1
|
Lu Y, Niu S, Zhang G, Guo Y, Fu B, Wang M, Liu J, Zhang H, Lu W, Zhang M. Antagonistic interaction between miR-143 and KRAS gene regulating male mouse germ cell apoptosis. Theriogenology 2025; 235:121-133. [PMID: 39823829 DOI: 10.1016/j.theriogenology.2024.12.024] [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: 08/13/2023] [Revised: 12/21/2024] [Accepted: 12/26/2024] [Indexed: 01/20/2025]
Abstract
Precisely regulated spermatocyte growth, differentiation, and apoptosis are crucial for sustainable male fertility. miR-143 has been demonstrated to regulate gene expression and cell apoptosis in various human cancers. However, the function of mmu-mir-143 (miR-143) in mammalian testes and its underlying mechanism remains unexplored. In this study, the expression of miR-143 was detected in C57BL/6 mice spermatocytes by in situ hybridization (ISH) and immunofluorescence (IF) co-staining and transfecting miR-143 inhibitor into GC-2 cells (mouse spermatogenic cells) shows that miR-143 inhibits cleaved Caspase 3 (CC3)-induced male germ cell death. The current study used IF co-staining of KI67 and γ-H2A.X in the testes of C57BL/6 mice at different developmental stages, revealing that active proliferation and apoptosis of spermatocytes occurred simultaneously in the testes at 14 day post-partum (dpp). Kras was predicted as a potential target of miR-143 in mice using of the online database TargetScan, verified by quantitative real-time PCR (qPCR), western blotting (WB), and Dual-luciferase reporter gene assay. Co-transfection of miR-143 inhibitor and Kras siRNA into GC-2 cells revealed an antagonistic correlation between miR-143 and Kras in regulating male germ cell death. Finally, miR-143 inhibitor and mimics were administered into the seminiferous tubule of 3-week-old C57BL/6 mice. The histomorphology, IF co-staining, and WB data indicated that the testes treated with the miR-143 inhibitor showed significantly aberrant phenotypes, including damaged seminiferous tubules, reduced spermatocyte quantity, and elevated levels of apoptosis. This study uncovered the mechanism by which miR-143 inhibits male germ cell apoptosis through the repression of Kras/KRAS levels and the inhibition of Caspase 3 activation, providing insight into the role of miRNA in spermatogenesis and the maintenance of male fertility.
Collapse
Affiliation(s)
- Yu Lu
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China
| | - Shudong Niu
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China
| | - Guisheng Zhang
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China
| | - Yanfeng Guo
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China
| | - Baotong Fu
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China
| | - Miaomiao Wang
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China
| | - Jianan Liu
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China
| | - Haili Zhang
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China
| | - Wen Lu
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China
| | - Ming Zhang
- College of Agriculture and Bioengineering, Heze University, Heze, 274000, China.
| |
Collapse
|
2
|
Zhao T, Liang SH. Novel Tricyclic KRAS Inhibitors for the Treatment of Cancer. ACS Med Chem Lett 2025; 16:178-179. [PMID: 39967651 PMCID: PMC11831557 DOI: 10.1021/acsmedchemlett.4c00609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Indexed: 02/20/2025] Open
Abstract
The invention described in this patent application relates to oral compounds represented by Formula I. These compounds are designed to specifically target and inhibit KRAS alleles, providing both therapeutic and prophylactic potential in mammals, particularly for the treatment of KRAS-driven cancers.
Collapse
Affiliation(s)
- Taoqian Zhao
- Department of Radiology and
Imaging Sciences, Emory University, Atlanta, Georgia 30322, United States
| | - Steven H. Liang
- Department of Radiology and
Imaging Sciences, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
3
|
Pandey D, Roy KK. Decoding KRAS dynamics: Exploring the impact of mutations and inhibitor binding. Arch Biochem Biophys 2025; 764:110279. [PMID: 39710177 DOI: 10.1016/j.abb.2024.110279] [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: 06/05/2024] [Revised: 11/25/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
KRAS (Kirsten rat sarcoma viral oncogene homologue), the most common mutated protein in human cancers, is the leading cause of morbidity and mortality. Before Sotorasib (AMG-510) was approved for non-small cell lung cancer treatment in 2020, the oncogenic KRAS mutations were believed to be non-druggable. High-resolution X-ray crystal structures of GDP-bound KRAS mutants with and without inhibitor are resolved and deposited in the Protein Data Bank (PDB). Nevertheless, to develop inhibitors targeting oncogenic KRAS mutants, understanding the dynamics of protein conformations and respective binding sites is crucial. In the present study, multiple molecular dynamics (MD) simulations were conducted on wild-type and mutant KRAS structures to understand how G12C or G12D mutations lead to the stabilization of the active state and how KRAS inhibitors lock the mutated conformations in their inactive state. The study found that the guanosine diphosphate (GDP)-bound KRAS mutants, G12C and G12D, were locked in the inactive state, in terms of stability, when the KRAS inhibitors, AMG-510 and MRTX1133, respectively, bind to the respective Switch-II (S-II) pocket. Covalent inhibitor AMG-510 locked the inactive GDP-bound KRASG12C mutant more efficiently when compared to the non-covalent inhibitor MRTX1133. The Cα atom distance between key highly dynamic amino acids from P-loop, Switch-I, and Switch-II domains, lying within 4 Å of the inhibitor, were stable in the KRAS mutant with bound inhibitors (AMG-510 or MRTX1133), but were varying largely in the absence of any inhibitor throughout the microsecond simulation. According to the per-residue energy decomposition results, S-II amino acids in inhibitor-free KRASG12C and KRASG12D mutants showed larger variations in energy values as compared to AMG-510-bound KRASG12C and MRTX1133-bound KRASG12D, respectively. For example, the inhibitor-free KRASG12C exhibited larger variations in energy values in the S-II residues, namely, Thr58, Gln61, Glu63, and Arg68, as compared to the AMG-510-bound KRASG12C. The study found that the higher stability of AMG-510 in torsion angles was due to its covalent nature of binding to the KRASG12C mutant. The S-II amino acids, namely, Thr58, Glu63, and Arg68 remained stable in AMG-510-bound KRASG12C. The study showed that AMG-510 binding significantly stabilizes the amino acids surrounding it, surpassing that of MRTX1133. The insights gained in the present study is expected to be useful in the design and development of new KRAS-targeted drugs.
Collapse
Affiliation(s)
- Divya Pandey
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, 248007, Uttarakhand, India
| | - Kuldeep K Roy
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, 248007, Uttarakhand, India.
| |
Collapse
|
4
|
Jafary B, Akbarzadeh-Khiavi M, Farzi-Khajeh H, Safary A, Adibkia K. EGFR-targeting RNase A-cetuximab antibody-drug conjugate induces ROS-mediated apoptosis to overcome drug resistance in KRAS mutant cancer cells. Sci Rep 2025; 15:1483. [PMID: 39789190 PMCID: PMC11718297 DOI: 10.1038/s41598-025-85856-9] [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: 08/24/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025] Open
Abstract
Antibody-drug conjugates (ADCs) are an emerging strategy in cancer therapy, enhancing precision and efficacy by linking targeted antibodies to potent cytotoxic agents. This study introduces a novel ADC that combines ribonuclease A (RNase A) with cetuximab (Cet), an anti-EGFR monoclonal antibody, through a polyethylene glycol (PEG) linker (RN-PEG-Cet), aimed to induce apoptosis in KRAS mutant colorectal cancer (CRC) via a ROS-mediated pathway. RN-PEG-Cet was successfully synthesized and characterized for its physicochemical properties, retaining full enzymatic activity in RNA degradation and high binding affinity to EGFR. In KRAS mutant SW-480 cells, RN-PEG-Cet significantly reduced cell viability at lower doses, with an IC50 of 11.7 µg/mL at 72 h. Compared to free Cet, RN-PEG-Cet demonstrated a ~ 2-fold increase in apoptosis and a ~ 3.5-fold increase in ROS production. The conjugate also disrupted the Nrf2/Keap1 pathway, with a significant upregulation of Keap1 (FC = 3.7, p ≤ 0.01) and downregulation of Nrf2 (FC = 3.3, p < 0.01), highlighting its role in impairing antioxidant defenses and promoting ROS-mediated cytotoxicity. These findings emphasize the potential of RN-PEG-Cet as a novel therapeutic approach for KRAS mutant CRC, offering superior apoptosis induction and targeted cytotoxicity compared to conventional therapies. This ADC could represent a new strategy for improving CRC treatment outcomes by effectively overcoming resistance mechanisms.
Collapse
Affiliation(s)
- Bita Jafary
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Akbarzadeh-Khiavi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hamed Farzi-Khajeh
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azam Safary
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Khosro Adibkia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Xiao C, Zhang X, Hou B, Wan P, Cao Z, Rao X. CYFIP2: potential pancreatic cancer biomarker and immunotherapeutic target. Discov Oncol 2024; 15:847. [PMID: 39739214 PMCID: PMC11685368 DOI: 10.1007/s12672-024-01650-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 11/28/2024] [Indexed: 01/02/2025] Open
Abstract
OBJECTIVE It has been shown that the CYFIP2 (Cytoplasmic FMR1-interacting protein 2) gene is apoptosis p53-dependent and is associated with poor prognosis in malignant tumors such as gastric cancer and other and cervical cancer. However, the prognostic potential of CYFIP2 in pancreatic cancer remains unclear. In this work, we first explain the great potential of CYFIP2 malignant progression from a broader perspective (pan-cancer) and confirm its oncogenic value in pancreatic cancer. METHODS The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx) database, CELL and GEO databases were utilized to explore the distribution of the CYFIP2 gene in human cancers. Data were analyzed using a variety of web-based platforms and software such as R (4.3.2), UCSC, MethSurv, Cytoscape (v3.10.2), UALAND, STRING, TISIDB, Harmonizome 3.0, TIMER 2.0, TCIA and TIDE. The R packages R packages ?limma? and ?ggplot2? were used to compare and visualize CYFIP2 mRNA expression. The R packages ?survminer? and ?survival? were used to statistically analyze the relationship between CYFIP2 expression and the survival and prognosis of tumor patients. the R package ?ss GSEA? was used to assess the correlation between CYFIP2 expression and immune infiltration. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Variation Analysis (GSVA) were used to explore the multiple biological functions and regulatory pathways in which CYFIP2 co-expressed genes co-engage in pancreatic cancer. The R package ?maftool? was used to explore somatic mutation information in pancreatic cancer, and the TIDE database and the R package ?oncoPredic? were used to explore immunotherapeutic responses and sensitive drugs. Pancreatic cancer cell lines were constructed with the specific expression of CYFIP2 mRNA, and their invasive and metastatic abilities were analysed using scratch and transwell assays. RESULTS Bioinformatics analysis and in vitro experiments confirmed that CYFIP2 was differentially expressed in a variety of tumors and correlated with clinical stage, and could be used as a potential marker for evaluating the prognosis and immunotherapy of a variety of tumors, including KIRC (Kidney renal clear cell carcinoma), PAAD (Pancreatic adenocarcinoma), PAAD (Pancreatic adenocarcinoma), SKCM (Skin Cutaneous Melanoma), and UCEC (Uterine Corpus Endometrial Carcinoma).Mutations in the CYFIP2 gene and methylation indices can affect the prognosis of tumor patients. In addition, we found that CYFIP2 expression values in pancreatic cancer were positively correlated with the expression of most immune cells, especially CD8?+?T Cells, and significantly negatively correlated with macrophages M0, and negatively correlated with the metastatic and invasive ability of pancreatic cancer cells, which may provide new strategies and ideas for pancreatic cancer immunotherapy.
Collapse
Affiliation(s)
- Cong Xiao
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Xiaojuan Zhang
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Bobo Hou
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Ping Wan
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Zhenjun Cao
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Xuefeng Rao
- The First Affiliated Hospital of Nanchang University, Nanchang University, 17 Yongwai Zhengjie, Donghu District, Nanchang, 330006, People's Republic of China.
| |
Collapse
|
6
|
Guo Y, Jin Y, Gao J, Wang D, Wang Y, Shan L, Yang M, Li X, Ma K. Bufadienolides from Chansu Injection Synergistically Enhances the Antitumor Effect of Erlotinib by Inhibiting the KRAS Pathway in Pancreatic Cancer. Pharmaceuticals (Basel) 2024; 17:1696. [PMID: 39770538 PMCID: PMC11677899 DOI: 10.3390/ph17121696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/01/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Background and Objectives: The Chansu injection (CSI), a sterile aqueous solution derived from Chansu, is applied in clinical settings to support antitumor and anti-radiation treatments. CSI's principal active components, bufadienolides (≥90%), demonstrate potential effects on pancreatic cancer (PDAC), but their underlying mechanisms remain unclear. This study aimed to elucidate the antitumor effects and pathways associated with CSI in PDAC. Methods: Network pharmacology and bioinformatics analyses explored CSI's mechanisms against PDAC. MTT, colony-formation, and migration assays evaluated CSI's impact on proliferation and migration in PANC-1 and MIA PACA-2 cells, both as a single agent and in combination with erlotinib (EGFR inhibitor). Cell cycle analysis employed flow cytometry. Animal experiments were performed on tumor-bearing mice, with targets and pathways assessed via molecular docking and western blotting. Results: CSI treatment suppressed PDAC cell proliferation and migration by inducing G2/M phase arrest. Network pharmacology, bioinformatics, and molecular docking indicated that CSI's anti-PDAC effects may involve EGFR pathway modulation, with CSI lowering p-EGFR/KRAS/p-ERK1/2 pathway expressions in PDAC cells. Additionally, sustained KRAS activation in mediating erlotinib resistance in PDAC and CSI potentiated erlotinib's antitumor effects through enhanced KRAS and p-ERK1/2 inhibition. CSI also enhanced erlotinib's efficacy in tumor-bearing mice without causing detectable toxicity in renal, cardiac, or hepatic tissues at therapeutic doses. Conclusions: CSI as an adjuvant used in antitumor and anti-radiation therapies enhanced erlotinib's antitumor effects through modulation of the KRAS pathway. CSI and erlotinib's synergistic interaction represents a promising approach for addressing erlotinib resistance in PDAC treatment.
Collapse
Affiliation(s)
- Yanli Guo
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China; (Y.G.); (Y.J.); (D.W.); (Y.W.); (L.S.); (M.Y.)
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Yu Jin
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China; (Y.G.); (Y.J.); (D.W.); (Y.W.); (L.S.); (M.Y.)
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Jie Gao
- Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China;
| | - Ding Wang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China; (Y.G.); (Y.J.); (D.W.); (Y.W.); (L.S.); (M.Y.)
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Pathophysiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Yanming Wang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China; (Y.G.); (Y.J.); (D.W.); (Y.W.); (L.S.); (M.Y.)
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Liya Shan
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China; (Y.G.); (Y.J.); (D.W.); (Y.W.); (L.S.); (M.Y.)
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Mengyu Yang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China; (Y.G.); (Y.J.); (D.W.); (Y.W.); (L.S.); (M.Y.)
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Xinzhi Li
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China; (Y.G.); (Y.J.); (D.W.); (Y.W.); (L.S.); (M.Y.)
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Pathophysiology, Shihezi University School of Medicine, Shihezi 832003, China
| | - Ketao Ma
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China; (Y.G.); (Y.J.); (D.W.); (Y.W.); (L.S.); (M.Y.)
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, First Affiliated Hospital, Shihezi University School of Medicine, Shihezi 832003, China
- Department of Physiology, Shihezi University School of Medicine, Shihezi 832003, China
| |
Collapse
|
7
|
Kidane M, Hoffman RM, Wolfe-Demarco JK, Huang TY, Teng CL, Samanta S, Gonzalez Lira LM, Lin-Jones J, Pallares G, Lamerdin JE, Servant NB, Lee CY, Yang CT, Bernatchez JA. Suite of Biochemical and Cell-Based Assays for the Characterization of Kirsten Rat Sarcoma (KRAS) Inhibitors and Degraders. ACS Pharmacol Transl Sci 2024; 7:3921-3934. [PMID: 39698278 PMCID: PMC11651172 DOI: 10.1021/acsptsci.4c00450] [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: 07/24/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024]
Abstract
KRAS is an important oncogenic driver which is mutated in numerous cancers. Recent advances in the selective targeting of KRAS mutants via small molecule inhibitors and targeted protein degraders have generated an increase in research activity in this area in recent years. As such, there is a need for new assay platforms to profile next generation inhibitors which improve on the potency and selectivity of existing drug candidates, while evading the emergence of resistance. Here, we describe the development of a new panel of biochemical and cell-based assays to evaluate the binding and function of known chemical entities targeting mutant KRAS. Our assay panels generated selectivity profiles and quantitative binding interaction dissociation constants for small molecules and degraders against wild type, G12C, G12D, and G12V KRAS, which were congruent with published data. These assays can be leveraged for additional mutants of interest beyond those described in this study, using both overexpressed cell-free systems and cell-based systems with endogenous protein levels.
Collapse
Affiliation(s)
- Medhanie Kidane
- Research
and Development and Technology Transfer, Eurofins DiscoverX, LLC, 11180 Roselle Street Suite D, San Diego, California 92121, United States
| | - Rene M. Hoffman
- Research
and Development, Eurofins DiscoverX Products,
LLC, 42501 Albrae Street, Fremont, California 94538, United States
| | - Jennifer K. Wolfe-Demarco
- Research
and Development and Technology Transfer, Eurofins DiscoverX, LLC, 11180 Roselle Street Suite D, San Diego, California 92121, United States
| | - Ting-Yu Huang
- Eurofins
Panlabs Discovery Services Taiwan, Ltd., 25 Wugong Sixth Road, Wugu District, New Taipei City 24891, Taiwan
| | - Chi-Ling Teng
- Eurofins
Panlabs Discovery Services Taiwan, Ltd., 25 Wugong Sixth Road, Wugu District, New Taipei City 24891, Taiwan
| | - Saheli Samanta
- Research
and Development, Eurofins DiscoverX Products,
LLC, 42501 Albrae Street, Fremont, California 94538, United States
| | - Luis M. Gonzalez Lira
- Research
and Development and Technology Transfer, Eurofins DiscoverX, LLC, 11180 Roselle Street Suite D, San Diego, California 92121, United States
| | - Jennifer Lin-Jones
- Research
and Development, Eurofins DiscoverX Products,
LLC, 42501 Albrae Street, Fremont, California 94538, United States
| | - Gabriel Pallares
- Research
and Development and Technology Transfer, Eurofins DiscoverX, LLC, 11180 Roselle Street Suite D, San Diego, California 92121, United States
| | - Jane E. Lamerdin
- Research
and Development, Eurofins DiscoverX Products,
LLC, 42501 Albrae Street, Fremont, California 94538, United States
| | - Nicole B. Servant
- Research
and Development and Technology Transfer, Eurofins DiscoverX, LLC, 11180 Roselle Street Suite D, San Diego, California 92121, United States
| | - Chun-Yao Lee
- Eurofins
Panlabs Discovery Services Taiwan, Ltd., 25 Wugong Sixth Road, Wugu District, New Taipei City 24891, Taiwan
| | - Chao-Tsung Yang
- Research
and Development, Eurofins DiscoverX Products,
LLC, 42501 Albrae Street, Fremont, California 94538, United States
| | - Jean A. Bernatchez
- Research
and Development and Technology Transfer, Eurofins DiscoverX, LLC, 11180 Roselle Street Suite D, San Diego, California 92121, United States
| |
Collapse
|
8
|
Liu J, Wang Z, Tian X, Xie B, Liu K. ETS1 Promotes Aerobic Glycolysis and Growth in Head and Neck Squamous Cell Carcinoma by Targeting RRAS2. Biochem Genet 2024:10.1007/s10528-024-10996-y. [PMID: 39661306 DOI: 10.1007/s10528-024-10996-y] [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: 06/04/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a prevalent malignancy with a five-year survival rate below 50%, highlighting the urgent need for novel therapeutic targets. This study explores the role of the small GTPase RRAS2 in HNSCC progression and its regulation of glycolysis. Analysis of data from the TCGA and GTEx databases revealed that RRAS2 is significantly upregulated in HNSCC tissues and is associated with poorer overall patient survival. Functional experiments demonstrated that silencing RRAS2 in HNSCC cell lines inhibits glycolytic activity and cell proliferation while promoting apoptosis, whereas overexpression of RRAS2 enhances glycolysis and cell growth. Additionally, bioinformatics and experimental approaches identified the transcription factor ETS1 as an upstream regulator of RRAS2. ETS1 binds to the RRAS2 promoter, facilitating its transcription and contributing to metabolic reprogramming in HNSCC cells. Rescue experiments confirmed that the ETS1-RRAS2 axis is crucial for maintaining the glycolytic phenotype and proliferative capacity of HNSCC cells. These findings suggest that the ETS1-RRAS2 pathway plays a critical role in HNSCC progression and metabolic adaptation, positioning RRAS2 as a potential therapeutic target for improving patient outcomes.
Collapse
Affiliation(s)
- Jianguo Liu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, P.R. China
| | - Zhi Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, P.R. China
| | - Xiaoyan Tian
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, P.R. China
| | - Bingbin Xie
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, P.R. China
| | - Ke Liu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, P.R. China.
| |
Collapse
|
9
|
Wang SF, Wu CH, Sung KF, Tsou YK, Lin CH, Lee MH, Liu NJ. The Distribution and Predictive Factor of Extra-Pancreatic Malignancy Occurrence in Patients with Pancreatic Intraductal Papillary Mucinous Neoplasm-A Ten-Year Follow-Up Case-Control Study in Taiwan. Cancers (Basel) 2024; 16:4102. [PMID: 39682288 DOI: 10.3390/cancers16234102] [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: 10/17/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND AND AIMS A higher incidence of extra-pancreatic malignancies (EPMs) in patients with pancreatic intraductal papillary mucinous neoplasm (IPMN) than in the general population has been shown in several studies. We suppose that EPMs also occur after IPMN has been diagnosed, but few reports have discussed the risk factors that have been identified, except for old age, which was only noted in one study. Our study aims to recognize the distribution of EPMs in Taiwanese patients with a longer duration of follow-up and investigate the risk factors to predict EPMs in IPMN patients. METHODS We retrospectively analyzed 114 patients with pancreatic IPMN from 1 January 2010 to 31 December 2014 in Chang Gung Memorial Hospital. The characteristics of the patients were all recorded. Different EPMs are demonstrated as occurring before, concurrently with, or after IPMN diagnosis. The risk factors were compared between patients with or without an EPM. RESULTS After an average follow-up duration of 10.45 years, 47 EPMs occurred in 42 patients (36.8%), and over half were found after IPMN was diagnosed (55.3%). The most common EPMs were colon cancer and lung cancer (21.3%). Moreover, cyst size progression was highly associated with EPM occurrence (p = 0.004) and predictive of EPM occurrence after IPMN (p = 0.002), with a cut-off value of 1 cm (accuracy: 79%; sensitivity: 88%; specificity: 58%). CONCLUSIONS Colon cancer and lung cancer account for the majority EPMs in Taiwan. EPMs were also frequently found after IPMN diagnosis when the follow-up duration was prolonged up to 10.45 years. Cyst size progression is a risk factor of EPM after IPMN diagnosis and we suggest a cut-off value of 1 cm for clinical utility.
Collapse
Affiliation(s)
- Sheng-Fu Wang
- Department of Gastroenterology and Hepatology, Chang-Gung Memorial Hospital, Linkou Medical Center, Taoyuan 333, Taiwan
- School of Medicine, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | - Chi-Huan Wu
- Department of Gastroenterology and Hepatology, Chang-Gung Memorial Hospital, Linkou Medical Center, Taoyuan 333, Taiwan
- School of Medicine, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | - Kai-Feng Sung
- Department of Gastroenterology and Hepatology, Chang-Gung Memorial Hospital, Linkou Medical Center, Taoyuan 333, Taiwan
- School of Medicine, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | - Yung-Kuan Tsou
- Department of Gastroenterology and Hepatology, Chang-Gung Memorial Hospital, Linkou Medical Center, Taoyuan 333, Taiwan
- School of Medicine, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | - Cheng-Hui Lin
- Department of Gastroenterology and Hepatology, Chang-Gung Memorial Hospital, Linkou Medical Center, Taoyuan 333, Taiwan
- School of Medicine, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | - Mu-Hsien Lee
- Department of Gastroenterology and Hepatology, Chang-Gung Memorial Hospital, Linkou Medical Center, Taoyuan 333, Taiwan
- School of Medicine, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | - Nai-Jen Liu
- Department of Gastroenterology and Hepatology, Chang-Gung Memorial Hospital, Linkou Medical Center, Taoyuan 333, Taiwan
- School of Medicine, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
10
|
Mo C, Chadha B, Kuang C. An Evolving Landscape: New Therapies for Metastatic Colorectal Cancer. Clin Colorectal Cancer 2024; 23:337-345. [PMID: 39332920 PMCID: PMC11608151 DOI: 10.1016/j.clcc.2024.08.003] [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: 07/20/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/29/2024]
Abstract
Substantial progress is being made in the development of novel therapies directed against colorectal cancer. The discovery of various molecular markers and advances in tumor profiling have facilitated the development of new targeted agents and immunotherapy. Not only have these drugs improved progression-free survival and even overall survival in some cases, but their related outcomes have also raised questions as to how to best combine or sequence therapies for even greater efficacy. Furthermore, we are beginning to understand how these combination therapies may yield for greater therapeutic response for patients with microsatellite stable colorectal cancer for which there is much need for improvement. In this article, we review recent trial data and explore the outcomes of various targeted therapies and immunotherapies for patient with advanced colorectal cancer.
Collapse
Affiliation(s)
- Christiana Mo
- Department of Oncology, Montefiore Einstein, Bronx, NY; Montefiore Einstein Comprehensive Cancer Center, Bronx, NY
| | - Bhawneet Chadha
- Department of Oncology, Montefiore Einstein, Bronx, NY; Montefiore Einstein Comprehensive Cancer Center, Bronx, NY
| | - Chaoyuan Kuang
- Department of Oncology, Montefiore Einstein, Bronx, NY; Department of Molecular Pharmacology, Montefiore Einstein, Bronx, NY.
| |
Collapse
|
11
|
López-Gil CI, Téllez-Jurado A, Velasco-Velázquez MA, Anducho-Reyes MA. Identification and Analysis of Anticancer Therapeutic Targets from the Polysaccharide Krestin (PSK) and Polysaccharopeptide (PSP) Using Inverse Docking. Molecules 2024; 29:5390. [PMID: 39598781 PMCID: PMC11596896 DOI: 10.3390/molecules29225390] [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: 09/23/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
The natural compounds PSK and PSP have antitumor and immunostimulant properties. These pharmacological benefits have been documented in vitro and in vivo, although there is no information in silico which describes the action mechanisms at the molecular level. In this study, the inverse docking method was used to identify the interactions of PSK and PSP with two local databases: BPAT with 66 antitumor proteins, and BPSIC with 138 surfaces and intracellular proteins. This led to the identification interactions and similarities of PSK and the AB680 inhibitor in the active site of CD73. It was also found that PSK binds to CD59, interacting with the amino acids APS22 and PHE23, which coincide with the rlLYd4 internalization inhibitor. With the isoform of the K-RAS protein, PSK bonded to the TYR32 amino acid at switch 1, while with BAK it bonded to the region of the α1 helix, while PSP bonded to the activation site and the C-terminal and N-terminal ends of that helix. In Bcl-2, PSK interacted at the binding site of the Venetoclax inhibitor, showing similarities with the amino acids ASP111, VAL133, LEU137, MET115, PHE112, and TYR108, while PSP had similarities with THR132, VAL133, LEU137, GLN118, MET115, APS111, PHE112, and PHE104.
Collapse
Affiliation(s)
- Carlos Iván López-Gil
- Department of Biotechnology, Universidad Politécnica de Pachuca, Zempoala 43830, Mexico; (C.I.L.-G.); (A.T.-J.)
| | - Alejandro Téllez-Jurado
- Department of Biotechnology, Universidad Politécnica de Pachuca, Zempoala 43830, Mexico; (C.I.L.-G.); (A.T.-J.)
| | | | - Miguel Angel Anducho-Reyes
- Department of Biotechnology, Universidad Politécnica de Pachuca, Zempoala 43830, Mexico; (C.I.L.-G.); (A.T.-J.)
| |
Collapse
|
12
|
Vithani N, Zhang S, Thompson JP, Patel LA, Demidov A, Xia J, Balaeff A, Mentes A, Arnautova YA, Kohlmann A, Lawson JD, Nicholls A, Skillman AG, LeBard DN. Exploration of Cryptic Pockets Using Enhanced Sampling Along Normal Modes: A Case Study of KRAS G12D. J Chem Inf Model 2024; 64:8258-8273. [PMID: 39419500 PMCID: PMC11558672 DOI: 10.1021/acs.jcim.4c01435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024]
Abstract
Identification of cryptic pockets has the potential to open new therapeutic opportunities by discovering ligand binding sites that remain hidden in static apo structures of a target protein. Moreover, allosteric cryptic pockets can become valuable for designing target-selective ligands when the natural ligand binding sites are conserved in variants of a protein. For example, before an allosteric cryptic pocket was discovered, KRAS was considered undruggable due to its smooth surface and conservation of the GDP/GTP binding pocket across the wild type and oncogenic isoforms. Recent identification of the Switch-II cryptic pocket in the KRASG12C mutant and FDA approval of anticancer drugs targeting this site underscores the importance of cryptic pockets in solving pharmaceutical challenges. Here, we present a newly developed approach for the exploration of cryptic pockets using weighted ensemble molecular dynamics simulations with inherent normal modes as progress coordinates applied to the wild type KRAS and the G12D mutant. We performed extensive all-atomic simulations (>400 μs) with and without several cosolvents (xenon, ethanol, benzene), and analyzed trajectories using three distinct methods to search for potential binding pockets. These methods have been applied as a proof-of-concept to KRAS and have shown they can predict known cryptic binding sites. Furthermore, we performed ligand-binding simulations of a known inhibitor (MRTX1133) to shed light on the nature of cryptic pockets in KRASG12D and the role of conformational selection vs induced-fit mechanism in the formation of these cryptic pockets.
Collapse
Affiliation(s)
- Neha Vithani
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | - She Zhang
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | - Jeffrey P. Thompson
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | - Lara A. Patel
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | - Alex Demidov
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | - Junchao Xia
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | - Alexander Balaeff
- Black
Diamond Therapeutics, Cambridge, Massachusetts 02142, United States
| | - Ahmet Mentes
- Black
Diamond Therapeutics, Cambridge, Massachusetts 02142, United States
| | | | - Anna Kohlmann
- Black
Diamond Therapeutics, Cambridge, Massachusetts 02142, United States
| | - J. David Lawson
- Mirati
Therapeutics, Inc., San Diego, California 92121, United States
| | - Anthony Nicholls
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| | | | - David N. LeBard
- OpenEye,
Cadence Molecular Sciences, Santa Fe, New Mexico 87508, United States
| |
Collapse
|
13
|
Skhoun H, El Fessikh M, Khattab M, Mchich B, Agadr A, Abilkassem R, Dakka N, Flatters D, Camproux AC, Ouzzif Z, El Baghdadi J. A Novel NRAS Variant Near the Splice Junction in Moroccan Childhood Acute Lymphoblastic Leukemia: A Molecular Dynamics Study. Biochem Genet 2024:10.1007/s10528-024-10968-2. [PMID: 39514082 DOI: 10.1007/s10528-024-10968-2] [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: 06/01/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
The RAS genes are importantly implicated in oncogenesis and are frequently mutated in childhood acute lymphoblastic leukemia. This study is the first to our knowledge, to determine the mutational status of NRAS and KRAS genes in Moroccan pediatric acute lymphoblastic leukemia (ALL). Polymerase chain reaction and Sanger sequencing were performed for 45 ALL samples to explore the coding exons. The functional effect of the mutation was evaluated using in silico prediction tools and molecular modeling. We identified a novel variant c.290 G > C p.Arg97Thr within NRAS gene in a patient with T-ALL, which is a rare missense point mutation affecting the last base of exon 3. Analyses revealed that p.Arg97Thr impairs the adjacent splice site efficiency. Moreover, it leads to structural modifications at local and global levels of the protein through the loss of hydrogen bonds. Additionally, the molecular dynamics (MD) simulation showed that it slightly increases the stability of NRAS protein by locally decreasing the flexibility of the mutated region. No variant was detected within KRAS gene. R97 at NRAS gene is an overlapping splice site residue. Our findings suggest that the NRAS p.Arg97Thr variant may disrupt the splicing machinery and functions of the protein, thus playing a vital role in leukemogenesis. In addition, the highly druggable pocket may possibly be studied for its therapeutic implications.
Collapse
Affiliation(s)
- Hanaa Skhoun
- Genetics Unit, Military Hospital Mohammed V, Rabat, Morocco
- Laboratory of Human Pathologies Biology and Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Meriem El Fessikh
- Genetics Unit, Military Hospital Mohammed V, Rabat, Morocco
- Laboratory of Human Pathologies Biology and Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Mohammed Khattab
- Pediatric Hematology and Oncology Center, Children's Hospital, Rabat, Morocco
- Centre of Childhood Care and Prevention, Cheikh Zaid International University Hospital, Rabat, Morocco
| | - Basma Mchich
- Unité de Biologie Fonctionnelle Et Adaptative, Université Paris Cité, CNRS, INSERM, Paris, France
| | - Aomar Agadr
- Department of Pediatrics, Military Hospital Mohammed V, Faculty of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco
| | - Rachid Abilkassem
- Department of Pediatrics, Military Hospital Mohammed V, Faculty of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco
| | - Nadia Dakka
- Laboratory of Human Pathologies Biology and Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Delphine Flatters
- Unité de Biologie Fonctionnelle Et Adaptative, Université Paris Cité, CNRS, INSERM, Paris, France
| | - Anne-Claude Camproux
- Unité de Biologie Fonctionnelle Et Adaptative, Université Paris Cité, CNRS, INSERM, Paris, France
| | - Zohra Ouzzif
- Laboratories Pole, Military Hospital Mohammed V, Rabat, Morocco
| | - Jamila El Baghdadi
- Genetics Unit, Military Hospital Mohammed V, Rabat, Morocco.
- Laboratories Pole, Military Hospital Mohammed V, Rabat, Morocco.
| |
Collapse
|
14
|
Chu T, Zhang R, Liu X, Lin L, Li Y, Niu Z, Quan H, Zhao Y, Li Y. Influence of recipient KRAS gene rs712 polymorphisms on the overall survival rate of hepatocellular carcinoma after hepatic transplantation. Clin Exp Med 2024; 24:246. [PMID: 39460812 PMCID: PMC11512907 DOI: 10.1007/s10238-024-01509-7] [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: 07/10/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Hepatocellular carcinoma (HCC) recurrence appears commonly after liver transplantation (LT), and it severely affected the long-term survival of patients. Previous studies have proved that Rap1A is involved in hepatocarcinogenesis and metastasis, and demonstrated the significant association between KRAS rs712 polymorphism and HCC. However, the relationship between KRAS rs712 polymorphism and HCC recurrence after LT remained unclear. A total of 93 HCC patients who underwent LT from March 2008 to Dec 2015 was analyzed. The genotypes of both donors and recipients had been confirmed as KRAS rs712. The independent risk factors that associated with HCC recurrence were investigated with univariate and multivariate logistic regression analysis. The recurrence-free (RFS) and overall survival (OS) were calculated with Cox regression analysis. The KRAS rs712 genotype frequencies were determined using the Χ2 test and the minor allele frequencies (MAFs) of KRAS rs712 genotypes were calculated by Hardy-Weinberg equilibrium. We found that the recipient KRAS rs712 polymorphism was significantly associated with HCC recurrence after LT. Moreover, the Milan criteria, microvascular invasion and recipient KRAS rs712 genotype were proved to be independent risk factors for HCC recurrence after LT. Patients with donor TG/TT genotypes had a significantly higher RFS and OS than TT genotype. The TNM stage, microvascular invasion, Milan criteria, treatment and recipient KRAS rs712 genotype were independent factors for the RFS of LT patients. Recipient KRAS rs712 polymorphism is associated with HCC recurrence after liver transplantation and plays as a promising bio-predictor of overall survival rate of HCC risks after hepatic transplantation.
Collapse
Affiliation(s)
- Tiancheng Chu
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
- Songjiang District Health Commission of Shanghai, Shanghai, China
| | - Rulin Zhang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaolei Liu
- Department of Clinical Oncology, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Li Lin
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanning Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ziguang Niu
- Department of Clinical Oncology, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Heng Quan
- Songjiang District Health Commission of Shanghai, Shanghai, China
| | - Yingying Zhao
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Yaohua Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
15
|
Mahran R, Kapp JN, Valtonen S, Champagne A, Ning J, Gillette W, Stephen AG, Hao F, Plückthun A, Härmä H, Pantsar T, Kopra K. Beyond KRAS(G12C): Biochemical and Computational Characterization of Sotorasib and Adagrasib Binding Specificity and the Critical Role of H95 and Y96. ACS Chem Biol 2024; 19:2152-2164. [PMID: 39283696 DOI: 10.1021/acschembio.4c00315] [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: 10/19/2024]
Abstract
Mutated KRAS proteins are frequently expressed in some of the most lethal human cancers and thus have been a target of intensive drug discovery efforts for decades. Lately, KRAS(G12C) switch-II pocket (SII-P)-targeting covalent small molecule inhibitors have finally reached clinical practice. Sotorasib (AMG-510) was the first FDA-approved covalent inhibitor to treat KRAS(G12C)-positive nonsmall cell lung cancer (NSCLC), followed soon by adagrasib (MRTX849). Both drugs target the GDP-bound state of KRAS(G12C), exploiting the strong nucleophilicity of acquired cysteine. Here, we evaluate the similarities and differences between sotorasib and adagrasib in their RAS SII-P binding by applying biochemical, cellular, and computational methods. Exact knowledge of SII-P engagement can enable targeting this site by reversible inhibitors for KRAS mutants beyond G12C. We show that adagrasib is strictly KRAS- but not KRAS(G12C)-specific due to its strong and unreplaceable interaction with H95. Unlike adagrasib, sotorasib is less dependent on H95 for its binding, making it a RAS isoform-agnostic compound, having a similar functionality also with NRAS and HRAS G12C mutants. Our results emphasize the accessibility of SII-P beyond oncogenic G12C and aid in understanding the molecular mechanism behind the clinically observed drug resistance, associated especially with secondary mutations on KRAS H95 and Y96.
Collapse
Affiliation(s)
- Randa Mahran
- Department of Chemistry, University of Turku, Henrikinkatu 2, 20500 Turku, Finland
| | - Jonas N Kapp
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Salla Valtonen
- Department of Chemistry, University of Turku, Henrikinkatu 2, 20500 Turku, Finland
| | - Allison Champagne
- NCI RAS Initiative, Frederick National Laboratory for Cancer Research, 8560 Progress Drive, Frederick, Maryland 21702, United States
| | - Jinying Ning
- KYinno Biotechnology Co., Ltd., Yizhuang Biomedical Park, No. 88 Kechuang Six Street, BDA, Beijing 101111, China
| | - William Gillette
- NCI RAS Initiative, Frederick National Laboratory for Cancer Research, 8560 Progress Drive, Frederick, Maryland 21702, United States
| | - Andrew G Stephen
- NCI RAS Initiative, Frederick National Laboratory for Cancer Research, 8560 Progress Drive, Frederick, Maryland 21702, United States
| | - Feng Hao
- KYinno Biotechnology Co., Ltd., Yizhuang Biomedical Park, No. 88 Kechuang Six Street, BDA, Beijing 101111, China
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Harri Härmä
- Department of Chemistry, University of Turku, Henrikinkatu 2, 20500 Turku, Finland
| | - Tatu Pantsar
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonrinne 3, 70210 Kuopio, Finland
| | - Kari Kopra
- Department of Chemistry, University of Turku, Henrikinkatu 2, 20500 Turku, Finland
| |
Collapse
|
16
|
Peng Y, Yang Q. Targeting KRAS in gynecological malignancies. FASEB J 2024; 38:e70089. [PMID: 39377766 DOI: 10.1096/fj.202401734r] [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: 07/28/2024] [Revised: 09/15/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024]
Abstract
Cervical, endometrial, and ovarian cancers stand prominently as the leading gynecological malignancies of the female reproductive system. The conventional therapeutic modalities for gynecological malignancies have predominantly encompassed surgery, chemotherapy, and radiotherapy. However, efficacy of these approaches remains limited in cases of relapse or drug resistance. KRAS is one of the most frequently mutated oncogenes in human cancers. The KRAS gene encodes a small guanosine triphosphatase protein that acts as a molecular switch for crucial intracellular signaling pathways. KRAS mutations are deeply involved in the occurrence and development of gynecological malignancies. The present review aims to expound upon the role of oncogenic KRAS as a biomarker, elucidating various therapeutic approaches under investigation targeting the KRAS pathway in gynecological tumors.
Collapse
Affiliation(s)
- Yuanyuan Peng
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
17
|
Tajiknia V, Pinho-Schwermann M, Srinivasan PR, Hernandez Borrero L, Zhang L, Huntington KE, El-Deiry WS. Synergistic anti-tumor activity, reduced pERK, and immuno-stimulatory cytokine profiles with 5-FU or ONC212 plus KRAS G12D inhibitor MRTX1133 in CRC and pancreatic cancer cells independent of G12D mutation. Am J Cancer Res 2024; 14:4523-4536. [PMID: 39417197 PMCID: PMC11477830 DOI: 10.62347/dvxl1377] [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: 10/08/2023] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
KRAS mutations occur in ~40-50% of mCRC and are associated with aggressive disease that is refractory to anti-EGFR therapies. Pancreatic cancer harbors ~90% KRAS driver gene mutation frequency. Small molecules targeting KRAS G12C gained FDA approval for KRAS G12C-mutated NSCLC. ONC212, a fluorinated imipridone with nM anti-cancer activity has preclinical efficacy against pancreatic cancer and other malignancies. MRTX1133, identified as a noncovalent selective KRAS G12D inhibitor that suppresses G12D signaling by binding to the switch II pocket thereby inhibiting protein-protein interactions. We investigated cell viability, drug synergies, pERK suppression and cytokine, chemokine or growth factor alterations following treatment with 5-Fluorouracil (5-FU) or ONC212 plus MRTX1133 in 6 human CRC and 4 human pancreatic cancer cell lines. IC50 sensitivities ranged from 7 to 12 µM for 5-FU, 0.2-0.8 µM for ONC212, and > 100 nM to > 5,000 nM for MRTX1133 (G12D N = 4: LS513 > 100, HPAF-II > 1,000, SNUC2B > 5,000, PANC-1 > 5,000). For non-G12D, the range of IC50 for MRTX1133 was > 1,000 to > 5,000 nM for CRC lines with G12V, G13D, or WT KRAS (N = 7). Synergies between MRTX1133 plus 5-FU or ONC212 were observed regardless of KRAS G12D mutation with combination indices of < 0.5 indicating strong synergy. Observed synergies were greater with MRTX1133 plus ONC212 compared to MRTX1133 plus 5-FU. pERK was suppressed with mutant but not wild-type KRAS at nM MRTX1133 doses. Immunostimulatory profiles included reduction in IL8/CXCL8, MICA, Angiopoietin 2, VEGF and TNF-alpha and increase in IL-18/IL-1F4 with MRTX treatments and combinations. Our studies reveal preclinical activity of MRTX1133 alone or synergies when combined with 5-FU or ONC212 against mCRC and pancreatic cancer cells regardless of KRAS G12D mutation. The results suggest that KRAS G12V and KRAS G13D should be further considered in clinical trials including combination therapies involving MRTX1133 and 5-FU or ONC212.
Collapse
Affiliation(s)
- Vida Tajiknia
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Maximilian Pinho-Schwermann
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Praveen R Srinivasan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Liz Hernandez Borrero
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Leiqing Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Kelsey E Huntington
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| |
Collapse
|
18
|
Pfeil AJ, Hale JD, Zhang TS, Wakayama K, Miyazaki I, Odintsov I, Somwar R. Preclinical evaluation of targeted therapies for central nervous system metastases. Dis Model Mech 2024; 17:dmm050836. [PMID: 39344915 PMCID: PMC11463968 DOI: 10.1242/dmm.050836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024] Open
Abstract
The central nervous system (CNS) represents a site of sanctuary for many metastatic tumors when systemic therapies that control the primary tumor cannot effectively penetrate intracranial lesions. Non-small cell lung cancers (NSCLCs) are the most likely of all neoplasms to metastasize to the brain, with up to 60% of patients developing CNS metastases during the disease process. Targeted therapies such as tyrosine kinase inhibitors (TKIs) have helped reduce lung cancer mortality but vary considerably in their capacity to control CNS metastases. The ability of these therapies to effectively target lesions in the CNS depends on several of their pharmacokinetic properties, including blood-brain barrier permeability, affinity for efflux transporters, and binding affinity for both plasma and brain tissue. Despite the existence of numerous preclinical models with which to characterize these properties, many targeted therapies have not been rigorously tested for CNS penetration during the discovery process, whereas some made it through preclinical testing despite poor brain penetration kinetics. Several TKIs have now been engineered with the characteristics of CNS-penetrant drugs, with clinical trials proving these efforts fruitful. This Review outlines the extent and variability of preclinical evidence for the efficacy of NSCLC-targeted therapies, which have been approved by the US Food and Drug Administration (FDA) or are in development, for treating CNS metastases, and how these data correlate with clinical outcomes.
Collapse
Affiliation(s)
- Alexander J. Pfeil
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- University of North Carolina School of Medicine, Chapel Hill, NC 27514, USA
| | - Joshua D. Hale
- University of North Carolina School of Medicine, Chapel Hill, NC 27514, USA
| | - Tiger S. Zhang
- University of North Carolina School of Medicine, Chapel Hill, NC 27514, USA
| | - Kentaro Wakayama
- Taiho Pharmaceutical Co. Ltd. 3, Okubo, Tsukuba, Ibaraki 300-2611, Japan
| | - Isao Miyazaki
- Taiho Pharmaceutical Co. Ltd. 3, Okubo, Tsukuba, Ibaraki 300-2611, Japan
| | - Igor Odintsov
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 021105, USA
| | - Romel Somwar
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
19
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [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: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
20
|
Cao C, Li A, Xu C, Wu B, Yao L, Liu Y. Engineering artificial non-coding RNAs for targeted protein degradation. Nat Chem Biol 2024:10.1038/s41589-024-01719-w. [PMID: 39215101 DOI: 10.1038/s41589-024-01719-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Targeted protein degradation has become a notable drug development strategy, but its application has been limited by the dependence on protein-based chimeras with restricted genetic manipulation capabilities. The use of long non-coding RNAs (lncRNAs) has emerged as a viable alternative, offering interactions with cellular proteins to modulate pathways and enhance degradation capabilities. Here we introduce a strategy employing artificial lncRNAs (alncRNAs) for precise targeted protein degradation. By integrating RNA aptamers and sequences from the lncRNA HOTAIR, our alncRNAs specifically target and facilitate the ubiquitination and degradation of oncogenic transcription factors and tumor-related proteins, such as c-MYC, NF-κB, ETS-1, KRAS and EGFR. These alncRNAs show potential in reducing malignant phenotypes in cells, both in vitro and in vivo, offering advantages in efficiency, adaptability and versatility. This research enhances knowledge of lncRNA-driven protein degradation and presents an effective method for targeted therapies.
Collapse
Affiliation(s)
- Congcong Cao
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Synthetic Biology Research Center, Health Science Center, Shenzhen University, Shenzhen, China
| | - Aolin Li
- Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China
| | - Chaojie Xu
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Baorui Wu
- Department of Urology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Lin Yao
- Department of Urology, Peking University First Hospital, Beijing, China.
| | - Yuchen Liu
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Synthetic Biology Research Center, Health Science Center, Shenzhen University, Shenzhen, China.
| |
Collapse
|
21
|
Ghazi PC, O'Toole KT, Srinivas Boggaram S, Scherzer MT, Silvis MR, Zhang Y, Bogdan M, Smith BD, Lozano G, Flynn DL, Snyder EL, Kinsey CG, McMahon M. Inhibition of ULK1/2 and KRAS G12C controls tumor growth in preclinical models of lung cancer. eLife 2024; 13:RP96992. [PMID: 39213022 PMCID: PMC11364435 DOI: 10.7554/elife.96992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Mutational activation of KRAS occurs commonly in lung carcinogenesis and, with the recent U.S. Food and Drug Administration approval of covalent inhibitors of KRASG12C such as sotorasib or adagrasib, KRAS oncoproteins are important pharmacological targets in non-small cell lung cancer (NSCLC). However, not all KRASG12C-driven NSCLCs respond to these inhibitors, and the emergence of drug resistance in those patients who do respond can be rapid and pleiotropic. Hence, based on a backbone of covalent inhibition of KRASG12C, efforts are underway to develop effective combination therapies. Here, we report that the inhibition of KRASG12C signaling increases autophagy in KRASG12C-expressing lung cancer cells. Moreover, the combination of DCC-3116, a selective ULK1/2 inhibitor, plus sotorasib displays cooperative/synergistic suppression of human KRASG12C-driven lung cancer cell proliferation in vitro and superior tumor control in vivo. Additionally, in genetically engineered mouse models of KRASG12C-driven NSCLC, inhibition of either KRASG12C or ULK1/2 decreases tumor burden and increases mouse survival. Consequently, these data suggest that ULK1/2-mediated autophagy is a pharmacologically actionable cytoprotective stress response to inhibition of KRASG12C in lung cancer.
Collapse
Affiliation(s)
- Phaedra C Ghazi
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Kayla T O'Toole
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Sanjana Srinivas Boggaram
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Michael T Scherzer
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Mark R Silvis
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
| | - Yun Zhang
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | | | | | - Guillermina Lozano
- Department of Genetics, The University of Texas MD Anderson Cancer CenterHoustonUnited States
| | | | - Eric L Snyder
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
- Department of Pathology, University of UtahSalt Lake CityUnited States
| | - Conan G Kinsey
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
- Department of Internal Medicine, Division of Medical Oncology, University of UtahSalt Lake CityUnited States
| | - Martin McMahon
- Department of Oncological Sciences, University of UtahSalt Lake CityUnited States
- Huntsman Cancer Institute, University of UtahSalt Lake CityUnited States
- Department of Dermatology, University of UtahSalt Lake CityUnited States
| |
Collapse
|
22
|
Prinsa, Saha S, Bulbul MZH, Ozeki Y, Alamri MA, Kawsar SMA. Flavonoids as potential KRAS inhibitors: DFT, molecular docking, molecular dynamics simulation and ADMET analyses. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024; 26:955-992. [PMID: 38647682 DOI: 10.1080/10286020.2024.2343821] [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: 12/01/2023] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
KRAS mutations linked with cancer. Flavonoids were docked against KRAS G12C and G12D receptors. Abyssinone III, alpha naphthoflavone, beta naphthoflavone, abyssinone I, abyssinone II and beta naphthoflavone, genistin, daidzin showed good docking scores against KRAS G12C and G12D receptors, respectively. The MD simulation data revealed that Rg, RMSD, RMSF, and SASA values were within acceptable limits. Alpha and beta naphthoflavone showed good binding energies with KRAS G12C and G12D receptors. DFT and MEP analysis highlighted the nucleophilic and electrophilic zones of best-docked flavonoids. A novel avenue for the control of KRAS G12C and G12D mutations is made possible by flavonoids.
Collapse
Affiliation(s)
- Prinsa
- Department of Pharmaceutical Chemistry, Siddhartha Institute of Pharmacy, Near IT-Park, Dehradun, Uttarakhand, India
| | - Supriyo Saha
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Md Zahidul Haque Bulbul
- Laboratory of Carbohydrate and Nucleoside Chemistry (LCNC), Department of Chemistry, Faculty of Science, University of Chittagong, Chittagong, Bangladesh
| | - Yasuhiro Ozeki
- Graduate School of Nanobio Sciences, Yokohama City University, Yokohama, Japan
| | - Mubarak A Alamri
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sarkar M A Kawsar
- Laboratory of Carbohydrate and Nucleoside Chemistry (LCNC), Department of Chemistry, Faculty of Science, University of Chittagong, Chittagong, Bangladesh
| |
Collapse
|
23
|
Qin C, Li T, Lin C, Zhao B, Li Z, Zhao Y, Wang W. The systematic role of pancreatic cancer exosomes: distant communication, liquid biopsy and future therapy. Cancer Cell Int 2024; 24:264. [PMID: 39054529 PMCID: PMC11271018 DOI: 10.1186/s12935-024-03456-5] [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: 05/02/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024] Open
Abstract
Pancreatic cancer remains one of the most lethal diseases worldwide. Cancer-derived exosomes, benefiting from the protective role of the lipid membrane, exhibit remarkable stability in the circulatory system. These exosomes, released by tumor microenvironment, contain various biomolecules such as proteins, RNAs, and lipids that plays a pivotal role in mediating distant communication between the local pancreatic tumor and other organs or tissues. They facilitate the transfer of oncogenic factors to distant sites, contributing to the compromised body immune system, distant metastasis, diabetes, cachexia, and promoting a microenvironment conducive to tumor growth and metastasis in pancreatic cancer patients. Beyond their intrinsic roles, circulating exosomes in peripheral blood can be detected to facilitate accurate liquid biopsy. This approach offers a novel and promising method for the diagnosis and management of pancreatic cancer. Consequently, circulating exosomes are not only crucial mediators of systemic cell-cell communication during pancreatic cancer progression but also hold great potential as precise tools for pancreatic cancer management and treatment. Exosome-based liquid biopsy and therapy represent promising advancements in the diagnosis and treatment of pancreatic cancer. Exosomes can serve as drug delivery vehicles, enhancing the targeting and efficacy of anticancer treatments, modulating the immune system, and facilitating gene editing to suppress tumor growth. Ongoing research focuses on biomarker identification, drug delivery systems, and clinical trials to validate the safety and efficacy of exosome-based therapies, offering new possibilities for early diagnosis and precision treatment in pancreatic cancer. Leveraging the therapeutic potential of exosomes, including their ability to deliver targeted drugs and modulate immune responses, opens new avenues for innovative treatment strategies.
Collapse
Affiliation(s)
- Cheng Qin
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianyu Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chen Lin
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bangbo Zhao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zeru Li
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yutong Zhao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weibin Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
24
|
Siegman A, Shaykevich A, Chae D, Silverman I, Goel S, Maitra R. Erlotinib Treatment in Colorectal Cancer Suppresses Autophagy Based on KRAS Mutation. Curr Issues Mol Biol 2024; 46:7530-7547. [PMID: 39057088 PMCID: PMC11276370 DOI: 10.3390/cimb46070447] [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: 06/14/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The KRAS gene is mutated in approximately 45% of colorectal cancer patients. There are currently very few targeted treatments or therapies equipped to directly inhibit KRAS due to its unusual structural intricacies. Erlotinib, an EGFR inhibitor, has previously been demonstrated to reduce cell viability by inducing autophagy in lung cancer cell lines with varying EGFR mutations. In contrast to lung cancer cells, evidence is provided herein for the first time that erlotinib treatment in colorectal cancer (CRC) cell lines reduces autophagy and still results in decreased cell viability. However, the effects of erlotinib in CRC cell lines containing a wildtype KRAS gene were different than in cells carrying a mutant KRAS gene. We show that there is significantly more downregulation of autophagy in KRAS mutant CRC cells compared to KRAS wildtype cells, both at transcriptional and translational levels, suggesting that the KRAS mutation is advantageous for cancer growth, even in the presence of erlotinib. Cell viability results determined that KRAS wildtype CRC cells had significantly more cell death compared to KRAS mutant cells. Using patient mRNA datasets, we showed that there was a significant correlation between the presence of the KRAS mutation and the expression of autophagy proteins. Additionally, through molecular dynamics simulations, we develop a blueprint for KRAS and autophagy protein interaction and the impact of the KRAS mutation on autophagy protein regulation. Overall, this is the first report of erlotinib treatment in CRC cells that assesses autophagy, and we demonstrate that autophagy activity is downregulated in these cells. This effect is not only greater in cells carrying a KRAS mutation compared to wildtype cells, but the KRAS mutant cells also have increased cell viability compared to wildtype cells. We hypothesize that the difference in cell viability and autophagy expression between KRAS mutant and KRAS wildtype cells after treatment with erlotinib can be of therapeutic value to treat CRC patients carrying KRAS mutations.
Collapse
Affiliation(s)
- Alexander Siegman
- Department of Biology, Yeshiva University, New York, NY 10033, USA (D.C.)
| | - Aaron Shaykevich
- Department of Biology, Yeshiva University, New York, NY 10033, USA (D.C.)
| | - Danbee Chae
- Department of Biology, Yeshiva University, New York, NY 10033, USA (D.C.)
| | - Isaac Silverman
- Department of Biology, Yeshiva University, New York, NY 10033, USA (D.C.)
| | - Sanjay Goel
- Department of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Radhashree Maitra
- Department of Biology, Yeshiva University, New York, NY 10033, USA (D.C.)
| |
Collapse
|
25
|
Fan H, Hu X, Cao F, Zhou L, Wen R, Shen H, Fu Y, Zhu X, Jia H, Liu Z, Wang G, Yu G, Chang W, Zhang W. WWP1 inhibition increases SHP2 inhibitor efficacy in colorectal cancer. NPJ Precis Oncol 2024; 8:144. [PMID: 39014007 PMCID: PMC11252267 DOI: 10.1038/s41698-024-00650-6] [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: 03/29/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
Protein tyrosine phosphatase SHP2 activates RAS signaling, which is a novel target for colorectal cancer (CRC) therapy. However, SHP2 inhibitor monotherapy is ineffective for metastatic CRC and a combination therapy is required. In this study, we aimed to improve the antitumor efficacy of SHP2 inhibition and try to explore the resistance mechanism of SHP2 inhibitor. Results showed that WWP1 promoted the proliferation of CRC cells. Genetic or pharmacological inhibition of WWP1 enhanced the effect of SHP2 inhibitor in suppressing tumor growth in vitro and in vivo. WWP1 may mediate feedback reactivation of AKT signaling following SHP2 inhibition. Furthermore, nomogram models constructed with IHC expression of WWP1 and SHP2 greatly improved the accuracy of prognosis prediction for patients with CRC. Our findings indicate that WWP1 inhibitor I3C can synergize with SHP2 inhibitor and is expected to be a new strategy for clinical trials in treating advanced CRC patients.
Collapse
Affiliation(s)
- Hao Fan
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xuefei Hu
- Department of Navy Environmental and Occupational Health, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Fuao Cao
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Leqi Zhou
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Rongbo Wen
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hao Shen
- Department of Navy Environmental and Occupational Health, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Yating Fu
- Department of Navy Environmental and Occupational Health, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Xiaoming Zhu
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hang Jia
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zixuan Liu
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Guimin Wang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Guanyu Yu
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China.
| | - Wenjun Chang
- Department of Navy Environmental and Occupational Health, Faculty of Naval Medicine, Naval Medical University, Shanghai, China.
| | - Wei Zhang
- Department of Colorectal Surgery, Changhai Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
26
|
Song Q, Zhang Q, Fan X, Kayaat F, Lv R, Li J, Wang Y. The discovery of novel imidazo[1,2- a]pyridine derivatives as covalent anticancer agents. Org Biomol Chem 2024; 22:5374-5384. [PMID: 38869445 DOI: 10.1039/d4ob00694a] [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: 06/14/2024]
Abstract
The success of targeted covalent inhibitors (TCIs) for treating cancers has spurred the search for novel scaffolds to install covalent warheads. In our endeavour, using a scaffold hopping strategy, we managed to utilize imidazo[1,2-a]pyridine as the core backbone and explored its potential for the development of covalent inhibitors, therefore, synthesizing a series of novel KRAS G12C inhibitors facilitated by the Groebke-Blackburn-Bienaymè reaction (GBB reaction). Preliminary bio-evaluation screening delivered compound I-11 as a potent anticancer agent for KRAS G12C-mutated NCI-H358 cells, whose effects were further clarified by a series of cellular, biochemical, and molecular docking experiments. These results not only indicate the potential of compound I-11 as a lead compound for the treatment of intractable cancers, but also validate the unique role of imidazo[1,2-a]pyridine as a novel scaffold suitable for the discovery of covalent anticancer agents.
Collapse
Affiliation(s)
- Qin Song
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, Qingdao 26003, Shandong, P. R. China.
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266200, P. R. China
| | - Qianer Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, Qingdao 26003, Shandong, P. R. China.
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266200, P. R. China
| | - Xuejing Fan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, Qingdao 26003, Shandong, P. R. China.
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266200, P. R. China
| | - Fatmata Kayaat
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, Qingdao 26003, Shandong, P. R. China.
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266200, P. R. China
| | - Ruicheng Lv
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, Qingdao 26003, Shandong, P. R. China.
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266200, P. R. China
| | - Jing Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, Qingdao 26003, Shandong, P. R. China.
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266200, P. R. China
| | - Yong Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education; School of Medicine and Pharmacy, Ocean University of China, Qingdao 26003, Shandong, P. R. China.
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao 266200, P. R. China
| |
Collapse
|
27
|
Song W, Hu H, Yuan Z, Yao H. A prognostic model for anoikis-related genes in pancreatic cancer. Sci Rep 2024; 14:15200. [PMID: 38956290 PMCID: PMC11220081 DOI: 10.1038/s41598-024-65981-7] [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: 04/18/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024] Open
Abstract
Anoikis, a distinct form of programmed cell death, is crucial for both organismal development and maintaining tissue equilibrium. Its role extends to the proliferation and progression of cancer cells. This study aimed to establish an anoikis-related prognostic model to predict the prognosis of pancreatic cancer (PC) patients. Gene expression data and patient clinical profiles were sourced from The Cancer Genome Atlas (TCGA-PAAD: Pancreatic Adenocarcinoma) and the International Cancer Genome Consortium (ICGC-PACA: Pancreatic Ductal Adenocarcinoma). Non-cancerous pancreatic tissue gene expression data were obtained from the Genotype-Tissue Expression (GTEx) project. The R package was used to construct anoikis-related PC prognostic models, which were later validated with the ICGC-PACA database. Survival analyses demonstrated a poorer prognosis for patients in the high-risk group, consistent across both TCGA-PAAD and ICGC-PACA datasets. A nomogram was designed as a predictive tool to estimate patient mortality. The study also analyzed tumor mutations and immune infiltration across various risk groups, uncovering notable differences in tumor mutation patterns and immune landscapes between high- and low-risk groups. In conclusion, this research successfully developed a prognostic model centered on anoikis-related genes, offering a novel tool for predicting the clinical trajectory of PC patients.
Collapse
Affiliation(s)
- Wenbin Song
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Tianjin Key Laboratory of Precise Vascular Reconstruction and Organ Function Repair, Tianjin, 300052, People's Republic of China
| | - Haiyang Hu
- Department of Cardiac Critical Care Medicine, Affiliated Hospital of Jining Medical University, Jining, 272007, People's Republic of China
| | - Zhengbo Yuan
- School of Medicine, Xiamen University, No.4221 Xiangan South Road, Xiangan District, Xiamen, 361102, People's Republic of China.
- Department of Neurosurgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, No.55 Zhenghai load, Siming District, Xiamen, 361001, People's Republic of China.
| | - Hao Yao
- Department of Hepatological Surgery, The Second Hospital of Tianjin Medical University, No.23 Pingjiang Road, Hexi District, Tianjin, 300211, People's Republic of China.
| |
Collapse
|
28
|
Yang J, Wang QL, Wang GN, Ye JC, Li ZQ, Wang JY, Liang ZH, Li SX, Sun C, Liao WT, Gao YJ, Wang J, Mao Y, Yu C, Feng GK, Zeng MS. A pan-KRAS degrader for the treatment of KRAS-mutant cancers. Cell Discov 2024; 10:70. [PMID: 38937452 PMCID: PMC11211324 DOI: 10.1038/s41421-024-00699-4] [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: 09/07/2023] [Accepted: 06/02/2024] [Indexed: 06/29/2024] Open
Abstract
KRAS mutations are highly prevalent in a wide range of lethal cancers, and these mutant forms of KRAS play a crucial role in driving cancer progression and conferring resistance to treatment. While there have been advancements in the development of small molecules to target specific KRAS mutants, the presence of undruggable mutants and the emergence of secondary mutations continue to pose challenges in the clinical treatment of KRAS-mutant cancers. In this study, we developed a novel molecular tool called tumor-targeting KRAS degrader (TKD) that effectively targets a wide range of KRAS mutants. TKD is composed of a KRAS-binding nanobody, a cell-penetrating peptide selectively targeting cancer cells, and a lysosome-binding motif. Our data revealed that TKD selectively binds to KRAS in cancer cells and effectively induces KRAS degradation via a lysosome-dependent process. Functionally, TKD suppresses tumor growth with no obvious side effects and enhances the antitumor effects of PD-1 antibody and cetuximab. This study not only provides a strategy for developing drugs targeting "undruggable" proteins but also reveals that TKD is a promising therapeutic for treating KRAS-mutant cancers.
Collapse
Affiliation(s)
- Jie Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Qiao-Li Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Guan-Nan Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jia-Cong Ye
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zi-Qian Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jing-Yun Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhao-Hui Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Shu-Xin Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Cong Sun
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Wen-Ting Liao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yi-Jun Gao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jing Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yong Mao
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Chunjing Yu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Guo-Kai Feng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
| |
Collapse
|
29
|
Tan WY, Nagabhyrava S, Ang-Olson O, Das P, Ladel L, Sailo B, He L, Sharma A, Ahuja N. Translation of Epigenetics in Cell-Free DNA Liquid Biopsy Technology and Precision Oncology. Curr Issues Mol Biol 2024; 46:6533-6565. [PMID: 39057032 PMCID: PMC11276574 DOI: 10.3390/cimb46070390] [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: 05/27/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/28/2024] Open
Abstract
Technological advancements in cell-free DNA (cfDNA) liquid biopsy have triggered exponential growth in numerous clinical applications. While cfDNA-based liquid biopsy has made significant strides in personalizing cancer treatment, the exploration and translation of epigenetics in liquid biopsy to clinical practice is still nascent. This comprehensive review seeks to provide a broad yet in-depth narrative of the present status of epigenetics in cfDNA liquid biopsy and its associated challenges. It highlights the potential of epigenetics in cfDNA liquid biopsy technologies with the hopes of enhancing its clinical translation. The momentum of cfDNA liquid biopsy technologies in recent years has propelled epigenetics to the forefront of molecular biology. We have only begun to reveal the true potential of epigenetics in both our understanding of disease and leveraging epigenetics in the diagnostic and therapeutic domains. Recent clinical applications of epigenetics-based cfDNA liquid biopsy revolve around DNA methylation in screening and early cancer detection, leading to the development of multi-cancer early detection tests and the capability to pinpoint tissues of origin. The clinical application of epigenetics in cfDNA liquid biopsy in minimal residual disease, monitoring, and surveillance are at their initial stages. A notable advancement in fragmentation patterns analysis has created a new avenue for epigenetic biomarkers. However, the widespread application of cfDNA liquid biopsy has many challenges, including biomarker sensitivity, specificity, logistics including infrastructure and personnel, data processing, handling, results interpretation, accessibility, and cost effectiveness. Exploring and translating epigenetics in cfDNA liquid biopsy technology can transform our understanding and perception of cancer prevention and management. cfDNA liquid biopsy has great potential in precision oncology to revolutionize conventional ways of early cancer detection, monitoring residual disease, treatment response, surveillance, and drug development. Adapting the implementation of liquid biopsy workflow to the local policy worldwide and developing point-of-care testing holds great potential to overcome global cancer disparity and improve cancer outcomes.
Collapse
Affiliation(s)
- Wan Ying Tan
- Department of Surgery, Yale School of Medicine, New Haven, CT 06520-8000, USA; (W.Y.T.); (P.D.); (L.L.); (B.S.); (L.H.)
- Department of Internal Medicine, Norwalk Hospital, Norwalk, CT 06850, USA
- Hematology & Oncology, Neag Comprehensive Cancer Center, UConn Health, Farmington, CT 06030, USA
| | | | - Olivia Ang-Olson
- Department of Surgery, Yale School of Medicine, New Haven, CT 06520-8000, USA; (W.Y.T.); (P.D.); (L.L.); (B.S.); (L.H.)
| | - Paromita Das
- Department of Surgery, Yale School of Medicine, New Haven, CT 06520-8000, USA; (W.Y.T.); (P.D.); (L.L.); (B.S.); (L.H.)
| | - Luisa Ladel
- Department of Surgery, Yale School of Medicine, New Haven, CT 06520-8000, USA; (W.Y.T.); (P.D.); (L.L.); (B.S.); (L.H.)
- Department of Internal Medicine, Norwalk Hospital, Norwalk, CT 06850, USA
| | - Bethsebie Sailo
- Department of Surgery, Yale School of Medicine, New Haven, CT 06520-8000, USA; (W.Y.T.); (P.D.); (L.L.); (B.S.); (L.H.)
| | - Linda He
- Department of Surgery, Yale School of Medicine, New Haven, CT 06520-8000, USA; (W.Y.T.); (P.D.); (L.L.); (B.S.); (L.H.)
| | - Anup Sharma
- Department of Surgery, Yale School of Medicine, New Haven, CT 06520-8000, USA; (W.Y.T.); (P.D.); (L.L.); (B.S.); (L.H.)
| | - Nita Ahuja
- Department of Surgery, Yale School of Medicine, New Haven, CT 06520-8000, USA; (W.Y.T.); (P.D.); (L.L.); (B.S.); (L.H.)
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520-8000, USA
- Biological and Biomedical Sciences Program (BBS), Yale University, New Haven, CT 06520-8084, USA
| |
Collapse
|
30
|
Giarratana AO, Prendergast CM, Salvatore MM, Capaccione KM. TGF-β signaling: critical nexus of fibrogenesis and cancer. J Transl Med 2024; 22:594. [PMID: 38926762 PMCID: PMC11201862 DOI: 10.1186/s12967-024-05411-4] [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: 02/01/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
The transforming growth factor-beta (TGF-β) signaling pathway is a vital regulator of cell proliferation, differentiation, apoptosis, and extracellular matrix production. It functions through canonical SMAD-mediated processes and noncanonical pathways involving MAPK cascades, PI3K/AKT, Rho-like GTPases, and NF-κB signaling. This intricate signaling system is finely tuned by interactions between canonical and noncanonical pathways and plays key roles in both physiologic and pathologic conditions including tissue homeostasis, fibrosis, and cancer progression. TGF-β signaling is known to have paradoxical actions. Under normal physiologic conditions, TGF-β signaling promotes cell quiescence and apoptosis, acting as a tumor suppressor. In contrast, in pathological states such as inflammation and cancer, it triggers processes that facilitate cancer progression and tissue remodeling, thus promoting tumor development and fibrosis. Here, we detail the role that TGF-β plays in cancer and fibrosis and highlight the potential for future theranostics targeting this pathway.
Collapse
Affiliation(s)
- Anna O Giarratana
- Northwell Health - Peconic Bay Medical Center, 1 Heroes Way, Riverhead, NY, 11901, USA.
| | | | - Mary M Salvatore
- Department of Radiology, Columbia University, New York, NY, 11032, USA
| | | |
Collapse
|
31
|
Salva de Torres C, Baraibar I, Saoudi González N, Ros J, Salva F, Rodríguez-Castells M, Alcaraz A, García A, Tabernero J, Élez E. Current and Emerging Treatment Paradigms in Colorectal Cancer: Integrating Hallmarks of Cancer. Int J Mol Sci 2024; 25:6967. [PMID: 39000083 PMCID: PMC11241496 DOI: 10.3390/ijms25136967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
The treatment of unresectable metastatic colorectal cancer has evolved over the last two decades, as knowledge of cancer biology has broadened and new targets have emerged. 'The Hallmarks of Cancer' illustrate the crucial capabilities acquired by cells to become malignant and represent the evolution of knowledge of tumor biology. This review integrates these novel targets and therapies into selected hallmarks: sustaining proliferative signaling, inducing vasculature, avoiding immune destruction, genome instability and mutation, reprogramming cellular metabolism, and resisting cell death. The different strategies and combinations under study are based on treatments with anti-EGFR, anti-VEGF, and anti-HER2 agents, KRAS G12C inhibitors, BRAF and MEK inhibitors, and immune checkpoint inhibitors. However, new approaches are emerging, including vaccines, WEE1 inhibitors, and PARP inhibitors, among others. The further deciphering of cancer biology will unravel new targets, develop novel therapies, and improve patients' outcomes.
Collapse
Affiliation(s)
| | - Iosune Baraibar
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| | - Nadia Saoudi González
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| | - Javier Ros
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| | - Francesc Salva
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| | - Marta Rodríguez-Castells
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| | - Adriana Alcaraz
- Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (A.A.); (A.G.)
| | - Ariadna García
- Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (A.A.); (A.G.)
| | - Josep Tabernero
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| | - Elena Élez
- Medical Oncology Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), E-08035 Barcelona, Spain; (I.B.); (N.S.G.); (J.R.); (F.S.); (M.R.-C.), (J.T.)
| |
Collapse
|
32
|
Malamos P, Papanikolaou C, Gavriatopoulou M, Dimopoulos MA, Terpos E, Souliotis VL. The Interplay between the DNA Damage Response (DDR) Network and the Mitogen-Activated Protein Kinase (MAPK) Signaling Pathway in Multiple Myeloma. Int J Mol Sci 2024; 25:6991. [PMID: 39000097 PMCID: PMC11241508 DOI: 10.3390/ijms25136991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
The DNA damage response (DDR) network and the mitogen-activated protein kinase (MAPK) signaling pathway are crucial mechanisms for the survival of all living beings. An accumulating body of evidence suggests that there is crosstalk between these two systems, thus favoring the appropriate functioning of multi-cellular organisms. On the other hand, aberrations within these mechanisms are thought to play a vital role in the onset and progression of several diseases, including cancer, as well as in the emergence of drug resistance. Here, we provide an overview of the current knowledge regarding alterations in the DDR machinery and the MAPK signaling pathway as well as abnormalities in the DDR/MAPK functional crosstalk in multiple myeloma, the second most common hematologic malignancy. We also present the latest advances in the development of anti-myeloma drugs targeting crucial DDR- and MAPK-associated molecular components. These data could potentially be exploited to discover new therapeutic targets and effective biomarkers as well as for the design of novel clinical trials. Interestingly, they might provide a new approach to increase the efficacy of anti-myeloma therapy by combining drugs targeting the DDR network and the MAPK signaling pathway.
Collapse
Affiliation(s)
- Panagiotis Malamos
- Institute of Chemical Biology, National Hellenic Research Foundation, 116 35 Athens, Greece; (P.M.); (C.P.)
| | - Christina Papanikolaou
- Institute of Chemical Biology, National Hellenic Research Foundation, 116 35 Athens, Greece; (P.M.); (C.P.)
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (M.G.); (M.A.D.); (E.T.)
| | - Meletios A. Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (M.G.); (M.A.D.); (E.T.)
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 115 28 Athens, Greece; (M.G.); (M.A.D.); (E.T.)
| | - Vassilis L. Souliotis
- Institute of Chemical Biology, National Hellenic Research Foundation, 116 35 Athens, Greece; (P.M.); (C.P.)
| |
Collapse
|
33
|
Zhang ZW, Zhang KX, Liao X, Quan Y, Zhang HY. Evolutionary screening of precision oncology biomarkers and its applications in prognostic model construction. iScience 2024; 27:109859. [PMID: 38799582 PMCID: PMC11126775 DOI: 10.1016/j.isci.2024.109859] [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: 10/10/2023] [Revised: 03/15/2024] [Accepted: 04/27/2024] [Indexed: 05/29/2024] Open
Abstract
Biomarker screening is critical for precision oncology. However, one of the main challenges in precision oncology is that the screened biomarkers often fail to achieve the expected clinical effects and are rarely approved by regulatory authorities. Considering the close association between cancer pathogenesis and the evolutionary events of organisms, we first explored the evolutionary feature underlying clinically approved biomarkers, and two evolutionary features of approved biomarkers (Ohnologs and specific evolutionary stages of genes) were identified. Subsequently, we utilized evolutionary features for screening potential prognostic biomarkers in four common cancers: head and neck squamous cell carcinoma, liver hepatocellular carcinoma, lung adenocarcinoma, and lung squamous cell carcinoma. Finally, we constructed an evolution-strengthened prognostic model (ESPM) for cancers. These models can predict cancer patients' survival time across different cancer cohorts effectively and perform better than conventional models. In summary, our study highlights the application potentials of evolutionary information in precision oncology biomarker screening.
Collapse
Affiliation(s)
- Zhi-Wen Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Ke-Xin Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Xuan Liao
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Yuan Quan
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Hong-Yu Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| |
Collapse
|
34
|
Ghazi PC, O'Toole KT, Srinivas Boggaram S, Scherzer MT, Silvis MR, Zhang Y, Bogdan M, Smith BD, Lozano G, Flynn DL, Snyder EL, Kinsey CG, McMahon M. Inhibition of ULK1/2 and KRAS G12C controls tumor growth in preclinical models of lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.06.579200. [PMID: 38370808 PMCID: PMC10871191 DOI: 10.1101/2024.02.06.579200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Mutational activation of KRAS occurs commonly in lung carcinogenesis and, with the recent FDA approval of covalent inhibitors of KRAS G12C such as sotorasib or adagrasib, KRAS oncoproteins are important pharmacological targets in non-small cell lung cancer (NSCLC). However, not all KRAS G12C -driven NSCLCs respond to these inhibitors, and the emergence of drug resistance in those patients that do respond can be rapid and pleiotropic. Hence, based on a backbone of covalent inhibition of KRAS G12C , efforts are underway to develop effective combination therapies. Here we report that inhibition of KRAS G12C signaling increases autophagy in KRAS G12C expressing lung cancer cells. Moreover, the combination of DCC-3116, a selective ULK1/2 inhibitor, plus sotorasib displays cooperative/synergistic suppression of human KRAS G12C -driven lung cancer cell proliferation in vitro and superior tumor control in vivo . Additionally, in genetically engineered mouse models of KRAS G12C -driven NSCLC, inhibition of either KRAS G12C or ULK1/2 decreases tumor burden and increases mouse survival. Consequently, these data suggest that ULK1/2-mediated autophagy is a pharmacologically actionable cytoprotective stress response to inhibition of KRAS G12C in lung cancer.
Collapse
|
35
|
Pandya DV, Parikh RV, Gena RM, Kothari NR, Parekh PS, Chorawala MR, Jani MA, Yadav MR, Shah PA. The scaffold protein disabled 2 (DAB2) and its role in tumor development and progression. Mol Biol Rep 2024; 51:701. [PMID: 38822973 DOI: 10.1007/s11033-024-09653-9] [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: 03/29/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Disabled 2 (DAB2) is a multifunctional protein that has emerged as a critical component in the regulation of tumor growth. Its dysregulation is implicated in various types of cancer, underscoring its importance in understanding the molecular mechanisms underlying tumor development and progression. This review aims to unravel the intricate molecular mechanisms by which DAB2 exerts its tumor-suppressive functions within cancer signaling pathways. METHODS AND RESULTS We conducted a comprehensive review of the literature focusing on the structure, expression, physiological functions, and tumor-suppressive roles of DAB2. We provide an overview of the structure, expression, and physiological functions of DAB2. Evidence supporting DAB2's role as a tumor suppressor is explored, highlighting its ability to inhibit cell proliferation, induce apoptosis, and modulate key signaling pathways involved in tumor suppression. The interaction between DAB2 and key oncogenes is examined, elucidating the interplay between DAB2 and oncogenic signaling pathways. We discuss the molecular mechanisms underlying DAB2-mediated tumor suppression, including its involvement in DNA damage response and repair, regulation of cell cycle progression and senescence, and modulation of epithelial-mesenchymal transition (EMT). The review explores the regulatory networks involving DAB2, covering post-translational modifications, interactions with other tumor suppressors, and integration within complex signaling networks. We also highlight the prognostic significance of DAB2 and its role in pre-clinical studies of tumor suppression. CONCLUSION This review provides a comprehensive understanding of the molecular mechanisms by which DAB2 exerts its tumor-suppressive functions. It emphasizes the significance of DAB2 in cancer signaling pathways and its potential as a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Disha V Pandya
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad, Gujarat, 380009, India
| | - Rajsi V Parikh
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad, Gujarat, 380009, India
| | - Ruhanahmed M Gena
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad, Gujarat, 380009, India
| | - Nirjari R Kothari
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad, Gujarat, 380009, India
| | - Priyajeet S Parekh
- Pharmacy Practice Division, AV Pharma LLC, 1545 University Blvd N Ste A, Jacksonville, FL, 32211, USA
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad, Gujarat, 380009, India.
| | - Maharsh A Jani
- Pharmacy Practice Division, Anand Niketan, Shilaj, Ahmedabad, Gujarat, 380059, India
| | - Mayur R Yadav
- Department of Pharmacy Practice and Administration, Western University of Health Science, 309 E Second St, Pomona, CA, 91766, USA
| | - Palak A Shah
- Department of Pharmacology and Pharmacy Practice, K. B. Institute of Pharmaceutical Education and Research, Gandhinagar, Gujarat, 382023, India
| |
Collapse
|
36
|
Fujimori H, Shima‐Nakamura M, Kanno S, Shibuya‐Takahashi R, Mochizuki M, Mizuma M, Unno M, Wakui Y, Abue M, Iwai W, Fukushi D, Satoh K, Yamaguchi K, Shindo N, Yasuda J, Tamai K. FAXC interacts with ANXA2 and SRC in mitochondria and promotes tumorigenesis in cholangiocarcinoma. Cancer Sci 2024; 115:1896-1909. [PMID: 38480477 PMCID: PMC11145136 DOI: 10.1111/cas.16140] [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: 10/11/2023] [Revised: 02/13/2024] [Accepted: 02/22/2024] [Indexed: 06/04/2024] Open
Abstract
Cholangiocarcinoma (CCA) is one of the most difficult malignancies to treat as the therapeutic options are limited. Although several driver genes have been identified, most remain unknown. In this study, we identified a failed axon connection homolog (FAXC), whose function is unknown in mammals, by analyzing serially passaged CCA xenograft models. Knockdown of FAXC reduced subcutaneous tumorigenicity in mice. FAXC was bound to annexin A2 (ANXA2) and c-SRC, which are tumor-promoting genes. The FAXC/ANXA2/c-SRC complex forms in the mitochondria. FAXC enhances SRC-dependent ANXA2 phosphorylation at tyrosine-24, and the C-terminal amino acid residues (351-375) of FAXC are required for ANXA2 phosphorylation. Transcriptome data from a xenografted CCA cell line revealed that FAXC correlated with epithelial-mesenchymal transition, hypoxia, and KRAS signaling genes. Collectively, these findings advance our understanding of CCA tumorigenesis and provide candidate therapeutic targets.
Collapse
Affiliation(s)
- Haruna Fujimori
- Division of Cancer Stem CellMiyagi Cancer Center Research InstituteNatoriJapan
| | - Mao Shima‐Nakamura
- Division of Cancer Stem CellMiyagi Cancer Center Research InstituteNatoriJapan
| | - Shin‐Ichiro Kanno
- IDAC Fellow Research Group for DNA Repair and Dynamic Proteome Institute of Development, Aging and Cancer (IDAC)Tohoku UniversitySendaiJapan
| | | | - Mai Mochizuki
- Division of Cancer Stem CellMiyagi Cancer Center Research InstituteNatoriJapan
| | - Masamichi Mizuma
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Michiaki Unno
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Yuta Wakui
- Department of GastroenterologyMiyagi Cancer CenterNatoriJapan
| | - Makoto Abue
- Department of GastroenterologyMiyagi Cancer CenterNatoriJapan
| | - Wataru Iwai
- Department of GastroenterologyMiyagi Cancer CenterNatoriJapan
| | - Daisuke Fukushi
- Division of GastroenterologyTohoku Medical and Pharmaceutical UniversitySendaiJapan
| | - Kennich Satoh
- Division of GastroenterologyTohoku Medical and Pharmaceutical UniversitySendaiJapan
| | - Kazunori Yamaguchi
- Division of Molecular and Cellular OncologyMiyagi Cancer Center Research InstituteNatoriJapan
| | - Norihisa Shindo
- Cancer Chromosome Biology UnitMiyagi Cancer Center Research InstituteNatoriJapan
| | - Jun Yasuda
- Division of Molecular and Cellular OncologyMiyagi Cancer Center Research InstituteNatoriJapan
| | - Keiichi Tamai
- Division of Cancer Stem CellMiyagi Cancer Center Research InstituteNatoriJapan
| |
Collapse
|
37
|
Rijmers J, Retmana IA, Bui V, Arguedas D, Lebre MC, Sparidans RW, Beijnen JH, Schinkel AH. ABCB1 attenuates brain exposure to the KRAS G12C inhibitor opnurasib whereas binding to mouse carboxylesterase 1c influences its plasma exposure. Biomed Pharmacother 2024; 175:116720. [PMID: 38733773 DOI: 10.1016/j.biopha.2024.116720] [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: 02/28/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
Opnurasib (JDQ443) is a newly developed oral KRASG12C inhibitor, with a binding mechanism distinct from the registered KRASG12C inhibitors sotorasib and adagrasib. Phase I and II clinical trials for opnurasib in NSCLC are ongoing. We evaluated the pharmacokinetic roles of the ABCB1 (P-gp/MDR1) and ABCG2 (BCRP) efflux and OATP1 influx transporters, and of the metabolizing enzymes CYP3A and CES1 in plasma and tissue disposition of oral opnurasib, using genetically modified cell lines and mouse models. In vitro, opnurasib was potently transported by human (h)ABCB1 and slightly by mouse (m)Abcg2. In Abcb1a/b- and Abcb1a/b;Abcg2-deficient mice, a significant ∼100-fold increase in brain-to-plasma ratios was observed. Brain penetration was unchanged in Abcg2-/- mice. ABCB1 activity in the blood-brain barrier may therefore potentially limit the efficacy of opnurasib against brain metastases. The Abcb1a/b transporter activity could be almost completely reversed by co-administration of elacridar, a dual ABCB1/ABCG2 inhibitor, increasing the brain penetration without any behavioral or postural signs of acute CNS-related toxicity. No significant pharmacokinetic roles of the OATP1 transporters were observed. Transgenic human CYP3A4 did not substantially affect the plasma exposure of opnurasib, indicating that opnurasib is likely not a sensitive CYP3A4 substrate. Interestingly, Ces1-/- mice showed a 4-fold lower opnurasib plasma exposure compared to wild-type mice, whereas no strong effect was seen on the tissue distribution. Plasma Ces1c therefore likely binds opnurasib, increasing its retention in plasma. The obtained pharmacokinetic insights may be useful for further optimization of the clinical efficacy and safety of opnurasib, and might reveal potential drug-drug interaction risks.
Collapse
Affiliation(s)
- Jamie Rijmers
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands
| | - Irene A Retmana
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands; Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, the Netherlands
| | - Viët Bui
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands
| | - Davinia Arguedas
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands
| | - Maria C Lebre
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands
| | - Rolf W Sparidans
- Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacology, Utrecht, the Netherlands
| | - Jos H Beijnen
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands; Utrecht University, Faculty of Science, Department of Pharmaceutical Sciences, Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht, the Netherlands; The Netherlands Cancer Institute, Division of Pharmacy and Pharmacology, Amsterdam, the Netherlands
| | - Alfred H Schinkel
- The Netherlands Cancer Institute, Division of Pharmacology, Amsterdam, the Netherlands.
| |
Collapse
|
38
|
Zhao F, Yao Z, Li Y, Zhao W, Sun Y, Yang X, Zhao Z, Huang B, Wang J, Li X, Chen A. Targeting the molecular chaperone CCT2 inhibits GBM progression by influencing KRAS stability. Cancer Lett 2024; 590:216844. [PMID: 38582394 DOI: 10.1016/j.canlet.2024.216844] [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: 12/08/2023] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 04/08/2024]
Abstract
Proper protein folding relies on the assistance of molecular chaperones post-translation. Dysfunctions in chaperones can cause diseases associated with protein misfolding, including cancer. While previous studies have identified CCT2 as a chaperone subunit and an autophagy receptor, its specific involvement in glioblastoma remains unknown. Here, we identified CCT2 promote glioblastoma progression. Using approaches of coimmunoprecipitation, mass spectrometry and surface plasmon resonance, we found CCT2 directly bound to KRAS leading to increased stability and upregulated downstream signaling of KRAS. Interestingly, we found that dihydroartemisinin, a derivative of artemisinin, exhibited therapeutic effects in a glioblastoma animal model. We further demonstrated direct binding between dihydroartemisinin and CCT2. Treatment with dihydroartemisinin resulted in decreased KRAS expression and downstream signaling. Highlighting the significance of CCT2, CCT2 overexpression rescued the inhibitory effect of dihydroartemisinin on glioblastoma. In conclusion, the study demonstrates that CCT2 promotes glioblastoma progression by directly binding to and enhancing the stability of the KRAS protein. Additionally, dihydroartemisinin inhibits glioblastoma by targeting the CCT2 and the following KRAS signaling. Our findings overcome the challenge posed by the undruggable nature of KRAS and offer potential therapeutic strategies for glioblastoma treatment.
Collapse
Affiliation(s)
- Feihu Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, PR China; Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250017, PR China
| | - Zhong Yao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, PR China
| | - Yaquan Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, PR China; Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250017, PR China
| | - Wenbo Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, PR China; Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250017, PR China
| | - Yanfei Sun
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, PR China; Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250017, PR China
| | - Xiaobing Yang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, PR China; Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250017, PR China
| | - Zhimin Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, PR China; Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250017, PR China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, PR China; Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250017, PR China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, PR China; Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250017, PR China; Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, PR China; Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250017, PR China.
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, 250012, PR China; Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function Remodeling, Jinan, 250017, PR China.
| |
Collapse
|
39
|
Wang Y, Sun N, He R, Wang Z, Jin J, Gao T, Qu J. Molecular characterization of m6A RNA methylation regulators with features of immune dysregulation in IgA nephropathy. Clin Exp Med 2024; 24:92. [PMID: 38693353 PMCID: PMC11062981 DOI: 10.1007/s10238-024-01346-8] [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: 12/10/2023] [Accepted: 04/02/2024] [Indexed: 05/03/2024]
Abstract
The role of RNA N6-methyladenosine (m6A) modification in immunity is being elucidated. This study aimed to explore the potential association between m6A regulators and the immune microenvironment in IgA nephropathy (IgAN). The expression profiles of 24 m6A regulators in 107 IgAN patients were obtained from the Gene Expression Omnibus (GEO) database. The least absolute shrinkage and selection operator (LASSO) regression and logistic regression analysis were utilized to construct a model for distinguishing IgAN from control samples. Based on the expression levels of m6A regulators, unsupervised clustering was used to identify m6A-induced molecular clusters in IgAN. Gene set enrichment analysis (GSEA) and immunocyte infiltration among different clusters were examined. The gene modules with the highest correlation for each of the three clusters were identified by weighted gene co-expression network analysis (WGCNA). A model containing 10 m6A regulators was developed using LASSO and logistic regression analyses. Three molecular clusters were determined using consensus clustering of 24 m6A regulators. A decrease in the expression level of YTHDF2 in IgAN samples was significantly negatively correlated with an increase in resting natural killer (NK) cell infiltration and was positively correlated with the abundance of M2 macrophage infiltration. The risk scores calculated by the nomogram were significantly higher for cluster-3, and the expression levels of m6A regulators in this cluster were generally low. Immunocyte infiltration and pathway enrichment results for cluster-3 differed significantly from those for the other two clusters. Finally, the expression of YTHDF2 was significantly decreased in IgAN based on immunohistochemical staining. This study demonstrated that m6A methylation regulators play a significant role in the regulation of the immune microenvironment in IgAN. Based on m6A regulator expression patterns, IgAN can be classified into multiple subtypes, which might provide additional insights into novel therapeutic methods for IgAN.
Collapse
Affiliation(s)
- Yihao Wang
- Department of Nephrology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Nan Sun
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Rui He
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Zida Wang
- Department of Emergency, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jingsi Jin
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ting Gao
- Department of Emergency, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Junwen Qu
- Department of Urology, Jiading Branch, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201899, China.
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
40
|
Gong Z, Xue L, Vlantis AC, van Hasselt CA, Chan JYK, Fang J, Wang R, Yang Y, Li D, Zeng X, Tong MCF, Chen GG. Brusatol attenuated proliferation and invasion induced by KRAS in differentiated thyroid cancer through inhibiting Nrf2. J Endocrinol Invest 2024; 47:1271-1280. [PMID: 38062319 DOI: 10.1007/s40618-023-02248-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/13/2023] [Indexed: 04/23/2024]
Abstract
BACKGROUND Poorly differentiated thyroid cancer (PDTC) and anaplastic thyroid cancer (ATC) can be developed from differentiated thyroid cancer, and this dedifferentiated transformation leads to poor prognosis and high mortality. The role of Nrf2 in the dedifferentiation of differentiated thyroid cancer (DTC) induced by KRAS remains unclear. METHODS AND MATERIALS In this study, two DTC cell lines, BCPAP and WRO, were used to evaluate the function of Nrf2 in the dedifferentiation caused by wild-type KRAS (KRAS-WT) and G12V point mutation KRAS (KRAS-G12V). RESULTS The overexpression of KRAS-WT and KRAS-G12V increased the proliferative and invasive ability of BCPAP and WRO cells. Aggressive morphology was observed in KRAS-WT and KRAS-G12V overexpressed WRO cells. These results suggested that overexpression of KRAS-WT or KRAS-G12V may induce dedifferentiation in DTC cells. The expression of Nrf2 was increased by KRAS-WT and KRAS-G12V in DTC cells. In addition, compared with normal thyroid tissues, the expression of Nrf2 protein was considerably higher in thyroid cancer tissues on immunohistochemistry (IHC) staining, and the increased expression of Nrf2 indicated a poor prognosis of thyroid cancer. These results indicated that Nrf2 is the KRAS downstream molecule in thyroid cancer. Functional studies showed that the Nrf2 inhibitor Brusatol counteracted the proliferative and invasive abilities induced by KRAS-WT and KRAS-G12V in BCPAP and WRO cells. In addition, the xenograft assay further confirmed that Brusatol inhibits tumor growth induced by KRAS-WT and KRAS-G12V. CONCLUSION Collectively, this study suggests that Nrf2 could be a promising therapeutic target in KRAS-mediated dedifferentiation of thyroid cancer.
Collapse
Affiliation(s)
- Z Gong
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - L Xue
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - A C Vlantis
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - C A van Hasselt
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - J Y K Chan
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - J Fang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Otolaryngology Head and Neck Surgery (Ministry of Education of China), Beijing Institute of Otolaryngology, Beijing, China
| | - R Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Otolaryngology Head and Neck Surgery (Ministry of Education of China), Beijing Institute of Otolaryngology, Beijing, China
| | - Y Yang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Key Laboratory of Otolaryngology Head and Neck Surgery (Ministry of Education of China), Beijing Institute of Otolaryngology, Beijing, China
| | - D Li
- Shenzhen Key Laboratory of ENT, Institute of ENT and Longgang ENT Hospital, Shenzhen, Guangdong, China
| | - X Zeng
- Shenzhen Key Laboratory of ENT, Institute of ENT and Longgang ENT Hospital, Shenzhen, Guangdong, China
| | - M C F Tong
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China.
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, China.
| | - G G Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China.
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, China.
| |
Collapse
|
41
|
Ríos-Hoyo A, Monzonís X, Vidal J, Linares J, Montagut C. Unveiling acquired resistance to anti-EGFR therapies in colorectal cancer: a long and winding road. Front Pharmacol 2024; 15:1398419. [PMID: 38711991 PMCID: PMC11070789 DOI: 10.3389/fphar.2024.1398419] [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: 03/09/2024] [Accepted: 04/03/2024] [Indexed: 05/08/2024] Open
Abstract
Emergence of acquired resistance limits the efficacy of the anti-EGFR therapies cetuximab and panitumumab in metastatic colorectal cancer. In the last decade, preclinical and clinical cohort studies have uncovered genomic alterations that confer a selective advantage to tumor cells under EGFR blockade, mainly downstream re-activation of RAS-MEK signaling and mutations in the extracellular domain of EGFR (EGFR-ECD). Liquid biopsies (genotyping of ctDNA) have been established as an excellent tool to easily monitor the dynamics of genomic alterations resistance in the blood of patients and to select patients for rechallenge with anti-EGFR therapies. Accordingly, several clinical trials have shown clinical benefit of rechallenge with anti-EGFR therapy in genomically-selected patients using ctDNA. However, alternative mechanisms underpinning resistance beyond genomics -mainly related to the tumor microenvironment-have been unveiled, specifically relevant in patients receiving chemotherapy-based multi-drug treatment in first line. This review explores the complexity of the multifaceted mechanisms that mediate secondary resistance to anti-EGFR therapies and potential therapeutic strategies to circumvent acquired resistance.
Collapse
Affiliation(s)
- Alejandro Ríos-Hoyo
- Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Xavier Monzonís
- Department of Medical Oncology, Hospital del Mar Research Institute, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Joana Vidal
- Department of Medical Oncology, Hospital del Mar Research Institute, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Jenniffer Linares
- Department of Medical Oncology, Hospital del Mar Research Institute, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Clara Montagut
- Department of Medical Oncology, Hospital del Mar Research Institute, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| |
Collapse
|
42
|
Hu J, Xu H, Wu T, Zhang C, Shen H, Dong R, Hu Q, Xiang Q, Chai S, Luo G, Chen X, Huang Y, Zhao X, Peng C, Wu X, Lin B, Zhang Y, Xu Y. Discovery of Highly Potent and Efficient CBP/p300 Degraders with Strong In Vivo Antitumor Activity. J Med Chem 2024. [PMID: 38649304 DOI: 10.1021/acs.jmedchem.3c02195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
The transcriptional coactivator cAMP response element binding protein (CREB)-binding protein (CBP) and its homologue p300 have emerged as attractive therapeutic targets for human cancers such as acute myeloid leukemia (AML). Herein, we report the design, synthesis, and biological evaluation of a series of cereblon (CRBN)-recruiting CBP/p300 proteolysis targeting chimeras (PROTACs) based on the inhibitor CCS1477. The representative compounds 14g (XYD190) and 14h (XYD198) potently inhibited the growth of AML cells with low nanomolar IC50 values and effectively degraded CBP and p300 proteins in a concentration- and time-dependent manner. Mechanistic studies confirmed that 14g and 14h can selectively bind to CBP/p300 bromodomains and induce CBP and p300 degradation in bromodomain family proteins in a CRBN- and proteasome-dependent manner. 14g and 14h displayed remarkable antitumor efficacy in the MV4;11 xenograft model (TGI = 88% and 93%, respectively). Our findings demonstrated that 14g and 14h are useful lead compounds and deserve further optimization and activity evaluation for the treatment of human cancers.
Collapse
Affiliation(s)
- Jiankang Hu
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, No. 19 Yuquan Road, Beijing 100049, China
| | - Hongrui Xu
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Guangzhou Medical University, Guangzhou 511436, China
| | - Tianbang Wu
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Cheng Zhang
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Hui Shen
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Ruibo Dong
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
- School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Qingqing Hu
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Qiuping Xiang
- Ningbo No. 2 Hospital, Ningbo, Zhejiang 315010, China
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, Zhejiang 315010, China
| | - Shuang Chai
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Guolong Luo
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Xiaoshan Chen
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, No. 19 Yuquan Road, Beijing 100049, China
| | - Yumin Huang
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, No. 19 Yuquan Road, Beijing 100049, China
| | - Xiaofan Zhao
- Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Guangzhou Medical University, Guangzhou 511436, China
| | - Chao Peng
- Jiangsu S&T Exchange Center with Foreign Countries, No. 175 Longpan Road, Nanjing 210042, China
| | - Xishan Wu
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Bin Lin
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yan Zhang
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
| | - Yong Xu
- China-New Zealand Joint Laboratory of Biomedicine and Health, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Biocomputing, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, No. 190 Kaiyuan Avenue, Guangzhou 510530, China
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
43
|
Ma F, Li Y, Cai M, Yang W, Wu Z, Dong J, Qin JJ. ML162 derivatives incorporating a naphthoquinone unit as ferroptosis/apoptosis inducers: Design, synthesis, anti-cancer activity, and drug-resistance reversal evaluation. Eur J Med Chem 2024; 270:116387. [PMID: 38593589 DOI: 10.1016/j.ejmech.2024.116387] [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: 01/09/2024] [Revised: 04/01/2024] [Accepted: 04/01/2024] [Indexed: 04/11/2024]
Abstract
Activating apoptosis has long been viewed as an anti-cancer process, but recently increasing evidence has accumulated that induction of ferroptosis has emerged as a promising strategy for cancer therapeutics. Glutathione peroxidase 4 (GPX4) is one of the pivotal factors regulating ferroptosis that targeted inhibition or degradation of GPX4 could effectively trigger ferroptosis. In this study, a series of ML162-quinone conjugates were constructed by using pharmacophore hybridization and bioisosterism strategies, with the aim of obtaining more active anticancer agents via the ferroptosis and apoptosis dual cell death processes. Of these compounds, GIC-20 was identified as the most active one that exhibited promising anticancer activity both in vitro and in vivo via ferroptosis and apoptosis dual-targeting processes, without obvious toxicity compared with ML162. On one hand, GIC-20 could trigger ferroptosis in cells by inducing intracellular lipid peroxide and ROS accumulation, and destroying mitochondrial structure. In addition to GPX4 inhibition, GIC-20 can also trigger ferroptosis via proteasomal-mediated degradation of GPX4, suggesting GIC-20 may function as a molecule glue degrader. On the other hand, GIC-20 can also induce apoptosis via upregulating the level of apoptotic protein Bax and downregulating the level of anti-apoptotic protein Bcl-2 in HT1080 cells. Furthermore, GIC-20 also enhanced the sensitivity of resistant MIA-PaCa-2-AMG510R cells to AMG510, suggesting the great potential of GIC-20 in overcoming the acquired resistance of KRASG12C inhibitors. Overall, GIC-20 represents a novel dual ferroptosis/apoptosis inducer warranting further development for cancer therapeutics and overcoming drug resistance.
Collapse
Affiliation(s)
- Furong Ma
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhoum, 310053, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Yulong Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhoum, 310053, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Maohua Cai
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhoum, 310053, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Wenyan Yang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China; College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Zumei Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhoum, 310053, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Jinyun Dong
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| | - Jiang-Jiang Qin
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| |
Collapse
|
44
|
Pagba CV, Gupta AK, Dilsha K, Sadrpour P, Jakubec J, Prakash P, van der Hoeven D, Cho KJ, Gilbertson S, Gorfe AA. Biophysical and Biochemical Characterization of Structurally Diverse Small Molecule Hits for KRAS Inhibition. Chembiochem 2024; 25:e202300827. [PMID: 38349283 DOI: 10.1002/cbic.202300827] [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: 12/12/2023] [Revised: 02/12/2024] [Indexed: 03/08/2024]
Abstract
We describe six compounds as early hits for the development of direct inhibitors of KRAS, an important anticancer drug target. We show that these compounds bind to KRAS with affinities in the low micromolar range and exert different effects on its interactions with binding partners. Some of the compounds exhibit selective binding to the activated form of KRAS and inhibit signal transduction through both the MAPK or the phosphatidylinositide 3-kinase PI3K-protein kinase B (AKT) pathway in cells expressing mutant KRAS. Most inhibit intrinsic and/or SOS-mediated KRAS activation while others inhibit RAS-effector interaction. We propose these compounds as starting points for the development of non-covalent allosteric KRAS inhibitors.
Collapse
Affiliation(s)
- Cynthia V Pagba
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, Texas, 77030, USA
| | - Amit K Gupta
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, Texas, 77030, USA
| | - Kasuni Dilsha
- Department of Chemistry, University of Houston, 3585 Cullen Blvd., Houston, TX 77204, USA
| | - Parisa Sadrpour
- Department of Biochemistry and Molecular Biology, Wright State University, 3640 Colonel Glenn Hwy, Dayton, OH 45435, USA
| | - Jacob Jakubec
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, Texas, 77030, USA
| | - Priyanka Prakash
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, Texas, 77030, USA
| | - Dharini van der Hoeven
- Department of Diagnostic and Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, 7500 Cambridge St., Houston, Texas, 77030, USA
| | - Kwang-Jin Cho
- Department of Biochemistry and Molecular Biology, Wright State University, 3640 Colonel Glenn Hwy, Dayton, OH 45435, USA
| | - Scott Gilbertson
- Department of Chemistry, University of Houston, 3585 Cullen Blvd., Houston, TX 77204, USA
| | - Alemayehu A Gorfe
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin St., Houston, Texas, 77030, USA
- Biochemistry and Cell Biology Program & Therapeutics and Pharmacology Program, UTHealth MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, 6431 Fannin St., Houston, Texas, 77030, USA
| |
Collapse
|
45
|
Yang C, Lin ZI, Zhang X, Xu Z, Xu G, Wang YM, Tsai TH, Cheng PW, Law WC, Yong KT, Chen CK. Recent Advances in Engineering Carriers for siRNA Delivery. Macromol Biosci 2024; 24:e2300362. [PMID: 38150293 DOI: 10.1002/mabi.202300362] [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: 08/09/2023] [Revised: 11/29/2023] [Indexed: 12/28/2023]
Abstract
RNA interference (RNAi) technology has been a promising treatment strategy for combating intractable diseases. However, the applications of RNAi in clinical are hampered by extracellular and intracellular barriers. To overcome these barriers, various siRNA delivery systems have been developed in the past two decades. The first approved RNAi therapeutic, Patisiran (ONPATTRO) using lipids as the carrier, for the treatment of amyloidosis is one of the most important milestones. This has greatly encouraged researchers to work on creating new functional siRNA carriers. In this review, the recent advances in siRNA carriers consisting of lipids, polymers, and polymer-modified inorganic particles for cancer therapy are summarized. Representative examples are presented to show the structural design of the carriers in order to overcome the delivery hurdles associated with RNAi therapies. Finally, the existing challenges and future perspective for developing RNAi as a clinical modality will be discussed and proposed. It is believed that the addressed contributions in this review will promote the development of siRNA delivery systems for future clinical applications.
Collapse
Affiliation(s)
- Chengbin Yang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Zheng-Ian Lin
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Xinmeng Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Zhourui Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Gaixia Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Yu-Min Wang
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Tzu-Hsien Tsai
- Division of Cardiology and Department of Internal Medicine, Ditmanson Medical Foundation Chiayi Christian Hospital, Chiayi, 60002, Taiwan
| | - Pei-Wen Cheng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 81362, Taiwan
- Department of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| | - Wing-Cheung Law
- Department of Industrial and Systems Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, P. R. China
| | - Ken-Tye Yong
- School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Chih-Kuang Chen
- Polymeric Biomaterials Laboratory, Department of Materials and Optoelectronic Science, National Sun Yat-Sen University, Kaohsiung, 80424, Taiwan
| |
Collapse
|
46
|
Kumarasamy V, Wang J, Frangou C, Wan Y, Dynka A, Rosenheck H, Dey P, Abel EV, Knudsen ES, Witkiewicz AK. The Extracellular Niche and Tumor Microenvironment Enhance KRAS Inhibitor Efficacy in Pancreatic Cancer. Cancer Res 2024; 84:1115-1132. [PMID: 38294344 PMCID: PMC10982648 DOI: 10.1158/0008-5472.can-23-2504] [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: 08/20/2023] [Revised: 11/28/2023] [Accepted: 01/25/2024] [Indexed: 02/01/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease that lacks effective treatment options, highlighting the need for developing new therapeutic interventions. Here, we assessed the response to pharmacologic inhibition of KRAS, the central oncogenic driver of PDAC. In a panel of PDAC cell lines, inhibition of KRASG12D with MRTX1133 yielded variable efficacy in suppressing cell growth and downstream gene expression programs in 2D cultures. On the basis of CRISPR-Cas9 loss-of-function screens, ITGB1 was identified as a target to enhance the therapeutic response to MRTX1133 by regulating mechanotransduction signaling and YAP/TAZ expression, which was confirmed by gene-specific knockdown and combinatorial drug synergy. Interestingly, MRTX1133 was considerably more efficacious in 3D cell cultures. Moreover, MRTX1133 elicited a pronounced cytostatic effect in vivo and controlled tumor growth in PDAC patient-derived xenografts. In syngeneic models, KRASG12D inhibition led to tumor regression that did not occur in immune-deficient hosts. Digital spatial profiling on tumor tissues indicated that MRTX1133-mediated KRAS inhibition enhanced IFNγ signaling and induced antigen presentation that modulated the tumor microenvironment. Further investigation of the immunologic response using single-cell sequencing and multispectral imaging revealed that tumor regression was associated with suppression of neutrophils and influx of effector CD8+ T cells. Together, these findings demonstrate that both tumor cell-intrinsic and -extrinsic events contribute to response to MRTX1133 and credential KRASG12D inhibition as a promising therapeutic strategy for a large percentage of patients with PDAC. SIGNIFICANCE Pharmacologic inhibition of KRAS elicits varied responses in pancreatic cancer 2D cell lines, 3D organoid cultures, and xenografts, underscoring the importance of mechanotransduction and the tumor microenvironment in regulating therapeutic responses.
Collapse
Affiliation(s)
- Vishnu Kumarasamy
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Jianxin Wang
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Costakis Frangou
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Yin Wan
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Andrew Dynka
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Hanna Rosenheck
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Prasenjit Dey
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Ethan V. Abel
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Erik S. Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Agnieszka K. Witkiewicz
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
47
|
Huang J, Zhang Y, Fang L, Xi F, Tang C, Ou K, Wang C. Chronic exposure to low levels of phenanthrene induces histological damage and carcinogenic risk in the uterus of female mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:22858-22869. [PMID: 38413531 DOI: 10.1007/s11356-024-32636-z] [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: 08/22/2023] [Accepted: 02/21/2024] [Indexed: 02/29/2024]
Abstract
Phenanthrene (Phe), a polycyclic aromatic hydrocarbon with low molecular weight, is detected in the environment at high frequency. To study the toxic effects of Phe on the uterine structure and function, female Kunming mice were exposed to Phe (0.05, 0.5, 5 ng/mL) for 270 days by drinking water. Pathological alterations and their action pathways were analyzed using immunohistochemical and biomolecular technology. Phe significantly increased the percentage of blood vessel area, the number of endometrial neutrophils (indicating the occurrence of inflammation), collagen deposition (indicating fibrosis), and the percentage of Ki-67-positive cells (indicating carcinogenesis) in the uterus. Transcriptome sequencing identified differentially expressed genes that were mainly enriched in some signaling pathways, including inflammation and carcinogenesis, suggesting a carcinogenic risk in the Phe-exposed uterus. Elevated serum estrogen levels and decreased progesterone levels exhibited a disturbance of steroid hormone balance, which might be related to uterine damage. Upregulated protein levels of uterine androgen receptor and estrogen receptor α were linked to the pathological effects. Most of the effects exhibited a nonmonotonic dose response, which might be attributed to the corresponding change in the serum levels of Phe. The results suggest that exposure to low levels of Phe could exert adverse effects on the uterus.
Collapse
Affiliation(s)
- Jie Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Ying Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Lu Fang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Feifei Xi
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Chen Tang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Kunlin Ou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China.
| |
Collapse
|
48
|
Aslam M, Rajbdad F, Azmat S, Li Z, Boudreaux JP, Thiagarajan R, Yao S, Xu J. A novel method for detection of pancreatic Ductal Adenocarcinoma using explainable machine learning. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 245:108019. [PMID: 38237450 DOI: 10.1016/j.cmpb.2024.108019] [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: 04/23/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/15/2024]
Abstract
BACKGROUND AND OBJECTIVE Pancreatic Ductal Adenocarcinoma (PDAC) is a form of pancreatic cancer that is one of the primary causes of cancer-related deaths globally, with less than 10 % of the five years survival rate. The prognosis of pancreatic cancer has remained poor in the last four decades, mainly due to the lack of early diagnostic mechanisms. This study proposes a novel method for detecting PDAC using explainable and supervised machine learning from Raman spectroscopic signals. METHODS An insightful feature set consisting of statistical, peak, and extended empirical mode decomposition features is selected using the support vector machine recursive feature elimination method integrated with a correlation bias reduction. Explicable features successfully identified mutations in Kirsten rat sarcoma viral oncogene homolog (KRAS) and tumor suppressor protein53 (TP53) in the fingerprint region for the first time in the literature. PDAC and normal pancreas are classified using K-nearest neighbor, linear discriminant analysis, and support vector machine classifiers. RESULTS This study achieved a classification accuracy of 98.5% using a nonlinear support vector machine. Our proposed method reduced test time by 28.5 % and saved 85.6 % memory utilization, which reduces complexity significantly and is more accurate than the state-of-the-art method. The generalization of the proposed method is assessed by fifteen-fold cross-validation, and its performance is evaluated using accuracy, specificity, sensitivity, and receiver operating characteristic curves. CONCLUSIONS In this study, we proposed a method to detect and define the fingerprint region for PDAC using explainable machine learning. This simple, accurate, and efficient method for PDAC detection in mice could be generalized to examine human pancreatic cancer and provide a basis for precise chemotherapy for early cancer treatment.
Collapse
Affiliation(s)
- Murtaza Aslam
- Department of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Fozia Rajbdad
- Department of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Shoaib Azmat
- Department of Electrical and Computer Engineering, COMSATS University Islamabad, Pakistan
| | - Zheng Li
- Department of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - J Philip Boudreaux
- Department of Surgery, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Ramcharan Thiagarajan
- Department of Surgery, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Shaomian Yao
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Jian Xu
- Department of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
49
|
Lv Y, Yang Z, Chen Y, Ma X, Guo M, Zhang C, Jiang X, Wang C, Li Z, Tai Z, Wang X, Zhang S, Ma S, Qin C. A Potent SOS1 PROTAC Degrader with Synergistic Efficacy in Combination with KRAS G12C Inhibitor. J Med Chem 2024; 67:2487-2511. [PMID: 38316747 DOI: 10.1021/acs.jmedchem.3c01598] [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: 02/07/2024]
Abstract
AMG510, as the first approved inhibitor for KRASG12C mutation, has shown promising efficacy in nonsmall-cell lung cancer and colorectal cancer harboring KRASG12C mutation. However, the moderate response rate and the rapid emergence of acquired resistance limit the therapeutic potential of AMG510, highlighting the need for the development of combination strategies. Here, we observed the suppression of RAS-MAPK signaling induced by AMG510 was prolonged and enhanced by SOS1 knockdown. Thus, we design, synthesize, and characterize a potent and specific SOS1 degrader 23. Compound 23 showed efficient SOS1 degradation in KRAS-driven cancer cells and achieved significant antiproliferative potency. Importantly, the combination of 23 with AMG510 suppressed RAS signaling feedback activation, showing synergistic effects against KRASG12C mutant cells in vitro and in vivo. Our findings demonstrated that KRASG12C inhibition plus SOS1 degradation as a potential therapeutic strategy to improve antitumor response and overcome acquired resistance to KRASG12C inhibitor.
Collapse
Affiliation(s)
- Yan Lv
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, China
| | - Zixuan Yang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, China
| | - Yiming Chen
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, China
| | - Xuepei Ma
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, China
| | - Mengqi Guo
- Center for Targeted Protein Degradation and Drug Discovery, Ocean University of China, Qingdao, Shandong 266003, China
| | - Chengwei Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, China
| | - Xiaolin Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, China
- Center for Targeted Protein Degradation and Drug Discovery, Ocean University of China, Qingdao, Shandong 266003, China
| | - Chengli Wang
- Center for Targeted Protein Degradation and Drug Discovery, Ocean University of China, Qingdao, Shandong 266003, China
| | - Zhuoyue Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, China
- Center for Targeted Protein Degradation and Drug Discovery, Ocean University of China, Qingdao, Shandong 266003, China
| | - Zhengfu Tai
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, China
- Center for Targeted Protein Degradation and Drug Discovery, Ocean University of China, Qingdao, Shandong 266003, China
| | - Xiao Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, China
- Center for Targeted Protein Degradation and Drug Discovery, Ocean University of China, Qingdao, Shandong 266003, China
| | - Siqi Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, China
- Center for Targeted Protein Degradation and Drug Discovery, Ocean University of China, Qingdao, Shandong 266003, China
| | - Shumin Ma
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, China
- Center for Targeted Protein Degradation and Drug Discovery, Ocean University of China, Qingdao, Shandong 266003, China
| | - Chong Qin
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, China
- Center for Targeted Protein Degradation and Drug Discovery, Ocean University of China, Qingdao, Shandong 266003, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, Shandong 266237, China
| |
Collapse
|
50
|
Jiang Z, Li Y, Zhou X, Wen J, Zheng P, Zhu W. Research progress on small molecule inhibitors targeting KRAS G12C with acrylamide structure and the strategies for solving KRAS inhibitor resistance. Bioorg Med Chem 2024; 100:117627. [PMID: 38310752 DOI: 10.1016/j.bmc.2024.117627] [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: 11/30/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/06/2024]
Abstract
KRAS (Kirsten-RAS) is a highly mutated gene in the RAS (rat sarcoma) gene family that acts as a critical switch in intracellular signaling pathways, regulating cell proliferation, differentiation, and survival. The continuous activation of KRAS protein resulting from mutations leads to the activation of multiple downstream signaling pathways, inducing the development of malignant tumors. Despite the significant role of KRAS in tumorigenesis, targeted drugs against KRAS gene mutations have failed, and KRAS was once considered an undruggable target. The development of KRAS G12C mutant conformational modulators and the introduction of Sotorasib (R&D code: AMG510) have been a breakthrough in this field, with its remarkable clinical outcomes. Consequently, there is now a great number of KRAS G12C mutations. Patent applications for mutant GTPase KRAS G12C inhibitors, which are said to be covalently modified by cysteine codon 12, have been submitted since 2014. This review classifies KRAS G12C inhibitors based on their chemical structure and evaluates their biological properties. Additionally, it discusses the obstacles encountered in KRAS inhibitor research and the corresponding solutions.
Collapse
Affiliation(s)
- Zhiyan Jiang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China
| | - Yan Li
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China
| | - Xin Zhou
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China
| | - Jie Wen
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China
| | - Pengwu Zheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China.
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang, Jiangxi 330013, China.
| |
Collapse
|