Rezatapopt (PC14586): A First-in-Class Small Molecule p53 Y220C Mutant Protein Stabilizer in Clinical Trials

IF 6.8 1区 医学 Q1 CHEMISTRY, MEDICINAL
Zonghui Ma, Qiang Shen, Jia Zhou
{"title":"Rezatapopt (PC14586): A First-in-Class Small Molecule p53 Y220C Mutant Protein Stabilizer in Clinical Trials","authors":"Zonghui Ma, Qiang Shen, Jia Zhou","doi":"10.1021/acs.jmedchem.5c00670","DOIUrl":null,"url":null,"abstract":"The p53 protein, an important transcription factor, plays a critical role in regulating cell responses to stress. (1,2) Coded by the <i>TP</i>53 gene, p53 is a well-recognized tumor suppressorprotein. Dysregulation of p53 function is closely associated with the initiation and development of various cancers. Although restoration of p53 function appears to be a promising therapeutic approach, p53 is considered to be a traditionally “undruggable” target owing to the lack of active binding pockets or frequent mutations. (3) Mutations in the <i>TP</i>53 gene can assist tumor cells to evade p53 suppressive effects, promoting cell proliferation, migration, and invasion, thereby aggravating tumorigenesis. <i>TP</i>53 genetic mutations present in more than 50% of human cancers, most of which are missense mutations occurring in the DNA-binding domain of p53 (amino acids 94–292). Y220C is the ninth most prevalent mutation among all <i>TP</i>53 mutations observed across various tumor types, accounting for 1.8%, and presents in ∼1% of human cancers. (4) Y220C mutation leads to tyrosine-to-cysteine substitution and creates a pocket in the p53 Y220C mutant protein. p53 Y220C protein is structurally unstable at physiological temperatures and can be ubiquitinated and delivered to proteasomal for degradation. Therefore, targeting p53 Y220C-mutant stabilization may be a viable therapeutic strategy for various associated cancers. Small molecule compounds targeting p53 Y220C stabilization are in active development for potential cancer therapeutics. (5) Rezatapopt (PC14586), discovered by PMV Pharmaceuticals, Inc., is a first-in-class small molecule stabilizer and reactivator of p53 Y220C currently in the Phase II clinical trial (NCT04585750) for treating various locally advanced or metastatic solid tumors harboring a p53 Y220C mutation. (6) In addition, rezatapopt is in a Phase Ib clinical trial (NCT06616636) in combination with azacitidine for patients with <i>TP</i>53 Y220C mutant myeloid malignancies (acute myeloid leukemia or myelodysplastic syndrome). Rezatapopt was designed to tightly occupy the pocket created by mutation-caused amino acid tyrosine-to-cysteine substitution in the p53 Y220C mutant protein. Rezatapopt was obtained based on structure-based drug design (SBDD) and systematical structural optimization, starting from hit compound <b>1</b> (Figure 1). The substrate concentration required to increase DNA binding by 1.5-fold (SC<sub>150</sub>) was determined by a time-resolved fluorescence resonance energy transfer (FRET) assay to assess the <i>in vitro</i> potency of the p53 Y220C reactivators. Hit compound <b>1</b> was obtained by scaffold combination of hit compounds PhiKan83 (SC<sub>150</sub> = 37.2 μM) with a carbazole scaffold (7) and PK1596 (SC<sub>150</sub> = 1.6 μM) with an iodophenol scaffold. (8) Compound <b>1</b> contains a widely recognized “privileged” indole scaffold in drug development, which exists in numerous natural products and bioactive molecules. Compound <b>1</b> has three anchors attached to the indole scaffold, including a hydrophobic ethyl group, a polar alkylamine extending to the solvent region, and a substituted acetylene occupying the adjacent subsite. Compound <b>1</b> (SC<sub>150</sub> = 13.6 μM) showed inadequate potency (SC<sub>150</sub> &gt; 100 nM) and poor metabolic stability in human liver microsome (<i>T</i><sub>1/2</sub> = 4 min). SBDD efforts around <b>1</b> (SC<sub>150</sub> = 13.6 μM) resulted in the lead compound <b>2</b> (PC-9859) with significantly improved DNA binding activity (SC<sub>150</sub> = 54 nM). The cocrystal structure (PDB code: 9BR4) of p53 Y220C mutant protein in complex with <b>2</b>, revealed that <b>2</b> locates at the pocket created by tyrosine-to-cysteine substitution in p53 Y220C mutant protein, with indole scaffold occupying this space (Figure 1). Trifluoromethyl group inserts deeply into the hydrophobic pocket, the 4-aminopiperidine ring extends toward the solvent-exposed region, and the pyridine ring is connected to indole core via an alkyne occupies the adjacent subsite. The pyridine ring forms a favorable CH-π stacking interaction with Pro153. Compound <b>2</b> forms two key hydrogen bonds with p53 Y220C mutant protein. One interaction is formed between the amino group on pyridine ring and the carbonyl of Cys220 while the other is formed between the amino group on 1-methylpiperidine ring and the carbonyl of Thr150 on the side chain. Further drug design and structural optimization around lead compound <b>2</b> has successfully led to the discovery of rezatapopt with a fluorine atom substitution on the piperidine ring and a polar amide group on the phenyl ring projected toward the solvent region. Figure 1. Drug discovery and development of clinical compound rezatapopt (PC14586), a first-in-class small molecule reactivator of p53 Y220C mutant protein. Cocrystal structure of lead compound <b>2</b> (PC-9859) complexed with p53 Y220C mutant protein (PDB code: 9BR4) is shown. The p53 Y220C mutant protein is shown as a cartoon. Hydrogen bonds formed between <b>2</b>, and the key residues within p53 Y220C mutant protein and water molecules are highlighted by red dashed lines. <b>2</b> is shown as green sticks. Key amino acid residues Cys220, Thr150, and Pro153 in p53 Y220C mutant protein are shown as yellow sticks. Water molecules are shown as red balls. As the optimal p53 Y220C reactivator, rezatapopt binds to p53 Y220C mutant in a similar mode to that of <b>2</b> and stabilizes its structure in the wide-type (WT) conformation, thereby restoring its tumor suppressive functions. The introduction of fluorine atom to the piperidine ring within rezatapopt further strengthened the interaction with p53 Y220C mutant protein, including increased hydrogen bond interaction with Thr150, enhanced conformational rigidity of piperidine ring, and reduced basicity. Moreover, the chiral conformation of fluorine-substituted piperidine ring showed obvious effect on protein binding and 3<i>S</i>,4<i>R</i> configuration is the optimal. Furthermore, the introduction of polar amide group to the phenyl ring remarkably improved the pharmacokinetic (PK) profiles. Rezatapopt significantly increases DNA binding to p53 Y220C (SC<sub>150</sub> = 9 nM). Furthermore, it potently suppresses the growth of NUGC-3 cells (a gastric cancer cell line with a <i>TP</i>53 Y220C mutation) with an IC<sub>50</sub> value of 504 nM, determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Moreover, rezatapopt (po, 50 mg/kg) displays desirable PK profiles in mouse (<i>C</i><sub>max</sub> = 16600 ng/mL and AUC<sub>0-last</sub> = 163342 ng.h/mL). Excitingly, rezatapopt (po, QD) at doses of both 25 mg/kg and 50 mg/kg, robustly suppresses the tumor growth in a NUGC-3 tumor xenograft mouse model (TGI = 33% and 71%, respectively). Moreover, rezatapopt causes a significant tumor growth suppression at a higher dose of 100 mg/kg, resulting in an impressive 80% tumor regression. Notably, in toxicological studies, rezatapopt presents a favorable safety profile. Given its potent <i>in vitro</i> and <i>in vivo</i> efficacy as well as favorable PK safety profiles, rezatapopt was advanced into human clinical trials. In a Phase I clinical trial (NCT04585750), rezatapopt displayed a favorable safety profile across all efficacious doses. (9) Moreover, rezatapopt, as a monotherapy, produced robust clinical efficacy in heavily pretreated patients with multiple solid tumor types harboring the <i>TP</i>53 Y220C mutation. (9) A Phase II clinical trial of rezatapopt (NCT04585750, https://clinicaltrials.gov) is currently recruiting, serving as a registrational study to investigate its safety, tolerability and efficacy as a monotherapy in participants with locally advanced or metastatic solid tumors harboring a <i>TP</i>53 Y220C mutation. In some cases, protein destabilization is the root cause of several classes of diseases, including tumor suppressive proteins (e.g., p53, p21, p57, FOXO3A, IRF3 and BAX), and mutated and misfolded proteins, such as ΔF508-cystic fibrosis transmembrane conductance regulator (ΔF508-CFTR) in cystic fibrosis, glucokinase in pancreatic cells in maturity-onset diabetes of the young type 2 (MODY2), and transthyretin (TTR) in cardiac amyloidosis (ATTR-CM). (10) Therefore, in such cases, target protein stabilization (TPS) rather than target protein degradation (TPD) would be therapeutically beneficial. TPS is attracting increasing attention of researchers from both academic and industrial settings and several molecule protein stabilizers have been developed. Inspiringly, two TTR stabilizers, including tafamidis developed by researchers at Pfizer and acoramidis developed by researchers at Stanford University, had been approved by the U.S. FDA for treating ATTR-CM in 2019 and 2024 respectively. (11) Notably, emergence of rezatapopt, the first-in-class stabilizer of p53 Y220C mutant in Phase II clinical trial, represents a great breakthrough in the field of drug discovery targeting p53, a traditionally considered “undruggable protein. Moreover, it also provides the basis for developing p53-based chemical inducers of proximity (CIPs). Besides monovalent stabilizers, several heterobifunctional stabilizers (CIPs) have been developed, including deubiquitinase-targeting chimeras (DUBTACs), (12) RESTORACs, and enhancement-targeting chimeras (ENTACs). (10) Biotech companies Vicinitas, Stablix, and Entact Bio are centering on the development of DUBTACs, RESTORACs and ENTACs, respectively. (13) With continued efforts, more and more protein stabilizers are anticipated to be developed as clinical trial drug candidates, conferring therapeutic benefits. In summary, the discovery of rezatapopt is an exciting breakthrough for drug discovery targeting p53 function restoration. Like drugs targeting KRAS G12C, rezatapopt targeting p53 Y220C shows potential to convert p53 from an “undruggable” to “druggable” target. Rezatapopt is currently in a phase II clinical trial (NCT04585750), benefiting patients with locally advanced or metastatic solid tumors harboring a <i>TP</i>53 Y220C mutation. More clinical results of rezatapopt will be achieved and released in the near future. This work was partially supported by R01CA226001 and R01CA231150 grants from the National Institutes of Health, Breast Cancer Research Program (BCRP) Breakthrough Awards W81XWH-17-1-0071 and W81XWH-17-1-0072 from the Department of Defense (DoD), the John D. Stobo, M.D. Distinguished Chair Endowment, and the Edith &amp; Robert Zinn Chair Endowment in Drug Discovery. This article references 13 other publications. This article has not yet been cited by other publications.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"183 1","pages":""},"PeriodicalIF":6.8000,"publicationDate":"2025-03-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of Medicinal Chemistry","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1021/acs.jmedchem.5c00670","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CHEMISTRY, MEDICINAL","Score":null,"Total":0}
引用次数: 0

Abstract

The p53 protein, an important transcription factor, plays a critical role in regulating cell responses to stress. (1,2) Coded by the TP53 gene, p53 is a well-recognized tumor suppressorprotein. Dysregulation of p53 function is closely associated with the initiation and development of various cancers. Although restoration of p53 function appears to be a promising therapeutic approach, p53 is considered to be a traditionally “undruggable” target owing to the lack of active binding pockets or frequent mutations. (3) Mutations in the TP53 gene can assist tumor cells to evade p53 suppressive effects, promoting cell proliferation, migration, and invasion, thereby aggravating tumorigenesis. TP53 genetic mutations present in more than 50% of human cancers, most of which are missense mutations occurring in the DNA-binding domain of p53 (amino acids 94–292). Y220C is the ninth most prevalent mutation among all TP53 mutations observed across various tumor types, accounting for 1.8%, and presents in ∼1% of human cancers. (4) Y220C mutation leads to tyrosine-to-cysteine substitution and creates a pocket in the p53 Y220C mutant protein. p53 Y220C protein is structurally unstable at physiological temperatures and can be ubiquitinated and delivered to proteasomal for degradation. Therefore, targeting p53 Y220C-mutant stabilization may be a viable therapeutic strategy for various associated cancers. Small molecule compounds targeting p53 Y220C stabilization are in active development for potential cancer therapeutics. (5) Rezatapopt (PC14586), discovered by PMV Pharmaceuticals, Inc., is a first-in-class small molecule stabilizer and reactivator of p53 Y220C currently in the Phase II clinical trial (NCT04585750) for treating various locally advanced or metastatic solid tumors harboring a p53 Y220C mutation. (6) In addition, rezatapopt is in a Phase Ib clinical trial (NCT06616636) in combination with azacitidine for patients with TP53 Y220C mutant myeloid malignancies (acute myeloid leukemia or myelodysplastic syndrome). Rezatapopt was designed to tightly occupy the pocket created by mutation-caused amino acid tyrosine-to-cysteine substitution in the p53 Y220C mutant protein. Rezatapopt was obtained based on structure-based drug design (SBDD) and systematical structural optimization, starting from hit compound 1 (Figure 1). The substrate concentration required to increase DNA binding by 1.5-fold (SC150) was determined by a time-resolved fluorescence resonance energy transfer (FRET) assay to assess the in vitro potency of the p53 Y220C reactivators. Hit compound 1 was obtained by scaffold combination of hit compounds PhiKan83 (SC150 = 37.2 μM) with a carbazole scaffold (7) and PK1596 (SC150 = 1.6 μM) with an iodophenol scaffold. (8) Compound 1 contains a widely recognized “privileged” indole scaffold in drug development, which exists in numerous natural products and bioactive molecules. Compound 1 has three anchors attached to the indole scaffold, including a hydrophobic ethyl group, a polar alkylamine extending to the solvent region, and a substituted acetylene occupying the adjacent subsite. Compound 1 (SC150 = 13.6 μM) showed inadequate potency (SC150 > 100 nM) and poor metabolic stability in human liver microsome (T1/2 = 4 min). SBDD efforts around 1 (SC150 = 13.6 μM) resulted in the lead compound 2 (PC-9859) with significantly improved DNA binding activity (SC150 = 54 nM). The cocrystal structure (PDB code: 9BR4) of p53 Y220C mutant protein in complex with 2, revealed that 2 locates at the pocket created by tyrosine-to-cysteine substitution in p53 Y220C mutant protein, with indole scaffold occupying this space (Figure 1). Trifluoromethyl group inserts deeply into the hydrophobic pocket, the 4-aminopiperidine ring extends toward the solvent-exposed region, and the pyridine ring is connected to indole core via an alkyne occupies the adjacent subsite. The pyridine ring forms a favorable CH-π stacking interaction with Pro153. Compound 2 forms two key hydrogen bonds with p53 Y220C mutant protein. One interaction is formed between the amino group on pyridine ring and the carbonyl of Cys220 while the other is formed between the amino group on 1-methylpiperidine ring and the carbonyl of Thr150 on the side chain. Further drug design and structural optimization around lead compound 2 has successfully led to the discovery of rezatapopt with a fluorine atom substitution on the piperidine ring and a polar amide group on the phenyl ring projected toward the solvent region. Figure 1. Drug discovery and development of clinical compound rezatapopt (PC14586), a first-in-class small molecule reactivator of p53 Y220C mutant protein. Cocrystal structure of lead compound 2 (PC-9859) complexed with p53 Y220C mutant protein (PDB code: 9BR4) is shown. The p53 Y220C mutant protein is shown as a cartoon. Hydrogen bonds formed between 2, and the key residues within p53 Y220C mutant protein and water molecules are highlighted by red dashed lines. 2 is shown as green sticks. Key amino acid residues Cys220, Thr150, and Pro153 in p53 Y220C mutant protein are shown as yellow sticks. Water molecules are shown as red balls. As the optimal p53 Y220C reactivator, rezatapopt binds to p53 Y220C mutant in a similar mode to that of 2 and stabilizes its structure in the wide-type (WT) conformation, thereby restoring its tumor suppressive functions. The introduction of fluorine atom to the piperidine ring within rezatapopt further strengthened the interaction with p53 Y220C mutant protein, including increased hydrogen bond interaction with Thr150, enhanced conformational rigidity of piperidine ring, and reduced basicity. Moreover, the chiral conformation of fluorine-substituted piperidine ring showed obvious effect on protein binding and 3S,4R configuration is the optimal. Furthermore, the introduction of polar amide group to the phenyl ring remarkably improved the pharmacokinetic (PK) profiles. Rezatapopt significantly increases DNA binding to p53 Y220C (SC150 = 9 nM). Furthermore, it potently suppresses the growth of NUGC-3 cells (a gastric cancer cell line with a TP53 Y220C mutation) with an IC50 value of 504 nM, determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Moreover, rezatapopt (po, 50 mg/kg) displays desirable PK profiles in mouse (Cmax = 16600 ng/mL and AUC0-last = 163342 ng.h/mL). Excitingly, rezatapopt (po, QD) at doses of both 25 mg/kg and 50 mg/kg, robustly suppresses the tumor growth in a NUGC-3 tumor xenograft mouse model (TGI = 33% and 71%, respectively). Moreover, rezatapopt causes a significant tumor growth suppression at a higher dose of 100 mg/kg, resulting in an impressive 80% tumor regression. Notably, in toxicological studies, rezatapopt presents a favorable safety profile. Given its potent in vitro and in vivo efficacy as well as favorable PK safety profiles, rezatapopt was advanced into human clinical trials. In a Phase I clinical trial (NCT04585750), rezatapopt displayed a favorable safety profile across all efficacious doses. (9) Moreover, rezatapopt, as a monotherapy, produced robust clinical efficacy in heavily pretreated patients with multiple solid tumor types harboring the TP53 Y220C mutation. (9) A Phase II clinical trial of rezatapopt (NCT04585750, https://clinicaltrials.gov) is currently recruiting, serving as a registrational study to investigate its safety, tolerability and efficacy as a monotherapy in participants with locally advanced or metastatic solid tumors harboring a TP53 Y220C mutation. In some cases, protein destabilization is the root cause of several classes of diseases, including tumor suppressive proteins (e.g., p53, p21, p57, FOXO3A, IRF3 and BAX), and mutated and misfolded proteins, such as ΔF508-cystic fibrosis transmembrane conductance regulator (ΔF508-CFTR) in cystic fibrosis, glucokinase in pancreatic cells in maturity-onset diabetes of the young type 2 (MODY2), and transthyretin (TTR) in cardiac amyloidosis (ATTR-CM). (10) Therefore, in such cases, target protein stabilization (TPS) rather than target protein degradation (TPD) would be therapeutically beneficial. TPS is attracting increasing attention of researchers from both academic and industrial settings and several molecule protein stabilizers have been developed. Inspiringly, two TTR stabilizers, including tafamidis developed by researchers at Pfizer and acoramidis developed by researchers at Stanford University, had been approved by the U.S. FDA for treating ATTR-CM in 2019 and 2024 respectively. (11) Notably, emergence of rezatapopt, the first-in-class stabilizer of p53 Y220C mutant in Phase II clinical trial, represents a great breakthrough in the field of drug discovery targeting p53, a traditionally considered “undruggable protein. Moreover, it also provides the basis for developing p53-based chemical inducers of proximity (CIPs). Besides monovalent stabilizers, several heterobifunctional stabilizers (CIPs) have been developed, including deubiquitinase-targeting chimeras (DUBTACs), (12) RESTORACs, and enhancement-targeting chimeras (ENTACs). (10) Biotech companies Vicinitas, Stablix, and Entact Bio are centering on the development of DUBTACs, RESTORACs and ENTACs, respectively. (13) With continued efforts, more and more protein stabilizers are anticipated to be developed as clinical trial drug candidates, conferring therapeutic benefits. In summary, the discovery of rezatapopt is an exciting breakthrough for drug discovery targeting p53 function restoration. Like drugs targeting KRAS G12C, rezatapopt targeting p53 Y220C shows potential to convert p53 from an “undruggable” to “druggable” target. Rezatapopt is currently in a phase II clinical trial (NCT04585750), benefiting patients with locally advanced or metastatic solid tumors harboring a TP53 Y220C mutation. More clinical results of rezatapopt will be achieved and released in the near future. This work was partially supported by R01CA226001 and R01CA231150 grants from the National Institutes of Health, Breast Cancer Research Program (BCRP) Breakthrough Awards W81XWH-17-1-0071 and W81XWH-17-1-0072 from the Department of Defense (DoD), the John D. Stobo, M.D. Distinguished Chair Endowment, and the Edith & Robert Zinn Chair Endowment in Drug Discovery. This article references 13 other publications. This article has not yet been cited by other publications.

Abstract Image

Rezatapopt (PC14586):临床试验中的一类小分子p53 Y220C突变蛋白稳定剂
p53蛋白是一种重要的转录因子,在调节细胞对应激的反应中起着关键作用。(1,2) p53由TP53基因编码,是一种公认的肿瘤抑制蛋白。p53功能失调与多种癌症的发生和发展密切相关。尽管恢复p53功能似乎是一种很有前途的治疗方法,但由于缺乏活性结合袋或频繁突变,p53被认为是传统上“不可药物”的靶标。(3) TP53基因突变可帮助肿瘤细胞逃避p53的抑制作用,促进细胞增殖、迁移和侵袭,从而加重肿瘤发生。TP53基因突变存在于超过50%的人类癌症中,其中大多数是发生在p53 dna结合区域的错义突变(氨基酸94-292)。Y220C是在各种肿瘤类型中观察到的所有TP53突变中的第九大常见突变,占1.8%,存在于约1%的人类癌症中。(4) Y220C突变导致酪氨酸到半胱氨酸的替代,并在p53 Y220C突变蛋白中产生一个口袋。p53 Y220C蛋白在生理温度下结构不稳定,可被泛素化并传递到蛋白酶体降解。因此,靶向p53 y220c突变体稳定可能是一种可行的治疗各种相关癌症的策略。靶向p53 Y220C稳定的小分子化合物正积极开发用于潜在的癌症治疗。(5) Rezatapopt (PC14586)由PMV Pharmaceuticals, Inc.发现,是一种小分子稳定剂和p53 Y220C再激活剂,目前处于II期临床试验(NCT04585750),用于治疗含有p53 Y220C突变的各种局部晚期或转移性实体肿瘤。(6)此外,rezatapopt正在进行一项Ib期临床试验(NCT06616636),联合阿扎胞苷治疗TP53 Y220C突变型髓系恶性肿瘤(急性髓系白血病或骨髓增生异常综合征)患者。Rezatapopt被设计成紧紧占据突变引起的p53 Y220C突变蛋白中氨基酸酪氨酸到半胱氨酸取代所产生的口袋。Rezatapopt是基于基于结构的药物设计(SBDD)和系统的结构优化获得的,从hit化合物1开始(图1)。通过时间分辨率荧光共振能量转移(FRET)测定将DNA结合增加1.5倍(SC150)所需的底物浓度,以评估p53 Y220C再激活剂的体外效力。Hit化合物1由Hit化合物PhiKan83 (SC150 = 37.2 μM)与咔唑支架(7)和PK1596 (SC150 = 1.6 μM)与碘酚支架组合而成。(8)化合物1含有药物开发中公认的“特权”吲哚支架,存在于许多天然产物和生物活性分子中。化合物1具有附着在吲哚支架上的三个锚点,包括疏水乙基、延伸至溶剂区域的极性烷基胺和占据相邻亚位的取代乙炔。化合物1 (SC150 = 13.6 μM)效价不足(SC150 &gt;100 nM),人肝微粒体代谢稳定性差(T1/2 = 4 min)。在1 (SC150 = 13.6 μM)附近进行SBDD处理,先导化合物2 (PC-9859)的DNA结合活性显著提高(SC150 = 54 nM)。p53 Y220C突变蛋白与2复合物的共晶结构(PDB代码:9BR4)显示,2位于p53 Y220C突变蛋白酪氨酸与半胱氨酸取代形成的口袋中,吲哚支架占据了这个空间(图1)。三氟甲基深入到疏水口袋中,4-氨基吡啶环向溶剂暴露区延伸,吡啶环通过一个占据邻近亚位的炔与吲哚核相连。吡啶环与Pro153形成良好的CH-π堆叠相互作用。化合物2与p53 Y220C突变蛋白形成两个关键氢键。吡啶环上的氨基与Cys220的羰基形成相互作用,1-甲基哌啶环上的氨基与侧链上的Thr150的羰基形成相互作用。围绕先导化合物2的进一步药物设计和结构优化成功地发现了rezatapopt,其哌啶环上有一个氟原子取代,苯基环上有一个极性酰胺基团指向溶剂区。图1所示。p53 Y220C突变蛋白小分子活化物rezatapopt (PC14586)的药物发现与开发显示了先导化合物2 (PC-9859)与p53 Y220C突变蛋白(PDB代码:9BR4)络合的共晶结构。p53 Y220C突变蛋白如图所示。2之间形成氢键,p53 Y220C突变蛋白和水分子内的关键残基用红色虚线表示。 2用绿色条表示。p53 Y220C突变蛋白中的关键氨基酸残基Cys220、Thr150和Pro153用黄色棒表示。水分子用红球表示。rezatapopt作为最理想的p53 Y220C再激活剂,以与2相似的方式与p53 Y220C突变体结合,使其结构稳定在宽型(WT)构象,从而恢复其抑瘤功能。rezatapopt内哌啶环引入氟原子,进一步加强了与p53 Y220C突变蛋白的相互作用,包括与Thr150的氢键相互作用增加,哌啶环构象刚性增强,碱度降低。此外,氟取代哌替啶环的手性构象对蛋白质结合有明显影响,3S、4R构象是最佳构象。此外,在苯环上引入极性酰胺基团显著改善了药代动力学(PK)谱。Rezatapopt显著增加p53 Y220C的DNA结合(SC150 = 9 nM)。此外,通过3-(4,5-二甲基噻唑-2-基)-2,5-二苯基溴化四唑(MTT)试验,它能有效抑制NUGC-3细胞(一种TP53 Y220C突变的胃癌细胞系)的生长,IC50值为504 nM。此外,rezatapopt (po, 50 mg/kg)在小鼠体内表现出良好的PK谱(Cmax = 16600 ng/mL, AUC0-last = 163342 ng.h/mL)。令人兴奋的是,25 mg/kg和50 mg/kg剂量的rezatapopt (po, QD)在NUGC-3肿瘤异种移植小鼠模型中均能显著抑制肿瘤生长(TGI分别为33%和71%)。此外,rezatapopt在100 mg/kg的较高剂量下可显著抑制肿瘤生长,导致令人印象深刻的80%肿瘤消退。值得注意的是,在毒理学研究中,rezatapopt表现出良好的安全性。鉴于rezatapopt在体外和体内的有效作用以及良好的PK安全性,rezatapopt已进入人体临床试验阶段。在一项I期临床试验(NCT04585750)中,rezatapopt在所有有效剂量中都显示出良好的安全性。(9)此外,rezatapopt作为一种单药治疗,在含有TP53 Y220C突变的多种实体肿瘤类型的重度预处理患者中具有强大的临床疗效。(9) rezatapopt (NCT04585750, https://clinicaltrials.gov)的II期临床试验目前正在招募,作为一项注册研究,研究其作为单药治疗TP53 Y220C突变的局部晚期或转移性实体瘤患者的安全性、耐受性和有效性。在某些情况下,蛋白质不稳定是几类疾病的根本原因,包括肿瘤抑制蛋白(如p53、p21、p57、FOXO3A、IRF3和BAX),以及突变和错误折叠的蛋白,如囊性纤维化中的ΔF508-cystic纤维化跨膜传导调节剂(ΔF508-CFTR),年轻2型糖尿病成熟期胰腺细胞中的葡萄糖激酶(MODY2),以及心脏淀粉样变性(atr - cm)中的转甲状腺素(TTR)。(10)因此,在这种情况下,靶蛋白稳定(TPS)而不是靶蛋白降解(TPD)将在治疗上有益。TPS越来越受到学术界和工业界研究人员的关注,已经开发出了几种分子蛋白稳定剂。令人鼓舞的是,两种TTR稳定剂,包括辉瑞研究人员开发的他法米迪斯和斯坦福大学研究人员开发的acoramidis,分别于2019年和2024年获得美国FDA批准用于治疗atr - cm。(11)值得注意的是,在II期临床试验中,p53 Y220C突变体的同类首个稳定剂rezatapopt的出现,代表了针对p53这个传统上被认为是“不可药物”的蛋白的药物发现领域的重大突破。这也为开发基于p53的化学邻近诱导剂(CIPs)提供了基础。除了单价稳定剂外,还开发了几种异双功能稳定剂(cip),包括去泛素酶靶向嵌合体(DUBTACs), (12) RESTORACs和靶向增强嵌合体(ENTACs)。(10) Vicinitas、Stablix、Entact Bio等生物技术公司正在集中开发dubtac、RESTORACs、ENTACs。(13)随着不断的努力,越来越多的蛋白质稳定剂有望作为临床试验候选药物开发出来,并带来治疗效果。总之,rezatapopt的发现是靶向p53功能恢复药物发现的一个令人兴奋的突破。与靶向KRAS G12C的药物一样,靶向p53 Y220C的rezatapopt显示出将p53从“不可药物”转化为“可药物”靶标的潜力。Rezatapopt目前处于II期临床试验(NCT04585750),使局部晚期或转移性实体肿瘤患者受益,这些肿瘤携带TP53 Y220C突变。在不久的将来,rezatapopt将获得更多的临床结果并发布。 这项工作得到了美国国立卫生研究院R01CA226001和R01CA231150拨款的部分支持,乳腺癌研究计划(BCRP)突破奖W81XWH-17-1-0071和W81XWH-17-1-0072来自国防部(DoD), John D. Stobo, M.D.杰出主席捐赠基金和Edith &amp;罗伯特·津恩,药物发现基金主席。本文引用了其他13个出版物。这篇文章尚未被其他出版物引用。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Journal of Medicinal Chemistry
Journal of Medicinal Chemistry 医学-医药化学
CiteScore
4.00
自引率
11.00%
发文量
804
审稿时长
1.9 months
期刊介绍: The Journal of Medicinal Chemistry is a prestigious biweekly peer-reviewed publication that focuses on the multifaceted field of medicinal chemistry. Since its inception in 1959 as the Journal of Medicinal and Pharmaceutical Chemistry, it has evolved to become a cornerstone in the dissemination of research findings related to the design, synthesis, and development of therapeutic agents. The Journal of Medicinal Chemistry is recognized for its significant impact in the scientific community, as evidenced by its 2022 impact factor of 7.3. This metric reflects the journal's influence and the importance of its content in shaping the future of drug discovery and development. The journal serves as a vital resource for chemists, pharmacologists, and other researchers interested in the molecular mechanisms of drug action and the optimization of therapeutic compounds.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信