1
|
Pla I, Szabolcs BL, Péter PN, Ujfaludi Z, Kim Y, Horvatovich P, Sanchez A, Pawlowski K, Wieslander E, Kuras M, Murillo JR, Guedes J, Pál DM, Ascsillán AA, Betancourt LH, Németh IB, Gil J, de Almeida NP, Szeitz B, Szadai L, Doma V, Woldmar N, Bartha Á, Pahi Z, Pankotai T, Győrffy B, Szasz AM, Domont G, Nogueira F, Kwon HJ, Appelqvist R, Kárpáti S, Fenyö D, Malm J, Marko‐Varga G, Kemény LV. Unbiased Drug Target Prediction Reveals Sensitivity to Ferroptosis Inducers, HDAC and RTK Inhibitors in Melanoma Subtypes. Int J Dermatol 2025; 64:870-881. [PMID: 39722169 PMCID: PMC12008611 DOI: 10.1111/ijd.17586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/12/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND The utilization of PD1 and CTLA4 inhibitors has revolutionized the treatment of malignant melanoma (MM). However, resistance to targeted and immune-checkpoint-based therapies still poses a significant problem. OBJECTIVE Here, we mine large-scale MM proteogenomic data to identify druggable targets and forecast treatment efficacy and resistance. METHODS Leveraging protein profiles from established MM subtypes and molecular structures of 82 cancer treatment drugs, we identified nine candidate hub proteins, mTOR, FYN, PIK3CB, EGFR, MAPK3, MAP4K1, MAP2K1, SRC, and AKT1, across five distinct MM subtypes. These proteins are potential drug targets applicable to one or multiple MM subtypes. Additionally, by integrating proteogenomic profiles obtained from MM subtypes with MM cell line dependency and drug sensitivity data, we identified a total of 162 potentially targetable genes. Lastly, we identified 20 compounds exhibiting potential drug impact in at least one melanoma subtype. RESULTS Employing these unbiased approaches, we have uncovered compounds targeting ferroptosis demonstrating a striking 30× fold difference in sensitivity among different subtypes. CONCLUSIONS Our results suggest innovative and novel therapeutic strategies by stratifying melanoma samples through proteomic profiling, offering a spectrum of novel therapeutic interventions and prospects for combination therapy.
Collapse
Affiliation(s)
- Indira Pla
- Department of Biomedical Engineering, Faculty of EngineeringLTH, Lund UniversityLundSweden
- European Cancer Moonshot Lund CenterLundSweden
| | - Botond L. Szabolcs
- HCEMM‐SU Translational Dermatology Research GroupSemmelweis UniversityBudapestHungary
- Department of Physiology, Faculty of MedicineSemmelweis UniversityBudapestHungary
- Department of Dermatology, Venereology and Dermatooncology, Faculty of MedicineSemmelweis UniversityBudapestHungary
- MTA‐SE Lendület “Momentum” Dermatology Research GroupHungarian Academy of Sciences and Semmelweis UniversityBudapestHungary
| | - Petra Nikolett Péter
- HCEMM‐SU Translational Dermatology Research GroupSemmelweis UniversityBudapestHungary
- Department of Physiology, Faculty of MedicineSemmelweis UniversityBudapestHungary
- Department of Dermatology, Venereology and Dermatooncology, Faculty of MedicineSemmelweis UniversityBudapestHungary
- Department of Dermatology and Allergology, Albert Szent‐Györgyi Medical SchoolUniversity of SzegedSzegedHungary
| | - Zsuzsanna Ujfaludi
- Department of Pathology, Albert Szent‐Györgyi Medical SchoolUniversity of SzegedSzegedHungary
- Competence Centre of the Life Sciences Cluster of the Centre of Excellence for Interdisciplinary Research, Development and InnovationUniversity of SzegedSzegedHungary
| | - Yonghyo Kim
- Drug Discovery Platform Research Center, Therapeutics and Biotechnology DivisionKorea Research Institute of Chemical TechnologyDaejeonRepublic of Korea
| | - Peter Horvatovich
- Groningen Research Institute of Pharmacy, Analytical Biochemistry, University of GroningenGroningenThe Netherlands
| | - Aniel Sanchez
- Section for Clinical Chemistry, Department of Translational MedicineSkåne University Hospital MalmöMalmöSweden
| | - Krzysztof Pawlowski
- Section for Clinical Chemistry, Department of Translational MedicineSkåne University Hospital MalmöMalmöSweden
- Department of Biochemistry and MicrobiologyWarsaw University of Life SciencesWarszawaPoland
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Elisabet Wieslander
- Section for Clinical Chemistry, Department of Translational MedicineSkåne University Hospital MalmöMalmöSweden
| | - Magdalena Kuras
- Department of Biomedical Engineering, Faculty of EngineeringLTH, Lund UniversityLundSweden
- European Cancer Moonshot Lund CenterLundSweden
| | | | - Jéssica Guedes
- European Cancer Moonshot Lund CenterLundSweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical EngineeringLund UniversityLundSweden
- Chemistry Institute Federal, University of Rio de JaneiroRio de JaneiroBrazil
| | - Dorottya M.P. Pál
- HCEMM‐SU Translational Dermatology Research GroupSemmelweis UniversityBudapestHungary
- Department of Physiology, Faculty of MedicineSemmelweis UniversityBudapestHungary
- Department of Dermatology, Venereology and Dermatooncology, Faculty of MedicineSemmelweis UniversityBudapestHungary
| | - Anna A. Ascsillán
- HCEMM‐SU Translational Dermatology Research GroupSemmelweis UniversityBudapestHungary
- Department of Physiology, Faculty of MedicineSemmelweis UniversityBudapestHungary
- Department of Dermatology, Venereology and Dermatooncology, Faculty of MedicineSemmelweis UniversityBudapestHungary
| | - Lazaro Hiram Betancourt
- European Cancer Moonshot Lund CenterLundSweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical EngineeringLund UniversityLundSweden
| | - István Balázs Németh
- Department of Dermatology and Allergology, Albert Szent‐Györgyi Medical SchoolUniversity of SzegedSzegedHungary
| | - Jeovanis Gil
- European Cancer Moonshot Lund CenterLundSweden
- Department of Translational MedicineLund UniversityLundSweden
| | - Natália Pinto de Almeida
- European Cancer Moonshot Lund CenterLundSweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical EngineeringLund UniversityLundSweden
- Chemistry Institute Federal, University of Rio de JaneiroRio de JaneiroBrazil
| | - Beáta Szeitz
- Division of Oncology, Department of Internal Medicine and OncologySemmelweis UniversityBudapestHungary
| | - Leticia Szadai
- Department of Dermatology and Allergology, Albert Szent‐Györgyi Medical SchoolUniversity of SzegedSzegedHungary
| | - Viktória Doma
- Department of Dermatology and Allergology, Albert Szent‐Györgyi Medical SchoolUniversity of SzegedSzegedHungary
| | - Nicole Woldmar
- European Cancer Moonshot Lund CenterLundSweden
- Thermo Fisher ScientificWalthamMAUSA
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical EngineeringLund UniversityLundSweden
| | - Áron Bartha
- Department of BioinformaticsSemmelweis UniversityBudapestHungary
- Research Centre for Natural SciencesInstitute of Molecular Life SciencesBudapestHungary
| | - Zoltan Pahi
- MTA‐SE Lendület “Momentum” Dermatology Research GroupHungarian Academy of Sciences and Semmelweis UniversityBudapestHungary
- Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Genome Integrity and DNA Repair Core GroupUniversity of SzegedSzegedHungary
| | - Tibor Pankotai
- Department of Pathology, Albert Szent‐Györgyi Medical SchoolUniversity of SzegedSzegedHungary
- Competence Centre of the Life Sciences Cluster of the Centre of Excellence for Interdisciplinary Research, Development and InnovationUniversity of SzegedSzegedHungary
- Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Genome Integrity and DNA Repair Core GroupUniversity of SzegedSzegedHungary
| | - Balázs Győrffy
- Division of Oncology, Department of Internal Medicine and OncologySemmelweis UniversityBudapestHungary
- Research Centre for Natural SciencesInstitute of Molecular Life SciencesBudapestHungary
| | - A. Marcell Szasz
- Division of Oncology, Department of Internal Medicine and OncologySemmelweis UniversityBudapestHungary
| | - Gilberto Domont
- Chemistry Institute Federal, University of Rio de JaneiroRio de JaneiroBrazil
| | - Fábio Nogueira
- Proteomics UnitInstitute of Chemistry and Research Center for Precision Medicine, Institute of Biophysics Carlos Chagas Filho, Federal Univesity of Rio de JaneiroRio de JaneiroBrazil
| | - Ho Jeong Kwon
- Chemical Genomics Leader Research Laboratory, Department of BiotechnologyCollege of Life Science and Biotechnology, Yonsei UniversitySeoulKorea
| | - Roger Appelqvist
- European Cancer Moonshot Lund CenterLundSweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical EngineeringLund UniversityLundSweden
| | - Sarolta Kárpáti
- Department of Dermatology, Venereology and Dermatooncology, Faculty of MedicineSemmelweis UniversityBudapestHungary
| | - David Fenyö
- Institute for Systems Genetics, NYU Grossman School of MedicineNew YorkNYUSA
- Department of Biochemistry and Molecular PharmacologyNYU Grossman School of MedicineNew YorkNYUSA
| | - Johan Malm
- Section for Clinical Chemistry, Department of Translational MedicineSkåne University Hospital MalmöMalmöSweden
| | - György Marko‐Varga
- European Cancer Moonshot Lund CenterLundSweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical EngineeringLund UniversityLundSweden
| | - Lajos V. Kemény
- HCEMM‐SU Translational Dermatology Research GroupSemmelweis UniversityBudapestHungary
- Department of Physiology, Faculty of MedicineSemmelweis UniversityBudapestHungary
- Department of Dermatology, Venereology and Dermatooncology, Faculty of MedicineSemmelweis UniversityBudapestHungary
- MTA‐SE Lendület “Momentum” Dermatology Research GroupHungarian Academy of Sciences and Semmelweis UniversityBudapestHungary
| |
Collapse
|
2
|
Chalepaki AM, Gkoris M, Chondrou I, Kourti M, Georgakopoulos-Soares I, Zaravinos A. A multi-omics analysis of effector and resting treg cells in pan-cancer. Comput Biol Med 2025; 189:110021. [PMID: 40088713 DOI: 10.1016/j.compbiomed.2025.110021] [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: 10/05/2024] [Revised: 02/09/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Regulatory T cells (Tregs) are critical for maintaining the stability of the immune system and facilitating tumor escape through various mechanisms. Resting T cells are involved in cell-mediated immunity and remain in a resting state until stimulated, while effector T cells promote immune responses. Here, we investigated the roles of two gene signatures, one for resting Tregs (FOXP3 and IL2RA) and another for effector Tregs (FOXP3, CTLA-4, CCR8 and TNFRSF9) in pan-cancer. Using data from The Cancer Genome Atlas (TCGA), The Cancer Proteome Atlas (TCPA) and Gene Expression Omnibus (GEO), we focused on the expression profile of the two signatures, the existence of single nucleotide variants (SNVs) and copy number variants (CNVs), methylation, infiltration of immune cells in the tumor and sensitivity to different drugs. Our analysis revealed that both signatures are differentially expressed across different cancer types, and correlate with patient survival. Furthermore, both types of Tregs influence important pathways in cancer development and progression, like apoptosis, epithelial-to-mesenchymal transition (EMT) and the DNA damage pathway. Moreover, a positive correlation was highlighted between the expression of gene markers in both resting and effector Tregs and immune cell infiltration in adrenocortical carcinoma, while mutations in both signatures correlated with enrichment of specific immune cells, mainly in skin melanoma and endometrial cancer. In addition, we reveal the existence of widespread CNVs and hypomethylation affecting both Treg signatures in most cancer types. Last, we identified a few correlations between the expression of CCR8 and TNFRSF9 and sensitivity to several drugs, including COL-3, Chlorambucil and GSK1070916, in pan-cancer. Overall, these findings highlight new evidence that both Treg signatures are crucial regulators of cancer progression, providing potential clinical outcomes for cancer therapy.
Collapse
Affiliation(s)
- Anna-Maria Chalepaki
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Marios Gkoris
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Irene Chondrou
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Malamati Kourti
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA.
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| |
Collapse
|
3
|
Mao Y, Shangguan D, Huang Q, Xiao L, Cao D, Zhou H, Wang YK. Emerging artificial intelligence-driven precision therapies in tumor drug resistance: recent advances, opportunities, and challenges. Mol Cancer 2025; 24:123. [PMID: 40269930 PMCID: PMC12016295 DOI: 10.1186/s12943-025-02321-x] [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: 02/07/2025] [Accepted: 04/02/2025] [Indexed: 04/25/2025] Open
Abstract
Drug resistance is one of the main reasons for cancer treatment failure, leading to a rapid recurrence/disease progression of the cancer. Recently, artificial intelligence (AI) has empowered physicians to use its powerful data processing and pattern recognition capabilities to extract and mine valuable drug resistance information from large amounts of clinical or omics data, to study drug resistance mechanisms, to evaluate and predict drug resistance, and to develop innovative therapeutic strategies to reduce drug resistance. In this review, we proposed a feasible workflow for incorporating AI into tumor drug resistance research, highlighted current AI-driven tumor drug resistance applications, and discussed the opportunities and challenges encountered in the process. Based on a comprehensive literature analysis, we systematically summarized the role of AI in tumor drug resistance research, including drug development, resistance mechanism elucidation, drug sensitivity prediction, combination therapy optimization, resistance phenotype identification, and clinical biomarker discovery. With the continuous advancement of AI technology and rigorous validation of clinical data, AI models are expected to fuel the development of precision oncology by improving efficacy, guiding therapeutic decisions, and optimizing patient prognosis. In summary, by leveraging clinical and omics data, AI models are expected to pioneer new therapy strategies to mitigate tumor drug resistance, improve efficacy and patient survival, and provide novel perspectives and tools for oncology treatment.
Collapse
Affiliation(s)
- Yuan Mao
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Lymphoma and Hematology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Dangang Shangguan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Qi Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ling Xiao
- Department of Histology and Embryology of Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Dongsheng Cao
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hui Zhou
- Department of Lymphoma and Hematology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China.
- Department of Lymphoma and Hematology, Hunan Cancer Hospital, Changsha, Hunan, People's Republic of China.
| | - Yi-Kun Wang
- Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
4
|
Zhang Y, Yang J, Gong Y, Liu Z, Yang Y, Song X, Gao Y, Xiong Y, Wang D, Fu K, Jia L, Shi X. RalB promotes lymph node metastasis in tongue squamous cell carcinoma. Genes Genomics 2025:10.1007/s13258-025-01628-9. [PMID: 40208483 DOI: 10.1007/s13258-025-01628-9] [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: 10/05/2024] [Accepted: 02/18/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Lymph nodes metastasis is the main metastasis mode of tongue squamous cell carcinoma (TSCC). Ras related GTP binding protein B (RalB) have been recently described that it was involved in tumor growth and metastasis, but the effect in TSCC is still ill-defined. OBJECTIVE This study provides insights into the role of RALB as a prognostic factor in head and neck squamous cell carcinoma (HNSCC) and demonstrates its involvement in promoting lymph node metastasis in TSCC. METHODS Firstly, the expression level of RALB and the relationship with clinical features were examined. Subsequently, RALB knockdown Cal-27 cells orthotopic xenotransplantation in the tongue of BALB/c nude mice were established. Finally, using Connectivity Map (CMAP) database to find possible drugs. RESULTS Firstly, RALB could not only predict the cancer patients' prognosis and survival and but also act as a potential prognostic factor, particularly in HNSCC by pan-cancer bioinformatics analysis. In addition, we found that RalB promoted tumor growth and lymph node metastasis. Finally, we identified Tirabrutinib (ONO-4059) targeting RalB with good binding properties. CONCLUSIONS RalB act as a prognostic gene in HNSCC, and promote lymph node metastasis in early stage of TSCC.
Collapse
Affiliation(s)
- Yuman Zhang
- Department of Otolaryngology Head and Neck Surgery, Chongqing General Hospital, Chongqing, 401147, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Jiali Yang
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yi Gong
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
| | - Zhihan Liu
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
| | - Yanguang Yang
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Xiaoyong Song
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
| | - Yuting Gao
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
| | - Yajun Xiong
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
| | - Dan Wang
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
| | - Kai Fu
- Department of Otolaryngology Head and Neck Surgery, The Fourth Hospital of Hebei Medical University, 12# Jiankang Road, Shijiazhuang, 050000, Hebei Province, China.
| | - Lifeng Jia
- Department of Otolaryngology Head and Neck Surgery, Chongqing General Hospital, Chongqing, 401147, China.
- Department of Otolaryngology Head and Neck Surgery, Chongqing General Hospital, No.118 Xingguang Avenue, Liangjiang New Area, Chongqing, 401147, China.
| | - Xinli Shi
- Department of Otolaryngology Head and Neck Surgery, Chongqing General Hospital, Chongqing, 401147, China.
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China.
| |
Collapse
|
5
|
Zhong Y, Zhang W, Zheng C, Wu H, Luo J, Yuan Z, Zhang H, Wang C, Feng H, Wang M, Zhang Q, Ju H, Wang G. Multi-omic analyses reveal PTPN6's impact on tumor immunity across various cancers. Sci Rep 2025; 15:11025. [PMID: 40164665 PMCID: PMC11958644 DOI: 10.1038/s41598-025-96302-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/27/2025] [Indexed: 04/02/2025] Open
Abstract
Protein Tyrosine Phosphatase Non-Receptor Type 6 (PTPN6) plays a crucial regulatory role in cellular processes and has been implicated in oncogenesis. This pan-cancer analysis aimed to elucidate PTPN6's involvement across various cancer types, with a particular emphasis on its association with tumor immunity. We analyzed PTPN6 expression data from open access databases using various statistical techniques, including survival analysis, genetic heterogeneity analysis, immune profiling, single-cell analysis, drug sensitivity analysis, and protein interaction analysis. We also conducted in vitro experiments utilizing colorectal cancer cell lines to validate PTPN6's functional role. PTPN6 exhibited distinct expression patterns across cancers, and its prognostic significance was apparent in several cancer types, particularly in glioblastoma, sarcoma, and melanoma. We observed correlations between PTPN6 and immune genes/cell infiltration in these cancers, suggesting a potential role in modulating the tumor immune microenvironment. Single-cell analysis revealed that PTPN6 is predominantly localized in macrophages, B cells, and dendritic cells within the tumor microenvironment, implying its involvement in regulating immune cell function. Enrichment analysis highlighted PTPN6's role in immune-related pathways. Drug sensitivity analysis identified specific drugs, including PAC-1, SNX-2112, BELINOSTAT, VORINOSTAT, TPCA-1, and PHA-893,888, whose efficacy may be influenced by PTPN6 expression. Knocking down PTPN6 expression inhibited the proliferation and migration of colorectal cancer cells in vitro, confirming its oncogenic role in this cancer type. This pan-cancer analysis establishes PTPN6's multifaceted influence on tumor immunity and its potential as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Yuchen Zhong
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China
| | - Weiyuan Zhang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Chaojing Zheng
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Hongyu Wu
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Jun Luo
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Ziming Yuan
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Hao Zhang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Chunlin Wang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Haiyang Feng
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Meng Wang
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Qian Zhang
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China.
| | - Haixing Ju
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China.
| | - Guiyu Wang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China.
| |
Collapse
|
6
|
Spirin P, Vedernikova V, Volkava T, Morozov A, Kleymenova A, Zemskaya A, Shyrokova L, Porozov Y, Glumakova K, Lebedev T, Kozlov M, Prassolov V. New and Effective Inhibitor of Class I HDACs, Eimbinostat, Reduces the Growth of Hematologic Cancer Cells and Triggers Apoptosis. Pharmaceutics 2025; 17:416. [PMID: 40284412 PMCID: PMC12030756 DOI: 10.3390/pharmaceutics17040416] [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: 02/21/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Histone deacetylases (HDACs) are critical epigenetic modulators involved in regulating various molecular mechanisms essential for cell development and growth. Alterations in HDAC activity have been linked to the progression of numerous cancers, including lymphoma. Over the past decade, the FDA has approved several HDAC inhibitors for lymphoma treatment, leading to heightened interest in this emerging class of drugs. Methods: In our research, we developed a novel HDAC inhibitor that exhibits high selectivity for class I HDACs. Results: Our in vitro findings indicate that treating lymphoma/leukemia cells with this inhibitor results in a marked suppression of cell growth and promotes apoptosis, while leaving the cell cycle unaffected. Conclusions: We propose that our new inhibitor, named eimbinostat, holds significant promise as a potential therapeutic agent for the treatment of hematologic malignancies such as lymphoma or leukemia.
Collapse
Affiliation(s)
- Pavel Spirin
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (V.V.); (K.G.); (T.L.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia
| | - Valeria Vedernikova
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (V.V.); (K.G.); (T.L.)
- Moscow Center for Advanced Studies, Kulakova 20, 123592 Moscow, Russia
| | - Tatsiana Volkava
- Faculty of Biology, Ludwig Maximilians University, Großhaderner Str. 2, 82152 Munich, Germany;
| | - Alexey Morozov
- Laboratory of Regulation of Intracellular Proteolysis, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia;
| | - Alla Kleymenova
- Laboratory of Molecular Basis of Action of Physiologically Active Compounds, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (A.K.); (A.Z.); (M.K.)
| | - Anastasia Zemskaya
- Laboratory of Molecular Basis of Action of Physiologically Active Compounds, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (A.K.); (A.Z.); (M.K.)
| | - Lena Shyrokova
- Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden;
| | - Yuri Porozov
- St. Petersburg School of Physics, Mathematics, and Computer Science, HSE University, 199106 Saint Petersburg, Russia;
- Advitam Laboratory, Mihaila Shushkaloviħa 13, 11030 Belgrade, Serbia
| | - Ksenia Glumakova
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (V.V.); (K.G.); (T.L.)
- Moscow Center for Advanced Studies, Kulakova 20, 123592 Moscow, Russia
| | - Timofey Lebedev
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (V.V.); (K.G.); (T.L.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia
| | - Maxim Kozlov
- Laboratory of Molecular Basis of Action of Physiologically Active Compounds, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (A.K.); (A.Z.); (M.K.)
| | - Vladimir Prassolov
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (V.V.); (K.G.); (T.L.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia
| |
Collapse
|
7
|
Zeltser N, Zhu C, Oh J, Li CH, Boutros PC. Sex Differences in Cancer Functional Genomics: Gene Dependency and Drug Sensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636540. [PMID: 39975298 PMCID: PMC11838570 DOI: 10.1101/2025.02.05.636540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Patient sex influences a wide range of cancer phenotypes, including prevalence, response to therapy and survival endpoints. Molecular sex differences have been identified at all levels of the central dogma. It is hypothesized that these molecular differences may drive the observed clinical sex differences. Yet despite a growing catalog of molecular sex differences in a range of cancer types, their specific functional consequences remain unclear. To directly assess how patient sex impacts cancer cell function, we evaluated 1,209 cell lines subjected to CRISPR knockout, RNAi knockdown or drug exposures. Despite limited statistical power, we identified pan- and per-cancer sex differences in gene essentiality in six sex-linked and fourteen autosomal genes, and in drug sensitivity for two compounds. These data fill a gap in our understanding of the link between sex-differential molecular effects and patient phenotypes. They call for much more careful and systematic consideration of sex-specific effects in mechanistic and functional studies.
Collapse
Affiliation(s)
- Nicole Zeltser
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Chenghao Zhu
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Jieun Oh
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
| | - Constance H. Li
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Paul C. Boutros
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Department of Urology, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Liu W, Li J, Tang Y, Zhao Y, Liu C, Song M, Ju Z, Kumar SV, Lu Y, Akbani R, Mills GB, Liang H. DrBioRight 2.0: an LLM-powered bioinformatics chatbot for large-scale cancer functional proteomics analysis. Nat Commun 2025; 16:2256. [PMID: 40050282 PMCID: PMC11885830 DOI: 10.1038/s41467-025-57430-4] [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: 03/11/2024] [Accepted: 02/24/2025] [Indexed: 03/09/2025] Open
Abstract
Functional proteomics provides critical insights into cancer mechanisms, facilitating the discovery of novel biomarkers and therapeutic targets. We have developed a comprehensive cancer functional proteomics resource using reverse phase protein arrays, incorporating data from nearly 8000 patient samples from The Cancer Genome Atlas and approximately 900 samples from the Cancer Cell Line Encyclopedia. Our dataset includes a curated panel of nearly 500 high-quality antibodies, covering all major cancer hallmark pathways. To enhance the accessibility and analytic power of this resource, we introduce DrBioRight 2.0 ( https://drbioright.org ), an intuitive bioinformatic platform powered by state-of-the-art large language models. DrBioRight enables researchers to explore protein-centric cancer omics data, perform advanced analyses, visualize results, and engage in interactive discussions using natural language. By streamlining complex proteogenomic analyses, this tool accelerates the translation of large-scale functional proteomics data into meaningful biomedical insights.
Collapse
Affiliation(s)
- Wei Liu
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jun Li
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yitao Tang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences Houston, Houston, TX, USA
| | - Yining Zhao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chaozhong Liu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shwetha V Kumar
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yiling Lu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rehan Akbani
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gordon B Mills
- Knight Cancer Institute and Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences Houston, Houston, TX, USA.
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
9
|
Juthi RT, Sazed SA, Mareboina M, Zaravinos A, Georgakopoulos-Soares I. Characterization of Exhausted T Cell Signatures in Pan-Cancer Settings. Int J Mol Sci 2025; 26:2311. [PMID: 40076932 PMCID: PMC11899893 DOI: 10.3390/ijms26052311] [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: 01/07/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
T cells play diverse roles in cancer immunology, acting as tumor suppressors, cytotoxic effectors, enhancers of cytotoxic T lymphocyte responses and immune suppressors; providing memory and surveillance; modulating the tumor microenvironment (TME); or activating innate immune cells. However, cancer cells can disrupt T cell function, leading to T cell exhaustion and a weakened immune response against the tumor. The expression of exhausted T cell (Tex) markers plays a pivotal role in shaping the immune landscape of multiple cancers. Our aim was to systematically investigate the role of known T cell exhaustion (Tex) markers across multiple cancers while exploring their molecular interactions, mutation profiles, and potential implications for immunotherapy. The mRNA expression profile of six Tex markers, LAG-3, PDCD1, TIGIT, HAVCR2, CXCL13, and LAYN was investigated in pan-cancer. Utilizing data from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), The Cancer Proteome Atlas (TCPA), and other repositories, we characterized the differential expression of the Tex markers, their association with the patients' survival outcome, and their mutation profile in multiple cancers. Additionally, we analyzed the effects on cancer-related pathways and immune infiltration within the TME, offering valuable insights into mechanisms of cancer immune evasion and progression. Finally, the correlation between their expression and sensitivity to multiple anti-cancer drugs was investigated extensively. Differential expression of all six markers was significantly associated with KIRC and poor prognosis in several cancers. They also played a potential activating role in apoptosis, EMT, and hormone ER pathways, as well as a potential inhibitory role in the DNA damage response and RTK oncogenic pathways. Infiltration of different immune cells was also found to be associated with the expression of the Tex-related genes in most cancer types. These findings underline that the reviving of exhausted T cells can be used to enhance the efficacy of immunotherapy in cancer patients.
Collapse
Affiliation(s)
- Rifat Tasnim Juthi
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh;
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (S.A.S.); (M.M.)
| | - Saiful Arefeen Sazed
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (S.A.S.); (M.M.)
| | - Manvita Mareboina
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (S.A.S.); (M.M.)
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, 22006, 1516 Nicosia, Cyprus
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), 22006, 1516 Nicosia, Cyprus
| | - Ilias Georgakopoulos-Soares
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (S.A.S.); (M.M.)
| |
Collapse
|
10
|
Yin J, Zhang H, Sun X, You N, Mou M, Lu M, Pan Z, Li F, Li H, Zeng S, Zhu F. Decoding Drug Response With Structurized Gridding Map-Based Cell Representation. IEEE J Biomed Health Inform 2025; 29:1702-1713. [PMID: 38090819 DOI: 10.1109/jbhi.2023.3342280] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025]
Abstract
A thorough understanding of cell-line drug response mechanisms is crucial for drug development, repurposing, and resistance reversal. While targeted anticancer therapies have shown promise, not all cancers have well-established biomarkers to stratify drug response. Single-gene associations only explain a small fraction of the observed drug sensitivity, so a more comprehensive method is needed. However, while deep learning models have shown promise in predicting drug response in cell lines, they still face significant challenges when it comes to their application in clinical applications. Therefore, this study proposed a new strategy called DD-Response for cell-line drug response prediction. First, a limitation of narrow modeling horizons was overcome to expand the model training domain by integrating multiple datasets through source-specific label binarization. Second, a modified representation based on a two-dimensional structurized gridding map (SGM) was developed for cell lines & drugs, avoiding feature correlation neglect and potential information loss. Third, a dual-branch, multi-channel convolutional neural network-based model for pairwise response prediction was constructed, enabling accurate outcomes and improved exploration of underlying mechanisms. As a result, the DD-Response demonstrated superior performance, captured cell-line characteristic variations, and provided insights into key factors impacting cell-line drug response. In addition, DD-Response exhibited scalability in predicting clinical patient responses to drug therapy. Overall, because of DD-response's excellent ability to predict drug response and capture key molecules behind them, DD-response is expected to greatly facilitate drug discovery, repurposing, resistance reversal, and therapeutic optimization.
Collapse
|
11
|
Sefer E. DRGAT: Predicting Drug Responses Via Diffusion-Based Graph Attention Network. J Comput Biol 2025; 32:330-350. [PMID: 39639802 DOI: 10.1089/cmb.2024.0807] [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] [Indexed: 12/07/2024] Open
Abstract
Accurately predicting drug response depending on a patient's genomic profile is critical for advancing personalized medicine. Deep learning approaches rise and especially the rise of graph neural networks leveraging large-scale omics datasets have been a key driver of research in this area. However, these biological datasets, which are typically high dimensional but have small sample sizes, present challenges such as overfitting and poor generalization in predictive models. As a complicating matter, gene expression (GE) data must capture complex inter-gene relationships, exacerbating these issues. In this article, we tackle these challenges by introducing a drug response prediction method, called drug response graph attention network (DRGAT), which combines a denoising diffusion implicit model for data augmentation with a recently introduced graph attention network (GAT) with high-order neighbor propagation (HO-GATs) prediction module. Our proposed approach achieved almost 5% improvement in the area under receiver operating characteristic curve compared with state-of-the-art models for the many studied drugs, indicating our method's reasonable generalization capabilities. Moreover, our experiments confirm the potential of diffusion-based generative models, a core component of our method, to mitigate the inherent limitations of omics datasets by effectively augmenting GE data.
Collapse
Affiliation(s)
- Emre Sefer
- Artificial Intelligence and Data Engineering Department, Ozyegin University, Istanbul, Turkey
| |
Collapse
|
12
|
Törzsök P, Santer FR, Kunz Y, van Creij NCH, Tymoszuk P, Klinglmair G, Culig Z, Pichler R. Biological and therapeutic implications of sex hormone-related gene clustering in testicular cancer. Basic Clin Androl 2025; 35:8. [PMID: 40011822 DOI: 10.1186/s12610-025-00254-5] [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: 08/06/2024] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Gonadotropin dysregulation seems to play a potential role in the carcinogenesis of testicular germ cell tumor (TGCT). The aim of this study was to explore the expression of specific genes related to sex hormone regulation, synthesis, and metabolism in TGCT and to define specific hormonal clusters. Two publicly available databases were used for this analysis (TCGA and GSE99420). By means of hard-threshold regularized KMEANS clustering, we assigned TGCT samples into four clusters defined in respect to different expression of the sex hormone-related genes. We analysed clinical data, protein and gene expression, signaling regarding hormonal clusters. Based on whole-transcriptome gene expression, prediction of anti-cancer drug response was made by RIDGE models. RESULTS Cluster #1 (12-16%) consisted primarily of non-seminomatous germ cell tumor (NSGCT), characterized by high expression of PRL, GNRH1, HSD17B2 and SRD5A1. Cluster #2 (42-50%) included predominantly seminomas with high expression of SRD5A3, being highly infiltrated by T and B cells. Cluster #3 (8.3-18%) comprised of NSGCT with high expression of CGA, CYP19A1, HSD17B12, HSD17B1, SHBG. Cluster #4 (23-30%), which consisted primarily of NSGCT with a small fraction of seminomas, was outlined by increased expression of STAR, POMC, CYP11A1, CYP17A1, HSD3B2 and HSD17B3. Elevated fibroblast levels and increased extracellular matrix- and growth factor signaling-related gene signature scores were described in cluster #1 and #3. In the combined model of progression-free survival, S2/S3 tumor marker status, hormonal cluster #1 or #3 and teratoma histology, were independently associated with 25-30% increase of progression risk. Based on the increased receptor tyrosine kinase and growth factor signaling, cluster #1, #3 and #4 were predicted to be sensitive to tyrosine kinase inhibitors, FGFR inhibitors or EGFR/ERBB inhibitors. Cluster #2 and #4 were responsive to compounds interfering with DNA synthesis, cytoskeleton, cell cycle and epigenetics. Response to apoptosis modulators was predicted only for cluster #2. CONCLUSIONS Hormonal cluster #1 or #3 is an independent prognostic factor regarding poor progression-free survival. Hormonal cluster assignment also affects the predicted drug response with cluster-dependent susceptibility to specific novel therapeutic compounds.
Collapse
Affiliation(s)
- Péter Törzsök
- Faculty of Health and Sport Sciences, Széchenyi István University, Győr, Hungary
| | - Frédéric R Santer
- Division of Experimental Urology, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Yannic Kunz
- Department of Urology, Medical University of Innsbruck, Comprehensive Cancer Center Innsbruck (CCCI), Anichstraße 35, Innsbruck, 6020, Austria
| | - Nils C H van Creij
- Division of Experimental Urology, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Gerald Klinglmair
- Department of Urology, Medical University of Innsbruck, Comprehensive Cancer Center Innsbruck (CCCI), Anichstraße 35, Innsbruck, 6020, Austria
| | - Zoran Culig
- Division of Experimental Urology, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Renate Pichler
- Department of Urology, Medical University of Innsbruck, Comprehensive Cancer Center Innsbruck (CCCI), Anichstraße 35, Innsbruck, 6020, Austria.
| |
Collapse
|
13
|
Lebedev T, Mikheeva A, Gasca V, Spirin P, Prassolov V. Systematic Comparison of FBS and Medium Variation Effect on Key Cellular Processes Using Morphological Profiling. Cells 2025; 14:336. [PMID: 40072065 PMCID: PMC11898771 DOI: 10.3390/cells14050336] [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: 01/31/2025] [Revised: 02/17/2025] [Accepted: 02/22/2025] [Indexed: 03/15/2025] Open
Abstract
Although every cell biologist knows the importance of selecting the right growth conditions and it is well known that the composition of growth medium may vary depending on a product brand or lot affecting many cellular processes, still those effects are poorly systematized. We addressed this issue by comparing the effect of 12 fetal bovine sera (FBS) and eight growth media from different brands on the morphological and functional parameters of five cell types: lung adenocarcinoma, neuroblastoma, glioblastoma, embryonic kidney, and colorectal cancer cells. Using high-throughput imaging, we compared cell proliferation; performed morphological profiling based on the imaging of 561,519 cells; measured extracellular regulated kinases (ERK1/2) activity, mitochondria potential, and lysosome accumulation; and compared cell sensitivity to drugs, response to EGF stimulation, and ability to differentiate. We found that changes in cell proliferation and morphology were independent, and morphological changes were associated with differences in mitochondria potential or the cell's ability to differentiate. Surprisingly, the most drastic differences were detected in serum-free conditions, where medium choice affected cell survival and response to EGF. Overall, our data may be used to improve the reproducibility of experiments involving cell cultures, and the effects of 28 growth conditions on proliferation and 44 morphological parameters can be explored through a Shinyapp.
Collapse
Affiliation(s)
- Timofey Lebedev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (A.M.); (V.G.); (P.S.); (V.P.)
| | | | | | | | | |
Collapse
|
14
|
Gollowitzer A, Pein H, Rao Z, Waltl L, Bereuter L, Loeser K, Meyer T, Jafari V, Witt F, Winkler R, Su F, Große S, Thürmer M, Grander J, Hotze M, Harder S, Espada L, Magnutzki A, Gstir R, Weinigel C, Rummler S, Bonn G, Pachmayr J, Ermolaeva M, Harayama T, Schlüter H, Kosan C, Heller R, Thedieck K, Schmitt M, Shimizu T, Popp J, Shindou H, Kwiatkowski M, Koeberle A. Attenuated growth factor signaling during cell death initiation sensitizes membranes towards peroxidation. Nat Commun 2025; 16:1774. [PMID: 40000627 PMCID: PMC11861335 DOI: 10.1038/s41467-025-56711-2] [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: 01/16/2019] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Cell death programs such as apoptosis and ferroptosis are associated with aberrant redox homeostasis linked to lipid metabolism and membrane function. Evidence for cross-talk between these programs is emerging. Here, we show that cytotoxic stress channels polyunsaturated fatty acids via lysophospholipid acyltransferase 12 into phospholipids that become susceptible to peroxidation under additional redox stress. This reprogramming is associated with altered acyl-CoA synthetase isoenzyme expression and caused by a decrease in growth factor receptor tyrosine kinase (RTK)-phosphatidylinositol-3-kinase signaling, resulting in suppressed fatty acid biosynthesis, for specific stressors via impaired Akt-SREBP1 activation. The reduced availability of de novo synthesized fatty acids favors the channeling of polyunsaturated fatty acids into phospholipids. Growth factor withdrawal by serum starvation mimics this phenotype, whereas RTK ligands counteract it. We conclude that attenuated RTK signaling during cell death initiation increases cells' susceptibility to oxidative membrane damage at the interface of apoptosis and alternative cell death programs.
Collapse
Affiliation(s)
- André Gollowitzer
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Helmut Pein
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Zhigang Rao
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Lorenz Waltl
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Leonhard Bereuter
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria
| | - Konstantin Loeser
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Tobias Meyer
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology Jena e.V., Member of Leibniz Health Technology, 07745, Jena, Germany
| | - Vajiheh Jafari
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Finja Witt
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - René Winkler
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich-Schiller-University Jena, 07745, Jena, Germany
- Josep Carreras Leukaemia Research Institute (IJC), Campus Can Ruti, 08916, Badalona, Spain
| | - Fengting Su
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria
| | - Silke Große
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745, Jena, Germany
| | - Maria Thürmer
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Julia Grander
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Madlen Hotze
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
| | - Sönke Harder
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Lilia Espada
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745, Jena, Germany
| | - Alexander Magnutzki
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Ronald Gstir
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Christina Weinigel
- Institute of Transfusion Medicine, University Hospital Jena, 07747, Jena, Germany
| | - Silke Rummler
- Institute of Transfusion Medicine, University Hospital Jena, 07747, Jena, Germany
| | - Günther Bonn
- ADSI-Austrian Drug Screening Institute, University of Innsbruck, 6020, Innsbruck, Austria
| | - Johanna Pachmayr
- Institute of Pharmacy, Paracelsus Medical University, 5020, Salzburg, Austria
| | - Maria Ermolaeva
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745, Jena, Germany
| | - Takeshi Harayama
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur - CNRS UMR7275 - Inserm U1323, 06560, Valbonne, France
| | - Hartmut Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Christian Kosan
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Friedrich-Schiller-University Jena, 07745, Jena, Germany
| | - Regine Heller
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745, Jena, Germany
| | - Kathrin Thedieck
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
- Department Metabolism, Senescence and Autophagy, Research Center One Health Ruhr, University Alliance Ruhr & University Hospital Essen, University Duisburg-Essen, 45141, Essen, Germany
- Freiburg Materials Research Center FMF, Albert-Ludwigs-University of Freiburg, 79104, Freiburg, Germany
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signaling, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, The Netherlands
- German Cancer Consortium (DKTK), partner site Essen/Duesseldorf, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, 45147, Essen, Germany
| | - Michael Schmitt
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Takao Shimizu
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
- Institute of Microbial Chemistry, Tokyo 141-0021, Japan
| | - Jürgen Popp
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich-Schiller-University Jena, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology Jena e.V., Member of Leibniz Health Technology, 07745, Jena, Germany
| | - Hideo Shindou
- Department of Lipid Life Science, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Medical Lipid Science, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Marcel Kwiatkowski
- Institute of Biochemistry and Center for Molecular Biosciences Innsbruck, University of Innsbruck, 6020, Innsbruck, Austria
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria.
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany.
- Institute of Pharmaceutical Sciences and Excellence Field BioHealth, University of Graz, Graz, Austria.
| |
Collapse
|
15
|
Wang Y, Haase S, Whitman A, Beltran A, Spanheimer PM, Brunk E. A Multimodal Framework to Uncover Drug-Responsive Subpopulations in Triple-Negative Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638274. [PMID: 40027670 PMCID: PMC11870422 DOI: 10.1101/2025.02.14.638274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Understanding how individual cancer cells adapt to drug treatment is a fundamental challenge limiting precision medicine cancer therapy strategies. While single-cell technologies have advanced our understanding of cellular heterogeneity, efforts to connect the behavior of individual cells to broader tumor drug responses and uncover global trends across diverse systems remain limited. There is a growing availability of single-cell and bulk omics data, but a lack of centralized tools and repositories makes it difficult to study drug response globally, especially at the level of single-cell adaptation. To address this, we present a multimodal framework that integrates bulk and single-cell treated and untreated transcriptomics data to identify drug responsive cell populations in triple-negative breast cancer (TNBC). Our framework leverages population-scale bulk transcriptomics data from TNBC samples to define seven main "identities", each representing unique combinations of biologically relevant genes. These identities are dynamic and trackable, allowing us to map them onto single cells and uncover global patterns of how cell populations respond to drug treatment. Unlike static classifications, this approach captures the evolving nature of cellular states, revealing that a select few identities dominate and drive population-level responses during treatment. Crucially, our ability to decode these trends through the inherent noise of single-cell data provides a clearer picture of how heterogeneous cell populations adapt to therapy. By identifying the dominant identities and their dynamics, we can better predict how entire tumors respond to treatment. This insight is essential for designing precise combination therapies tailored to the unique heterogeneity of patient tumors, addressing the single-cell variations that ultimately determine therapeutic outcomes.
Collapse
|
16
|
Sgobba MN, Musio B, Pastrana CI, Todisco S, Schlosserovà N, Mastropirro F, Favia M, Radesco A, Duarte IF, De Grassi A, Volpicella M, Gallo V, Pierri CL, Ciani E, Guerra L. Serum Starvation Enhances the Antitumor Activity of Natural Matrices: Insights into Bioactive Molecules from Dromedary Urine Extracts. Molecules 2025; 30:821. [PMID: 40005133 PMCID: PMC11858132 DOI: 10.3390/molecules30040821] [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: 12/30/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Natural matrices have historically been a cornerstone in drug discovery, offering a rich source of structurally diverse and biologically active compounds. However, research on natural products often faces significant challenges due to the complexity of natural matrices, such as urine, and the limitations of bioactivity assessment assays. To ensure reliable insights, it is crucial to optimize experimental conditions to reveal the bioactive potential of samples, thereby improving the validity of statistical analyses. Approaches in metabolomics further strengthen this process by identifying and focusing on the most promising compounds within natural matrices, enhancing the precision of bioactive metabolite prioritization. In this study, we assessed the bioactivity of 17 dromedary urine samples on human renal cells under serum-reduced conditions (1%FBS) in order to minimize possible FBS-derived interfering factors. Using viability assays and Annexin V/PI staining, we found that the tumor renal cell lines Caki-1 and RCC-Shaw were more sensitive to the cytotoxic effects of the small molecules present in dromedary urine compared to non-tumor HK-2 cells. Employing NMR metabolomics analysis combined with detected in vitro activity, our statistical model highlights the presence of bioactive compounds in dromedary urine, such as azelaic acid and phenylacetyl glycine, underscoring its potential as a sustainable source of bioactive molecules within the framework of green chemistry and circular economy initiatives.
Collapse
Affiliation(s)
- Maria Noemi Sgobba
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy (F.M.); (A.D.G.); (M.V.); (E.C.); (L.G.)
| | - Biagia Musio
- Department of Civil, Environmental, Land, Building Engineering and Chemistry (DICATECh), Polytechnic University of Bari, Via Orabona 4, 70125 Bari, Italy; (B.M.); (S.T.); (V.G.)
| | - Carlos Iglesias Pastrana
- Faculty of Veterinary Sciences, Department of Genetics, University of Córdoba, 14071 Córdoba, Spain;
| | - Stefano Todisco
- Department of Civil, Environmental, Land, Building Engineering and Chemistry (DICATECh), Polytechnic University of Bari, Via Orabona 4, 70125 Bari, Italy; (B.M.); (S.T.); (V.G.)
| | - Nikola Schlosserovà
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy (F.M.); (A.D.G.); (M.V.); (E.C.); (L.G.)
| | - Federica Mastropirro
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy (F.M.); (A.D.G.); (M.V.); (E.C.); (L.G.)
| | - Maria Favia
- Department of Translational Biomedicine and Neurosciences (DiBraiN), University of Bari “Aldo Moro”, Piazza Giulio Cesare, 70124 Bari, Italy;
| | - Antonio Radesco
- Istituto Tumori “Giovanni Paolo II” I.R.C.C.S., Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Iola F. Duarte
- Department of Chemistry, CICECO—Aveiro Institute of Materials and LAQV-REQUIMTE, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Anna De Grassi
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy (F.M.); (A.D.G.); (M.V.); (E.C.); (L.G.)
| | - Mariateresa Volpicella
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy (F.M.); (A.D.G.); (M.V.); (E.C.); (L.G.)
| | - Vito Gallo
- Department of Civil, Environmental, Land, Building Engineering and Chemistry (DICATECh), Polytechnic University of Bari, Via Orabona 4, 70125 Bari, Italy; (B.M.); (S.T.); (V.G.)
- Innovative Solutions S.r.l.—Spin-Off Company of the Polytechnic University of Bari, Zona H 150/B, 70015 Noci, Italy
| | - Ciro Leonardo Pierri
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy
| | - Elena Ciani
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy (F.M.); (A.D.G.); (M.V.); (E.C.); (L.G.)
| | - Lorenzo Guerra
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, Via Orabona 4, 70125 Bari, Italy (F.M.); (A.D.G.); (M.V.); (E.C.); (L.G.)
| |
Collapse
|
17
|
Singhal A, Zhao X, Wall P, So E, Calderini G, Partin A, Koussa N, Vasanthakumari P, Narykov O, Zhu Y, Jones SE, Abbas-Aghababazadeh F, Nair SK, Bélisle-Pipon JC, Jayaram A, Parker BA, Yeung KT, Griffiths JI, Weil R, Nath A, Haibe-Kains B, Ideker T. The Hallmarks of Predictive Oncology. Cancer Discov 2025; 15:271-285. [PMID: 39760657 PMCID: PMC11969157 DOI: 10.1158/2159-8290.cd-24-0760] [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: 05/25/2024] [Revised: 08/30/2024] [Accepted: 10/16/2024] [Indexed: 01/07/2025]
Abstract
SIGNIFICANCE As the field of artificial intelligence evolves rapidly, these hallmarks are intended to capture fundamental, complementary concepts necessary for the progress and timely adoption of predictive modeling in precision oncology. Through these hallmarks, we hope to establish standards and guidelines that enable the symbiotic development of artificial intelligence and precision oncology.
Collapse
Affiliation(s)
- Akshat Singhal
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Xiaoyu Zhao
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Patrick Wall
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Emily So
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Guido Calderini
- Faculty of Health Science, Simon Fraser University, Burnaby, BC, Canada
- École de santé publique, Université de Montréal, Montréal, QC, Canada
| | - Alexander Partin
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
| | - Natasha Koussa
- Cancer Data Science Initiatives, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Oleksandr Narykov
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
| | - Yitan Zhu
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
| | - Sara E. Jones
- Cancer Data Science Initiatives, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | | | | | | - Barbara A. Parker
- Moores Cancer Center, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Kay T. Yeung
- Moores Cancer Center, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jason I. Griffiths
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute, City of Hope National Medical Center, Monrovia, CA, USA
| | - Ryan Weil
- Cancer Data Science Initiatives, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Aritro Nath
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute, City of Hope National Medical Center, Monrovia, CA, USA
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Medical Biophysics, University of Toronto, Toronto, Canada
- Vector Institute for Artificial Intelligence, Toronto, Canada
- Department of Biostatistics, Dalla Lana School of Public Health, Toronto, Canada
| | - Trey Ideker
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
18
|
Cai Y, Xiao H, Zhou Q, Lin J, Liang X, Xu W, Cao Y, Zhang X, Wang H. Comprehensive Analyses of PANoptosome with Potential Implications in Cancer Prognosis and Immunotherapy. Biochem Genet 2025; 63:331-353. [PMID: 38436818 PMCID: PMC11832696 DOI: 10.1007/s10528-024-10687-8] [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: 07/22/2023] [Accepted: 01/04/2024] [Indexed: 03/05/2024]
Abstract
Cell death resistance significantly contributes to poor therapeutic outcomes in various cancers. PANoptosis, a unique inflammatory programmed cell death (PCD) pathway activated by specific triggers and regulated by the PANoptosome, possesses key features of apoptosis, pyroptosis, and necroptosis, but these cannot be accounted for by any of the three PCD pathways alone. While existing studies on PANoptosis have predominantly centered on infectious and inflammatory diseases, its role in cancer malignancy has been understudied. In this comprehensive investigation, we conducted pan-cancer analyses of PANoptosome component genes across 33 cancer types. We characterized the genetic, epigenetic, and transcriptomic landscapes, and introduced a PANoptosome-related potential index (PANo-RPI) for evaluating the intrinsic PANoptosome assembly potential in cancers. Our findings unveil PANo-RPI as a prognostic factor in numerous cancers, including KIRC, LGG, and PAAD. Crucially, we established a significant correlation between PANo-RPI and tumor immune responses, as well as the infiltration of diverse lymphoid and myeloid cell subsets across nearly all cancer types. Moreover, a high PANo-RPI was consistently associated with improved immunotherapy response and efficacy, as evidenced by re-analysis of multiple immunotherapy cohorts. In conclusion, our study suggests that targeting PANoptosome components and modulating PANoptosis may hold tremendous therapeutic potential in the context of cancer.
Collapse
Affiliation(s)
- Yonghua Cai
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Heng Xiao
- Southern Medical School, No. 1023, South Shatai Road, Baiyun District, Guangzhou, 510515, Guangdong, China
| | - Qixiong Zhou
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Jie Lin
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Xianqiu Liang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Wei Xu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Yongfu Cao
- Department of Neurosurgery, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Xian Zhang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China.
| | - Hai Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China.
| |
Collapse
|
19
|
Wang Y, Hu M, Cao J, Wang F, Han JR, Wu TW, Li L, Yu J, Fan Y, Xie G, Lian H, Cao Y, Naowarojna N, Wang X, Zou Y. ACSL4 and polyunsaturated lipids support metastatic extravasation and colonization. Cell 2025; 188:412-429.e27. [PMID: 39591965 DOI: 10.1016/j.cell.2024.10.047] [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: 09/26/2023] [Revised: 04/13/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024]
Abstract
Metastatic dissemination to distant organs demands that cancer cells possess high morphological and metabolic adaptability. However, contributions of the cellular lipidome to metastasis remain elusive. Here, we uncover a correlation between metastasis potential and ferroptosis susceptibility in multiple cancers. Metastases-derived cancer cells exhibited higher ferroptosis sensitivity and polyunsaturated fatty acyl (PUFA)-lipid contents than primary-tumor-derived cells from ovarian cancer patients. Metabolism-focused CRISPR screens in a mouse model for ovarian cancer distant metastasis established via two rounds of in vivo selection revealed the PUFA-lipid biosynthesis enzyme acyl-coenzyme A (CoA) synthetase long-chain family member 4 (ACSL4) as a pro-hematogenous metastasis factor. ACSL4 promotes metastatic extravasation by enhancing membrane fluidity and cellular invasiveness. While promoting metastasis, the high PUFA-lipid state creates dependencies on abhydrolase-domain-containing 6, acylglycerol lipase (ABHD6), enoyl-CoA delta isomerase 1 (ECI1), and enoyl-CoA hydratase 1 (ECH1)-rate-limiting enzymes preparing unsaturated fatty acids (UFAs) for β-oxidation. ACSL4/ECH1 co-inhibition achieved potent suppression of metastasis. Our work establishes the dual functions of PUFA-lipids in tumor progression and metastasis that may be exploitable for therapeutic development.
Collapse
Affiliation(s)
- Yuqi Wang
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Mangze Hu
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Jian Cao
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China
| | - Fengxiang Wang
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingrong Regina Han
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; School of Life Sciences, Fudan University, Shanghai, China
| | - Tianshu William Wu
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China; College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Luxiao Li
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Jinshi Yu
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Yujing Fan
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Guanglei Xie
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Bioinformatics and Genomics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Heyuan Lian
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Yueying Cao
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, Jiangsu, China
| | - Nathchar Naowarojna
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Xi Wang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Westlake Bioinformatics and Genomics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| | - Yilong Zou
- Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
20
|
Wu Y, Xie L. AI-driven multi-omics integration for multi-scale predictive modeling of genotype-environment-phenotype relationships. Comput Struct Biotechnol J 2025; 27:265-277. [PMID: 39886532 PMCID: PMC11779603 DOI: 10.1016/j.csbj.2024.12.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/22/2024] [Accepted: 12/26/2024] [Indexed: 02/01/2025] Open
Abstract
Despite the wealth of single-cell multi-omics data, it remains challenging to predict the consequences of novel genetic and chemical perturbations in the human body. It requires knowledge of molecular interactions at all biological levels, encompassing disease models and humans. Current machine learning methods primarily establish statistical correlations between genotypes and phenotypes but struggle to identify physiologically significant causal factors, limiting their predictive power. Key challenges in predictive modeling include scarcity of labeled data, generalization across different domains, and disentangling causation from correlation. In light of recent advances in multi-omics data integration, we propose a new artificial intelligence (AI)-powered biology-inspired multi-scale modeling framework to tackle these issues. This framework will integrate multi-omics data across biological levels, organism hierarchies, and species to predict genotype-environment-phenotype relationships under various conditions. AI models inspired by biology may identify novel molecular targets, biomarkers, pharmaceutical agents, and personalized medicines for presently unmet medical needs.
Collapse
Affiliation(s)
- You Wu
- Ph.D. Program in Computer Science, The Graduate Center, The City University of New York, New York, NY, USA
| | - Lei Xie
- Ph.D. Program in Computer Science, The Graduate Center, The City University of New York, New York, NY, USA
- Ph.D. Program in Biology and Biochemistry, The Graduate Center, The City University of New York, New York, NY, USA
- Department of Computer Science, Hunter College, The City University of New York, New York, NY, USA
- Helen & Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, Cornell University, New York, NY, USA
| |
Collapse
|
21
|
Ngoi NYL, Gallo D, Torrado C, Nardo M, Durocher D, Yap TA. Synthetic lethal strategies for the development of cancer therapeutics. Nat Rev Clin Oncol 2025; 22:46-64. [PMID: 39627502 DOI: 10.1038/s41571-024-00966-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2024] [Indexed: 12/20/2024]
Abstract
Synthetic lethality is a genetic phenomenon whereby the simultaneous presence of two different genetic alterations impairs cellular viability. Importantly, targeting synthetic lethal interactions offers potential therapeutic strategies for cancers with alterations in pathways that might otherwise be considered undruggable. High-throughput screening methods based on modern CRISPR-Cas9 technologies have emerged and become crucial for identifying novel synthetic lethal interactions with the potential for translation into biologically rational cancer therapeutic strategies as well as associated predictive biomarkers of response capable of guiding patient selection. Spurred by the clinical success of PARP inhibitors in patients with BRCA-mutant cancers, novel agents targeting multiple synthetic lethal interactions within DNA damage response pathways are in clinical development, and rational strategies targeting synthetic lethal interactions spanning alterations in epigenetic, metabolic and proliferative pathways have also emerged and are in late preclinical and/or early clinical testing. In this Review, we provide a comprehensive overview of established and emerging technologies for synthetic lethal drug discovery and development and discuss promising therapeutic strategies targeting such interactions.
Collapse
Affiliation(s)
- Natalie Y L Ngoi
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Gallo
- Repare Therapeutics, Inc., Montreal, Quebec, Canada
| | - Carlos Torrado
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mirella Nardo
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel Durocher
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
22
|
Gunji D, Abe Y, Muraoka S, Narumi R, Isoyama J, Ikemoto N, Ishida M, Shinkura A, Tomonaga T, Nagayama S, Takahashi Y, Fukunaga Y, Sakai Y, Obama K, Adachi J. Longitudinal phosphoproteomics reveals the PI3K-PAK1 axis as a potential target for recurrent colorectal liver metastases. Cell Rep 2024; 43:115061. [PMID: 39689713 DOI: 10.1016/j.celrep.2024.115061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/07/2024] [Accepted: 11/21/2024] [Indexed: 12/19/2024] Open
Abstract
The resistance of colorectal cancer liver metastases (CRLMs) to 5-fluorouracil (5-FU) chemotherapy remains a significant global health challenge. We investigated the phosphoproteomic dynamics of serial tissue sections obtained from initial metastases and recurrent tumors collected from 24 patients to address this unmet need for innovative therapeutic strategies for patients with CRLM with a poor prognosis. Our analysis revealed the activation of PAK kinase in patients with CRLM with a poor prognosis. Using an unbiased computational approach, we conducted a correlation analysis between PAK1 kinase activity and 545 drug sensitivity profiles across 35 colorectal cancer cell lines and identified PI3K inhibitors as potential therapeutic candidates. The efficacy of the FDA-approved PI3K inhibitor copanlisib was validated in 5-FU-resistant cell lines with high PAK1 kinase activity both in vitro and in vivo. This study presents an effective strategy for drug target discovery based on kinase activity, and the concept of this approach is widely applicable.
Collapse
Affiliation(s)
- Daigo Gunji
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Yuichi Abe
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; Immunoproteomics Laboratory, Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
| | - Satoshi Muraoka
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Ryohei Narumi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Junko Isoyama
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Narumi Ikemoto
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Mimiko Ishida
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Akina Shinkura
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Takeshi Tomonaga
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan
| | - Satoshi Nagayama
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; Department of Surgery, Uji-Tokusyukai Medical Center, Kyoto 611-0041, Japan
| | - Yu Takahashi
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Yosuke Fukunaga
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Yoshiharu Sakai
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Kazutaka Obama
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; Laboratory of Proteomics and Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
23
|
Xu P, Zhang Q, Zhai J, Chen P, Deng X, Miao L, Zhang X. Systematic pan-cancer analysis identifies ZBTB11 as a potential pan-cancer biomarker and immunotherapy target in multiple tumor types. Discov Oncol 2024; 15:830. [PMID: 39715911 DOI: 10.1007/s12672-024-01697-4] [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: 06/13/2024] [Accepted: 12/11/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND ZBTB11 is a putative transcription factor with an N-terminal BTB domain and tandem C-terminal zinc finger motifs. Recent studies have suggested a potential role for ZBTB11 in tumorigenesis. However, the biological significance of ZBTB11 in different cancer types remains uncertain. METHODS The expression levels, prognostic values, genetic mutations, and DNA promoter methylation of ZBTB11 across tumor types were explored via various online websites and databases, including TIMER2.0, GEPIA2, cBioPortal, UALCAN, GSCA, CancerSEA, and others. Additionally, a competing lncRNA-miRNA network of ZBTB11 was constructed, and its interaction with chemicals and genes was investigated. RESULTS Our findings revealed that ZBTB11 was aberrantly expressed in a multitude of tumor types and exhibited variability across various tumor stages. A survival analysis revealed that ZBTB11 predicted a poor prognosis in BRCA, KIRP, LIHC, PCPG, PRAD, SARC, UCEC, and a good prognosis in CHOL, ESCA, GBM, KIRC, and READ. We also found that the most frequent genetic alterations type of ZBTB11 was mutation, and the DNA methylation level of ZBTB11 decreased in various cancers. Furthermore, ZBTB11 expression correlated with immune cells infiltration and genetic markers of immunodulators in cancers. Moreover, the results of single-cell sequencing demonstrated that ZBTB11 could regulate several tumor biological behaviors, including apoptosis, DNA damage, and angiogenesis. A lncRNA-miRNA network regulating ZBTB11 expression in tumor development and progression was constructed. It is of particular significance that ZBTB11 demonstrated a correlation with the CTRP and GDSC drug sensitivity, and that it served as a mediator between chemicals and cancers. CONCLUSION These findings demonstrate that ZBTB11 is associated with multiple tumor types and disease prognosis. ZBTB11 may represent a potential key biomarker and therapeutic target in cancers.
Collapse
Affiliation(s)
- Peiyi Xu
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Gulou District, Nanjing, Jiangsu, China
| | - Qiuyan Zhang
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Gulou District, Nanjing, Jiangsu, China
| | - Jing Zhai
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Gulou District, Nanjing, Jiangsu, China
| | - Pu Chen
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Gulou District, Nanjing, Jiangsu, China
| | - Xueting Deng
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Gulou District, Nanjing, Jiangsu, China
| | - Lin Miao
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Gulou District, Nanjing, Jiangsu, China
| | - Xiuhua Zhang
- Department of Gastroenterology, Second Affiliated Hospital, Nanjing Medical University, 121 Jiangjiayuan Road, Gulou District, Nanjing, Jiangsu, China.
| |
Collapse
|
24
|
Gu C, Chen X, Wu J, Zhang Y, Zhong L, Luo H, Luo W, Yang F. SOCS1: A potential diagnostic and prognostic marker for aggressive gliomas and a new target for immunotherapy. Medicine (Baltimore) 2024; 103:e40632. [PMID: 39654174 PMCID: PMC11630960 DOI: 10.1097/md.0000000000040632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Gliomas, the most common and deadly cancers of the central nervous system, present a unique immunological barrier that severely undermines the effectiveness of immunotherapies. Suppressor of cytokine signaling 1 (SOCS1), belonging to the SOCS protein family and playing a pivotal role in various cancer treatment strategies and is abundant in high-grade gliomas. This study conducted a comparative analysis of SOCS1 and glioma immune checkpoints. It underscores the feasibility of leveraging SOCS1 as a promising diagnostic and prognostic marker for aggressive gliomas, thus offering novel targets for glioma immunotherapy. Comprehensive gene expression analyses and clinical data validations were performed across multiple databases. The expression and biological functions of SOCS1 were examined through an array of techniques including pan-cancer analysis, functional enrichment, gene set variation analysis, and immune microenvironment examination. This was done alongside a comparison of the similarities between SOCS1 and various glioma immune checkpoints. Utilizing clinical information from patients, a bespoke predictive model was developed to further corroborate the prognostic capabilities of SOCS1. The investigation revealed considerable similarities between SOCS1 and several immune checkpoints such as CTLA4, demonstrating SOCS1's role as an independent prognostic factor positively influencing glioma patient outcomes. The inclusion of SOCS1 in the developed predictive model significantly enhanced its precision. Our findings highlight SOCS1's potential as an innovative target for glioma immunotherapy, providing a novel strategy to overcome the immunological barriers posed by gliomas. Furthermore, identifying SOCS1 as a viable diagnostic marker for aggressive gliomas improves the accuracy of prognostic predictions for affected patients.
Collapse
Affiliation(s)
- Chuanshen Gu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Xinyi Chen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiayan Wu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Yiwen Zhang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Linyu Zhong
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Han Luo
- College of Acupuncture and Tuina, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Wenshu Luo
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Fuxia Yang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
25
|
Jiménez-Santos M, García-Martín S, Rubio-Fernández M, Gómez-López G, Al-Shahrour F. Spatial transcriptomics in breast cancer reveals tumour microenvironment-driven drug responses and clonal therapeutic heterogeneity. NAR Cancer 2024; 6:zcae046. [PMID: 39703753 PMCID: PMC11655296 DOI: 10.1093/narcan/zcae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024] Open
Abstract
Breast cancer patients are categorized into three subtypes with distinct treatment approaches. Precision oncology has increased patient outcomes by targeting the specific molecular alterations of tumours, yet challenges remain. Treatment failure persists due to the coexistence of several malignant subpopulations with different drug sensitivities within the same tumour, a phenomenon known as intratumour heterogeneity (ITH). This heterogeneity has been extensively studied from a tumour-centric view, but recent insights underscore the role of the tumour microenvironment in treatment response. Our research utilizes spatial transcriptomics data from breast cancer patients to predict drug sensitivity. We observe diverse response patterns across tumour, interphase and microenvironment regions, unveiling a sensitivity and functional gradient from the tumour core to the periphery. Moreover, we find tumour therapeutic clusters with different drug responses associated with distinct biological functions driven by unique ligand-receptor interactions. Importantly, we identify genetically identical subclones with different responses depending on their location within the tumour ducts. This research underscores the significance of considering the distance from the tumour core and microenvironment composition when identifying suitable treatments to target ITH. Our findings provide critical insights into optimizing therapeutic strategies, highlighting the necessity of a comprehensive understanding of tumour biology for effective cancer treatment.
Collapse
Affiliation(s)
- María José Jiménez-Santos
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Santiago García-Martín
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Marcos Rubio-Fernández
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
- Lung-H120 Group, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Gonzalo Gómez-López
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Fátima Al-Shahrour
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Calle Melchor Fernández Almagro, 3, Madrid 28029, Spain
| |
Collapse
|
26
|
Cai Z, Apolinário S, Baião AR, Pacini C, Sousa MD, Vinga S, Reddel RR, Robinson PJ, Garnett MJ, Zhong Q, Gonçalves E. Synthetic augmentation of cancer cell line multi-omic datasets using unsupervised deep learning. Nat Commun 2024; 15:10390. [PMID: 39614072 PMCID: PMC11607321 DOI: 10.1038/s41467-024-54771-4] [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: 12/21/2023] [Accepted: 11/18/2024] [Indexed: 12/01/2024] Open
Abstract
Integrating diverse types of biological data is essential for a holistic understanding of cancer biology, yet it remains challenging due to data heterogeneity, complexity, and sparsity. Addressing this, our study introduces an unsupervised deep learning model, MOSA (Multi-Omic Synthetic Augmentation), specifically designed to integrate and augment the Cancer Dependency Map (DepMap). Harnessing orthogonal multi-omic information, this model successfully generates molecular and phenotypic profiles, resulting in an increase of 32.7% in the number of multi-omic profiles and thereby generating a complete DepMap for 1523 cancer cell lines. The synthetically enhanced data increases statistical power, uncovering less studied mechanisms associated with drug resistance, and refines the identification of genetic associations and clustering of cancer cell lines. By applying SHapley Additive exPlanations (SHAP) for model interpretation, MOSA reveals multi-omic features essential for cell clustering and biomarker identification related to drug and gene dependencies. This understanding is crucial for developing much-needed effective strategies to prioritize cancer targets.
Collapse
Affiliation(s)
- Zhaoxiang Cai
- ProCan®, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Sofia Apolinário
- INESC-ID, 1000-029, Lisboa, Portugal
- Instituto Superior Técnico (IST), Universidade de Lisboa, 1049-001, Lisboa, Portugal
| | - Ana R Baião
- INESC-ID, 1000-029, Lisboa, Portugal
- Instituto Superior Técnico (IST), Universidade de Lisboa, 1049-001, Lisboa, Portugal
| | - Clare Pacini
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Miguel D Sousa
- INESC-ID, 1000-029, Lisboa, Portugal
- Instituto Superior Técnico (IST), Universidade de Lisboa, 1049-001, Lisboa, Portugal
| | - Susana Vinga
- INESC-ID, 1000-029, Lisboa, Portugal
- Instituto Superior Técnico (IST), Universidade de Lisboa, 1049-001, Lisboa, Portugal
| | - Roger R Reddel
- ProCan®, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Phillip J Robinson
- ProCan®, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Mathew J Garnett
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1SA, UK
| | - Qing Zhong
- ProCan®, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia.
| | - Emanuel Gonçalves
- INESC-ID, 1000-029, Lisboa, Portugal.
- Instituto Superior Técnico (IST), Universidade de Lisboa, 1049-001, Lisboa, Portugal.
| |
Collapse
|
27
|
Paulson AL, Gruener RF, Lee AM, Huang RS. Discovery, Validation and Mechanistic Study of XPO1 Inhibition in the Treatment of Triple-Negative Breast Cancer. Cancers (Basel) 2024; 16:3980. [PMID: 39682167 DOI: 10.3390/cancers16233980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer with limited treatment options. The nuclear export protein XPO1 has emerged as a potential therapeutic target in cancer, but its role in TNBC has not been fully characterized. This study investigates the potential of repurposing selinexor, an FDA-approved XPO1 inhibitor, as a novel therapeutic options for TNBC. Methods: A computational drug repurposing pipeline was used to predict patient tumor responses to hundreds of drugs. We identified XPO1 inhibitors as a candidate drug and validated its efficacy on an independent patient dataset and across various TNBC cell lines. RNA-sequencing after longitudinal XPO1 inhibition and further mechanistic studies were performed to explore and confirm the leading causes of TNBC cell sensitivity to XPO1 inhibition. Results: Selinexor significantly reduce the viability of a variety of TNBC cell lines. Mechanistically, selinexor induces TNBC cell death by inhibiting the NF-kB pathway through nuclear retention of NFKBIA. This effect was consistent across multiple TNBC cell lines. Conclusions: XPO1 inhibitors show promise as targeted therapies for TNBC patients. New mechanistic insight into the causes leading to TNBC sensitivity to XPO1-inhibition-mediated cell death warrant further clinical trials to evaluate the safety and efficacy in TNBC.
Collapse
Affiliation(s)
- Amy L Paulson
- Department of Molecular Pharmacology and Therapeutics, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA
| | - Robert F Gruener
- Department of Experimental and Clinical Pharmacology, University of Minnesota School of Pharmacy, Minneapolis, MN 55455, USA
| | - Adam M Lee
- Department of Experimental and Clinical Pharmacology, University of Minnesota School of Pharmacy, Minneapolis, MN 55455, USA
| | - R Stephanie Huang
- Department of Molecular Pharmacology and Therapeutics, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA
- Department of Experimental and Clinical Pharmacology, University of Minnesota School of Pharmacy, Minneapolis, MN 55455, USA
| |
Collapse
|
28
|
Gazola AA, Lautert-Dutra W, Archangelo LF, Reis RBD, Squire JA. Precision oncology platforms: practical strategies for genomic database utilization in cancer treatment. Mol Cytogenet 2024; 17:28. [PMID: 39543667 PMCID: PMC11566986 DOI: 10.1186/s13039-024-00698-w] [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: 07/29/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
In recent years, the expansion of molecularly targeted cancer therapies has significantly advanced precision oncology. Parallel developments in next-generation sequencing (NGS) technologies have also improved precision oncology applications, making genomic analysis of tumors more affordable and accessible. Targeted NGS panels now enable the rapid identification of diverse actionable mutations, requiring clinicians to efficiently assess the predictive value of cancer biomarkers for specific treatments. The urgency for timely and accurate decision-making in oncology emphasizes the importance of reliable precision oncology software. Online clinical decision-making tools and associated cancer databases have been designed by consolidating genomic data into standardized, accessible formats. These new platforms are highly integrated and crucial for identifying actionable somatic genomic biomarkers essential for tumor survival, determining corresponding drug targets, and selecting appropriate treatments based on the mutational profile of each patient's tumor. To help oncologists and translational cancer researchers unfamiliar with these tools, we review the utility, accuracy, and comprehensiveness of several commonly used precision medicine software options currently available. Our analysis categorized selected genomic databases based on their primary content, utility, and how well they provide practical guidance for interpreting somatic biomarker data. We identified several comprehensive, mostly open-access platforms that are easy to use for genetic biomarker searches, each with unique features and limitations. Among the precision oncology tools we evaluated, we found MyCancerGenome and OncoKB to be the first choice, offering comprehensive, accurate up-to-date information on the clinical significance of somatic mutations. To illustrate the application of these precision oncology tools in clinical settings, we evaluated three case studies to see how use of the platforms could have influenced treatment planning. Most of the precision oncology software evaluated could be easily streamlined into clinical workflows to provide updated information on approved drugs and clinical trials related the actionable mutations detected. Some platforms were very intuitive and easy to use, while others, often developed in smaller academic settings, were more difficult to navigate and may not be updated consistently. Future enhancements, incorporating artificial intelligence algorithms, are likely to improve integration of the platforms with diverse big data sources, enabling more accurate predictions of potential therapeutic responses.
Collapse
Affiliation(s)
- Antonia A Gazola
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul - PUCRS, Av. Ipiranga, 668, Porto Alegre, RS, 90619-900, Brazil
| | - William Lautert-Dutra
- Department of Genetics, Medical School of Ribeirao Preto, University of Sao Paulo - USP, Ribeirao Preto, SP, 14049-900, Brazil
| | - Leticia Frohlich Archangelo
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Medical School of Ribeirao Preto, University of Sao Paulo (FMRP-USP), Ribeirao Preto, SP, 14049-900, Brazil
| | - Rodolfo B Dos Reis
- Division of Urology, Department of Surgery and Anatomy, Medical School of Ribeirao Preto, University of Sao Paulo - USP, Ribeirao Preto, SP, 14049-900, Brazil
| | - Jeremy A Squire
- Department of Genetics, Medical School of Ribeirao Preto, University of Sao Paulo - USP, Ribeirao Preto, SP, 14049-900, Brazil.
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, K7L3N6, Canada.
| |
Collapse
|
29
|
Qian L, Sun R, Aebersold R, Bühlmann P, Sander C, Guo T. AI-empowered perturbation proteomics for complex biological systems. CELL GENOMICS 2024; 4:100691. [PMID: 39488205 PMCID: PMC11605689 DOI: 10.1016/j.xgen.2024.100691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/02/2024] [Accepted: 10/06/2024] [Indexed: 11/04/2024]
Abstract
The insufficient availability of comprehensive protein-level perturbation data is impeding the widespread adoption of systems biology. In this perspective, we introduce the rationale, essentiality, and practicality of perturbation proteomics. Biological systems are perturbed with diverse biological, chemical, and/or physical factors, followed by proteomic measurements at various levels, including changes in protein expression and turnover, post-translational modifications, protein interactions, transport, and localization, along with phenotypic data. Computational models, employing traditional machine learning or deep learning, identify or predict perturbation responses, mechanisms of action, and protein functions, aiding in therapy selection, compound design, and efficient experiment design. We propose to outline a generic PMMP (perturbation, measurement, modeling to prediction) pipeline and build foundation models or other suitable mathematical models based on large-scale perturbation proteomic data. Finally, we contrast modeling between artificially and naturally perturbed systems and highlight the importance of perturbation proteomics for advancing our understanding and predictive modeling of biological systems.
Collapse
Affiliation(s)
- Liujia Qian
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
| | - Rui Sun
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | | | - Chris Sander
- Harvard Medical School, Boston, MA, USA; Broad Institute of Harvard and MIT, Boston, MA, USA; Ludwig Center at Harvard, Boston, MA, USA.
| | - Tiannan Guo
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
30
|
Wu M, Yan J, Qin S, Fu L, Sun S, Li W, Lv J, Chen L. Connections Between Endoplasmic Reticulum Stress and Prognosis of Hepatocarcinoma. Bioengineering (Basel) 2024; 11:1136. [PMID: 39593796 PMCID: PMC11591847 DOI: 10.3390/bioengineering11111136] [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: 09/26/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
Endoplasmic reticulum (ER) stress is a state in which misfolded or unfolded proteins accumulate in the lumen of the ER as a result of some exogenous or endogenous factors. It plays a crucial role in the pathogenesis of malignancies, affecting cell survival, proliferation, and metastasis in cancer. ER stress genes could provide new ideas for potential therapeutic targets in cancer. In our study, we aimed to construct an ER stress-related genes (ERGs) model for hepatocellular carcinoma (HCC). ERGs with differential expression and significant survival were screened to construct a prognostic model. The effectiveness of the model was successfully validated by external datasets. High and low-risk groups were classified based on risk scores. Functional analysis showed risk groups involved in the unfolded protein response, DNA repair, and other differential pathways. When compared to patients with low risk, the prognosis for HCC patients in the high-risk group might be worsened by disruptions in these pathways. Importantly, we considered genomic druggability and predicted drugs. Sorafenib-induced autophagy in HCC cells through an ES stress mechanism. Sorafenib was more sensitive for high-risk patients. In brief, our model predicted the prognosis of HCC and provided novel treatment strategies for the study of other cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Junjie Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (M.W.); (J.Y.); (S.Q.); (L.F.); (S.S.); (W.L.)
| | - Lina Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (M.W.); (J.Y.); (S.Q.); (L.F.); (S.S.); (W.L.)
| |
Collapse
|
31
|
Luo Y, Liang H. Developmental-status-aware transcriptional decomposition establishes a cell state panorama of human cancers. Genome Med 2024; 16:124. [PMID: 39468667 PMCID: PMC11514945 DOI: 10.1186/s13073-024-01393-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 10/03/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Cancer cells evolve under unique functional adaptations that unlock transcriptional programs embedded in adult stem and progenitor-like cells for progression, metastasis, and therapeutic resistance. However, it remains challenging to quantify the stemness-aware cell state of a tumor based on its gene expression profile. METHODS We develop a developmental-status-aware transcriptional decomposition strategy using single-cell RNA-sequencing-derived tissue-specific fetal and adult cell signatures as anchors. We apply our method to various biological contexts, including developing human organs, adult human tissues, experimentally induced differentiation cultures, and bulk human tumors, to benchmark its performance and to reveal novel biology of entangled developmental signaling in oncogenic processes. RESULTS Our strategy successfully captures complex dynamics in developmental tissue bulks, reveals remarkable cellular heterogeneity in adult tissues, and resolves the ambiguity of cell identities in in vitro transformations. Applying it to large patient cohorts of bulk RNA-seq, we identify clinically relevant cell-of-origin patterns and observe that decomposed fetal cell signals significantly increase in tumors versus normal tissues and metastases versus primary tumors. Across cancer types, the inferred fetal-state strength outperforms published stemness indices in predicting patient survival and confers substantially improved predictive power for therapeutic responses. CONCLUSIONS Our study not only provides a general approach to quantifying developmental-status-aware cell states of bulk samples but also constructs an information-rich, biologically interpretable, cell-state panorama of human cancers, enabling diverse translational applications.
Collapse
Affiliation(s)
- Yikai Luo
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Han Liang
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- Institute for Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
32
|
Johnson SJ, Johnson HL, Powell RT, Stephan C, Stossi F, Cooper TA. Small Molecule Screening Identifies HSP90 as a Modifier of RNA Foci in Myotonic Dystrophy Type 1. Mol Cell Biol 2024:1-13. [PMID: 39415708 DOI: 10.1080/10985549.2024.2408025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a multisystemic disorder caused by a CTG triplet repeat expansion within the 3' untranslated region of the DMPK gene. Expression of the expanded allele generates RNA containing long tracts of CUG repeats (CUGexp RNA) that form hairpin structures and accumulate in nuclear RNA foci; however, the factors that control DMPK expression and the formation of CUGexp RNA foci remain largely unknown. We performed an unbiased small molecule screen in an immortalized human DM1 skeletal muscle myoblast cell line and identified HSP90 as a modifier of endogenous RNA foci. Small molecule inhibition of HSP90 leads to enhancement of RNA foci and upregulation of DMPK mRNA levels. Knockdown and overexpression of HSP90 in undifferentiated DM1 myoblasts validated the impact of HSP90 with upregulation and downregulation of DMPK mRNA, respectively. Furthermore, we identified p-STAT3 as a downstream mediator of HSP90 impacting levels of DMPK mRNA and RNA foci. Interestingly, differentiated cells exhibited an opposite effect of HSP90 inhibition displaying downregulation of DMPK mRNA through a mechanism independent of p-STAT3 involvement. This study has revealed a novel mediator for DMPK mRNA and foci regulation in DM1 cells with the potential to identify targets for future therapeutic intervention.
Collapse
Affiliation(s)
- Sara J Johnson
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Hannah L Johnson
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Reid T Powell
- Department of Translational Medical Science, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas, USA
| | - Clifford Stephan
- Department of Translational Medical Science, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas, USA
| | - Fabio Stossi
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Thomas A Cooper
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
33
|
Yang J, Shen L, Zhou J, Wu J, Yue C, Wang T, Chai S, Cai Y, Xu D, Lei Y, Zhao J, Zhou Y, Mei Z, Xiong N. A Novel Mitochondrial-Related Gene Signature for the Prediction of Prognosis and Therapeutic Efficacy in Lower-Grade Glioma. Biochem Genet 2024:10.1007/s10528-024-10928-w. [PMID: 39356352 DOI: 10.1007/s10528-024-10928-w] [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: 03/16/2024] [Accepted: 09/15/2024] [Indexed: 10/03/2024]
Abstract
Lower-grade glioma (LGG) is a common primary brain tumor with a highly heterogeneous clinical presentation, and its prognosis cannot be accurately predicted by current histopathology. It has been found that mitochondria play an important role in hypoxia, angiogenesis, and energy metabolism in glioma, and mitochondrial function may have an important impact on LGG prognosis. The goal of this study was to develop a novel prognostic model based on Mitochondrial-related genes (MRGs). We first analyzed the somatic alterations profiles of MRGs in patients with LGG and found that somatic alterations were common in LGG and correlated with prognosis. Using RNA-seq data from TCGA and CGGA, 12 prognosis-related MRGs were identified to construct a mitochondrial activation score (MiAS) model by combining univariate regression and LASSO regression analysis. The model and nomogram were evaluated using the area under the ROC curve with AUC = 0.910. The model was closely correlated with the clinical characteristics of LGG patients and performed well in predicting the prognosis of LGG patients with significantly shorter overall survival (OS) time in the high-MiAS group. GSVA and GSEA results showed that oxidative stress, pro-cancer, and immune-related pathways were significantly enriched in the high-MiAS group. CIBERSORT results showed that MiAS was significantly associated with immune cell infiltration in LGG. Macrophage M1 and follicular helper T cells had increased infiltration in the high-MiAS group. TIDE predicted a better immunotherapy outcome in patients in the low-MiAS group. Finally, using data from the CTRPv2 and GDSC2 datasets to assess chemotherapy response in LGG, it was predicted that the chemotherapeutic agents AZD6482, MG-132, and PLX-4720 might be potential agents for patients in the high-MiAS group of LGG. In addition, we performed in vitro experiments and found that knockdown of OCIAD2 expression reduced the abilities of glioma cells to proliferate, migrate, and invade. In contrast, overexpression of OCIAD2 enhanced these abilities of glioma cells. This study found that MRGs were correlated with LGG patient prognosis, which is expected to provide new treatment strategies for LGG patients with different MiAS.
Collapse
Affiliation(s)
- Jingyi Yang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China
| | - Lei Shen
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China
| | - Jiabin Zhou
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China
| | - Ji Wu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China
| | - Chuqiao Yue
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China
| | - Tiansheng Wang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China
| | - Songshan Chai
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China
| | - Yuankun Cai
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China
| | - Dongyuan Xu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China
| | - Yu Lei
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China
| | - Jingwei Zhao
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China
| | - Yixuan Zhou
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China
| | - Zhimin Mei
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China
| | - Nanxiang Xiong
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, No.169, Donghu Road, Wuhan, 430071, Hubei, China.
| |
Collapse
|
34
|
Sahu D, Shi J, Segura Rueda IA, Chatrath A, Dutta A. Development of a polygenic score predicting drug resistance and patient outcome in breast cancer. NPJ Precis Oncol 2024; 8:219. [PMID: 39358487 PMCID: PMC11447244 DOI: 10.1038/s41698-024-00714-7] [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: 01/21/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024] Open
Abstract
Gene expression profiles of hundreds of cancer cell-lines and the cell-lines' response to drug treatment were analyzed to identify genes whose expression correlated with drug resistance. In the GDSC dataset of 809 cancer cell lines, expression of 36 genes were associated with drug resistance (increased IC50) to many anti-cancer drugs. This was validated in the CTRP dataset of 860 cell lines. A polygenic score derived from the correlation coefficients of the 36 genes in cancer cell lines, UAB36, predicted resistance of cell lines to Tamoxifen. Although the 36 genes were selected from cell line behaviors, UAB36 successfully predicted survival of breast cancer patients in three different cohorts of patients treated with Tamoxifen. UAB36 outperforms two existing predictive gene signatures and is a predictor of outcome of breast cancer patients independent of the known clinical co-variates that affect outcome. This approach should provide promising polygenic biomarkers for resistance in many cancer types against specific drugs.
Collapse
Affiliation(s)
- Divya Sahu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jeffrey Shi
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22903, USA
| | | | - Ajay Chatrath
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22903, USA
| | - Anindya Dutta
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22903, USA.
| |
Collapse
|
35
|
Zhang Y, Tu J, Wang J, Dai T, Zheng L, Sun S, Tu C, Li H, Qian L. NFKBIE is a predictive factor of survival and is correlated with immune infiltration and antigen processing and presentation in hepatocellular carcinoma. Oncol Lett 2024; 28:480. [PMID: 39161335 PMCID: PMC11332585 DOI: 10.3892/ol.2024.14613] [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: 05/21/2023] [Accepted: 02/21/2024] [Indexed: 08/21/2024] Open
Abstract
The important role of the nuclear factor κB (NFκB) pathway in tumour development has long been recognized; however, the role of the NFκB inhibitor family in liver cancer has not been elucidated. Hepatocellular carcinoma (HCC) is a serious public health burden with a high incidence, poor prognosis, and early detection, especially in Asia, where hepatitis is prevalent. In the present study, the mRNA expression level of the NFκB inhibitor family was assessed in HCC and normal tissues using the Metabolic Gene Rapid Visualizer, University of Alabama at Birmingham Cancer Data Analysis Portal, and the Tumor Immune Estimation Resource database (TIMER). Survival curves of nuclear factor of κ light polypeptide gene enhancer in B-cells inhibitor (NFKBI)E were obtained using the Kaplan-Meier method. Genes co-expressed with NFKBIE in HCC samples were studied using data from the LinkedOmics and the Hepatocellular Carcinoma Databases. Protein-protein interaction networks, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment pathway analyses were used to assess the NFKBIE mechanism in HCC. Using the TIMER database, the association between immune infiltration and NFKBIE was determined. RNA-sequencing (RNA-seq) was used to evaluate the function of NFKBIE in HCC and its impact on proliferation and migration. Western blotting was used to confirm the expression of NFKBIE in HCC cell lines. In addition, NFKBIE overexpression in HCC was demonstrated using tissue microarrays encompassing 80 pairs of HCC and normal liver tissues. NFKBIE was the only NFκB inhibitor with high expression and an improved prognosis in HCC compared with other NFκB inhibitors. NFKBIE was correlated with clinical characteristics, such as tumour grade, tumour protein P53 mutation status and tumour stage. Data obtained from Gene Set Cancer Analysis suggested that NFKBIE may inhibit the PI3K/AKT, RAS/MAPK, RTK and TSC/mTOR pathways. In addition, NFKBIE was significantly associated with B-cell immune infiltration and the RNA-seq data demonstrated that knockdown of NFKBIE significantly affected 'Antigen processing and presentation' and 'hepatocellular carcinoma' pathways. Immunohistochemistry of microarrays of tissue samples revealed that NFKBIE was overexpressed in several stages of HCC. Finally, inhibition of NFKBIE decreased the proliferation and migration of HCC cells. In conclusion, due to its prognostic value and overexpression in HCC, NFKBIE distinguished itself from other NFκB inhibitors. As such, it may provide a novel prognostic indicator and immunotherapeutic target for HCC.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Comprehensive Surgery, The First Affiliated Hospital of University of Science and Technology of China West District, Hefei, Anhui 230031, P.R. China
| | - Jinqi Tu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui 241001, P.R. China
| | - Jian Wang
- Department of Comprehensive Surgery, The First Affiliated Hospital of University of Science and Technology of China West District, Hefei, Anhui 230031, P.R. China
| | - Tiancheng Dai
- Department of Medical Laboratory Technology, The First Clinical College of Anhui Medical University, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Lin Zheng
- Department of Comprehensive Surgery, The First Affiliated Hospital of University of Science and Technology of China West District, Hefei, Anhui 230031, P.R. China
| | - Sinan Sun
- Department of Radiation Oncology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230031, P.R. China
| | - Conyin Tu
- Department of Comprehensive Surgery, The First Affiliated Hospital of University of Science and Technology of China West District, Hefei, Anhui 230031, P.R. China
| | - Heng Li
- Department of Comprehensive Surgery, The First Affiliated Hospital of University of Science and Technology of China West District, Hefei, Anhui 230031, P.R. China
| | - Liting Qian
- Department of Radiation Oncology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230031, P.R. China
| |
Collapse
|
36
|
Qiu J, Fu Y, Liu T, Wang J, Liu Y, Zhang Z, Ye Z, Cao Z, Su D, Luo W, Tao J, Weng G, Ye L, Zhang F, Liang Z, Zhang T. Single-cell RNA-seq reveals heterogeneity in metastatic renal cell carcinoma and effect of anti-angiogenesis therapy in the pancreas metastatic lesion. Cancer Lett 2024; 601:217193. [PMID: 39159881 DOI: 10.1016/j.canlet.2024.217193] [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: 06/10/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024]
Abstract
Metastatic clear cell renal cell carcinoma has heterogenous tumor microenvironment (TME). Among the metastatic lesions, pancreas metastasis is rare and controversy in treatment approaches. Here, extensive primary and metastatic lesion samples were included by single-cell RNA-seq to decipher the distinct metastasis TME. The hypoxic and inflammatory TME of pancreas metastasis was decoded in this study, and the activation of PAX8-myc signaling, and metabolic reprogramming were observed. The active components including endothelial cells, fibroblasts and T cells were profiled. Meanwhile, we also evaluated the effect of anti-angiogenesis treatment in the pancreas metastasis patient. The potential mechanisms of pancreatic tropism, instability of genome, and the response of immunotherapy were also discussed in this work. Taken together, our findings suggest a clue to the heterogeneity in metastasis TME and provide evidence for the treatment of pancreas metastasis in renal cell carcinoma patients.
Collapse
Affiliation(s)
- Jiangdong Qiu
- General Surgery Department, 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, 100730, China.
| | - Yifan Fu
- General Surgery Department, 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, 100730, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Tao Liu
- General Surgery Department, 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, 100730, China.
| | - Jun Wang
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yueze Liu
- General Surgery Department, 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, 100730, China.
| | - Zeyu Zhang
- General Surgery Department, 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, 100730, China.
| | - Ziwen Ye
- Department of Urology, The Fist Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Zhe Cao
- General Surgery Department, 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, 100730, China.
| | - Dan Su
- General Surgery Department, 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, 100730, China.
| | - Wenhao Luo
- General Surgery Department, 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, 100730, China.
| | - Jinxin Tao
- General Surgery Department, 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, 100730, China.
| | - Guihu Weng
- General Surgery Department, 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, 100730, China.
| | - Liyuan Ye
- General Surgery Department, 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, 100730, China.
| | - Feifan Zhang
- Department of Computer Science, University College London, UK.
| | - Zhiyong Liang
- Department of Pathology, State Key Laboratory of Complex Severe and Rare Disease, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Taiping Zhang
- General Surgery Department, 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, 100730, China.
| |
Collapse
|
37
|
Xu J, Hao J, Liao X, Shang X, Li X. SSCI: Self-Supervised Deep Learning Improves Network Structure for Cancer Driver Gene Identification. Int J Mol Sci 2024; 25:10351. [PMID: 39408682 PMCID: PMC11476395 DOI: 10.3390/ijms251910351] [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: 08/27/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
The pathogenesis of cancer is complex, involving abnormalities in some genes in organisms. Accurately identifying cancer genes is crucial for the early detection of cancer and personalized treatment, among other applications. Recent studies have used graph deep learning methods to identify cancer driver genes based on biological networks. However, incompleteness and the noise of the networks will weaken the performance of models. To address this, we propose a cancer driver gene identification method based on self-supervision for graph convolutional networks, which can efficiently enhance the structure of the network and further improve predictive accuracy. The reliability of SSCI is verified by the area under the receiver operating characteristic curves (AUROC), the area under the precision-recall curves (AUPRC), and the F1 score, with respective values of 0.966, 0.964, and 0.913. The results show that our method can identify cancer driver genes with strong discriminative power and biological interpretability.
Collapse
Affiliation(s)
- Jialuo Xu
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China; (J.X.); (J.H.); (X.L.); (X.S.)
| | - Jun Hao
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China; (J.X.); (J.H.); (X.L.); (X.S.)
| | - Xingyu Liao
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China; (J.X.); (J.H.); (X.L.); (X.S.)
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China; (J.X.); (J.H.); (X.L.); (X.S.)
| | - Xingyi Li
- School of Computer Science, Northwestern Polytechnical University, Xi’an 710072, China; (J.X.); (J.H.); (X.L.); (X.S.)
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518063, China
| |
Collapse
|
38
|
Kamble P, Nagar PR, Bhakhar KA, Garg P, Sobhia ME, Naidu S, Bharatam PV. Cancer pharmacoinformatics: Databases and analytical tools. Funct Integr Genomics 2024; 24:166. [PMID: 39294509 DOI: 10.1007/s10142-024-01445-5] [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: 07/29/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
Cancer is a subject of extensive investigation, and the utilization of omics technology has resulted in the generation of substantial volumes of big data in cancer research. Numerous databases are being developed to manage and organize this data effectively. These databases encompass various domains such as genomics, transcriptomics, proteomics, metabolomics, immunology, and drug discovery. The application of computational tools into various core components of pharmaceutical sciences constitutes "Pharmacoinformatics", an emerging paradigm in rational drug discovery. The three major features of pharmacoinformatics include (i) Structure modelling of putative drugs and targets, (ii) Compilation of databases and analysis using statistical approaches, and (iii) Employing artificial intelligence/machine learning algorithms for the discovery of novel therapeutic molecules. The development, updating, and analysis of databases using statistical approaches play a pivotal role in pharmacoinformatics. Multiple software tools are associated with oncoinformatics research. This review catalogs the databases and computational tools related to cancer drug discovery and highlights their potential implications in the pharmacoinformatics of cancer.
Collapse
Affiliation(s)
- Pradnya Kamble
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Prinsa R Nagar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Kaushikkumar A Bhakhar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Prabha Garg
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - M Elizabeth Sobhia
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India
| | - Srivatsava Naidu
- Center of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
| | - Prasad V Bharatam
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India.
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Punjab, India.
| |
Collapse
|
39
|
Connell W, Garcia K, Goodarzi H, Keiser MJ. Learning chemical sensitivity reveals mechanisms of cellular response. Commun Biol 2024; 7:1149. [PMID: 39278951 PMCID: PMC11402971 DOI: 10.1038/s42003-024-06865-4] [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: 12/11/2023] [Accepted: 09/06/2024] [Indexed: 09/18/2024] Open
Abstract
Chemical probes interrogate disease mechanisms at the molecular level by linking genetic changes to observable traits. However, comprehensive chemical screens in diverse biological models are impractical. To address this challenge, we develop ChemProbe, a model that predicts cellular sensitivity to hundreds of molecular probes and drugs by learning to combine transcriptomes and chemical structures. Using ChemProbe, we infer the chemical sensitivity of cancer cell lines and tumor samples and analyze how the model makes predictions. We retrospectively evaluate drug response predictions for precision breast cancer treatment and prospectively validate chemical sensitivity predictions in new cellular models, including a genetically modified cell line. Our model interpretation analysis identifies transcriptome features reflecting compound targets and protein network modules, identifying genes that drive ferroptosis. ChemProbe is an interpretable in silico screening tool that allows researchers to measure cellular response to diverse compounds, facilitating research into molecular mechanisms of chemical sensitivity.
Collapse
Affiliation(s)
- William Connell
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Kristle Garcia
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Hani Goodarzi
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Michael J Keiser
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA.
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA.
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
40
|
Manen-Freixa L, Antolin AA. Polypharmacology prediction: the long road toward comprehensively anticipating small-molecule selectivity to de-risk drug discovery. Expert Opin Drug Discov 2024; 19:1043-1069. [PMID: 39004919 DOI: 10.1080/17460441.2024.2376643] [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: 03/15/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024]
Abstract
INTRODUCTION Small molecules often bind to multiple targets, a behavior termed polypharmacology. Anticipating polypharmacology is essential for drug discovery since unknown off-targets can modulate safety and efficacy - profoundly affecting drug discovery success. Unfortunately, experimental methods to assess selectivity present significant limitations and drugs still fail in the clinic due to unanticipated off-targets. Computational methods are a cost-effective, complementary approach to predict polypharmacology. AREAS COVERED This review aims to provide a comprehensive overview of the state of polypharmacology prediction and discuss its strengths and limitations, covering both classical cheminformatics methods and bioinformatic approaches. The authors review available data sources, paying close attention to their different coverage. The authors then discuss major algorithms grouped by the types of data that they exploit using selected examples. EXPERT OPINION Polypharmacology prediction has made impressive progress over the last decades and contributed to identify many off-targets. However, data incompleteness currently limits most approaches to comprehensively predict selectivity. Moreover, our limited agreement on model assessment challenges the identification of the best algorithms - which at present show modest performance in prospective real-world applications. Despite these limitations, the exponential increase of multidisciplinary Big Data and AI hold much potential to better polypharmacology prediction and de-risk drug discovery.
Collapse
Affiliation(s)
- Leticia Manen-Freixa
- Oncobell Division, Bellvitge Biomedical Research Institute (IDIBELL) and ProCURE Department, Catalan Institute of Oncology (ICO), Barcelona, Spain
| | - Albert A Antolin
- Oncobell Division, Bellvitge Biomedical Research Institute (IDIBELL) and ProCURE Department, Catalan Institute of Oncology (ICO), Barcelona, Spain
- Center for Cancer Drug Discovery, The Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| |
Collapse
|
41
|
Rintala TJ, Napolitano F, Fortino V. Multi-task deep latent spaces for cancer survival and drug sensitivity prediction. Bioinformatics 2024; 40:ii182-ii189. [PMID: 39230696 PMCID: PMC11520233 DOI: 10.1093/bioinformatics/btae388] [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] [Indexed: 09/05/2024] Open
Abstract
MOTIVATION Cancer is a very heterogeneous disease that can be difficult to treat without addressing the specific mechanisms driving tumour progression in a given patient. High-throughput screening and sequencing data from cancer cell-lines has driven many developments in drug development, however, there are important aspects crucial to precision medicine that are often overlooked, namely the inherent differences between tumours in patients and the cell-lines used to model them in vitro. Recent developments in transfer learning methods for patient and cell-line data have shown progress in translating results from cell-lines to individual patients in silico. However, transfer learning can be forceful and there is a risk that clinically relevant patterns in the omics profiles of patients are lost in the process. RESULTS We present MODAE, a novel deep learning algorithm to integrate omics profiles from cell-lines and patients for the purposes of exploring precision medicine opportunities. MODAE implements patient survival prediction as an additional task in a drug-sensitivity transfer learning schema and aims to balance autoencoding, domain adaptation, drug-sensitivity prediction, and survival prediction objectives in order to better preserve the heterogeneity between patients that is relevant to survival. While burdened with these additional tasks, MODAE performed on par with baseline survival models, but struggled in the drug-sensitivity prediction task. Nevertheless, these preliminary results were promising and show that MODAE provides a novel AI-based method for prioritizing drug treatments for high-risk patients. AVAILABILITY AND IMPLEMENTATION https://github.com/UEFBiomedicalInformaticsLab/MODAE.
Collapse
Affiliation(s)
- Teemu J Rintala
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio 70210, Finland
| | - Francesco Napolitano
- Department of Science and Technology, University of Sannio, Benevento 82100, Italy
| | - Vittorio Fortino
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio 70210, Finland
| |
Collapse
|
42
|
Schwab A, Rao Z, Zhang J, Gollowitzer A, Siebenkäs K, Bindel N, D'Avanzo E, van Roey R, Hajjaj Y, Özel E, Armstark I, Bereuter L, Su F, Grander J, Bonyadi Rad E, Groenewoud A, Engel FB, Bell GW, Henry WS, Angeli JPF, Stemmler MP, Brabletz S, Koeberle A, Brabletz T. Zeb1 mediates EMT/plasticity-associated ferroptosis sensitivity in cancer cells by regulating lipogenic enzyme expression and phospholipid composition. Nat Cell Biol 2024; 26:1470-1481. [PMID: 39009641 PMCID: PMC11392809 DOI: 10.1038/s41556-024-01464-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 06/20/2024] [Indexed: 07/17/2024]
Abstract
Therapy resistance and metastasis, the most fatal steps in cancer, are often triggered by a (partial) activation of the epithelial-mesenchymal transition (EMT) programme. A mesenchymal phenotype predisposes to ferroptosis, a cell death pathway exerted by an iron and oxygen-radical-mediated peroxidation of phospholipids containing polyunsaturated fatty acids. We here show that various forms of EMT activation, including TGFβ stimulation and acquired therapy resistance, increase ferroptosis susceptibility in cancer cells, which depends on the EMT transcription factor Zeb1. We demonstrate that Zeb1 increases the ratio of phospholipids containing pro-ferroptotic polyunsaturated fatty acids over cyto-protective monounsaturated fatty acids by modulating the differential expression of the underlying crucial enzymes stearoyl-Co-A desaturase 1 (SCD), fatty acid synthase (FASN), fatty acid desaturase 2 (FADS2), elongation of very long-chain fatty acid 5 (ELOVL5) and long-chain acyl-CoA synthetase 4 (ACSL4). Pharmacological inhibition of selected lipogenic enzymes (SCD and FADS2) allows the manipulation of ferroptosis sensitivity preferentially in high-Zeb1-expressing cancer cells. Our data are of potential translational relevance and suggest a combination of ferroptosis activators and SCD inhibitors for the treatment of aggressive cancers expressing high Zeb1.
Collapse
Affiliation(s)
- Annemarie Schwab
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Zhigang Rao
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Jie Zhang
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - André Gollowitzer
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Katharina Siebenkäs
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Nino Bindel
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Elisabetta D'Avanzo
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ruthger van Roey
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Yussuf Hajjaj
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ece Özel
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Isabell Armstark
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Leonhard Bereuter
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Fengting Su
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Julia Grander
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Ehsan Bonyadi Rad
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Arwin Groenewoud
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - George W Bell
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Whitney S Henry
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Dept. of Biology, MIT, Cambridge, MA, USA
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Marc P Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Simone Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences (CMBI), University of Innsbruck, Innsbruck, Austria.
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany.
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| |
Collapse
|
43
|
Kojima Y, Fujieda S, Zhou L, Takikawa M, Kuramochi K, Furuya T, Mizumoto A, Kagaya N, Kawahara T, Shin‐ya K, Dan S, Tomida A, Ishikawa F, Sadaie M. Cytochrome P450 2J2 is required for the natural compound austocystin D to elicit cancer cell toxicity. Cancer Sci 2024; 115:3054-3066. [PMID: 39009033 PMCID: PMC11462933 DOI: 10.1111/cas.16289] [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: 03/12/2024] [Revised: 06/25/2024] [Accepted: 07/04/2024] [Indexed: 07/17/2024] Open
Abstract
Austocystin D is a natural compound that induces cytochrome P450 (CYP) monooxygenase-dependent DNA damage and growth inhibition in certain cancer cell lines. Cancer cells exhibiting higher sensitivity to austocystin D often display elevated CYP2J2 expression. However, the essentiality and the role of CYP2J2 for the cytotoxicity of this compound remain unclear. In this study, we demonstrate that CYP2J2 depletion alleviates austocystin D sensitivity and DNA damage induction, while CYP2J2 overexpression enhances them. Moreover, the investigation into genes involved in austocystin D cytotoxicity identified POR and PGRMC1, positive regulators for CYP activity, and KAT7, a histone acetyltransferase. Through genetic manipulation and analysis of multiomics data, we elucidated a role for KAT7 in CYP2J2 transcriptional regulation. These findings strongly suggest that CYP2J2 is crucial for austocystin D metabolism and its subsequent cytotoxic effects. The potential use of austocystin D as a therapeutic prodrug is underscored, particularly in cancers where elevated CYP2J2 expression serves as a biomarker.
Collapse
Affiliation(s)
- Yukiko Kojima
- Department of Applied Biological Science, Faculty of Science and TechnologyTokyo University of ScienceNoda, ChibaJapan
| | - Saki Fujieda
- Department of Applied Biological Science, Faculty of Science and TechnologyTokyo University of ScienceNoda, ChibaJapan
| | - Liya Zhou
- Department of Applied Biological Science, Faculty of Science and TechnologyTokyo University of ScienceNoda, ChibaJapan
| | - Masahiro Takikawa
- Department of Applied Biological Science, Faculty of Science and TechnologyTokyo University of ScienceNoda, ChibaJapan
| | - Kouji Kuramochi
- Department of Applied Biological Science, Faculty of Science and TechnologyTokyo University of ScienceNoda, ChibaJapan
| | - Toshiki Furuya
- Department of Applied Biological Science, Faculty of Science and TechnologyTokyo University of ScienceNoda, ChibaJapan
| | - Ayaka Mizumoto
- Department of Gene Mechanisms, Graduate School of BiostudiesKyoto UniversityKyotoJapan
| | - Noritaka Kagaya
- National Institute of Advanced Industrial Science and Technology (AIST)TokyoJapan
| | | | - Kazuo Shin‐ya
- National Institute of Advanced Industrial Science and Technology (AIST)TokyoJapan
| | - Shingo Dan
- Cancer Chemotherapy CenterJapanese Foundation for Cancer Research (JFCR)TokyoJapan
| | - Akihiro Tomida
- Cancer Chemotherapy CenterJapanese Foundation for Cancer Research (JFCR)TokyoJapan
| | - Fuyuki Ishikawa
- Department of Gene Mechanisms, Graduate School of BiostudiesKyoto UniversityKyotoJapan
| | - Mahito Sadaie
- Department of Applied Biological Science, Faculty of Science and TechnologyTokyo University of ScienceNoda, ChibaJapan
- Department of Gene Mechanisms, Graduate School of BiostudiesKyoto UniversityKyotoJapan
| |
Collapse
|
44
|
Lu B, Liu Y, Yao Y, Zhu D, Zhang X, Dong K, Xu X, Lv D, Zhao Z, Zhang H, Yang X, Fu W, Huang R, Cao J, Chu J, Pan X, Cui X. Unveiling the unique role of TSPAN7 across tumors: a pan-cancer study incorporating retrospective clinical research and bioinformatic analysis. Biol Direct 2024; 19:72. [PMID: 39175035 PMCID: PMC11340126 DOI: 10.1186/s13062-024-00516-8] [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: 04/28/2024] [Accepted: 08/08/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND TSPAN7 is an important factor in tumor progression. However, the precise function of TSPAN7 and its role in pan-cancer are not clear. METHODS Based on Xinhua cohort incorporating 370 patients with kidney neoplasm, we conducted differential expression analysis by immunohistochemistry between tumor and normal tissues, and explored correlations of TSPAN7 with patients' survival. Subsequently, we conducted a pan-cancer study, and successively employed differential expression analysis, competing endogenous RNA (ceRNA) analysis, protein-protein interaction (PPI) analysis, correlation analysis of TSPAN7 with clinical characteristics, tumor purity, tumor genomics, tumor immunity, and drug sensitivity. Last but not least, gene set enrichment analysis was applied to identify enriched pathways of TSPAN7. RESULTS In Xinhua cohort, TSPAN7 expression was significantly up-regulated (P-value = 0.0019) in tumor tissues of kidney neoplasm patients. High TSPAN7 expression was associated with decreases in overall survival (OS) (P-value = 0.009) and progression-free survival (P-value = 0.009), and it was further revealed as an independent risk factor for OS (P-value = 0.0326, HR = 5.66, 95%CI = 1.155-27.8). In pan-cancer analysis, TSPAN7 expression was down-regulated in most tumors, and it was associated with patients' survival, tumor purity, tumor genomics, tumor immunity, and drug sensitivity. The ceRNA network and PPI network of TSPAN7 were also constructed. Last but not least, the top five enriched pathways of TSPAN7 in various tumors were identified. CONCLUSION TSPAN7 served as a promising biomarker of various tumors, especially kidney neoplasms, and it was closely associated with tumor purity, tumor genomics, tumor immunology, and drug sensitivity in pan-cancer level.
Collapse
Affiliation(s)
- Bingnan Lu
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Yifan Liu
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Yuntao Yao
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Dawei Zhu
- Department of Urology, the Second People's Hospital of Pinghu, Zhejiang, 314200, China
| | - Xiangmin Zhang
- Department of Urology, Shanghai Baoshan Luodian Hospital, Shanghai, 201908, China
| | - Keqin Dong
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Xiao Xu
- Department of Urology, the Second People's Hospital of Pinghu, Zhejiang, 314200, China
| | - Donghao Lv
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Zihui Zhao
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Haoyu Zhang
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Xinyue Yang
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Wenjia Fu
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China
| | - Runzhi Huang
- Department of Burn Surgery, the First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| | - Jianwei Cao
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China.
- Department of Urology, the Second People's Hospital of Pinghu, Zhejiang, 314200, China.
| | - Jian Chu
- Department of Urology, Shanghai Baoshan Luodian Hospital, Shanghai, 201908, China.
| | - Xiuwu Pan
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China.
| | - Xingang Cui
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
45
|
Vis DJ, Jaaks P, Aben N, Coker EA, Barthorpe S, Beck A, Hall C, Hall J, Lightfoot H, Lleshi E, Mironenko T, Richardson L, Tolley C, Garnett MJ, Wessels LFA. A pan-cancer screen identifies drug combination benefit in cancer cell lines at the individual and population level. Cell Rep Med 2024; 5:101687. [PMID: 39168097 PMCID: PMC11384948 DOI: 10.1016/j.xcrm.2024.101687] [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: 01/22/2024] [Revised: 05/10/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024]
Abstract
Combining drugs can enhance their clinical efficacy, but the number of possible combinations and inter-tumor heterogeneity make identifying effective combinations challenging, while existing approaches often overlook clinically relevant activity. We screen one of the largest cell line panels (N = 757) with 51 clinically relevant combinations and identify responses at the level of individual cell lines and tissue populations. We establish three response classes to model cellular effects beyond monotherapy: synergy, Bliss additivity, and independent drug action (IDA). Synergy is rare (11% of responses) and frequently efficacious (>50% viability reduction), whereas Bliss and IDA are more frequent but less frequently efficacious. We introduce "efficacious combination benefit" (ECB) to describe high-efficacy responses classified as either synergy, Bliss, or IDA. We identify ECB biomarkers in vitro and show that ECB predicts response in patient-derived xenografts better than synergy alone. Our work here provides a valuable resource and framework for preclinical evaluation and the development of combination treatments.
Collapse
Affiliation(s)
- Daniel J Vis
- Department of EEMCS, Delft University of Technology, the Netherlands
| | | | - Nanne Aben
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | | | | | | | - James Hall
- Wellcome Sanger Institute, Cambridge, UK
| | | | | | | | | | | | | | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands.
| |
Collapse
|
46
|
Ye J, Wei B, Zhou G, Xu Y, He Y, Hu X, Chen X, Zhang G, Liu H. Multi-dimensional characterization of apoptosis in the tumor microenvironment and therapeutic relevance in melanoma. Cell Oncol (Dordr) 2024; 47:1333-1353. [PMID: 38502270 PMCID: PMC11322377 DOI: 10.1007/s13402-024-00930-0] [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] [Accepted: 02/23/2024] [Indexed: 03/21/2024] Open
Abstract
PURPOSE Melanoma is widely utilized as a prominent model for the development of immunotherapy, thought an inadequate immune response can occur. Moreover, the development of apoptosis-related therapies and combinations with other therapeutic strategies is impeded by the limited understanding of apoptosis's role within diverse tumor immune microenvironments (TMEs). METHODS Here, we constructed an apoptosis-related tumor microenvironment signature (ATM) and employ multi-dimensional analysis to understand the roles of apoptosis in tumor microenvironment. We further assessed the clinical applications of ATM in nine independent cohorts, and anticipated the impact of ATM on cellular drug response in cultured cells. RESULTS Our ATM model exhibits robust performance in survival prediction in multiple melanoma cohorts. Different ATM groups exhibited distinct molecular signatures and biological processes. The low ATM group exhibited significant enrichment in B cell activation-related pathways. What's more, plasma cells showed the lowest ATM score, highlighting their role as pivotal contributors in the ATM model. Mechanistically, the analysis of the interplay between plasma cells and other immune cells elucidated their crucial role in orchestrating an effective anti-tumor immune response. Significantly, the ATM signature exhibited associations with therapeutic efficacy of immune checkpoint blockade and the drug sensitivity of various agents, including FDA-approved and clinically utilized drugs targeting the VEGF signaling pathway. Finally, ATM was associated with tertiary lymphoid structures (TLS), exhibiting stronger patient stratification ability compared to classical "hot tumors". CONCLUSION Our findings indicate that ATM is a prognostic factor and is associated with the immune response and drug sensitivity in melanoma.
Collapse
Affiliation(s)
- Jing Ye
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China
| | - Benliang Wei
- Big Data Institute, Central South University, Changsha, Hunan, 410008, China
| | - Guowei Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China
| | - Yantao Xu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China
| | - Yi He
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China
| | - Xiheng Hu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China.
- Furong Laboratory, Changsha, Hunan, China.
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China.
- Furong Laboratory, Changsha, Hunan, China.
| | - Guanxiong Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China.
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, 410008, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, 410008, China.
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, 410008, China.
- Xiangya Clinical Research Center for Cancer Immunotherapy, Changsha, Hunan, 410008, China.
- Big Data Institute, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
47
|
Liu Y, Cai L, Wang H, Yao L, Zhang K, Chen G, Zhou Y. Novel mitochondrial-related gene signature predicts prognosis and immunological status in glioma. Transl Cancer Res 2024; 13:3338-3353. [PMID: 39145059 PMCID: PMC11319993 DOI: 10.21037/tcr-23-2072] [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: 11/09/2023] [Accepted: 06/04/2024] [Indexed: 08/16/2024]
Abstract
Background Mitochondria are the center of cellular metabolism. The relationship between mitochondria and diseases has also been studied for a long time. However, the prognostic role of mitochondrial-related genes (MRGs) in patients with glioma and their biological effects are still unclear. The aim of the study was to construct a mitochondria-related model to assess prognosis and potential biological effects like immune infiltration, gene pathway and mutation, and give some predictive chemotherapeutic agents. Methods The data of 675 patients from The Cancer Genome Atlas (TCGA) database were used to identify MRG signature and construct a prognostic model. After validating its robustness in Chinese Glioma Genome Atlas (CGGA), two risk groups derived from the prognostic model were then conducted with Gene Set Enrichment Analysis (GSEA), immune status, mutation status and chemotherapeutic agents prediction. Results The prognostic model built from six gene signatures can successfully predict the prognosis and reflect clinicopathological characteristics. Patients in high-risk group displayed significantly worse overall survival (OS), immunosuppression effects, and mutation markers with worse prognosis. Twelve chemotherapeutic agents with strongly correlated sensitivity and risk scores were selected as potential agents. Conclusions The novel MRG signatures (TYMP, TSFM, MGME1, BOLA3, TRMT5, NDUFA9) can predict prognosis and immunological status in glioma.
Collapse
Affiliation(s)
- Yongsheng Liu
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lize Cai
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Wang
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lin Yao
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Kai Zhang
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guangliang Chen
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Youxin Zhou
- Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
48
|
Mohammadzadeh-Vardin T, Ghareyazi A, Gharizadeh A, Abbasi K, Rabiee HR. DeepDRA: Drug repurposing using multi-omics data integration with autoencoders. PLoS One 2024; 19:e0307649. [PMID: 39058696 PMCID: PMC11280260 DOI: 10.1371/journal.pone.0307649] [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: 05/13/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Cancer treatment has become one of the biggest challenges in the world today. Different treatments are used against cancer; drug-based treatments have shown better results. On the other hand, designing new drugs for cancer is costly and time-consuming. Some computational methods, such as machine learning and deep learning, have been suggested to solve these challenges using drug repurposing. Despite the promise of classical machine-learning methods in repurposing cancer drugs and predicting responses, deep-learning methods performed better. This study aims to develop a deep-learning model that predicts cancer drug response based on multi-omics data, drug descriptors, and drug fingerprints and facilitates the repurposing of drugs based on those responses. To reduce multi-omics data's dimensionality, we use autoencoders. As a multi-task learning model, autoencoders are connected to MLPs. We extensively tested our model using three primary datasets: GDSC, CTRP, and CCLE to determine its efficacy. In multiple experiments, our model consistently outperforms existing state-of-the-art methods. Compared to state-of-the-art models, our model achieves an impressive AUPRC of 0.99. Furthermore, in a cross-dataset evaluation, where the model is trained on GDSC and tested on CCLE, it surpasses the performance of three previous works, achieving an AUPRC of 0.72. In conclusion, we presented a deep learning model that outperforms the current state-of-the-art regarding generalization. Using this model, we could assess drug responses and explore drug repurposing, leading to the discovery of novel cancer drugs. Our study highlights the potential for advanced deep learning to advance cancer therapeutic precision.
Collapse
Affiliation(s)
- Taha Mohammadzadeh-Vardin
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
| | - Amin Ghareyazi
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
| | - Ali Gharizadeh
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
| | - Karim Abbasi
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
- Faculty of Mathematics and Computer Science, Kharazmi University, Tehran, Iran
| | - Hamid R. Rabiee
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
49
|
Oikonomou A, Watrin T, Valsecchi L, Scharov K, Savino AM, Schliehe-Diecks J, Bardini M, Fazio G, Bresolin S, Biondi A, Borkhardt A, Bhatia S, Cazzaniga G, Palmi C. Synergistic drug interactions of the histone deacetylase inhibitor givinostat (ITF2357) in CRLF2-rearranged pediatric B-cell precursor acute lymphoblastic leukemia identified by high-throughput drug screening. Heliyon 2024; 10:e34033. [PMID: 39071567 PMCID: PMC11277435 DOI: 10.1016/j.heliyon.2024.e34033] [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: 11/20/2023] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Combining multiple drugs broadens the window of therapeutic opportunities and is crucial for diseases that are currently lacking fully curative treatments. A powerful emerging tool for selecting effective drugs and combinations is the high-throughput drug screening (HTP). The histone deacetylase inhibitor (HDACi) givinostat (ITF2357) has been shown to act effectively against CRLF2-rearranged pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL), a subtype characterized by poor outcome and enriched in children with Down Syndrome, very fragile patients with a high susceptibility to treatment-related toxicity. The aim of this study is to investigate possible synergies with givinostat for these difficult-to-treat patients by performing HTP screening with a library of 174 drugs, either approved or in preclinical studies. By applying this approach to the CRLF2-r MHH-CALL-4 cell line, we identified 19 compounds with higher sensitivity in combination with givinostat compared to the single treatments. Next, the synergy between givinostat and the promising candidates was further validated in CRLF2r cell lines with a broad matrix of concentrations. The combinations with trametinib (MEKi) or venetoclax (BCL2i) were found to be the most effective and with the greatest synergy across three metrics (ZIP, HAS, Bliss). Their efficacy was confirmed in primary blasts treated ex vivo at concentration ranges with a safe profile on healthy cells. Finally, we described givinostat-induced modifications in gene expression of MAPK and BCL-2 family members, supporting the observed synergistic interactions. Overall, our study represents a model of drug repurposing strategy using HTP screening for identifying synergistic, efficient, and safe drug combinations.
Collapse
Affiliation(s)
| | - Titus Watrin
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Heinrich-Heine University Dusseldorf, Medical Faculty, Düsseldorf, Germany
| | - Luigia Valsecchi
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Katerina Scharov
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Heinrich-Heine University Dusseldorf, Medical Faculty, Düsseldorf, Germany
| | - Angela Maria Savino
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Italy
| | - Julian Schliehe-Diecks
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Heinrich-Heine University Dusseldorf, Medical Faculty, Düsseldorf, Germany
| | - Michela Bardini
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Grazia Fazio
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Silvia Bresolin
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Women and Child Health Department, Padua University and Hospital, Padua, Italy
- Onco-Hematology, Stem Cell Transplant and Gene Therapy, Istituto di Ricerca Pediatrica Foundation - Città della Speranza, Padua, Italy
| | - Andrea Biondi
- School of Medicine and Surgery, University of Milano-Bicocca, Italy
- Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Arndt Borkhardt
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Heinrich-Heine University Dusseldorf, Medical Faculty, Düsseldorf, Germany
| | - Sanil Bhatia
- Department of Paediatric Oncology, Haematology and Clinical Immunology, Heinrich-Heine University Dusseldorf, Medical Faculty, Düsseldorf, Germany
| | - Giovanni Cazzaniga
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Italy
| | - Chiara Palmi
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
50
|
Wu X, Yuan H, Wu Q, Gao Y, Duan T, Yang K, Huang T, Wang S, Yuan F, Lee D, Taori S, Plute T, Heissel S, Alwaseem H, Isay-Del Viscio M, Molina H, Agnihotri S, Hsu DJ, Zhang N, Rich JN. Threonine fuels glioblastoma through YRDC-mediated codon-biased translational reprogramming. NATURE CANCER 2024; 5:1024-1044. [PMID: 38519786 PMCID: PMC11552442 DOI: 10.1038/s43018-024-00748-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 02/23/2024] [Indexed: 03/25/2024]
Abstract
Cancers commonly reprogram translation and metabolism, but little is known about how these two features coordinate in cancer stem cells. Here we show that glioblastoma stem cells (GSCs) display elevated protein translation. To dissect underlying mechanisms, we performed a CRISPR screen and identified YRDC as the top essential transfer RNA (tRNA) modification enzyme in GSCs. YRDC catalyzes the formation of N6-threonylcarbamoyladenosine (t6A) on ANN-decoding tRNA species (A denotes adenosine, and N denotes any nucleotide). Targeting YRDC reduced t6A formation, suppressed global translation and inhibited tumor growth both in vitro and in vivo. Threonine is an essential substrate of YRDC. Threonine accumulated in GSCs, which facilitated t6A formation through YRDC and shifted the proteome to support mitosis-related genes with ANN codon bias. Dietary threonine restriction (TR) reduced tumor t6A formation, slowed xenograft growth and augmented anti-tumor efficacy of chemotherapy and anti-mitotic therapy, providing a molecular basis for a dietary intervention in cancer treatment.
Collapse
Affiliation(s)
- Xujia Wu
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Department of Neurosurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, China
| | - Huairui Yuan
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Qiulian Wu
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Yixin Gao
- Department of Neurosurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, China
| | - Tingting Duan
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
| | - Tengfei Huang
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Shuai Wang
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Fanen Yuan
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Derrick Lee
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Suchet Taori
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Tritan Plute
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- John G. Rangos Sr. Research Center, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Søren Heissel
- Proteomics Resource Center, the Rockefeller University, New York, NY, USA
| | - Hanan Alwaseem
- Proteomics Resource Center, the Rockefeller University, New York, NY, USA
| | | | - Henrik Molina
- Proteomics Resource Center, the Rockefeller University, New York, NY, USA
| | - Sameer Agnihotri
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- John G. Rangos Sr. Research Center, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Dennis J Hsu
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nu Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, China.
| | - Jeremy N Rich
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|