amino acid-based PROTACs, termed AATacs, hindered proliferation and induced cell cycle arrest and apoptosis of NSCLC cells in vitro. Compared to other PROTACs, AATacs are small, interchangeable but with different degradation efficiency. Our study further expands the repertoire of E3 ligases and their ligands for PROTAC application, improving the versatility and utility of targeted protein degradation for therapeutic purposes.
Collapse
Affiliation(s)
- Jianchao Zhang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiao Chen
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Congli Chen
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
| | - Fengming Li
- Department of Chemistry, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaoxiao Song
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Chaowei Liu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Kefan Liao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Ming-Yuan Su
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen 518055, China
| | - Chris Soon Heng Tan
- Department of Chemistry, Southern University of Science and Technology, Shenzhen 518055, China
| | - Lijing Fang
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials, CAS Key Laboratory of Biomedical Imaging Science and System, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen 518055, China
| | - Hai Rao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
42
|
Feng Y, Hu X, Wang X. Targeted protein degradation in hematologic malignancies: clinical progression towards novel therapeutics. Biomark Res 2024; 12:85. [PMID: 39169396 PMCID: PMC11340087 DOI: 10.1186/s40364-024-00638-1] [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: 06/30/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024] Open
Abstract
Targeted therapies, such as small molecule kinase inhibitors, have made significant progress in the treatment of hematologic malignancies by directly modulating protein activity. However, issues such as drug toxicity, drug resistance due to target mutations, and the absence of key active sites limit the therapeutic efficacy of these drugs. Targeted protein degradation (TPD) presents an emergent and rapidly evolving therapeutic approach that selectively targets proteins of interest (POI) based on endogenous degradation processes. With an event-driven pharmacology of action, TPD achieves efficacy with catalytic amounts, avoiding drug-related toxicity. Furthermore, TPD has the unique mode of degrading the entire POI, such that resistance derived from mutations in the targeted protein has less impact on its degradation function. Proteolysis-targeting chimeras (PROTACs) and molecular glue degraders (MGDs) are the most maturely developed TPD techniques. In this review, we focus on both preclinical experiments and clinical trials to provide a comprehensive summary of the safety and clinical effectiveness of PROTACs and MGDs in hematologic malignancies over the past two decades. In addition, we also delineate the challenges and opportunities associated with these burgeoning degradation techniques. TPD, as an approach to the precise degradation of specific proteins, provides an important impetus for its future application in the treatment of patients with hematologic malignancies.
Collapse
Affiliation(s)
- Yupiao Feng
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Xinting Hu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
| |
Collapse
|
43
|
Mitchell CL, Kurouski D. Novel strategies in Parkinson's disease treatment: a review. Front Mol Neurosci 2024; 17:1431079. [PMID: 39183754 PMCID: PMC11341544 DOI: 10.3389/fnmol.2024.1431079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
An unprecedented extension of life expectancy observed during the past century drastically increased the number of patients diagnosed with Parkinson's diseases (PD) worldwide. Estimated costs of PD alone reached $52 billion per year, making effective neuroprotective treatments an urgent and unmet need. Current treatments of both AD and PD focus on mitigating the symptoms associated with these pathologies and are not neuroprotective. In this review, we discuss the most advanced therapeutic strategies that can be used to treat PD. We also critically review the shift of the therapeutic paradigm from a small molecule-based inhibition of protein aggregation to the utilization of natural degradation pathways and immune cells that are capable of degrading toxic amyloid deposits in the brain of PD patients.
Collapse
Affiliation(s)
- Charles L. Mitchell
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| | - Dmitry Kurouski
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| |
Collapse
|
44
|
Koirala M, DiPaola M. Overcoming Cancer Resistance: Strategies and Modalities for Effective Treatment. Biomedicines 2024; 12:1801. [PMID: 39200265 PMCID: PMC11351918 DOI: 10.3390/biomedicines12081801] [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: 07/11/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Resistance to cancer drugs is a complex phenomenon that poses a significant challenge in the treatment of various malignancies. This review comprehensively explores cancer resistance mechanisms and discusses emerging strategies and modalities to overcome this obstacle. Many factors contribute to cancer resistance, including genetic mutations, activation of alternative signaling pathways, and alterations in the tumor microenvironment. Innovative approaches, such as targeted protein degradation, immunotherapy combinations, precision medicine, and novel drug delivery systems, hold promise for improving treatment outcomes. Understanding the intricacies of cancer resistance and leveraging innovative modalities are essential for advancing cancer therapy.
Collapse
|
45
|
Agarwal P, Reid DL, Amiji M. CNS delivery of targeted protein degraders. J Control Release 2024; 372:661-673. [PMID: 38936742 DOI: 10.1016/j.jconrel.2024.06.057] [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: 06/03/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
Heterobifunctional small molecule degraders are a subset of targeted protein degraders (TPDs), consisting of two ligands joined by a linker to induce proteasomal degradation of a target protein. As compared to traditional small molecules these compounds generally demonstrate inflated physicochemical properties, which may require innovative formulation strategies to enable their delivery and exert pharmacodynamic effect. The blood brain barrier (BBB) serves an essential function in human physiology, but its presence requires advanced approaches for treating central nervous system (CNS) diseases. By integrating emerging modalities like TPDs with conventional concepts of drug delivery, novel strategies to overcome the BBB can be developed. Amongst the available routes, lipid and polymer-based long-acting delivery seems to be the most amenable to TPDs, due to their ability to encapsulate lipophilic cargo and potential to be functionalized for targeted delivery. Another key consideration will be understanding E3 ligase expression in the different regions of the brain. Discovery of new brain or CNS disease specific E3 ligases could help overcome some of the barriers currently associated with CNS delivery of TPDs. This review discusses the current strategies that exist to overcome and improve therapeutic delivery of TPDs to the CNS.
Collapse
Affiliation(s)
- Prashant Agarwal
- Drug Product Technologies, Process Development, Amgen, Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, United States of America; Department of Chemical Engineering, College of Engineering, Northeastern University, 360 Huntington Ave, Boston, MA 02115, United States of America
| | - Darren L Reid
- Drug Product Technologies, Process Development, Amgen, Inc., 360 Binney St, Cambridge, MA 02142, United States of America
| | - Mansoor Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, 360 Huntington Ave, Boston, MA 02115, United States of America; Department of Chemical Engineering, College of Engineering, Northeastern University, 360 Huntington Ave, Boston, MA 02115, United States of America.
| |
Collapse
|
46
|
Zou ZF, Yang L, Nie HJ, Gao J, Lei SM, Lai Y, Zhang F, Wagner E, Yu HJ, Chen XH, Xu ZA. Tumor-targeted PROTAC prodrug nanoplatform enables precise protein degradation and combination cancer therapy. Acta Pharmacol Sin 2024; 45:1740-1751. [PMID: 38609561 PMCID: PMC11272941 DOI: 10.1038/s41401-024-01266-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/12/2024] [Indexed: 04/14/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) have emerged as revolutionary anticancer therapeutics that degrade disease-causing proteins. However, the anticancer performance of PROTACs is often impaired by their insufficient bioavailability, unsatisfactory tumor specificity and ability to induce acquired drug resistance. Herein, we propose a polymer-conjugated PROTAC prodrug platform for the tumor-targeted delivery of the most prevalent von Hippel-Lindau (VHL)- and cereblon (CRBN)-based PROTACs, as well as for the precise codelivery of a degrader and conventional small-molecule drugs. The self-assembling PROTAC prodrug nanoparticles (NPs) can specifically target and be activated inside tumor cells to release the free PROTAC for precise protein degradation. The PROTAC prodrug NPs caused more efficient regression of MDA-MB-231 breast tumors in a mouse model by degrading bromodomain-containing protein 4 (BRD4) or cyclin-dependent kinase 9 (CDK9) with decreased systemic toxicity. In addition, we demonstrated that the PROTAC prodrug NPs can serve as a versatile platform for the codelivery of a PROTAC and chemotherapeutics for enhanced anticancer efficiency and combination benefits. This study paves the way for utilizing tumor-targeted protein degradation for precise anticancer therapy and the effective combination treatment of complex diseases.
Collapse
Affiliation(s)
- Zhi-Feng Zou
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
- State Key Laboratory of Chemistry Biology & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Lei Yang
- State Key Laboratory of Chemistry Biology & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hui-Jun Nie
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jing Gao
- State Key Laboratory of Chemistry Biology & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Shu-Min Lei
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yi Lai
- State Key Laboratory of Chemistry Biology & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Fan Zhang
- Department of Chemistry, Fudan University, Shanghai, 20043, China
| | - Ernst Wagner
- Department of Pharmacy, Ludwig-Maximilians-Universität, 81377, München, Germany
| | - Hai-Jun Yu
- State Key Laboratory of Chemistry Biology & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiao-Hua Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Zhi-Ai Xu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
47
|
Mareczek L, Mueller LK, Halstenberg L, Geiger TM, Walz M, Zheng M, Hausch F. Use of Poly(vinyl alcohol) in Spray-Dried Dispersions: Enhancing Solubility and Stability of Proteolysis Targeting Chimeras. Pharmaceutics 2024; 16:924. [PMID: 39065621 PMCID: PMC11279962 DOI: 10.3390/pharmaceutics16070924] [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: 06/07/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
PROTACs, proteolysis targeting chimeras, are bifunctional molecules inducing protein degradation through a unique proximity-based mode of action. While offering several advantages unachievable by classical drugs, PROTACs have unfavorable physicochemical properties that pose challenges in application and formulation. In this study, we show the solubility enhancement of two PROTACs, ARV-110 and SelDeg51, using Poly(vinyl alcohol). Hereby, we apply a three-fluid nozzle spray drying set-up to generate an amorphous solid dispersion with a 30% w/w drug loading with the respective PROTACs and the hydrophilic polymer. Dissolution enhancement was achieved and demonstrated for t = 0 and t = 4 weeks at 5 °C using a phosphate buffer with a pH of 6.8. A pH shift study on ARV-110-PVA is shown, covering transfer from simulated gastric fluid (SGF) at pH 2.0 to fasted-state simulated intestinal fluid (FaSSIF) at pH 6.5. Additionally, activity studies and binding assays of the pure SelDeg51 versus the spray-dried SelDeg51-PVA indicate no difference between both samples. Our results show how modern enabling formulation technologies can partially alleviate challenging physicochemical properties, such as the poor solubility of increasingly large 'small' molecules.
Collapse
Affiliation(s)
| | | | | | - Thomas M. Geiger
- Department of Chemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Michael Walz
- Department of Chemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Min Zheng
- Department of Chemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
| | - Felix Hausch
- Department of Chemistry, Technical University of Darmstadt, 64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technical University of Darmstadt, 64287 Darmstadt, Germany
| |
Collapse
|
48
|
Grigoreva TA, Novikova DS, Melino G, Barlev NA, Tribulovich VG. Ubiquitin recruiting chimera: more than just a PROTAC. Biol Direct 2024; 19:55. [PMID: 38978100 PMCID: PMC11232244 DOI: 10.1186/s13062-024-00497-8] [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: 05/21/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024] Open
Abstract
Ubiquitinylation of protein substrates results in various but distinct biological consequences, among which ubiquitin-mediated degradation is most well studied for its therapeutic application. Accordingly, artificially targeted ubiquitin-dependent degradation of various proteins has evolved into the therapeutically relevant PROTAC technology. This tethered ubiquitinylation of various targets coupled with a broad assortment of modifying E3 ubiquitin ligases has been made possible by rational design of bi-specific chimeric molecules that bring these proteins in proximity. However, forced ubiquitinylation inflicted by the binary warheads of a chimeric PROTAC molecule should not necessarily result in protein degradation but can be used to modulate other cellular functions. In this respect it should be noted that the ubiquitinylation of a diverse set of proteins is known to control their transport, transcriptional activity, and protein-protein interactions. This review provides examples of potential PROTAC usage based on non-degradable ubiquitinylation.
Collapse
Affiliation(s)
- Tatyana A Grigoreva
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), St. Petersburg, 190013, Russia.
| | - Daria S Novikova
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), St. Petersburg, 190013, Russia
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, 00133, Italy
| | - Nick A Barlev
- Institute of Cytology RAS, Saint-Petersburg, 194064, Russia
- Department of Biomedical Studies, School of Medicine, Nazarbayev University, Astana, 010000, Kazakhstan
| | - Vyacheslav G Tribulovich
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), St. Petersburg, 190013, Russia.
| |
Collapse
|
49
|
Zhang S, Nie S, Ma G, Shen M, Kong L, Zuo Z, Li Y. Identification of novel GSPT1 degraders by virtual screening and bioassay. Eur J Med Chem 2024; 273:116524. [PMID: 38795517 DOI: 10.1016/j.ejmech.2024.116524] [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: 04/08/2024] [Revised: 05/11/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024]
Abstract
GSPT1 plays crucial physiological functions, such as terminating protein translation, overexpressed in various tumors. It is a promising anti-tumor target, but is also considered as an "undruggable" protein. Recent studies have found that a class of small molecules can degrade GSPT1 through the "molecular glue" mechanism with strong antitumor activity, which is expected to become a new therapy for hematological malignancies. Currently available GSPT1 degraders are mostly derived from the scaffold of immunomodulatory imide drug (IMiD), thus more active compounds with novel structure remain to be found. In this work, using computer-assisted multi-round virtual screening and bioassay, we identified a non-IMiD acylhydrazone compound, AN5782, which can reduce the protein level of GPST1 and obviously inhibit the proliferation of tumor cells. Some analogs were obtained by a substructure search of AN5782. The structure-activity relationship analysis revealed possible interactions between these compounds and CRBN-GSPT1. Further biological mechanistic studies showed that AN5777 decreased GSPT1 remarkably through the ubiquitin-proteasome system, and its effective cytotoxicity was CRBN- and GSPT1-dependent. Furthermore, AN5777 displayed good antiproliferative activities against U937 and OCI-AML-2 cells, and dose-dependently induced G1 phase arrest and apoptosis. The structure found in this work could be good start for antitumor drug development.
Collapse
Affiliation(s)
- Shuqun Zhang
- Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Shiyun Nie
- Key Laboratory of Medicinal Chemistry for Natural Resource, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, Ministry of Education, Yunnan University, Kunming, 650500, China
| | - Guangchao Ma
- Key Laboratory of Medicinal Chemistry for Natural Resource, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, Ministry of Education, Yunnan University, Kunming, 650500, China
| | - Meiling Shen
- Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lingmei Kong
- Key Laboratory of Medicinal Chemistry for Natural Resource, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, Ministry of Education, Yunnan University, Kunming, 650500, China
| | - Zhili Zuo
- Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yan Li
- Key Laboratory of Phytochemistry and Natural Medicines, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Key Laboratory of Medicinal Chemistry for Natural Resource, Yunnan Key Laboratory of Research and Development for Natural Products, School of Pharmacy, Ministry of Education, Yunnan University, Kunming, 650500, China.
| |
Collapse
|
50
|
Thomas M, Ahmad M, Tresadern G, de Fabritiis G. PromptSMILES: prompting for scaffold decoration and fragment linking in chemical language models. J Cheminform 2024; 16:77. [PMID: 38965600 PMCID: PMC11225391 DOI: 10.1186/s13321-024-00866-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 06/04/2024] [Indexed: 07/06/2024] Open
Abstract
SMILES-based generative models are amongst the most robust and successful recent methods used to augment drug design. They are typically used for complete de novo generation, however, scaffold decoration and fragment linking applications are sometimes desirable which requires a different grammar, architecture, training dataset and therefore, re-training of a new model. In this work, we describe a simple procedure to conduct constrained molecule generation with a SMILES-based generative model to extend applicability to scaffold decoration and fragment linking by providing SMILES prompts, without the need for re-training. In combination with reinforcement learning, we show that pre-trained, decoder-only models adapt to these applications quickly and can further optimize molecule generation towards a specified objective. We compare the performance of this approach to a variety of orthogonal approaches and show that performance is comparable or better. For convenience, we provide an easy-to-use python package to facilitate model sampling which can be found on GitHub and the Python Package Index.Scientific contributionThis novel method extends an autoregressive chemical language model to scaffold decoration and fragment linking scenarios. This doesn't require re-training, the use of a bespoke grammar, or curation of a custom dataset, as commonly required by other approaches.
Collapse
Affiliation(s)
- Morgan Thomas
- Computational Science Laboratory, Universitat Pompeu Fabra, Barcelona Biomedical Research Park (PRBB), C Dr. Aguiader 88, 08003, Barcelona, Spain.
| | - Mazen Ahmad
- In Silico Discovery, Janssen Pharmaceutica N. V., Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Gary Tresadern
- In Silico Discovery, Janssen Pharmaceutica N. V., Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Gianni de Fabritiis
- Computational Science Laboratory, Universitat Pompeu Fabra, Barcelona Biomedical Research Park (PRBB), C Dr. Aguiader 88, 08003, Barcelona, Spain.
- Acellera Labs, C Dr. Trueta 183, 08005, Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig Lluis Companys 23, 08010, Barcelona, Spain.
| |
Collapse
|