4.8 Article

Cancer Selective Target Degradation by Folate-Caged PROTACs

Journal

JOURNAL OF THE AMERICAN CHEMICAL SOCIETY
Volume 143, Issue 19, Pages 7380-7387

Publisher

AMER CHEMICAL SOC
DOI: 10.1021/jacs.1c00451

Keywords

-

Funding

  1. NIH [R35CA253027, R01CA218600, R01CA230854, R01GM122749, R01CA260666, P30CA196521]
  2. National Institutes of Health SIG [1S10OD025132-01A1]

Ask authors/readers for more resources

PROTACs are emerging as a promising therapeutic modality for degrading intracellular protein targets, but potential toxicity in normal cells limits their applications. By conjugating a folate group to a ligand of the VHL E3 ubiquitin ligase, a cancer cell selective delivery strategy for PROTACs was developed to achieve targeted degradation of proteins of interest in cancer cells. Folate-PROTACs provide a platform for selective degradation of proteins of interest in cancer cells.
PROTACs (proteolysis targeting chimeras) are an emerging class of promising therapeutic modalities that degrade intracellular protein targets by hijacking the cellular ubiquitin-proteasome system. However, potential toxicity of PROTACs in normal cells due to the off-tissue on-target degradation effect limits their clinical applications. Precise control of a PROTAC's on-target degradation activity in a tissue-selective manner could minimize potential toxicity/side-effects. To this end, we developed a cancer cell selective delivery strategy for PROTACs by conjugating a folate group to a ligand of the VHL E3 ubiquitin ligase, to achieve targeted degradation of proteins of interest (POIs) in cancer cells versus noncancerous normal cells. We show that our folate-PROTACs, including BRD PROTAC (folate-ARV-771), MEK PROTAC (folate-MS432), and ALK PROTAC (folate-MS99), are capable of degrading BRDs, MEKs, and ALK, respectively, in a folate receptor-dependent manner in cancer cells. This design provides a generalizable platform for PROTACs to achieve selective degradation of POIs in cancer cells.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

Article Chemistry, Medicinal

Folate-Guided Protein Degradation by Immunomodulatory Imide Drug-Based Molecular Glues and Proteolysis Targeting Chimeras

He Chen, Jing Liu, H. Umit Kaniskan, Wenyi Wei, Jian Jin

Summary: This study presents a general strategy for delivering IMiD-based molecular glues and PROTACs to FOLR1-expressing cancer cells, effectively degrading fusion proteins in cancer cells with the potential to ameliorate toxicity.

JOURNAL OF MEDICINAL CHEMISTRY (2021)

Article Chemistry, Multidisciplinary

TF-PROTACs Enable Targeted Degradation of Transcription Factors

Jing Liu, He Chen, H. Umit Kaniskan, Ling Xie, Xian Chen, Jian Jin, Wenyi Wei

Summary: A new therapeutic platform TF-PROTAC has been developed with selectivity to degrade target TFs and demonstrate anti-proliferative effects in cells. This platform holds promise as a universal strategy for targeting undruggable TFs.

JOURNAL OF THE AMERICAN CHEMICAL SOCIETY (2021)

Article Biochemistry & Molecular Biology

Functional analysis of the emerging roles for the KISS1/KISS1R signaling pathway in cancer metastasis

Zhenxi Li, Jing Liu, Hiroyuki Inuzuka, Wenyi Wei

Summary: Kisspeptins, encoded by the KISS1 gene, play an important role in suppressing metastasis in certain types of cancers, but their physiological effects on cancer metastasis are context-dependent and still controversial. This article discusses the epigenetic mechanism regulating KISS1 gene expression, the context-dependent role of KISS1/KISS1R, the signaling pathways promoting or inhibiting metastasis, and the potential of targeting KISS1/KISS1R for anticancer therapeutics.

JOURNAL OF GENETICS AND GENOMICS (2022)

Correction Biochemistry & Molecular Biology

Prostate-specific oncogene OTUD6A promotes prostatic tumorigenesis via deubiquitinating and stabilizing c-Myc (Feb, 10.1038/s41418-022-00960-x, 2022)

Yunhua Peng, Jing Liu, Zhen Wang, Chunping Cui, Tiantian Zhang, Shuangxi Zhang, Peipei Gao, Zhanwu Hou, Huadong Liu, Jianping Guo, Jinfang Zhang, Yurong Wen, Wenyi Wei, Lingqiang Zhang, Jiankang Liu, Jiangang Long

CELL DEATH AND DIFFERENTIATION (2023)

Article Biochemistry & Molecular Biology

Prostate-specific oncogene OTUD6A promotes prostatic tumorigenesis via deubiquitinating and stabilizing c-Myc

Yunhua Peng, Jing Liu, Zhen Wang, Chunping Cui, Tiantian Zhang, Shuangxi Zhang, Peipei Gao, Zhanwu Hou, Huadong Liu, Jianping Guo, Jinfang Zhang, Yurong Wen, Wenyi Wei, Lingqiang Zhang, Jiankang Liu, Jiangang Long

Summary: OTUD6A is a deubiquitinase that is specifically amplified and overexpressed in prostate cancer (PrCa), and it promotes prostatic tumorigenesis by deubiquitinating and stabilizing c-Myc oncoprotein. Genetic ablation of OTUD6A efficiently represses PrCa tumorigenesis in both cells and mouse models.

CELL DEATH AND DIFFERENTIATION (2022)

Article Chemistry, Medicinal

Discovery of a First-in-Class Degrader for Nuclear Receptor Binding SET Domain Protein 2 (NSD2) and Ikaros/Aiolos Published as part of the Journal of Medicinal Chemistry special issue Epigenetics 2022.

Fanye Meng, Chenxi Xu, Kwang-Su Park, H. Umit Kaniskan, Gang Greg Wang, Jian Jin

Summary: In this study, a first-in-class NSD2 proteolysis targeting chimera (PROTAC) degrader was discovered, which effectively degraded NSD2 and CRBN. This compound was more effective in suppressing cancer cell growth compared to existing inhibitors and showed bioavailability in mice.

JOURNAL OF MEDICINAL CHEMISTRY (2022)

Article Chemistry, Multidisciplinary

TF-DUBTACs Stabilize Tumor Suppressor Transcription Factors

Jing Liu, Xufen Yu, He Chen, H. Umit Kaniskan, Ling Xie, Xian Chen, Jian Jin, Wenyi Wei

Summary: A platform named TF-DUBTAC has been developed to selectively stabilize tumor suppressor transcription factors through linking a DNA oligonucleotide to a covalent ligand of the deubiquitinase OTUB1. This method successfully stabilized FOXO3A, p53, and IRF3 in cells, providing a potential therapeutic means to suppress tumorigenesis.

JOURNAL OF THE AMERICAN CHEMICAL SOCIETY (2022)

Article Chemistry, Medicinal

Discovery of the First Lactate Dehydrogenase Proteolysis Targeting Chimera Degrader for the Treatment of Pancreatic Cancer

Ning Sun, Md Kabir, Youngeun Lee, Ling Xie, Xiaoping Hu, Julia Velez, Xian Chen, H. Umit Kaniskan, Jian Jin

Summary: Lactate dehydrogenase (LDH) is a key glycolytic enzyme and biomarker of aggressive cancers. LDHA/B inhibitors have been developed, but here, researchers report the discovery of a LDH proteolysis targeting chimera (PROTAC) degrader, compound 22 (MS6105), which potently degrades LDHA and LDHB in a time- and ubiquitin-proteasome system dependent manner. Compound 22 shows stronger inhibitory effects on pancreatic cancer cell growth compared to the parent LDH inhibitor, and it is bioavailable in mice through intraperitoneal injection. Overall, compound 22 could be a valuable tool for studying the pathophysiological functions of LDH in vitro and in vivo.

JOURNAL OF MEDICINAL CHEMISTRY (2023)

Article Chemistry, Multidisciplinary

Telomere Targeting Chimera Enables Targeted Destruction of Telomeric Repeat-Binding Factor Proteins

Zhen Wang, Jing Liu, He Chen, Xing Qiu, Ling Xie, H. Umit Kaniskan, Xian Chen, Jian Jin, Wenyi Wei

Summary: In this study, a nucleotide-based proteolysis-targeting chimera (PROTAC) was developed to degrade TRF1/2, which are key components in regulating telomere length. The prototype telomere-targeting chimeras (TeloTACs) efficiently degraded TRF1/2 and resulted in telomere shortening, inhibiting cancer cell proliferation.

JOURNAL OF THE AMERICAN CHEMICAL SOCIETY (2023)

Article Multidisciplinary Sciences

PRMT1 mediated methylation of cGAS suppresses anti-tumor immunity

Jing Liu, Xia Bu, Chen Chu, Xiaoming Dai, John M. Asara, Piotr Sicinski, Gordon J. Freeman, Wenyi Wei

Summary: The protein arginine methyltransferase PRMT1 suppresses the anti-tumor immune response by methylating cGAS and preventing its dimerization. Inhibition of PRMT1 activates the cGAS/STING-dependent DNA sensing signaling and increases the expression of interferon response genes, tumor-infiltrating lymphocytes, and tumoral PD-L1. Combination therapy of PRMT1 inhibitor with anti-PD-1 antibody enhances the anti-tumor therapeutic efficacy.

NATURE COMMUNICATIONS (2023)

Article Chemistry, Multidisciplinary

Methylated Nucleotide-Based Proteolysis-Targeting Chimera Enables Targeted Degradation of Methyl-CpG-Binding Protein 2

Zhen Wang, Jing Liu, Xing Qiu, Dingpeng Zhang, Hiroyuki Inuzuka, Li Chen, He Chen, Ling Xie, H. Umit Kaniskan, Xian Chen, Jian Jin, Wenyi Wei

Summary: This study presents a novel strategy, methyl-proteolysis-targeting chimera (methyl-PROTAC), for manipulating epigenetic factors by designing a modified nucleotide-based degradation approach. It has the potential to degrade undruggable epigenetic regulatory proteins.

JOURNAL OF THE AMERICAN CHEMICAL SOCIETY (2023)

Article Chemistry, Multidisciplinary

Acetylation Targeting Chimera Enables Acetylation of the Tumor Suppressor p53

Md Kabir, Ning Sun, Xiaoping Hu, Tiphaine C. Martin, Jingjie Yi, Yue Zhong, Yan Xiong, H. Umit Kaniskan, Wei Gu, Ramon Parsons, Jian Jin

Summary: With the novel Acetylation Targeting Chimera (AceTAC) strategy, the p53 tumor suppressor protein can be acetylated successfully. The first p53Y220C AceTAC, MS78, recruits histone acetyltransferase p300/CBP to acetylate the p53Y220C mutant. MS78 effectively suppresses the proliferation and clonogenicity of cancer cells with the p53Y220C mutation and induces upregulation of TRAIL apoptotic genes and downregulation of DNA damage response pathways. In conclusion, the AceTAC strategy provides a generalizable platform for targeting proteins, such as tumor suppressors, via acetylation.

JOURNAL OF THE AMERICAN CHEMICAL SOCIETY (2023)

Review Chemistry, Multidisciplinary

Chemically induced degradation of epigenetic targets

Md Kabir, Xufen Yu, H. Umit Kaniskan, Jian Jin

Summary: Proteolysis-targeting chimeras (PROTACs) are small molecules that induce targeted degradation of proteins by forming ternary complexes with E3 ligases. They have the advantage of targeting both canonical and noncanonical functions of epigenetic targets, resulting in greater therapeutic efficacy. This review analyzes published PROTAC degraders of epigenetic writer, reader, and eraser proteins and discusses their mechanism of action and advantages in cancer treatment. Overall, pharmacological degradation of epigenetic targets has emerged as an effective strategy against cancer.

CHEMICAL SOCIETY REVIEWS (2023)

Article Chemistry, Medicinal

Targeting Triple-Negative Breast Cancer by a Novel Proteolysis Targeting Chimera Degrader of Enhancer of Zeste Homolog 2

Brandon Dale, Chris Anderson, Kwang-Su Park, H. Umit Kaniskan, Anqi Ma, Yudao Shen, Chengwei Zhang, Ling Xie, Xian Chen, Xufen Yu, Jian Jin

Summary: EZH2 overexpression is associated with poor prognosis in TNBC. A novel EZH2 PROTAC degrader, MS8815, has been discovered and shown to effectively suppress the growth of TNBC cells.

ACS PHARMACOLOGY & TRANSLATIONAL SCIENCE (2022)

Review Oncology

Advancing targeted protein degradation for cancer therapy

Brandon Dale, Meng Cheng, Kwang-Su Park, H. Umit Kaniskan, Yue Xiong, Jian Jin

Summary: The development of small-molecule degraders such as proteolysis-targeting chimeras (PROTACs) has enabled the targeting of previously considered undruggable oncoproteins. This review discusses recent advances in the field, with a focus on opportunities and challenges for future development.

NATURE REVIEWS CANCER (2021)

No Data Available