Skip to main content

An Unprecedented Covalent Binding Mechanism of ML210 to GPX4


GPX4 is a selenoprotein that plays a critical role in protecting cells against oxidative stress and ferroptosis. The nucleophilic selenocysteine residue (Sec) located in the catalytic center of GPX4 can be exploited to design covalent inhibitors. Previously reported covalent GPX4 inhibitors always contain reactive alkyl chloride groups, conferring poor selectivities. Recently, the Schreiber group reported that ML210, a direct covalent inhibitor targeting GPX4 in cells, exhibits much more potency and remarkable selectivity compared with chloroacetamide inhibitors. More interestingly, an unprecedented covalent binding mechanism is involved in the ML210 targeting GPX4 in cells.

ML210 was previously reported as a ferroptosis inducer. Due to lack of an apparent covalent reactive group and inability to react with small molecule thiols directly, the covalent binding action of ML210 with its target proteins has never been disclosed. To identify that ML210 acts as a direct covalent inhibitor targeting GPX4, pulldown and competitive pulldown experiments were performed, suggesting that ML210 interacts with GPX4 in cells. The cellular thermal shift assay (CETSA) also revealed that ML210 can stabilize GPX4U46C instead of GPX4U46A, which further confirmed that the covalent interaction may be dependent on the selenocysteine residue of GPX4.

Intriguingly, ML210 can only bind GPX4 in cells instead of the recombinant counterpart. This is distinctly different from other reported chloroacetamide GPX4 inhibitors, which can covalently bind both GPX4 in intact cells and in a purified form. Intact protein mass spectrometry-based assay showed that ML210-GPX4 complex (+434 Da) can be detected after cells treated with ML210. The binding feature and the loss of 41 Da mass suggested that ML210 maybe acts as a prodrug, which can be metabolized in cells to form new intermediates targeting GPX4 via a covalent bond.

This unusual covalent binding action prompted researchers to further investigate the detailed mechanism. Previous studies have demonstrated the nitroisoxazole group is likely hydrolyzed to release a carboxylic acid and α-nitroketoxime under basic conditions. The SAR studies of the isoxazole group reveled that both isoxazole and nitro groups are essential for the activity of ML210. The 5-position substitution has little effect on the cellular potency of ML210. Similar to ML210, isopropyl-ML210 also induced the same ML210-GPX4 (+434 Da) complex formation in cells. This is consistent with the hypothesis of nitroisoxazole group hydrolysis.

Therefore, JKE-1674 was proposed as a key intermediate after the hydrolysis of ML210 in cells. Indeed, after incubating ML210 in cells, the formation of JKE-1674 and the consumption of ML210 were correlated in a time-dependent manner. JKE-1674 showed a similar activity to ML210 in terms of the cellular potency and proteome-wide selectivity. However, JKE-1674 still cannot react with the purified GPX4, suggesting that an additional metabolite acts as the key intermediate to react with GPX4 directly. Subsequently, the hypothesized nitrile-oxide electrophile JKE-1777 was synthesized. The nitrile-oxide JKE-1777 showed considerable cellular activity and can react with purified GPX4 directly. Additionally, the molecular weight of JKE-1777 is consistent with the +434 Da mass of ML210-GPX4 complex. In contrast to JKE-1777, no activity was observed in cells for its precursor 39, suggesting that JKE-1777 was generated through other precursor in cells, a possible precursor is the furoxan. Collectively, JKE-1777 may be the final electrophile that forms the covalent bond with the selenocysteine residue of GPX4 in cells.

Based on the findings, the researchers further expanded the diversity of the masked nitrile-oxide electrophile. Compound 43, JKE-1708, JKE-1716 and diacylfuroxan showed pretty good potency in ferroptosis-inducing and fer-1 rescue experiments. It would be interesting to see whether the potency and selectivity will be improved if replacing the chloride of RSL3 and ML162 with the masked nitrile-oxide electrophiles.

Although the in vivo activities of these masked nitrile-oxide compounds remain to be evaluated, this novel covalent binding mechanism in cells indicates that masked nitrile-oxide electrophiles may be potentially designed as covalent inhibitors for targeting other (seleno)cysteine residues in proteins.

Reference:
1. Eaton J. K., et.al. Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles. Nat. Chem. Biol.  2020, 16, 497. https://doi.org/10.1038/s41589-020-0501-5
2.  Eaton J. K., et.al. Diacylfuroxans Are Masked Nitrile Oxides That Inhibit GPX4 Covalently. J. Am. Chem. Soc. 2019, 141, 20407.



Comments

Popular posts from this blog

Photoswitchable azopyrazoles Flipping the Switches of Ion Channels

Abnormally activated Ca 2+ channels are related to many human diseases, including Stormorken Syndrome, which makes Ca 2+ release-activated Ca 2+ (CRAC) channel a very promising therapeutic target. Several small molecule therapeutics targeting CRAC channels have been developed, including the GSKs series and Synta 66 . Those compounds have relatively high specificity. Meanwhile, a controllable system that can be activated with a switch can probably serve as a convenient tool for further related research.    Photoswitchable chemistry has been applied to a lot of bioactive targets such as ion channels, receptors enzymes and nucleic acids. Recently, the Li group developed CRAC channel inhibitors that can be turned ‘on’ and ‘off’ by UV-light exposure. Scheme 1 . C onverting CRAC channel inhibitors, GSKs , into photoswitchable derivatives, piCRACs . Starting from the well-established GSK-based CRAC inhibitor ( Scheme 1 ), the authors developed a series of nitrosaniline

Proteosome Inhibition Taps Into RNA Splicing

Multiple myeloma (MM) is a malignancy of white blood cells called plasma cells that reside mainly in the bone marrow and is the second most common blood cancer. With increased understanding of biology, the current use of immunomodulatory (IM) drugs and proteasome inhibitors (PI) have taken over the therapeutic landscape for MM. The combination of bortezomib (PI) with lenalidomiede (IM) and dexamethasone is commonly the initial treatment of choice. Early intervention seems to provide a good outcome, but unfortunately, many patients eventually relapse. A major goal in MM treatment is to increase the efficacy of proteasome inhibitors and prevent relapse. To investigate this, Huang et al. used unbiased mass spectrometry-based phosphoproteomics to identify potential vulnerabilities after treatment with the PI carfilzomib and discovered that splicing related proteins had significant changes in phosphorylation that is undetectable upon examination of RNA and protein abundance. Treatment w

One Fluorine Atom Fixes Poor KRAS Inhibitors

KRAS  is the most frequently mutated oncogene in human cancer. In the past few decades, the KRAS oncoprotein had always been deemed as an “undruggable target” due to lack of binding surface and tightly binding to its substrate GTP. In 2013, the Shokat group identified that the mutant cysteine KRAS G12C creates a new allosteric pocket “switch-II pocket” which can be exploited to design covalent inhibitors. KRAS G12C accounts for more than 50% of the incidences of KRAS mutations, involving in many cancers, such as non-small cell lung cancer (NSCLC), colorectal adenocarcinomas and pancreatic cancer. KARS G12C has been an attractive target for drug discovery and development in both academia and industry. MRTX849 is a potent, orally available covalent inhibitor of KRAS G12C developed by Mirati Therapeutics and currently undergoing Phase I/II clinical trials. Recently, a paper published in Journal of Medicinal Chemistry reported the design and optimization of MRTX849 . The medi