1
|
Wang K, Xiang J, Zhou J, Chen C, Wang Z, Qin N, Zhu M, Bi L, Gong L, Yang L, Chen Y, Xu X, Dai J, Ma H, Hu Z, Li W, Wang C, Jin G, Shen H. Development and validation of a transcription factor regulatory network-based signature for individualized prognostic risk in lung adenocarcinoma. Int J Cancer 2025; 156:2440-2451. [PMID: 39960662 DOI: 10.1002/ijc.35375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/28/2025] [Accepted: 02/05/2025] [Indexed: 03/17/2025]
Abstract
Despite significant progress in diagnostic and therapeutic modalities, lung adenocarcinoma (LUAD) still exhibits a high recurrence risk and a low 5-year survival rate. Reliable prognostic signatures are imperative for risk stratification in LUAD patients. This study encompassed 2740 patients from 23 LUAD cohorts, including one single-cell RNA sequencing (scRNA-seq) dataset, five bulk RNA-seq datasets, and 17 microarray datasets. Using scRNA-seq dataset, we defined a group of epithelial-specific transcription factors significantly over-represented in the epithelial-to-mesenchymal transition (EMT) gene set (enrichment ratio [ER] = 5.80, Fisher's exact test p < .001), and the corresponding target genes were significantly enriched in the cancer driver gene set (ER = 2.74, p < .001), indicating of their crucial roles in the EMT process and tumor progression. We constructed a single-cell gene pairs (scGPS) signature, composed of 3521 gene pairs derived from the epithelial cell-specific transcription factor regulatory network, to predict overall survival (OS) of LUAD. High-risk patients identified by scGPS in the discovery cohort exhibited significantly worse OS compared to low-risk patients (Hazard ratio [HR] = 1.78, 95% CI: 1.29-2.46, log-rank p = 1.80 × 10-4). The scGPS outperformed other established gene signatures and demonstrated robust prognostic stratification across various independent datasets, including microarray data and even early-stage LUAD patients. It remained an independent prognostic factor after adjusting for clinical and pathologic factors. In addition, combining scGPS with tumor stage further enhanced prognostic accuracy compared to using stage alone. The scGPS signature offers individualized prognosis estimations, showing significant potential for practical application in clinical settings.
Collapse
Affiliation(s)
- Kai Wang
- Department of Epidemiology, School of Public Health, Southeast University, Nanjing, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Xiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Zhou
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Congcong Chen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhoufeng Wang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Na Qin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Meng Zhu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lingfeng Bi
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Linnan Gong
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liu Yang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yingjia Chen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xianfeng Xu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Juncheng Dai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongxia Ma
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhibin Hu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guangfu Jin
- Department of Epidemiology, School of Public Health, Southeast University, Nanjing, Jiangsu, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Hongbing Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
- Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
2
|
Shen Y, Chen JQ, Li XP. Differences between lung adenocarcinoma and lung squamous cell carcinoma: Driver genes, therapeutic targets, and clinical efficacy. Genes Dis 2025; 12:101374. [PMID: 40083325 PMCID: PMC11904499 DOI: 10.1016/j.gendis.2024.101374] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/15/2024] [Accepted: 06/22/2024] [Indexed: 03/16/2025] Open
Abstract
With the rapid advancements in second-generation gene sequencing technologies, a growing number of driver genes and associated therapeutic targets have been unveiled for lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC). While they are clinically classified as non-small cell lung cancer (NSCLC), they display distinct genomic features and substantial variations in clinical efficacy, underscoring the need for particular attention. Hence, this review provides a comprehensive overview of the latest advancements in driver genes, epigenetic targets, chemotherapy, targeted therapy, and immunotherapy for LUAD and LUSC. Additionally, it delves into the distinctions in signaling pathways and pivotal facets of clinical management specific to these two categories of lung cancer. Moreover, we furnish pertinent details regarding clinical trials pertaining to driver genes and epigenetics, thus establishing a theoretical foundation for the realization of precision treatments for LUAD and LUSC.
Collapse
Affiliation(s)
- Yue Shen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jie-Qi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiang-Ping Li
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
3
|
Brumage L, Best S, Hippe DS, Grunblatt E, Chanana P, Wu F, Lee MC, Ying Z, Ibrahim A, Chung JH, Vigil A, Fatherree J, Beronja S, Paddison P, Sullivan L, Nabet B, MacPherson D. In vivo functional screens reveal KEAP1 loss as a driver of chemoresistance in small cell lung cancer. SCIENCE ADVANCES 2025; 11:eadq7084. [PMID: 40267200 PMCID: PMC12017300 DOI: 10.1126/sciadv.adq7084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 03/18/2025] [Indexed: 04/25/2025]
Abstract
Exquisitely chemosensitive initially, small cell lung cancer (SCLC) exhibits dismal outcomes owing to rapid transition to chemoresistance. Elucidating the genetic underpinnings has been challenging owing to limitations with cellular models. As SCLC patient-derived xenograft (PDX) models mimic therapeutic responses, we perform genetic screens in chemosensitive PDX models to identify drivers of chemoresistance. cDNA overexpression screens identify MYC, MYCN, and MYCL, while CRISPR deletion screens identify KEAP1 loss as driving chemoresistance. Deletion of KEAP1 switched a chemosensitive SCLC PDX model to become chemoresistant and resulted in sensitivity to inhibition of glutamine metabolism. Data from the IMpower133 clinical trial revealed ~6% of patients with extensive-stage SCLC exhibit KEAP1 genetic alterations, with activation of a KEAP1/NRF2 transcriptional signature associated with reduced survival upon chemotherapy treatment. While roles for KEAP1/NRF2 have been unappreciated in SCLC, our genetic screens revealed KEAP1 loss as a driver of chemoresistance, while patient genomic analyses demonstrate clinical importance.
Collapse
Affiliation(s)
- Lauren Brumage
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington Seattle, Seattle, WA, USA
| | - Scott Best
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington Seattle, Seattle, WA, USA
| | - Daniel S. Hippe
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Eli Grunblatt
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Pritha Chanana
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Feinan Wu
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Zhe Ying
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ali Ibrahim
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jae Heun Chung
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Anna Vigil
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jackson Fatherree
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Slobodan Beronja
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Patrick Paddison
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lucas Sullivan
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - David MacPherson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
4
|
García-Vázquez N, González-Robles TJ, Lane E, Spasskaya D, Zhang Q, Kerzhnerman MA, Jeong Y, Collu M, Simoneschi D, Ruggles KV, Róna G, Kaisari S, Pagano M. Stabilization of GTSE1 by cyclin D1-CDK4/6-mediated phosphorylation promotes cell proliferation with implications for cancer prognosis. eLife 2025; 13:RP101075. [PMID: 40272409 PMCID: PMC12021411 DOI: 10.7554/elife.101075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025] Open
Abstract
In healthy cells, cyclin D1 is expressed during the G1 phase of the cell cycle, where it activates CDK4 and CDK6. Its dysregulation is a well-established oncogenic driver in numerous human cancers. The cancer-related function of cyclin D1 has been primarily studied by focusing on the phosphorylation of the retinoblastoma (RB) gene product. Here, using an integrative approach combining bioinformatic analyses and biochemical experiments, we show that GTSE1 (G-Two and S phases expressed protein 1), a protein positively regulating cell cycle progression, is a previously unrecognized substrate of cyclin D1-CDK4/6 in tumor cells overexpressing cyclin D1 during G1 and subsequent phases. The phosphorylation of GTSE1 mediated by cyclin D1-CDK4/6 inhibits GTSE1 degradation, leading to high levels of GTSE1 across all cell cycle phases. Functionally, the phosphorylation of GTSE1 promotes cellular proliferation and is associated with poor prognosis within a pan-cancer cohort. Our findings provide insights into cyclin D1's role in cell cycle control and oncogenesis beyond RB phosphorylation.
Collapse
Affiliation(s)
- Nelson García-Vázquez
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Tania J González-Robles
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
- Department of Medicine, New York University Grossman School of MedicineNew YorkUnited States
- Howard Hughes Medical Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Ethan Lane
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Daria Spasskaya
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Qingyue Zhang
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Marc A Kerzhnerman
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - YeonTae Jeong
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Marta Collu
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Daniele Simoneschi
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Kelly V Ruggles
- Department of Medicine, New York University Grossman School of MedicineNew YorkUnited States
| | - Gergely Róna
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
- Howard Hughes Medical Institute, New York University Grossman School of MedicineNew YorkUnited States
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural SciencesBudapestHungary
| | - Sharon Kaisari
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
- Howard Hughes Medical Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
- Howard Hughes Medical Institute, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
5
|
Mine H, Sugimoto K, Kobayashi M, Takagi H, Okabe N, Muto S, Kobayashi Y, Hashimoto Y, Suzuki H, Chiba H. Abnormal phosphorylation of human LRH1 at Ser510 predicts poor prognosis and promotes cell viability in lung squamous cell carcinoma. BMC Cancer 2025; 25:764. [PMID: 40269773 PMCID: PMC12016208 DOI: 10.1186/s12885-025-14160-6] [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: 07/23/2024] [Accepted: 04/15/2025] [Indexed: 04/25/2025] Open
Abstract
The nuclear receptor liver receptor homolog 1 (LRH1)/NR5A2 is aberrantly expressed in diverse cancer types, including liver and lung cancers. Since we previously showed that excessive phosphorylation of human LRH1 at S510 (hLRH1pS510-high) is predictable of hepatocellular carcinoma recurrence, we here clarified the clinicopathological and biological significance of hLRH1pS510-high in lung cancer. By immunohistochemistry using an anti-hLRH1pS510 monoclonal antibody, we evaluated the hLRH1pS510 signals in 151 and 150 cases of lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) tissues, respectively, and performed clinicopathological analysis. hLRH1pS510 was localized in the nucleus of tumor cells in LUAD and LUSC tissues with different intensity and proportions among the patients. Of note, the strong hLRH1pS510 signal was occasionally detectable in LUAD and LUSC cells at the expanding tumor edges. A semi-quantitative analysis revealed that 28 (18.4%) and 36 (24.0%) of LUAD and LUSC cases, respectively, exhibited hLRH1pS510-high. Kaplan-Meier plots showed significant differences in the disease-free survival (DFS) between the hLRH1pS510-high and hLRH1pS510-low groups in LUSC, but not in LUAD patients. hLRH1pS510-high was also significantly correlated with recurrence in LUSC patients. Additionally, by multivariate analysis, hLRH1pS510-high represented an independent biomarker for the DFS of LUSC patients. Furthermore, the impact of hLRH1pS510 on the viability of LUSC cells was evaluated by comparing phenotypes among two distinct LUSC cell lines expressing wild-type LRH1, LRH1S510A, and LRH1S510E. Consequently, we demonstrated that phosphorylation of hLRH1S510 accelerates the viability of LUSC cells. Thus, hLRH1pS510 is attractive not only as the predictive biomarker for LUSC but also as the potential therapeutic target.
Collapse
Affiliation(s)
- Hayato Mine
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, 960- 1295, Japan
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima, 960- 1295, Japan
| | - Kotaro Sugimoto
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, 960- 1295, Japan.
| | - Makoto Kobayashi
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, 960- 1295, Japan
| | - Hironori Takagi
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima, 960- 1295, Japan
| | - Naoyuki Okabe
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima, 960- 1295, Japan
| | - Satoshi Muto
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima, 960- 1295, Japan
| | - Yasuyuki Kobayashi
- Department of Diagnostic Pathology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Yuko Hashimoto
- Department of Diagnostic Pathology, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Hiroyuki Suzuki
- Department of Chest Surgery, Fukushima Medical University School of Medicine, Fukushima, 960- 1295, Japan
| | - Hideki Chiba
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, 960- 1295, Japan.
| |
Collapse
|
6
|
Bipasha M, Deepali V, Prabal D, Supriya K, Megha B. Ferroptosis: A Mechanism of Cell Death With Potential Scope in Cancer Therapy. Asia Pac J Clin Oncol 2025. [PMID: 40235436 DOI: 10.1111/ajco.14172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/30/2024] [Accepted: 04/02/2025] [Indexed: 04/17/2025]
Abstract
Ferroptosis is a type of regulated cell death caused by oxidative imbalance of the intracellular microenvironment. This causes the accumulation of toxic lipid peroxides, depicted by iron overload and lipid peroxidation, which results in disease development. The affected cell population displays unique morphological and biochemical features, which are distinct from other modes of cell death, like apoptosis, pyroptosis, and necroptosis. The individual pathways of each of these modes are interrelated and tend to counterbalance each other in the mechanism of cell death. The process of ferroptosis is associated with disturbances in iron metabolism, in conjunction with glutathione peroxidase and lipid peroxidation, culminating in a reduction of antioxidant capacity and accumulation of lipid peroxides in the dying cell. It has been observed that even excess cellular levels of iron can cause cell death, where ferroptosis is initiated by diminishing the levels of glutathione and glutathione peroxidase 4, and thus leading to excess build-up of lipid reactive oxygen species (ROS). In the case of a neoplastic cell, ferroptosis along with its regulators tends to orchestrate cell death and also affects cancer progression by modulation of proliferation activity, apoptosis suppression, metastasis, and drug resistance. Comprehending the complex network of molecular processes implicated in ferroptosis regulation is vital for developing targeted therapies for diseases where ferroptosis plays a significant role.
Collapse
Affiliation(s)
- Mukherjee Bipasha
- Department of Biochemistry, Dr DY Patil Medical College, Navi Mumbai, India
| | - Vidhate Deepali
- Department of Biochemistry, Dr DY Patil Medical College, Navi Mumbai, India
| | - Deb Prabal
- Sultan Qaboos Comprehensive Cancer Care & Research Centre, University Medical City, Muscat, Sultanate of Oman
| | - Khillare Supriya
- Department of Biochemistry, Dr DY Patil Medical College, Navi Mumbai, India
| | - Bangar Megha
- Department of Biochemistry, Dr DY Patil Medical College, Navi Mumbai, India
| |
Collapse
|
7
|
Tawfiq RK, de Camargo Correia GS, Li S, Zhao Y, Lou Y, Manochakian R. Targeting Lung Cancer with Precision: The ADC Therapeutic Revolution. Curr Oncol Rep 2025:10.1007/s11912-025-01655-5. [PMID: 40238068 DOI: 10.1007/s11912-025-01655-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2025] [Indexed: 04/18/2025]
Abstract
PURPOSE OF REVIEW This review explores the evolving role of antibody-drug conjugates (ADCs) in lung cancer treatment, with a focus on their application in non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). It highlights advancements in ADC design, mechanisms of action, and key outcomes from recent clinical trials. RECENT FINDINGS ADCs have introduced a new level of precision in oncology by targeting tumor-specific antigens such as HER2, HER3, and TROP-2. Recent clinical trials of agents like trastuzumab deruxtecan, datopotamab deruxtecan, and sacituzumab govitecan have demonstrated meaningful objective response rates and manageable toxicity, offering hope for patients with advanced NSCLC and SCLC. ADCs are transforming the treatment landscape for lung cancer, offering a blend of targeted delivery and potent therapeutic effects. With ongoing efforts to improve safety, efficacy, and antigen targeting, ADCs have the potential to become a cornerstone of lung cancer therapy and pave the way for innovative multimodal approaches in the future.
Collapse
Affiliation(s)
- Reema Kamal Tawfiq
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic Florida, 4500 San Pablo Rd S, Jacksonville, FL, 32224, USA
| | - Guilherme Sacchi de Camargo Correia
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic Florida, 4500 San Pablo Rd S, Jacksonville, FL, 32224, USA
| | - Shenduo Li
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic Florida, 4500 San Pablo Rd S, Jacksonville, FL, 32224, USA
| | - Yujie Zhao
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic Florida, 4500 San Pablo Rd S, Jacksonville, FL, 32224, USA
| | - Yanyan Lou
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic Florida, 4500 San Pablo Rd S, Jacksonville, FL, 32224, USA
| | - Rami Manochakian
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic Florida, 4500 San Pablo Rd S, Jacksonville, FL, 32224, USA.
| |
Collapse
|
8
|
Kuo P, Elboudwarej E, Zavodovskaya M, Lin KW, Lee CV, Diehl L, Patel J, Mekan S, Jürgensmeier JM. Trop-2 expression in non-small cell lung cancer. PLoS One 2025; 20:e0321555. [PMID: 40233061 PMCID: PMC11999141 DOI: 10.1371/journal.pone.0321555] [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: 10/14/2024] [Accepted: 03/07/2025] [Indexed: 04/17/2025] Open
Abstract
Trophoblast cell-surface antigen 2 (Trop-2) is highly expressed in non-small cell lung cancer (NSCLC) and has become an attractive target for antibody-drug conjugates (ADCs). ADC tumor target expression is essential in investigating the predictive value of Trop-2 and Trop-2 ADC efficacy. Although Trop-2 mRNA expression in NSCLC has been described, protein-level expression is poorly understood. We investigated Trop-2 expression landscape across multiple data and sample sets to characterize mRNA expression and address the gap in protein-expression profiling. Trop-2 expression was analyzed using available mRNA, mutation, and protein data in three datasets: (1) The Cancer Genome Atlas (TCGA) included clinical-pathological and survival data in NSCLC adenocarcinoma and squamous cell carcinoma; (2) sample set 1 (adenocarcinoma) and (3) sample set 2 (adenocarcinoma, squamous cell carcinoma) underwent sequencing and immunohistochemistry for Trop-2 RNA, protein (Robust Prototype Assay, SP295 clone) and mutation analysis. Trop-2 was highly expressed in NSCLC and expression was similar in adenocarcinoma and squamous cell carcinoma and across baseline characteristics including patient age, sex, and tumor stage. Trop-2 expression was not associated with clinically relevant genetic alterations. Trop-2 was not a prognostic factor in NSCLC (TCGA survival data). High Trop-2 expression in NSCLC was independent of evaluated baseline characteristics, histology, and driver alterations. Trop-2 protein expression at any level was observed in 82% to 90% of NSCLC across sample sets; similar proportions of adenocarcinoma and squamous cell carcinoma expressed Trop-2. These data support broad Trop-2 ADC use in NSCLC.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Female
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Male
- Gene Expression Regulation, Neoplastic
- Middle Aged
- Aged
- Mutation
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
Collapse
Affiliation(s)
- Peiwen Kuo
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Emon Elboudwarej
- Gilead Sciences, Inc., Foster City, California, United States of America
| | | | - Kai-Wen Lin
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Chingwei V. Lee
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Lauri Diehl
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Jilpa Patel
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Sabeen Mekan
- Gilead Sciences, Inc., Morris Plains, New Jersey, United States of America
| | | |
Collapse
|
9
|
Mellor P, Kendall S, Hammond SA, Plett R, Kyrylenko L, Saxena A, Anderson DH. Expression of CREB3L1 blocks key cancer pathways and suppresses metastasis of lung squamous cell carcinoma cells. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167845. [PMID: 40228676 DOI: 10.1016/j.bbadis.2025.167845] [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: 01/06/2025] [Revised: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
Lung cancer is the leading cause of death due to cancer, with higher mortality rates than cancers of the colon, breast and prostate combined. About one quarter of lung cancers are lung squamous cell carcinomas (LUSC), with a five-year survival rate of only 16 %. We discovered that the majority of LUSCs have reduced expression of a key transcription factor CREB3L1 (cAMP responsive element binding protein 3 like 1), known to function as a metastasis suppressor in breast, bladder and ovarian cancers. In this report, we set out to determine if CREB3L1 functions as a metastasis suppressor in LUSCs. A differential gene expression analysis showed that ectopic expression of CREB3L1 in NCI-H2170 and NCI-1703 cells caused significant reductions in many signaling pathway genes involved in promoting cell viability, survival, migration and angiogenesis. Expression of CREB3L1 was able to reduce cell migration and anchorage-independent growth in soft agar in NCI-H2170, NCI-H1703 and NCI-H226 LUSC cells. Expression of CREB3L1 had less impact on the growth of primary xenograft tumors for NCI-H2170 and NCI-H1703 cells, the latter of which formed atypical masses filled with blood. In contrast, xenografts of NCI-H226 expressing CREB3L1 showed significant reductions in primary tumor growth. Finally, in a mouse metastasis assay, expression of CREB3L1 in NCI-H2170 cells significantly reduced the formation of liver metastases and in NCI-H226 cells, lung metastases, as compared to their respective CREB3L1-deficient parental LUSC cells. Taken together, these results strongly support a role for CREB3L1 as a metastasis suppressor in lung squamous cell carcinoma cells.
Collapse
Affiliation(s)
- Paul Mellor
- Cancer Research Group, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Stephanie Kendall
- Cancer Research Group, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - S Austin Hammond
- Cancer Research Group, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Riley Plett
- Cancer Research Group, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Liliia Kyrylenko
- Cancer Research Group, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Anurag Saxena
- Department of Pathology and Lab Medicine, Royal University Hospital, 2841 - 103 Hospital Drive, Saskatoon, SK S7N 0W8, Canada
| | - Deborah H Anderson
- Cancer Research Group, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada; Department of Oncology, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada; Discovery and Translational Research, Saskatchewan Cancer Agency, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5E5, Canada.
| |
Collapse
|
10
|
Nandi D, Janardhanan R, Hannenhalli S, Agrawal P. Molecular signatures bidirectionally link myocardial infarction and lung cancer. Front Med (Lausanne) 2025; 12:1576375. [PMID: 40270498 PMCID: PMC12014433 DOI: 10.3389/fmed.2025.1576375] [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: 02/14/2025] [Accepted: 03/21/2025] [Indexed: 04/25/2025] Open
Abstract
Myocardial Infarction (MI) and lung cancers are major contributors to mortality worldwide. While seemingly diverse, the two share common risk factors, such as smoking and hypertension. There is a pressing need to identify bidirectional molecular signatures that link MI and lung cancer, in order to improve clinical outcomes for patients. In this study, we identified common differentially expressed genes between MI and lung cancer. Specifically, we identified 1,496 upregulated and 1,482 downregulated genes in the MI datasets. By focusing on the 1,000 most upregulated and downregulated genes in Lung Adenocarcinoma (LUAD) and Lung Squamous Cell Carcinoma (LUSC), we identified 35 genes that are common across MI, LUAD, and LUSC. Functional enrichment analysis revealed shared biological processes, such as "inflammatory response" and "cell differentiation." The Cox proportional hazards model demonstrated a significant association between the shared genes and overall survival in lung cancer patients, as well as with smoking history in these patients. In addition, a machine learning model based on the expression of the shared genes distinguished MI patients from controls, achieving an AUROC of 0.72 and an AUPRC of 0.86. Finally, based on drug repurposing analysis, we proposed FDA-approved drugs potentially targeting the upregulated genes as novel therapeutic options for the co-occurring conditions of MI and lung cancer. Overall, our findings highlight the similarities in molecular makeup between lung cancer and MI.
Collapse
Affiliation(s)
- Dhruva Nandi
- Division of Medical Research, SRM Medical College Hospital & Research Centre, SRMIST, Kattankulathur, Chennai, Tamil Nadu, India
| | - Rajiv Janardhanan
- Division of Medical Research, SRM Medical College Hospital & Research Centre, SRMIST, Kattankulathur, Chennai, Tamil Nadu, India
| | | | - Piyush Agrawal
- Division of Medical Research, SRM Medical College Hospital & Research Centre, SRMIST, Kattankulathur, Chennai, Tamil Nadu, India
| |
Collapse
|
11
|
Preda AC, Todor N, Cârlan B, Kubelac-Varro AD, Iancu DI, Mocan C, Vasilica MB, Kubelac MP, Vlad C, Ciuleanu TE. Prospective Upfront Next-Generation Sequencing for Advanced Non-Small Cell Lung Cancer: Real-World Outcomes from the Ion Chiricuță Oncology Institute. Int J Mol Sci 2025; 26:3403. [PMID: 40244261 PMCID: PMC11989902 DOI: 10.3390/ijms26073403] [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: 03/16/2025] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Upfront Next-Generation Sequencing (NGS) is increasingly recommended in advanced NSCLC to guide targeted therapy. This prospective single-center study in Romania evaluated routine, upfront NGS in advanced NSCLC at baseline (tissue and/or liquid) and progression (liquid). Baseline FoundationOne NGS (tissue/liquid) was performed in 119 consecutive stage IV NSCLC patients, along with PD-L1 immunohistochemistry (IHC, SP263). Liquid biopsy was repeated at progression. Turnaround time (TAT), the prevalence of actionable targets, and clinical utility were assessed. Patients were predominantly male (68.1%) with a median age of 62 years (range 30-86). Most had ECOG PS 0-1 (79%) and non-squamous histology (67.2%). Never-smokers accounted for 25.2%. The median TAT for the NGS results was 9 days (range 5-21). Overall, 671 genetic alterations were detected in 149 genes. The mean number of distinct mutations per patient dropped from 5.6 at baseline to 4.3 at progression. Tissue samples yielded more alterations (6 per patient) than baseline liquid biopsies (4.6). Squamous tumors had more alterations (7.1 vs. 4.8 in non-squamous), and the number of smokers exceeded that of never-smokers (6 vs. 4.5). TP53 was the most frequent (70.59%). Actionable variants were found in 74.8% of patients, though only 35.3% received personalized therapy, largely due to performance status deterioration, reimbursement, or trial availability barriers. Common targets in non-squamous tumors included EGFR (21%), KRAS G12C (11%), NF1 (11%), and ERBB2 (6%); in squamous tumors, common targets included NF1 (24%), PIK3CA (18%), and ERBB2 (8%). Among smokers, driver mutations were often NF1 (15%), PIK3CA (11%), KRAS G12C (9%), and ERBB2 (8%); never-smokers were dominated by EGFR (45%), NF1 (15%), and KRAS G12C (8%). TMB ≥ 10 mut/Mb was seen in 26.9%; no patients were MSI-H. PD-L1 TPS was <1% in 33% of patients, 1-49% in 20%, ≥50% in 18%, and unknown in 29%. Upfront NGS offers rapid, comprehensive genomic data, guiding tailored therapies and trials in advanced NSCLC. Liquid rebiopsy at progression further refines treatment decisions.
Collapse
Affiliation(s)
- Alexandra Cristina Preda
- Oncology Institute “Prof. Dr. Ion Chiricuță” 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
- Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania
| | - Nicolae Todor
- Oncology Institute “Prof. Dr. Ion Chiricuță” 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Bogdan Cârlan
- Medlife Oncology Hospital, 65A Carierei Street, 500062 Brașov, Romania
| | - Adelina-Dadiana Kubelac-Varro
- Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania
| | - Dana Ioana Iancu
- Oncology Institute “Prof. Dr. Ion Chiricuță” 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Cristina Mocan
- Oncology Institute “Prof. Dr. Ion Chiricuță” 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Mariana Bandi Vasilica
- Oncology Institute “Prof. Dr. Ion Chiricuță” 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Milan-Paul Kubelac
- Oncology Institute “Prof. Dr. Ion Chiricuță” 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
- STAR Institute, Babeș-Bolyai University, 1 Mihail Kogălniceanu Street, 400347 Cluj-Napoca, Romania
| | - Cătălin Vlad
- Oncology Institute “Prof. Dr. Ion Chiricuță” 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
- Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 8 Victor Babeș Street, 400012 Cluj-Napoca, Romania
| | - Tudor Eliade Ciuleanu
- Oncology Institute “Prof. Dr. Ion Chiricuță” 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| |
Collapse
|
12
|
Pérez Malla CU, Kalla J, Tiefenbacher A, Wasinger G, Kluge K, Egger G, Sheibani-Tezerji R. Goistrat: gene-of-interest-based sample stratification for the evaluation of functional differences. BMC Bioinformatics 2025; 26:97. [PMID: 40188042 PMCID: PMC11971790 DOI: 10.1186/s12859-025-06109-0] [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] [Received: 03/11/2024] [Accepted: 03/10/2025] [Indexed: 04/07/2025] Open
Abstract
PURPOSE Understanding the impact of gene expression in pathological processes, such as carcinogenesis, is crucial for understanding the biology of cancer and advancing personalised medicine. Yet, current methods lack biologically-informed-omics approaches to stratify cancer patients effectively, limiting our ability to dissect the underlying molecular mechanisms. RESULTS To address this gap, we present a novel workflow for the stratification and further analysis of multi-omics samples with matched RNA-Seq data that relies on MSigDB curated gene sets, graph machine learning and ensemble clustering. We compared the performance of our workflow in the top 8 TCGA datasets and showed its clear superiority in separating samples for the study of biological differences. We also applied our workflow to analyse nearly a thousand prostate cancer samples, focusing on the varying expression of the FOLH1 gene, and identified specific pathways such as the PI3K-AKT-mTOR gene sets as well as signatures linked to prostate tumour aggressiveness. CONCLUSION Our comprehensive approach provides a novel tool to identify disease-relevant functions of genes of interest (GOI) in large datasets. This integrated approach offers a valuable framework for understanding the role of the expression variation of a GOI in complex diseases and for informing on targeted therapeutic strategies.
Collapse
Affiliation(s)
- Carlos Uziel Pérez Malla
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Ludwig Boltzmann Gesellschaft, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Jessica Kalla
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Andreas Tiefenbacher
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Gabriel Wasinger
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Kilian Kluge
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Christian Doppler Laboratory for Applied Metabolomics, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Ludwig Boltzmann Gesellschaft, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Raheleh Sheibani-Tezerji
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria.
- Ludwig Boltzmann Institute Applied Diagnostics, Ludwig Boltzmann Gesellschaft, Währinger Gürtel 18-20, Vienna, 1090, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria.
| |
Collapse
|
13
|
Ogden J, Sellers R, Sahoo S, Oojageer A, Chaturvedi A, Dive C, Lopez-Garcia C. A human model to deconvolve genotype-phenotype causations in lung squamous cell carcinoma. Nat Commun 2025; 16:3215. [PMID: 40185723 PMCID: PMC11971459 DOI: 10.1038/s41467-025-58343-y] [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: 02/26/2024] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
Tractable, patient-relevant models are needed to investigate cancer progression and heterogeneity. Here, we report an alternative in vitro model of lung squamous cell carcinoma (LUSC) using primary human bronchial epithelial cells (hBECs) from three healthy donors. The co-operation of ubiquitous alterations (TP53 and CDKN2A loss) and components of commonly deregulated pathways including squamous differentiation (SOX2), PI3K signalling (PTEN) and the oxidative stress response (KEAP1) is investigated by generating hBECs harbouring cumulative alterations. Our analyses confirms that SOX2-overexpression initiates early preinvasive LUSC stages, and co-operation with the oxidative stress response and PI3K pathways to drive more aggressive phenotypes, with expansion of cells expressing LUSC biomarkers and invasive properties. This cooperation is consistent with the classical LUSC subtype. Importantly, we connect pathway dysregulation with gene expression changes associated with cell-intrinsic processes and immunomodulation. Our approach constitutes a powerful system to model LUSC and unravel genotype-phenotype causations of clinical relevance.
Collapse
Affiliation(s)
- Julia Ogden
- Cancer Research UK Manchester Institute, Wilmslow Road, M20 4BX, Manchester, United Kingdom
| | - Robert Sellers
- Cancer Research UK Manchester Institute, Wilmslow Road, M20 4BX, Manchester, United Kingdom
| | - Sudhakar Sahoo
- Cancer Research UK Manchester Institute, Wilmslow Road, M20 4BX, Manchester, United Kingdom
| | - Anthony Oojageer
- Cancer Research UK Manchester Institute, Wilmslow Road, M20 4BX, Manchester, United Kingdom
| | - Anshuman Chaturvedi
- Department of Histopathology, The Christie Hospital, Wilmslow Road, Manchester, M20 4BX, United Kingdom
| | - Caroline Dive
- Cancer Research UK Manchester Institute, Wilmslow Road, M20 4BX, Manchester, United Kingdom
- Cancer Research UK, National Biomarker Centre, Wilmslow Road, M20 4BX, Manchester, United Kingdom
- Cancer Research UK Lung Cancer Centre of Excellence, Wilmslow Road, M20 4BX, Manchester, United Kingdom
| | - Carlos Lopez-Garcia
- Cancer Research UK Manchester Institute, Wilmslow Road, M20 4BX, Manchester, United Kingdom.
- Cancer Research UK Lung Cancer Centre of Excellence, Wilmslow Road, M20 4BX, Manchester, United Kingdom.
| |
Collapse
|
14
|
Chen YC, Bazewicz CG, Dinavahi SS, Huntington ND, Schell TD, Robertson GP. Emerging Role of the p53 Pathway in Modulating NK Cell-Mediated Immunity. Mol Cancer Ther 2025; 24:523-535. [PMID: 39470047 DOI: 10.1158/1535-7163.mct-24-0325] [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: 05/10/2024] [Revised: 09/03/2024] [Accepted: 10/25/2024] [Indexed: 10/30/2024]
Abstract
The p53 pathway plays an important role in role in cancer immunity. Mutation or downregulation of the proteins in the p53 pathway are prevalent in many cancers, contributing to tumor progression and immune dysregulation. Recent findings suggest that the activity of p53 within tumor cells, immune cells, and the tumor microenvironment can play an important role in modulating NK cell-mediated immunity. Consequently, efforts to restore p53 pathway activity are being actively pursued to modulate this form of immunity. This review focuses on p53 activity regulating the infiltration and activation of NK cells in the tumor immune microenvironment. Furthermore, the impact of p53 and its regulation of NK cells on immunogenic cell death within solid tumors and the abscopal effect are reviewed. Finally, future avenues for therapeutically restoring p53 activity to improve NK cell-mediated antitumor immunity and optimize the effectiveness of cancer therapies are discussed.
Collapse
Affiliation(s)
- Yu-Chi Chen
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Christopher G Bazewicz
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Saketh S Dinavahi
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Nicholas D Huntington
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Victoria, Australia
- oNKo-Innate Pty Ltd. Moonee Ponds, Victoria, Australia
| | - Todd D Schell
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- The Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
- Penn State Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
15
|
Feng X, Zeng R, Lyu M, Chen X, Xu Z, Hu Y, Bao Z, Sun X, Zhao J, Zhou L, Zhou J, Gao B, Dong L, Xiang Y. Clinical and molecular characteristics, therapeutic strategies, and prognosis of non-small cell lung cancer patients harboring primary and acquired BRAF mutations. Front Oncol 2025; 15:1514653. [PMID: 40242250 PMCID: PMC11999832 DOI: 10.3389/fonc.2025.1514653] [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: 10/21/2024] [Accepted: 03/14/2025] [Indexed: 04/18/2025] Open
Abstract
Background The differences in clinical characteristics and treatment prognosis in NSCLC patients harboring primary and acquired BRAF mutations are still poorly understood. Methods From Oct 2017 to Dec 2023, 10, 211 lung cancer patients at Shanghai Ruijin Hospital were reviewed. 88 primary and 15 acquired BRAF-mutated NSCLC patients resistant to EGFR TKIs were included in the study. Results Primary BRAF-mutated patients preferentially occurred in the elderly (median age: 67 vs 61, p=0.015), males (53.4% vs 26.7%, p=0.056), former/current smokers (36.5% vs 6.7%, p=0.033), non-adenocarcinoma (11.4% vs 0%, P=0.351) compared to acquired BRAF-mutated patients. Significant differences in gender (33.3% vs 62.3%, p=0.012), smoking history (22.2% vs 43.1%, p=0.063), and adenocarcinomas (100% vs 83.6%, p=0.028) were observed between primary BRAF/EGFR co-mutated and non-co-mutated groups. While primary and acquired BRAF/EGFR co-mutated patients had similar clinical characteristics, with EGFR mutations being the most common coexisting oncogene (30.7% and 93.3%). The genotype of EGFR mutations differed, with acquired BRAF-mutated cases showing more complexity and a higher rate of dual EGFR mutations (35.7%) compared to primary cases. For primary BRAF/EGFR co-mutated patients, no matter what kinds of therapies, the EGFR 19del patients had a better prognosis than non-19del patients, and the first line mPFS was NR and 9.0 months (95% CI: 7.7-10.3 months) (p=0.0062), respectively. Dabrafenib and trametinib plus 3rd EGFR TKIs improved the prognosis of primary BRAF/EGFR non-19del co-mutated patients, achieving ORR and mPFS of 100% (3/3) and 12 months. For acquired co-mutated patients, the mPFS for 5 patients was 8.6 months (95% CI: 5.4-11.8 months). No new safety concerns and > grade 3 AEs were noted. Conclusion Together, our study demonstrates that primary and acquired BRAF-mutant patients show distinct differences in some clinical and molecular characteristics, but acquired BRAF/EGFR co-mutated and primary BRAF/EGFR non-19del co-mutated patients may both respond to triple-targeted therapy.
Collapse
Affiliation(s)
- Xiangran Feng
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ran Zeng
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengchen Lyu
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Chen
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziwei Xu
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Hu
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyao Bao
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis, and Treatment of Respiratory Infectious Diseases, Shanghai, China
| | - Xianwen Sun
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis, and Treatment of Respiratory Infectious Diseases, Shanghai, China
| | - Jingya Zhao
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis, and Treatment of Respiratory Infectious Diseases, Shanghai, China
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis, and Treatment of Respiratory Infectious Diseases, Shanghai, China
| | - Jun Zhou
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis, and Treatment of Respiratory Infectious Diseases, Shanghai, China
| | - Beili Gao
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis, and Treatment of Respiratory Infectious Diseases, Shanghai, China
| | - Lei Dong
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Xiang
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis, and Treatment of Respiratory Infectious Diseases, Shanghai, China
| |
Collapse
|
16
|
Xiao‐ru M, Xiao‐Xiong S, Qian G, Ya‐jing Q, Li‐li H, Yan‐Yan G, Peng‐peng X, Guan‐nan M, Gui‐bing R. A Case of Lung Squamous Cell Carcinoma Harboring TP53 Mutation and PLPP5-FGFR1 Fusion Gene. THE CLINICAL RESPIRATORY JOURNAL 2025; 19:e70074. [PMID: 40242904 PMCID: PMC12004084 DOI: 10.1111/crj.70074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025]
Abstract
Lung squamous cell carcinoma (LUSC) is one of the most common subtype of lung cancer and is associated with the poor prognoses. The fibroblast growth factor receptor (FGFR) family is known to be activated through fusions with various partners across multiple cancer types, including nonsmall cell lung cancer (NSCLC). FGFR inhibitors are currently undergoing clinical evaluation for the treatment of tumors harboring these fusions. While FGFR1 amplification has been well-documented in numerous NSCLC datasets, the characterization of specific FGFR fusion variants remains limited. In this study, we identified a novel PLPP5-FGFR1 fusion in a 65-year-old male patient with lung squamous cell carcinoma through targeted RNA sequencing. The fusion junction was located between exon 1 of PLPP5 and exon 5 of FGFR1, and the result was validated by Sanger sequencing. To our knowledge, this is the first reported case of a PLPP5-FGFR1 fusion coexisting with a TP53 mutation in LUSC. These findings broaden the spectrum of potential translocation partners in FGFR1 fusions, and the clinical implications of this novel fusion on treatment outcomes and prognosis warrant further investigation and long-term follow-up.
Collapse
Affiliation(s)
- Meng Xiao‐ru
- Oncology DepartmentCharacteristic Medical Center of Chinese People's Armed Police ForceTianjinChina
| | - Shi Xiao‐Xiong
- Oncology DepartmentCharacteristic Medical Center of Chinese People's Armed Police ForceTianjinChina
| | - Gao Qian
- Medical Research Center Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang ProvinceHangzhouChina
| | - Qu Ya‐jing
- Oncology DepartmentCharacteristic Medical Center of Chinese People's Armed Police ForceTianjinChina
| | - Huo Li‐li
- Oncology DepartmentCharacteristic Medical Center of Chinese People's Armed Police ForceTianjinChina
| | - Gao Yan‐Yan
- Oncology DepartmentCharacteristic Medical Center of Chinese People's Armed Police ForceTianjinChina
| | - Xu Peng‐peng
- Oncology DepartmentCharacteristic Medical Center of Chinese People's Armed Police ForceTianjinChina
| | - Ma Guan‐nan
- Medical Research Center Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang ProvinceHangzhouChina
| | - Ren Gui‐bing
- Oncology DepartmentCharacteristic Medical Center of Chinese People's Armed Police ForceTianjinChina
| |
Collapse
|
17
|
Scalora N, DeWane G, Drebot Y, Khan AA, Sinha S, Ghosh K, Robinson D, Cogswell P, Bellizzi AM, Snow AN, Breheny P, Chimenti MS, Tanas MR. EHE cell cultures: a platform for mechanistic and therapeutic investigation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.644191. [PMID: 40196670 PMCID: PMC11974726 DOI: 10.1101/2025.03.24.644191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Epithelioid hemangioendothelioma (EHE) is a difficult to treat vascular sarcoma defined by TAZ- CAMTA1 or YAP-TFE3 fusion proteins. Human cell lines needed to further understand the pathogenesis of EHE have been lacking. Herein, we describe a method to generate EHE extended primary cell cultures. An integrated multi -omic and functional approach was used to characterize these cultures. The cell cultures, relatively homogenous by single cell RNA-Seq, demonstrated established characteristics of EHE including increased proliferation, anchorage independent growth, as well as the overall gene expression profile and secondary genetic alterations seen in EHE. Whole genome sequencing (WGS) identified links to epigenetic modifying complexes, metabolic processes, and pointed to the importance of the extracellular matrix (ECM) in these tumors. Bulk RNA-Seq demonstrated upregulation of pathways including PI3K-Akt signaling, ECM/ECM receptor interaction, and the Hippo signaling pathway. Development of these extended primary cell cultures allowed for single-cell profiling which demonstrated different cell compartments within the cultures. Furthermore, the cultures served as a therapeutic platform to test the efficacy of TEAD inhibitors in vitro . Overall, the development of EHE primary cell cultures will aid in the mechanistic understanding of this sarcoma and serve as a model system to test new therapeutic approaches.
Collapse
|
18
|
Berry D, Moldoveanu D, Rajkumar S, Lajoie M, Lin T, Tchelougou D, Sakthivel S, Sharon I, Bernard A, Pelletier S, Ripstein Y, Spatz A, Miller WH, Jamal R, Lapointe R, Mes-Masson AM, Petrecca K, Meguerditchian AN, Richardson K, Wang B, Chergui M, Guiot MC, Watters K, Stagg J, Schmeing TM, Rodier F, Turcotte S, Mihalcioiu C, Meterissian S, Watson IR. The NF1 tumor suppressor regulates PD-L1 and immune evasion in melanoma. Cell Rep 2025; 44:115365. [PMID: 40023845 DOI: 10.1016/j.celrep.2025.115365] [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: 10/20/2023] [Revised: 11/24/2024] [Accepted: 02/06/2025] [Indexed: 03/04/2025] Open
Abstract
Hotspot BRAF, hotspot NRAS, and NF1 loss-of-function mutations are found in approximately 50%, 25%, and 15% of cutaneous melanomas, respectively. Compared to mutant BRAF and NRAS, the role of NF1 loss in melanoma remains understudied. NF1 has a RAS GTPase-activating protein (GAP) function; however, studies also support NF1 RAS-independent tumor-suppressor functions. Recent reports indicate that patients with NF1 mutant melanoma have high response rates to anti-PD-1 immune checkpoint inhibitors (ICIs) for reasons that are not entirely clear. Here, we present data demonstrating that NF1 interacts with PD-L1. Furthermore, NF1 loss in melanoma lines increases PD-L1 cell surface expression through a RAS-GAP-independent mechanism. Co-culture experiments demonstrate that NF1 depletion in melanoma increases resistance to T cell killing, which can be abrogated with anti-PD-1/PD-L1 ICIs. These results support a model whereby NF1 loss leads to immune evasion through the PD-L1/PD-1 axis, providing support for the examination of anti-PD-1 therapies in other NF1 mutant cancers.
Collapse
Affiliation(s)
- Diana Berry
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Dan Moldoveanu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Shivshankari Rajkumar
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Mathieu Lajoie
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada
| | - Tiffany Lin
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Daméhan Tchelougou
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Samridhi Sakthivel
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Itai Sharon
- Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Antoine Bernard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Sandy Pelletier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Yael Ripstein
- Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Alan Spatz
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada; Lady Davis Institute, McGill University, Montréal, QC H3T 1E1, Canada
| | - Wilson H Miller
- Lady Davis Institute, McGill University, Montréal, QC H3T 1E1, Canada
| | - Rahima Jamal
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Réjean Lapointe
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Montreal Neurological Institute and Hospital, Montréal, QC H3A 2B4, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Kevin Petrecca
- Montreal Neurological Institute and Hospital, Montréal, QC H3A 2B4, Canada
| | | | | | - Beatrice Wang
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - May Chergui
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | | | - Kevin Watters
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - T Martin Schmeing
- Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Francis Rodier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Simon Turcotte
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | | | | | - Ian R Watson
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada; McGill University Health Centre, Montréal, QC H4A 3J1, Canada.
| |
Collapse
|
19
|
Xu L, Ma X, Yang Y, Ding Z, Luo Y. Successful treatment of an elderly patient with lung squamous cell carcinoma by tislelizumab and chemotherapy: a case report with novel imaging findings. Front Immunol 2025; 16:1543114. [PMID: 40196110 PMCID: PMC11973309 DOI: 10.3389/fimmu.2025.1543114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
The advent of immunotherapy has transformed the therapeutic landscape for inoperable, locally advanced Non-Small cell lung cancer (NSCLC), particularly for lung squamous cell carcinoma (LUSC) with a predominance of negative driver genes. Based on the results of clinical trials such as KEYNOTE-042 and KEYNOTE-407, PD-1/PD-L1 inhibitors are now recognized as the standard of care for first-line or second-line treatment in many countries. Among the 17 immune checkpoint inhibitors sanctioned in China, tislelizumab, a domestically developed PD-1 inhibitor, enjoys broad application. Here, we present a case of a patient with LUSC who attained complete remission by cyst formation with the combination of tislelizumab and chemotherapy. Despite the absence of expression data for this patient, imaging studies revealed a reduction in the primary lesion size and the emergence of an uncommon cystic alteration post-treatment with sequential immunochemotherapy and tislelizumab monotherapy. As per the most recent follow-up, the lesion has vanished entirely. This outcome holds significant implications for the treatment of driver gene-negative LUSC by tislelizumab.
Collapse
Affiliation(s)
- Lufan Xu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinxin Ma
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Yang
- Nantong Tumor Hospital, Tumor Hospital Affiliated to Nantong University, Nantong, China
| | - Zhiqiang Ding
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi Luo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
20
|
Zhang J, He P, Wang W, Wang Y, Yang H, Hu Z, Song Y, Chang J, Yu B. Structure-Based Design of New LSD1/EGFR L858R/T790M Dual Inhibitors for Treating EGFR Mutant NSCLC Cancers. J Med Chem 2025; 68:5954-5972. [PMID: 40015914 DOI: 10.1021/acs.jmedchem.5c00267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Epigenetic changes, such as LSD1 dysregulation, contribute to acquired resistance in EGFR mutant NSCLCs and reduce the effectiveness of current therapeutics. To address the challenges, we herein reported the structure-based design of new LSD1/EGFR dual inhibitors, of which ZJY-54 represents the shortlisted lead compound with high potency, selectivity, and unique dual modes of action (namely irreversibly binding to EGFR but reversibly binding to LSD1). ZJY-54 effectively inhibited growth in both parent- and TKI-resistant NSCLC cells. In H1975 cells, ZJY-54 induced accumulation of H3K4me2 and H3K9me2, as well as inhibited phosphorylation of EGFR signaling. ZJY-54 showed favorable PK profiles and effectively inhibited tumor growth in the H1975 xenograft model. ZJY-54 represents the best-in-class LSD1/EGFR dual inhibitor and warrants further preclinical development for treating NSCLCs. These findings highlight the therapeutic potential of LSD1/EGFR dual inhibitors in drug-resistant cancers where EGFR and LSD1 were dysregulated.
Collapse
Affiliation(s)
- Jingya Zhang
- College of Chemistry, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Pengxing He
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenwen Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yuxing Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Han Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhaoxin Hu
- College of Chemistry, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, China
| | - Yihui Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Junbiao Chang
- College of Chemistry, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, China
| | - Bin Yu
- College of Chemistry, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
21
|
Xie X, Macknight HP, Lu AL, Chalfant CE. RNA splicing variants of the novel long non-coding RNA, CyKILR, possess divergent biological functions in non-small cell lung cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102412. [PMID: 39807365 PMCID: PMC11728077 DOI: 10.1016/j.omtn.2024.102412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 12/03/2024] [Indexed: 01/16/2025]
Abstract
The CDKN2A gene, responsible for encoding the tumor suppressors p16(INK4A) and p14(ARF), is frequently inactivated in non-small cell lung cancer (NSCLC). Herein, an uncharacterized long non-coding RNA (lncRNA) (ENSG00000267053) on chromosome 19p13.12 was found to be overexpressed in NSCLC cells with an active, wild-type CDKN2A gene. This lncRNA, named cyclin-dependent kinase inhibitor 2A-regulated lncRNA (CyKILR), also correlated with an active WT STK11 gene, which encodes the tumor suppressor, liver kinase B1. CyKILR displayed two splice variants, CyKILRa (exon 3 included) and CyKILRb (exon 3 excluded), which are cooperatively regulated by CDKN2A and STK11 as knockdown of both tumor suppressor genes was required to induce a significant loss of exon 3 inclusion in mature CyKILR RNA. CyKILRa localized to the nucleus, and its downregulation using antisense RNA oligonucleotides enhanced cellular proliferation, migration, clonogenic survival, and tumor incidence. In contrast, CyKILRb localized to the cytoplasm, and its downregulation using small interfering RNA reduced cell proliferation, migration, clonogenic survival, and tumor incidence. Transcriptomics analyses revealed the enhancement of apoptotic pathways with concomitant suppression of key cell-cycle pathways by CyKILRa demonstrating its tumor-suppressive role. CyKILRb inhibited tumor suppressor miRNAs indicating an oncogenic nature. These findings elucidate the intricate roles of lncRNAs in cell signaling and tumorigenesis.
Collapse
Affiliation(s)
- Xiujie Xie
- Department of Medicine, Division of Hematology & Oncology, University of Virginia, Charlottesville, VA 22903, USA
| | - H. Patrick Macknight
- Department of Medicine, Division of Hematology & Oncology, University of Virginia, Charlottesville, VA 22903, USA
| | - Amy L. Lu
- Department of Medicine, Division of Hematology & Oncology, University of Virginia, Charlottesville, VA 22903, USA
| | - Charles E. Chalfant
- Department of Medicine, Division of Hematology & Oncology, University of Virginia, Charlottesville, VA 22903, USA
- Department of Cell Biology, University of Virginia, Charlottesville, VA 22903, USA
- Program in Cancer Biology, University of Virginia NCI Comprehensive Cancer Center, Charlottesville, VA 22903, USA
- Research Service, Richmond Veterans Administration Medical Center, Richmond, VA 23298, USA
| |
Collapse
|
22
|
Trelford CB, Shepherd TG. Insights into targeting LKB1 in tumorigenesis. Genes Dis 2025; 12:101402. [PMID: 39735555 PMCID: PMC11681833 DOI: 10.1016/j.gendis.2024.101402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/03/2024] [Accepted: 06/22/2024] [Indexed: 12/31/2024] Open
Abstract
Genetic alterations to serine-threonine kinase 11 (STK11) have been implicated in Peutz-Jeghers syndrome and tumorigenesis. Further exploration of the context-specific roles of liver kinase B1 (LKB1; encoded by STK11) observed that it regulates AMP-activated protein kinase (AMPK) and AMPK-related kinases. Given that both migration and proliferation are enhanced with the loss of LKB1 activity combined with the prevalence of STK11 genetic alterations in cancer biopsies, LKB1 was marked as a tumor suppressor. However, the role of LKB1 in tumorigenesis is paradoxical as LKB1 activates autophagy and reactive oxygen species scavenging while dampening anoikis, which contribute to cancer cell survival. Due to the pro-tumorigenic properties of LKB1, targeting LKB1 pathways is now relevant for cancer treatment. With the recent successes of targeting LKB1 signaling in research and clinical settings, and enhanced cytotoxicity of chemical compounds in LKB1-deficient tumors, there is now a need for LKB1 inhibitors. However, validating LKB1 inhibitors is challenging as LKB1 adaptor proteins, nucleocytoplasmic shuttling, and splice variants all manipulate LKB1 activity. Furthermore, STE-20-related kinase adaptor protein (STRAD) and mouse protein 25 dictate LKB1 cellular localization and kinase activity. For these reasons, prior to assessing the efficacy and potency of pharmacological candidates, the functional status of LKB1 needs to be defined. Therefore, to improve the understanding of LKB1 in physiology and oncology, this review highlights the role of LKB1 in tumorigenesis and addresses the therapeutic relevancy of LKB1 inhibitors.
Collapse
Affiliation(s)
- Charles B. Trelford
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON N6A 4L6, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Trevor G. Shepherd
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON N6A 4L6, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
23
|
Hu GS, Zheng ZZ, He YH, Wang DC, Nie RC, Liu W. Integrated Analysis of Proteome and Transcriptome Profiling Reveals Pan-Cancer-Associated Pathways and Molecular Biomarkers. Mol Cell Proteomics 2025; 24:100919. [PMID: 39884577 PMCID: PMC11907456 DOI: 10.1016/j.mcpro.2025.100919] [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: 06/12/2024] [Revised: 01/02/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025] Open
Abstract
Understanding dysregulated genes and pathways in cancer is critical for precision oncology. Integrating mass spectrometry-based proteomic data with transcriptomic data presents unique opportunities for systematic analyses of dysregulated genes and pathways in pan-cancer. Here, we compiled a comprehensive set of datasets, encompassing proteomic data from 2404 samples and transcriptomic data from 7752 samples across 13 cancer types. Comparisons between normal or adjacent normal tissues and tumor tissues identified several dysregulated pathways including mRNA splicing, interferon pathway, fatty acid metabolism, and complement coagulation cascade in pan-cancer. Additionally, pan-cancer upregulated and downregulated genes (PCUGs and PCDGs) were also identified. Notably, RRM2 and ADH1B, two genes which belong to PCUGs and PCDGs, respectively, were identified as robust pan-cancer diagnostic biomarkers. TNM stage-based comparisons revealed dysregulated genes and biological pathways involved in cancer progression, among which the dysregulation of complement coagulation cascade and epithelial-mesenchymal transition are frequent in multiple types of cancers. A group of pan-cancer continuously upregulated and downregulated proteins in different tumor stages (PCCUPs and PCCDPs) were identified. We further constructed prognostic risk stratification models for corresponding cancer types based on dysregulated genes, which effectively predict the prognosis for patients with these cancers. Drug prediction based on PCUGs and PCDGs as well as PCCUPs and PCCDPs revealed that small molecule inhibitors targeting CDK, HDAC, MEK, JAK, PI3K, and others might be effective treatments for pan-cancer, thereby supporting drug repurposing. We also developed web tools for cancer diagnosis, pathologic stage assessment, and risk evaluation. Overall, this study highlights the power of combining proteomic and transcriptomic data to identify valuable diagnostic and prognostic markers as well as drug targets and treatments for cancer.
Collapse
Affiliation(s)
- Guo-Sheng Hu
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou, China; State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zao-Zao Zheng
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yao-Hui He
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Du-Chuang Wang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Rui-Chao Nie
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wen Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
24
|
Wang Y, Safi M, Hirsch FR, Lu S, Peters S, Govindan R, Rosell R, Park K, Zhang JJ. Immunotherapy for advanced-stage squamous cell lung cancer: the state of the art and outstanding questions. Nat Rev Clin Oncol 2025; 22:200-214. [PMID: 39762577 DOI: 10.1038/s41571-024-00979-8] [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: 12/06/2024] [Indexed: 02/26/2025]
Abstract
Immune-checkpoint inhibitors (ICIs) have transformed the treatment paradigm for advanced-stage squamous non-small-cell lung cancer (LUSC), a histological subtype associated with inferior outcomes compared with lung adenocarcinoma. However, only a subset of patients derive durable clinical benefit. In the first-line setting, multiple ICI regimens are available, including anti-PD-(L)1 antibodies as monotherapy, in combination with chemotherapy, or with an anti-CTLA4 antibody with or without chemotherapy. Several important questions persist regarding the optimal regimen for individual patients, particularly how to identify patients who might benefit from adding chemotherapy and/or anti-CTLA4 antibodies to anti-PD-(L)1 antibodies. An urgent need exists for predictive biomarkers beyond PD-L1 to better guide precision oncology approaches. Deeper knowledge of the underlying molecular biology of LUSC and its implications for response to ICIs will be important in this regard. Integration of this knowledge into multi-omics methods coupled with artificial intelligence might enable the development of more robust biomarkers. Finally, several novel therapeutic strategies, including novel ICIs, bispecific antibodies and personalized cancer vaccines, are emerging. Addressing these unresolved questions through innovative clinical trials and translational research will be crucial to further improving the outcomes of patients with LUSC. In this Review, we provide a comprehensive overview of current immunotherapeutic approaches, unresolved challenges and emerging strategies for patients with LUSC.
Collapse
Affiliation(s)
- Yibei Wang
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mohammed Safi
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Fred R Hirsch
- Center for Thoracic Oncology, Tisch Cancer Institute and Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Shun Lu
- Department of Medical Oncology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Solange Peters
- Oncology Department, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | | | - Rafael Rosell
- Dr. Rosell Oncology Institute, Dexeus University Hospital, Barcelona, Spain
| | - Keunchil Park
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
- Division of Hematology/Oncology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Jianjun J Zhang
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, the University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
25
|
Suo Y, Song Y, Wang Y, Liu Q, Rodriguez H, Zhou H. Advancements in proteogenomics for preclinical targeted cancer therapy research. BIOPHYSICS REPORTS 2025; 11:56-76. [PMID: 40070661 PMCID: PMC11891078 DOI: 10.52601/bpr.2024.240053] [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: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 03/14/2025] Open
Abstract
Advancements in molecular characterization technologies have accelerated targeted cancer therapy research at unprecedented resolution and dimensionality. Integrating comprehensive multi-omic molecular profiling of a tumor, proteogenomics, marks a transformative milestone for preclinical cancer research. In this paper, we initially provided an overview of proteogenomics in cancer research, spanning genomics, transcriptomics, and proteomics. Subsequently, the applications were introduced and examined from different perspectives, including but not limited to genetic alterations, molecular quantifications, single-cell patterns, different post-translational modification levels, subtype signatures, and immune landscape. We also paid attention to the combined multi-omics data analysis and pan-cancer analysis. This paper highlights the crucial role of proteogenomics in preclinical targeted cancer therapy research, including but not limited to elucidating the mechanisms of tumorigenesis, discovering effective therapeutic targets and promising biomarkers, and developing subtype-specific therapies.
Collapse
Affiliation(s)
- Yuying Suo
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanli Song
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuqiu Wang
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Qian Liu
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Rockville, MD 20850, USA
| | - Hu Zhou
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
26
|
Mazzilli SA, Rahal Z, Rouhani MJ, Janes SM, Kadara H, Dubinett SM, Spira AE. Translating premalignant biology to accelerate non-small-cell lung cancer interception. Nat Rev Cancer 2025:10.1038/s41568-025-00791-1. [PMID: 39994467 DOI: 10.1038/s41568-025-00791-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/02/2025] [Indexed: 02/26/2025]
Abstract
Over the past decade, substantial progress has been made in the development of targeted and immune-based therapies for patients with advanced non-small-cell lung cancer. To further improve outcomes for patients with lung cancer, identifying and intercepting disease at the earliest and most curable stages are crucial next steps. With the recent implementation of low-dose computed tomography scan screening in populations at high risk, there is an emerging unmet need for new diagnostic, prognostic and therapeutic tools to help treat patients suspected of harbouring premalignant lesions and minimally invasive non-small-cell lung cancer. Continued advances in the identification of the earliest drivers of lung carcinogenesis are poised to address these unmet needs. Employing multimodal approaches to chart the temporal and spatial maps of the molecular events driving lung premalignant lesion progression will refine our understanding of early carcinogenesis. Elucidating the molecular drivers of premalignancy is critical to the development of biomarkers to detect those incubating a premalignant lesion, to stratify risk for progression to invasive cancer and to identify novel therapeutic targets to intercept that process. In this Review, we summarize emerging insights into the earliest cellular and molecular events associated with lung squamous and adenocarcinoma carcinogenesis and highlight the growing opportunity for translating these insights into clinical tools for early detection and disease interception to transform the outcomes for those at risk for lung cancer.
Collapse
Affiliation(s)
- Sarah A Mazzilli
- Sectional Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - Zahraa Rahal
- Division of Pathology-Lab Medicine, Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Maral J Rouhani
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Humam Kadara
- Division of Pathology-Lab Medicine, Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Steven M Dubinett
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, and Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Avrum E Spira
- Sectional Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Johnson & Johnson Innovative Medicine, Boston, MA, USA.
| |
Collapse
|
27
|
Jiang M, Sun J, Hu C, Wu L, Fan Y, Wang Z, Liu L, Wu C, Wu F, Gao G, Li F, Wang L, Li X, Cheng L, Peng B, Zhou H, Zhou C. A tumor cornification and immune-infiltration-based scheme for anti-PD-1 plus chemotherapy response in advanced squamous cell lung carcinoma. MED 2025; 6:100516. [PMID: 39395411 DOI: 10.1016/j.medj.2024.09.005] [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: 12/26/2023] [Revised: 07/29/2024] [Accepted: 09/13/2024] [Indexed: 10/14/2024]
Abstract
BACKGROUND Anti-PD-1 immunotherapy plus chemotherapy (combo) exhibits significantly prolonged survival for squamous cell lung cancer (LUSC). An exploration of predictive biomarkers is still needed. METHODS High-throughput RNA sequencing (RNA-seq) of 349 LUSC samples from the randomized, multi-center, phase 3 trial ORIENT-12 (ClinicalTrials.gov: NCT03629925) was conducted for biomarker discovery, followed by flow cytometry and multiplex immunohistochemistry (mIHC) in additional clinical cohorts, and in vitro experiments were performed for verification. RESULTS A high abundance of activated CD8+ T and CD56bright natural killer (NK) cells benefited patients' outcomes (progression-free survival [PFS]; overall survival [OS]) with combo treatment. Tumor cornification level remarkably affected the infiltration of the two crucial immune cells. Thus, a novel scheme of LUSC immune infiltration and cornification characterization-based classification (LICC) was established for combo efficacy prediction. Patients who received combo treatment achieved significant PFS improvements in LICC1 (hazard ratio [HR] = 0.43, 95% confidence interval [CI]: 0.25-0.75, p = 0.0029) and LICC2 (HR = 0.32, 95% CI: 0.17-0.58, p = 0.0002) subtypes but not in the LICC3 subtype (HR = 0.86, 95% CI: 0.60-1.23, p = 0.4053). Via single-cell RNA-seq analysis, the tumor cornification signal was mainly mapped to SPRR3+ tumor cells, whose relationships with activated CD8+ T or CD56bright NK cells were verified using flow cytometry and mIHC. Our data suggest that SPRR3+ tumor cells might evade immune surveillance via the CD24-SIGLEC10 (M2 macrophage) axis to maintain a suppressive tumor microenvironment. CONCLUSIONS Tumor cornification greatly impacts immune infiltration, and the LICC scheme may guide clinical medication of anti-PD-1+chemo treatment in patients with LUSC. FUNDING The study was funded by the National Key R&D Program of China, the National Natural Science Foundation of China, Shanghia Multidisplinary Cooperation Building Project for Diagnosis and Treatment of Major Disease, and Innovent Biologics, Inc.
Collapse
Affiliation(s)
- Minlin Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China; Medical School, Tongji University, Shanghai 200433, China
| | - Jiya Sun
- Innovent Biologics, Inc., Suzhou, Jiangsu 215123, China
| | - Congli Hu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China; Medical School, Tongji University, Shanghai 200433, China
| | - Lin Wu
- Thoracic Medicine Department II, Hunan Cancer Hospital, Changsha, Hunan 410031, China
| | - Yun Fan
- Oncology Department, Cancer Hospital of the University of Chinese Academy of Science, Hangzhou, Zhejiang 310005, China
| | - Zhehai Wang
- Respiratory Department, Shandong Cancer Hospital, Jinan, Shandong 250117, China
| | - Lianke Liu
- Oncology Department, Jiangsu Province Hospital, Nanjing, Jiangsu 210029, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No. 507 Zhengmin Road, Shanghai 200433, China
| | - Fengying Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Guanghui Gao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Fei Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Frontier Innovation Center, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Lei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Xuefei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Lei Cheng
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Bo Peng
- Innovent Biologics, Inc., Suzhou, Jiangsu 215123, China
| | - Hui Zhou
- Innovent Biologics, Inc., Suzhou, Jiangsu 215123, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai East Hospital, Shanghai 200120, China.
| |
Collapse
|
28
|
Cooper WA, Amanuel B, Cooper C, Fox SB, Graftdyk JWA, Jessup P, Klebe S, Lam WS, Leong TYM, Lwin Z, Roberts-Thomson R, Solomon BJ, Tay RY, Trowman R, Wale JL, Pavlakis N. Molecular testing of lung cancer in Australia: consensus best practice recommendations from the Royal College of Pathologists of Australasia in collaboration with the Thoracic Oncology Group of Australasia. Pathology 2025:S0031-3025(25)00066-2. [PMID: 40102144 DOI: 10.1016/j.pathol.2025.02.001] [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: 12/18/2024] [Revised: 02/07/2025] [Accepted: 02/09/2025] [Indexed: 03/20/2025]
Abstract
Molecular testing plays a critical role in guiding optimal treatment decisions for lung cancer patients across a variety of clinical settings. While guidelines for biomarker testing exist in other jurisdictions, to date no best practice guidelines have been developed for the Australian setting. To address this need, the Royal College of Pathologists of Australasia collaborated with the Thoracic Oncology Group of Australasia to identify state-based pathologists, oncologists and consumer representatives to develop consensus best practice recommendations. Sixteen recommendations were established encompassing appropriate biomarkers, lung cancer subtype, tumour stage, specimen types, assay selection and quality assurance protocols that can inform and standardise best practice in molecular testing of lung cancer. These multidisciplinary evidence-based recommendations are designed to standardise and enhance molecular testing practices for lung cancers and should help ensure laboratories provide high-quality molecular testing of lung cancer for all Australians, including those from regional or remote communities.
Collapse
Affiliation(s)
- Wendy A Cooper
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; School of Medicine, Western Sydney University, Sydney, NSW, Australia.
| | - Benhur Amanuel
- Anatomical Pathology, PathWest, WA, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia; School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, WA, Australia
| | - Caroline Cooper
- Anatomical Pathology, Pathology Queensland, Princess Alexandra Hospital, Woolloongabba, Qld, Australia; Faculty of Medicine, The University of Queensland, St Lucia, Qld, Australia
| | - Stephen B Fox
- Pathology, Peter MacCallum Cancer Centre, Parkville, Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology and the Collaborative Centre for Genomic Cancer Medicine, University of Melbourne, Parkville, Vic, Australia
| | | | - Peter Jessup
- Anatomical Pathology, Royal Hobart Hospital, Hobart, Tas, Australia
| | - Sonja Klebe
- Anatomical Pathology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; SA Pathology, Adelaide, SA, Australia
| | - Wei-Sen Lam
- Department of Medical Oncology, Fiona Stanley Hospital, Perth, WA, Australia; WA Regional Clinical Trial Coordinating Centre, WA Country Health Service, WA, Australia
| | - Trishe Y-M Leong
- Anatomical Pathology, Melbourne Pathology, Sonic Healthcare, Melbourne, Vic, Australia; Department of Clinical Pathology, University of Melbourne, Melbourne, Vic, Australia
| | - Zarnie Lwin
- Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, Qld, Australia; The Prince Charles Hospital, University of Queensland, Chermside, Qld, Australia
| | | | - Benjamin J Solomon
- Sir Peter MacCallum Department of Oncology and the Collaborative Centre for Genomic Cancer Medicine, University of Melbourne, Parkville, Vic, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Rebecca Y Tay
- Department of Medical Oncology, Royal Hobart Hospital. Hobart, Tas, Australia
| | - Rebecca Trowman
- Independent Health Technology Assessment Specialist, Perth, WA, Australia
| | - Janney L Wale
- Independent Consumer Advocate, Melbourne, Vic, Australia; Chair of the RCPA Community Advisory Committee, Sydney, NSW, Australia
| | - Nick Pavlakis
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia; The Thoracic Oncology Group of Australasia, Thornbury, Vic, Australia
| |
Collapse
|
29
|
Ellrott K, Wong CK, Yau C, Castro MAA, Lee JA, Karlberg BJ, Grewal JK, Lagani V, Tercan B, Friedl V, Hinoue T, Uzunangelov V, Westlake L, Loinaz X, Felau I, Wang PI, Kemal A, Caesar-Johnson SJ, Shmulevich I, Lazar AJ, Tsamardinos I, Hoadley KA, Robertson AG, Knijnenburg TA, Benz CC, Stuart JM, Zenklusen JC, Cherniack AD, Laird PW. Classification of non-TCGA cancer samples to TCGA molecular subtypes using compact feature sets. Cancer Cell 2025; 43:195-212.e11. [PMID: 39753139 PMCID: PMC11949768 DOI: 10.1016/j.ccell.2024.12.002] [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: 08/16/2024] [Revised: 08/26/2024] [Accepted: 12/05/2024] [Indexed: 02/12/2025]
Abstract
Molecular subtypes, such as defined by The Cancer Genome Atlas (TCGA), delineate a cancer's underlying biology, bringing hope to inform a patient's prognosis and treatment plan. However, most approaches used in the discovery of subtypes are not suitable for assigning subtype labels to new cancer specimens from other studies or clinical trials. Here, we address this barrier by applying five different machine learning approaches to multi-omic data from 8,791 TCGA tumor samples comprising 106 subtypes from 26 different cancer cohorts to build models based upon small numbers of features that can classify new samples into previously defined TCGA molecular subtypes-a step toward molecular subtype application in the clinic. We validate select classifiers using external datasets. Predictive performance and classifier-selected features yield insight into the different machine-learning approaches and genomic data platforms. For each cancer and data type we provide containerized versions of the top-performing models as a public resource.
Collapse
Affiliation(s)
- Kyle Ellrott
- Oregon Health and Science University, Portland, OR 97239, USA.
| | - Christopher K Wong
- Biomolecular Engineering Department, School of Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Christina Yau
- University of California, San Francisco, Department of Surgery, San Francisco, CA 94158, USA; Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Mauro A A Castro
- Bioinformatics and Systems Biology Laboratory, Federal University of Paraná, Curitiba, PR 81520-260, Brazil
| | - Jordan A Lee
- Oregon Health and Science University, Portland, OR 97239, USA
| | | | - Jasleen K Grewal
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Vincenzo Lagani
- JADBio Gnosis DA, GR-700 13 Heraklion, Crete, Greece; Institute of Chemical Biology, Ilia State University, Tbilisi 0162, Georgia
| | - Bahar Tercan
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA 98109, USA
| | - Verena Friedl
- Biomolecular Engineering Department, School of Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Toshinori Hinoue
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Vladislav Uzunangelov
- Biomolecular Engineering Department, School of Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Lindsay Westlake
- The Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Xavier Loinaz
- The Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Ina Felau
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Peggy I Wang
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Anab Kemal
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - Ilya Shmulevich
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA 98109, USA
| | - Alexander J Lazar
- Departments of Pathology & Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ioannis Tsamardinos
- JADBio Gnosis DA, GR-700 13 Heraklion, Crete, Greece; Department of Computer Science, University of Crete, GR-700 13 Heraklion, Crete, Greece; Institute of Applied and Computational Mathematics, Foundation for Research and Technology Hellas (FORTH), GR-700 13 Heraklion, Crete, Greece
| | - Katherine A Hoadley
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27519, USA
| | - A Gordon Robertson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Theo A Knijnenburg
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA 98109, USA
| | | | - Joshua M Stuart
- Biomolecular Engineering Department, School of Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jean C Zenklusen
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Andrew D Cherniack
- The Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Harvard Medical School, Boston, MA 02115, USA.
| | - Peter W Laird
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
30
|
Wang C, Li J, Chen J, Wang Z, Zhu G, Song L, Wu J, Li C, Qiu R, Chen X, Zhang L, Li W. Multi-omics analyses reveal biological and clinical insights in recurrent stage I non-small cell lung cancer. Nat Commun 2025; 16:1477. [PMID: 39929832 PMCID: PMC11811181 DOI: 10.1038/s41467-024-55068-2] [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: 06/27/2022] [Accepted: 11/26/2024] [Indexed: 02/13/2025] Open
Abstract
Post-operative recurrence rates of stage I non-small cell lung cancer (NSCLC) range from 20% to 40%. Nonetheless, the molecular mechanisms underlying recurrence hitherto remain largely elusive. Here, we generate genomic, epigenomic and transcriptomic profiles of paired tumors and adjacent tissues from 122 stage I NSCLC patients, among which 57 patients develop recurrence after surgery during follow-up. Integrated analyses illustrate that the presence of predominantly solid or micropapillary histological subtypes, increased genomic instability, and APOBEC-related signature are associated with recurrence. Furthermore, TP53 missense mutation in DNA-binding domain could contribute to shorter time to recurrence. DNA hypomethylation is pronounced in recurrent NSCLC, and PRAME is the significantly hypomethylated and overexpressed gene in recurrent lung adenocarcinoma (LUAD). Mechanistically, hypomethylation at TEAD1 binding site facilitates the transcriptional activation of PRAME. Inhibition of PRAME restrains the tumor metastasis via downregulation of epithelial-mesenchymal transition-related genes. We also identify that enrichment of AT2 cells with higher copy number variation burden, exhausted CD8 + T cells and Macro_SPP1, along with the reduced interaction between AT2 and immune cells, is essential for the formation of ecosystem in recurrent LUAD. Finally, multi-omics clustering could stratify the NSCLC patients into 4 subclusters with varying recurrence risk and subcluster-specific therapeutic vulnerabilities. Collectively, this study constitutes a promising resource enabling insights into the biological mechanisms and clinical management for post-operative recurrence of stage I NSCLC.
Collapse
Affiliation(s)
- Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Jingwei Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingyao Chen
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhoufeng Wang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guonian Zhu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lujia Song
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiayang Wu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Changshu Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong Qiu
- Department of Respiratory and Critical Care Medicine, Suining Central Hospital, Suining, China
| | - Xuelan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Chengdu, Sichuan, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
31
|
Henick BS, Taylor AM, Nakagawa H, Wong KK, Diehl JA, Rustgi AK. Squamous cell cancers of the aero-upper digestive tract: A unified perspective on biology, genetics, and therapy. Cancer Cell 2025; 43:178-194. [PMID: 39933897 PMCID: PMC11875029 DOI: 10.1016/j.ccell.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/23/2024] [Accepted: 01/10/2025] [Indexed: 02/13/2025]
Abstract
Squamous cell cancers (SCCs) of the head and neck, esophagus, and lung, referred to as aero-upper digestive SCCs, are prevalent in the United States and worldwide. Their incidence and mortality are projected to increase at alarming rates, posing diagnostic, prognostic, and therapeutic challenges. These SCCs share certain epigenetic, genomic, and genetic alterations, immunologic properties, environmental exposures, as well as lifestyle and nutritional risk factors, which may underscore common complex gene-environmental interactions across them. This review focuses upon the frequent shared epigenetic, genomic, and genetic alterations, emerging preclinical model systems, and how this collective knowledge can be leveraged into perspectives on standard of care therapies and mechanisms of resistance, nominating new potential directions in translational therapeutics.
Collapse
Affiliation(s)
- Brian S Henick
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Division of Hematology-Oncology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Alison M Taylor
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Hiroshi Nakagawa
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Kwok-Kin Wong
- Division of Hematology-Oncology, Department of Medicine, NYU Perlmutter Cancer Center, New York, NY, USA
| | - J Alan Diehl
- Department of Biochemistry, Case Western Reserve Comprehensive Cancer Center, Cleveland, OH, USA
| | - Anil K Rustgi
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
32
|
Barbeau LMO, Beelen NA, Savelkouls KG, Keulers TGH, Wieten L, Rouschop KMA. MAP1LC3C repression reduces CIITA- and HLA class II expression in non-small cell lung cancer. PLoS One 2025; 20:e0316716. [PMID: 39928678 PMCID: PMC11809862 DOI: 10.1371/journal.pone.0316716] [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: 06/12/2024] [Accepted: 12/15/2024] [Indexed: 02/12/2025] Open
Abstract
In the last decade, advancements in understanding the genetic landscape of lung squamous cell carcinoma (LUSC) have significantly impacted therapy development. Immune checkpoint inhibitors (ICI) have shown great promise, improving overall and progression-free survival in approximately 25% of the patients. However, challenges remain, such as the lack of predictive biomarkers, difficulties in patient stratification, and identifying mechanisms that cancers use to become immune-resistant ("immune-cold"). Analysis of TCGA datasets reveals reduced MAP1LC3C expression in cancer. Further analysis indicates that low MAP1LC3C is associated with reduced CIITA and HLA expression and with decreased immune cell infiltration. In tumor cells, silencing MAP1LC3C inhibits CIITA expression and suppresses HLA class II production. These findings suggest that cancer cells are selected for low MAP1LC3C expression to evade efficient immune responses.
Collapse
Affiliation(s)
- Lydie M. O. Barbeau
- Department of Radiation Oncology (Maastro), GROW - School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Nicky A. Beelen
- Department of Internal Medicine, GROW - School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Transplantation Immunology, GROW - School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Kim G. Savelkouls
- Department of Radiation Oncology (Maastro), GROW - School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Tom G. H. Keulers
- Department of Radiation Oncology (Maastro), GROW - School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Lotte Wieten
- Department of Transplantation Immunology, GROW - School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Kasper M. A. Rouschop
- Department of Radiation Oncology (Maastro), GROW - School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
33
|
Ciesielska K, Wawrzyniak D, Dutkiewicz G, Kubicki M, Jankowski W, Hoffmann M, Kamel K, Rolle K, Pluskota-Karwatka D. Diastereoselective synthesis and biological evaluation of new fluorine-containing α-aminophosphonates as anticancer agents and scaffold to human urokinase plasminogen activator inhibitors. Eur J Med Chem 2025; 283:117116. [PMID: 39637829 DOI: 10.1016/j.ejmech.2024.117116] [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: 09/24/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Phosphonate analogues of α-amino acids are increasingly valued for their significant potential in medicinal chemistry. Fluorine is a "magic" element that plays a huge role in modulating the properties of organic compounds. In this work, we combined the two pharmacophores in the synthesis of three series of new α-aminophosphonates. These compounds were obtained by diastereoselective hydrophosphonylation of imines prepared by an environmentally friendly mechanochemical approach. Results of computational SwissADME analysis suggested favorable drug-like properties of the α-aminophosphonates and indicated their potential for interaction with diverse biological targets including proteases, showing promising pharmacokinetic profiles compared to 5-fluoro-2'-deoxyuridine (FdU) used as a standard anticancer drug. Screening against ten cancer cell lines from seven types of cancer showed that five of the twenty compounds tested (1c, 2a, 2h, 3e, and 3f) exhibited superior activity against the HeLa cell line and lower cytotoxicity against normal MRC-5 cells than FdU. Compound 3e showed notable inhibitory effect on the MDA-MB-231 cell line, while 3a, 3h, and 3g demonstrated significant cytotoxic activity against U-87 MG and U-251 MG lines. Molecular docking highlighted the strong binding of compound 2a to the urokinase-type plasminogen activator (uPA) protein, with a binding affinity of -6.41 kcal/mol, suggesting the anti-metastatic potential of the compound. These findings enable to position the newly synthesized α-aminophosphonates as promising scaffolds for developing targeted anticancer therapies for metastatic cancers characterized by elevated uPA expression.
Collapse
Affiliation(s)
- Karolina Ciesielska
- Adam Mickiewicz University, Faculty of Chemistry, Uniwersytetu Poznańskiego 8, 61-614, Poznań, Poland
| | - Dariusz Wawrzyniak
- Adam Mickiewicz University, Faculty of Chemistry, Uniwersytetu Poznańskiego 8, 61-614, Poznań, Poland; Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704, Poznań, Poland.
| | - Grzegorz Dutkiewicz
- Adam Mickiewicz University, Faculty of Chemistry, Uniwersytetu Poznańskiego 8, 61-614, Poznań, Poland
| | - Maciej Kubicki
- Adam Mickiewicz University, Faculty of Chemistry, Uniwersytetu Poznańskiego 8, 61-614, Poznań, Poland
| | - Wojciech Jankowski
- Adam Mickiewicz University, Faculty of Chemistry, Uniwersytetu Poznańskiego 8, 61-614, Poznań, Poland
| | - Marcin Hoffmann
- Adam Mickiewicz University, Faculty of Chemistry, Uniwersytetu Poznańskiego 8, 61-614, Poznań, Poland
| | - Karol Kamel
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704, Poznań, Poland
| | - Katarzyna Rolle
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704, Poznań, Poland
| | - Donata Pluskota-Karwatka
- Adam Mickiewicz University, Faculty of Chemistry, Uniwersytetu Poznańskiego 8, 61-614, Poznań, Poland.
| |
Collapse
|
34
|
Li S, Zhan Y, Wang Y, Li W, Wang X, Wang H, Sun W, Cao X, Li Z, Ye F. One-step diagnosis of infection and lung cancer using metagenomic sequencing. Respir Res 2025; 26:48. [PMID: 39905469 DOI: 10.1186/s12931-025-03127-7] [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: 07/31/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Traditional detection methods face challenges in meeting the diverse clinical needs for diagnosing both lung cancer and infections within a single test. Onco-mNGS has emerged as a promising solution capable of accurately identifying infectious pathogens and tumors simultaneously. However, critical evidence is still lacking regarding its diagnostic performance in distinguishing between pulmonary infections, tumors, and non-infectious, non-tumor conditions in real clinical settings. METHODS In this study, data were gathered from 223 participants presenting symptoms of lung infection or tumor who underwent Onco-mNGS testing. Patients were categorized into four groups based on clinical diagnoses: infection, tumor, tumor with infection, and non-infection-non-tumor. Comparisons were made across different groups, subtypes, and stages of lung cancer regarding copy number variation (CNV) patterns, microbiome compositions, and clinical detection indices. RESULTS Compared to conventional infection testing methods, Onco-mNGS demonstrates superior infection detection performance, boasting a sensitivity of 81.82%, specificity of 72.55%, and an overall accuracy of 77.58%. In lung cancer diagnosis, Onco-mNGS showcases excellent diagnostic capabilities with sensitivity, specificity, accuracy, positive predictive value, and negative predictive value reaching 88.46%, 100%, 91.41%, 100%, and 90.98%, respectively. In bronchoalveolar lavage fluid (BALF) samples, these values stand at 87.5%, 100%, 94.74%, 100%, and 91.67%, respectively. Notably, more abundant CNV mutation types and higher mutation rates were observed in adenocarcinoma (ADC) compared to squamous cell carcinoma (SCC). Concurrently, onco-mNGS data revealed specific enrichment of Capnocytophaga sputigeria in the ADC group and Candida parapsilosis in the SCC group. These species exhibited significant correlations with C reaction protein (CRP) and CA153 values. Furthermore, Haemophilus influenzae was enriched in the early-stage SCC group and significantly associated with CRP values. CONCLUSIONS Onco-mNGS has exhibited exceptional efficiencies in the detection and differentiation of infection and lung cancer. This study provides a novel technological option for achieving single-step precise and swift detection of lung cancer.
Collapse
Affiliation(s)
- Shaoqiang Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Yangqing Zhan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Yan Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Weilong Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Xidong Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Haoru Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Wenjun Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Xuefang Cao
- MatriDx Biotechnology Co., Ltd, Hangzhou, 311112, China
| | - Zhengtu Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, People's Republic of China.
| | - Feng Ye
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, People's Republic of China.
| |
Collapse
|
35
|
Dammak S, Cecchini MJ, Coats J, Baranova K, Ward AD. Predicting cancer content in tiles of lung squamous cell carcinoma tumours with validation against pathologist labels. Comput Biol Med 2025; 185:109489. [PMID: 39637460 DOI: 10.1016/j.compbiomed.2024.109489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND A growing body of research is using deep learning to explore the relationship between treatment biomarkers for lung cancer patients and cancer tissue morphology on digitized whole slide images (WSIs) of tumour resections. However, these WSIs typically contain non-cancer tissue, introducing noise during model training. As digital pathology models typically start with splitting WSIs into tiles, we propose a model that can be used to exclude non-cancer tiles from the WSIs of lung squamous cell carcinoma (SqCC) tumours. METHODS We obtained 116 WSIs of tumours from 35 different centres from the Cancer Genome Atlas. A pathologist completed or reviewed cancer contours in four regions of interest (ROIs) within each WSIs. We then split the ROIs into tiles labelled with the percentage of cancer tissue within them and trained VGG16 to predict this value, and then we calculated regression error. To measure classification performance and visualize the classification results, we thresholded the predictions and calculated the area under the receiver operating characteristic curve (AUC). RESULTS The model's median regression error was 4% with a standard deviation of 35%. At a cancer threshold of 50%, the model had an AUC of 0.83. False positives tended to be in tissues that surround cancer, tiles with <50% cancer, and areas with high immune activity. False negatives tended to be microtomy defects. CONCLUSIONS With further validation for each specific research application, the model we describe in this paper could facilitate the development of more effective research pipelines for predicting treatment biomarkers for lung SqCC.
Collapse
Affiliation(s)
- Salma Dammak
- Baines Imaging Research Laboratory, London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada; School of Biomedical Engineering, Western University, London, Ontario, Canada
| | - Matthew J Cecchini
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jennifer Coats
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Katherina Baranova
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Aaron D Ward
- Baines Imaging Research Laboratory, London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada; School of Biomedical Engineering, Western University, London, Ontario, Canada; Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
36
|
Islam ME, Debnath KC, Moniruzzaman R, Okuyama K, Islam S, Dongre HN. Biological implications of decoding the extracellular matrix of vulva cancer. Oncol Rep 2025; 53:19. [PMID: 39670289 DOI: 10.3892/or.2024.8852] [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: 05/28/2024] [Accepted: 08/15/2024] [Indexed: 12/14/2024] Open
Abstract
The present review aimed to elucidate the roles of extracellular matrix (ECM) components in the progression of vulvar squamous cell carcinoma (VSCC) and explore potential therapeutic avenues for this type of malignancy. This exploration holds promise for identifying precise molecular targets within the ECM milieu, thus facilitating the development of innovative therapeutic modalities tailored to disrupt these interactions and ultimately improve patient outcomes in VSCC. The dysregulated ECM serves as a potent driver of SCC tumor progression, orchestrating key processes such as angiogenesis, inflammation and stromal cell behavior. Yet, the exploration of ECM role in VSCC is still in its early stages. Recent research highlights the critical role of ECM organization and expression within the tumor microenvironment (TME) in influencing key aspects of VSCC, including tumor staging, grading, metastasis, invasion and patient survival. Cancer‑associated fibroblasts play a pivotal role in this dynamic by engaging in reciprocal interactions with VSCC cells, leading to significant ECM alterations and creating an immune‑suppressive TME. This hinders antitumor immunity and fosters therapeutic resistance in VSCC treatment. The dysregulated ECM in VSCC drives tumor progression, metastasis and affects patient survival. Targeting ECM, along with emerging therapies such as immune checkpoint blockade, offers promise for improved VSCC treatment outcomes.
Collapse
Affiliation(s)
- Mohammad Emranul Islam
- Department of Oral and Maxillofacial Surgery, City Dental College and Hospital, 1229 Dhaka, Bangladesh
| | - Kala Chand Debnath
- Department of Head and Neck Surgery, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rohan Moniruzzaman
- Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kohei Okuyama
- Department of Head and Neck Surgery, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shajedul Islam
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Harsh Nitin Dongre
- Center for Cancer Biomarkers and Gade Laboratory for Pathology, Institute of Clinical Medicine, University of Bergen, 5021 Bergen, Norway
| |
Collapse
|
37
|
Roggero CM, Ghosh AB, Devineni A, Ma S, Blatt E, Raj GV, Yin Y. CDK4/6 inhibitors promote PARP1 degradation and synergize with PARP inhibitors in non-small cell lung cancer. Transl Oncol 2025; 52:102231. [PMID: 39662449 PMCID: PMC11683282 DOI: 10.1016/j.tranon.2024.102231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/02/2024] [Accepted: 11/28/2024] [Indexed: 12/13/2024] Open
Abstract
Despite widespread deregulation of CDK4/6 activity in non-small cell lung cancer (NSCLC), clinical trials with CDK4/6 inhibitor (CDK4/6i) as a monotherapy have shown poor antitumor activity. Preclinical studies indicate that CDK4/6i may collaborate by influencing DNA damage repair pathways during radiotherapy. Since PARP1 expression was also significantly upregulated in NSCLC, we analyzed the efficacy of combining PARP1 and CDK4/6 inhibition in NSCLC models. We found that CDK4/6is synergize with PARP1 inhibitors (PARPis) to inhibit the clonogenic growth of RB-proficient NSCLC models. This synergy correlates with increased accumulation of DNA damage, interrupted cell-cycle checkpoints, and enhanced apoptotic cell death. Mechanistically, we showed that CDK4/6is promote PARP1 protein degradation, which lead to decreased availability of DNA repair factors involved in homologous recombination and suppression of DNA repair competency. Furthermore, we showed that PARP trapping is engaged in this synergy. We then confirmed that combining PARPi and CDK4/6i blocked the growth of NSCLC xenografts in vivo and patient-derived explant models ex vivo. Our data reveal a previously uncharacterized impact of CDK4/6i on PARP1 levels in RB-proficient NSCLC models and the engagement of PARP trapping in the synergy between CDK4/6i and PARPi. Our findings suggest combining CDK4/6i with PARPi could be a viable therapeutic strategy for patients with RB-proficient NSCLC.
Collapse
Affiliation(s)
- Carlos M Roggero
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States; Instituto de Histología y Embriología de Mendoza (IHEM)-CONICET-Universidad Nacional de Cuyo, Argentina
| | - Anwesha B Ghosh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Anvita Devineni
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Shihong Ma
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Eliot Blatt
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Ganesh V Raj
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Yi Yin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States; Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States.
| |
Collapse
|
38
|
Huang X, Chen W, Wang Y, Shytikov D, Wang Y, Zhu W, Chen R, He Y, Yang Y, Guo W. Canonical and noncanonical NOTCH signaling in the nongenetic resistance of cancer: distinct and concerted control. Front Med 2025; 19:23-52. [PMID: 39745621 DOI: 10.1007/s11684-024-1107-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/18/2024] [Indexed: 02/27/2025]
Abstract
Therapeutic resistance in cancer is responsible for numerous cancer deaths in clinical practice. While target mutations are well recognized as the basis of genetic resistance to targeted therapy, nontarget mutation resistance (or nongenetic resistance) remains poorly characterized. Despite its complex and unintegrated mechanisms in the literature, nongenetic resistance is considered from our perspective to be a collective response of innate or acquired resistant subpopulations in heterogeneous tumors to therapy. These subpopulations, e.g., cancer stem-like cells, cancer cells with epithelial-to-mesenchymal transition, and drug-tolerant persisters, are protected by their resistance traits at cellular and molecular levels. This review summarizes recent advances in the research on resistant populations and their resistance traits. NOTCH signaling, as a central regulator of nongenetic resistance, is discussed with a special focus on its canonical maintenance of resistant cancer cells and noncanonical regulation of their resistance traits. This novel view of canonical and noncanonical NOTCH signaling pathways is translated into our proposal of reshaping therapeutic strategies targeting NOTCH signaling in resistant cancer cells. We hope that this review will lead researchers to study the canonical and noncanonical arms of NOTCH signaling as an integrated resistant mechanism, thus promoting the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xianzhe Huang
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Wenwei Chen
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Yanyan Wang
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Dmytro Shytikov
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Yanwen Wang
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Wangyi Zhu
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Ruyi Chen
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Yuwei He
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Yanjia Yang
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China
| | - Wei Guo
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Jiaxing, 314400, China.
- First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- Biomedical and Health Translational Research Center of Zhejiang Province, Jiaxing, 314400, China.
| |
Collapse
|
39
|
Wang YW, Gao YH, Wang C, Zhang PF, Wang M, Lan L, Liu JY, Shi L, Sun LP. Design, synthesis, and biological evaluation of novel FGFR1 PROTACs. Bioorg Chem 2025; 155:108109. [PMID: 39756204 DOI: 10.1016/j.bioorg.2024.108109] [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: 10/14/2024] [Revised: 12/07/2024] [Accepted: 12/27/2024] [Indexed: 01/07/2025]
Abstract
Dysregulation of the fibroblast growth factor receptor 1 (FGFR1) signaling has prompted efforts to develop therapeutic agents, which is a carcinogenic driver of many cancers, including breast, prostate, bladder, and chronic myeloid leukemia. Despite significant progress in the development of potent and selective FGFR inhibitors, the long-term efficacy of these drugs in cancer therapy has been hampered by the rapid onset of acquired resistance. Therefore, more drug discovery strategies are needed to promote the development of FGFR-targeted drugs. Here, we discovered compound S2h, a compound that selectively and effectively degrades FGFR1 at nanomolar concentrations in KG1a cells (IC50 = 26.81 nM; DC50 = 39.78 nM), which incorporates an essential, nine atom-long linkers. The importance of linker length, composition, and tethering site proteolysis-targeting chimeras (PROTACs) design is emphasized, and slight modifications can significantly affect degradation potency. Meanwhile, it was verified that the degradation of FGFR1 protein at compound S2h was concentration- and time-dependent and that the protein degradation occurred through the ubiquitin-proteasome system (UPS). In summary, the newly designed heterobifunctional FGFR1 degrader, compound S2h, provides new ideas and references for the research of FGFR small-molecule degraders.
Collapse
Affiliation(s)
- Yu-Wei Wang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yu-Hui Gao
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, PR China
| | - Cheng Wang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, PR China
| | - Ping-Fan Zhang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, PR China
| | - Min Wang
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, PR China
| | - Li Lan
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, PR China
| | - Jing-Ying Liu
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, PR China
| | - Lei Shi
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Li-Ping Sun
- Jiangsu Key Laboratory of Drug Design & Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
40
|
Oskomić M, Tomić A, Barbarić L, Matić A, Kindl DC, Matovina M. KEAP1-NRF2 Interaction in Cancer: Competitive Interactors and Their Role in Carcinogenesis. Cancers (Basel) 2025; 17:447. [PMID: 39941813 PMCID: PMC11816071 DOI: 10.3390/cancers17030447] [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: 12/23/2024] [Revised: 01/21/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
An American Cancer Society report estimates the emergence of around 2 million new cancer cases in the US in 2024 [...].
Collapse
Affiliation(s)
| | | | | | | | | | - Mihaela Matovina
- Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (M.O.); (A.T.); (L.B.); (A.M.); (D.C.K.)
| |
Collapse
|
41
|
García-Vázquez N, González-Robles TJ, Lane E, Spasskaya D, Zhang Q, Kerzhnerman M, Jeong Y, Collu M, Simoneschi D, Ruggles KV, Rona G, Kaisari S, Pagano M. Stabilization of GTSE1 by cyclin D1-CDK4/6-mediated phosphorylation promotes cell proliferation: relevance in cancer prognosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.26.600797. [PMID: 38979260 PMCID: PMC11230433 DOI: 10.1101/2024.06.26.600797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
In healthy cells, cyclin D1 is expressed during the G1 phase of the cell cycle, where it activates CDK4 and CDK6. Its dysregulation is a well-established oncogenic driver in numerous human cancers. The cancer-related function of cyclin D1 has been primarily studied by focusing on the phosphorylation of the retinoblastoma (RB) gene product. Here, using an integrative approach combining bioinformatic analyses and biochemical experiments, we show that GTSE1 (G-Two and S phases expressed protein 1), a protein positively regulating cell cycle progression, is a previously unrecognized substrate of cyclin D1-CDK4/6 in tumor cells overexpressing cyclin D1 during G1 and subsequent phases. The phosphorylation of GTSE1 mediated by cyclin D1-CDK4/6 inhibits GTSE1 degradation, leading to high levels of GTSE1 across all cell cycle phases. Functionally, the phosphorylation of GTSE1 promotes cellular proliferation and is associated with poor prognosis within a pan-cancer cohort. Our findings provide insights into cyclin D1's role in cell cycle control and oncogenesis beyond RB phosphorylation.
Collapse
Affiliation(s)
- Nelson García-Vázquez
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Tania J González-Robles
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
- Department of Medicine, New York University Grossman School of Medicine, NYC, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, NYC, NY, USA
| | - Ethan Lane
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Daria Spasskaya
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Qingyue Zhang
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Marc Kerzhnerman
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - YeonTae Jeong
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Marta Collu
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Daniele Simoneschi
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
| | - Kelly V Ruggles
- Department of Medicine, New York University Grossman School of Medicine, NYC, NY, USA
| | - Gergely Rona
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, NYC, NY, USA
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Sharon Kaisari
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, NYC, NY, USA
| | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYC, NY, USA
- Howard Hughes Medical Institute, New York University Grossman School of Medicine, NYC, NY, USA
| |
Collapse
|
42
|
Durfee C, Bergstrom EN, Díaz-Gay M, Zhou Y, Temiz NA, Ibrahim MA, Nandi SP, Wang Y, Liu X, Steele CD, Proehl J, Vogel RI, Argyris PP, Alexandrov LB, Harris RS. Tobacco smoke carcinogens exacerbate APOBEC mutagenesis and carcinogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.18.633716. [PMID: 39896515 PMCID: PMC11785121 DOI: 10.1101/2025.01.18.633716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Mutations in somatic cells are inflicted by both extrinsic and intrinsic sources and contribute over time to cancer. Tobacco smoke contains chemical carcinogens that have been causatively implicated with cancers of the lung and head & neck1,2. APOBEC family DNA cytosine deaminases have emerged as endogenous sources of mutation in cancer, with hallmark mutational signatures (SBS2/SBS13) that often co-occur in tumors of tobacco smokers with an equally diagnostic mutational signature (SBS4)3,4. Here we challenge the dogma that mutational processes are thought to occur independently and with additive impact by showing that 4-nitroquinoline 1-oxide (NQO), a model carcinogen for tobacco exposure, sensitizes cells to APOBEC3B (A3B) mutagenesis and leads to synergistic increases in both SBS2 mutation loads and oral carcinomas in vivo. NQO-exposed/A3B-expressing animals exhibit twice as many head & neck lesions as carcinogen-exposed wildtype animals. This increase in carcinogenesis is accompanied by a synergistic increase in mutations from APOBEC signature SBS2, but not from NQO signature SBS4. Interestingly, a large proportion of A3B-catalyzed SBS2 mutations occurs as strand-coordinated pairs within 32 nucleotides of each other in transcribed regions, suggesting a mechanism in which removal of NQO-DNA adducts by nucleotide excision repair exposes short single-stranded DNA tracts to enzymatic deamination. These highly enriched pairs of APOBEC signature mutations are termed didyma (Greek for twins) and are mechanistically distinct from other types of clustered mutation (omikli and kataegis). Computational analyses of lung and head & neck tumor genomes show that both APOBEC mutagenesis and didyma are elevated in cancers from smokers compared to non-smokers. APOBEC signature mutations and didyma are also elevated in normal lung tissues in smokers prior to cancer initiation. Collectively, these results indicate that DNA adducting mutagens in tobacco smoke can amplify DNA damage and mutagenesis by endogenous APOBEC enzymes and, more broadly, suggest that mutational mechanisms can interact synergistically in both cancer initiation and promotion.
Collapse
Affiliation(s)
- Cameron Durfee
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas, USA, 78229
| | - Erik N. Bergstrom
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA, 92093
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA, 92093
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA, 92093
| | - Marcos Díaz-Gay
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA, 92093
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA, 92093
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA, 92093
- Digital Genomics Group, Structural Biology Program, Spanish National Cancer Research Center (CNIO), Madrid, Spain, 28029
| | - Yufan Zhou
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas, USA, 78229
| | - Nuri Alpay Temiz
- Institute for Health Informatics, University of Minnesota, Minneapolis, Minnesota, USA, 55455
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA, 55455
| | - Mahmoud A. Ibrahim
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas, USA, 78229
| | - Shuvro P. Nandi
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA, 92093
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA, 92093
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA, 92093
| | - Yaxi Wang
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas, USA, 78229
| | - Xingyu Liu
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas, USA, 78229
| | - Christopher D. Steele
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA, 92093
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA, 92093
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA, 92093
| | - Joshua Proehl
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas, USA, 78229
| | - Rachel I. Vogel
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA, 55455
- Department of Obstetrics, Gynecology, and Women’s Health, University of Minnesota, Minneapolis, Minnesota, USA, 55455
| | - Prokopios P. Argyris
- Division of Oral and Maxillofacial Pathology, College of Dentistry, Ohio State University, Columbus, Ohio, USA, 43210
| | - Ludmil B. Alexandrov
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, USA, 92093
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA, 92093
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA, 92093
| | - Reuben S. Harris
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas, USA, 78229
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, Texas, USA, 78229
| |
Collapse
|
43
|
Zhang C, Huo Y, Fu J, Liu Y, Zhou Q, Hou M, Duan X, Lv Y, Hu J. Design, synthesis and antitumour activity of pyrimidine derivatives as novel selective EGFR kinase inhibitors. Mol Divers 2025:10.1007/s11030-024-11048-8. [PMID: 39832084 DOI: 10.1007/s11030-024-11048-8] [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: 09/06/2024] [Accepted: 11/06/2024] [Indexed: 01/22/2025]
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, often linked to overexpression or abnormal activation of the epidermal growth factor receptor (EGFR). The issue of developing resistance to third-generation EGFR kinase inhibitors, such as osimertinib, underscores the urgent need for new therapies to overcome this resistance. Our findings revealed that compound A8 exhibits 88.01% kinase inhibition efficacy against the EGFRL858R/T790M mutation at a concentration of 0.1 μM, with an IC50 value of 5.0 nM. Moreover, its selectivity for this double mutation is 29.5, surpassing that of osimertinib. Most notably, A8 demonstrates an inhibitory activity of 2.9 nM against the EGFRL858R/T790M/C797S triple mutation, outperforming the benchmark drug osimertinib. Furthermore, compound A8 has demonstrated strong antiproliferative effects against H1975 cells, and its activity was better than osimertinib. The mechanism by which compound A8 operates as a selective EGFRL858R/T790M inhibitor was confirmed through a series of cell migration, apoptosis, and cell cycle assays. This lays the foundation for the development of a new structural type of EGFR kinase inhibitors.
Collapse
Affiliation(s)
- Cheng Zhang
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, Shandong, People's Republic of China
| | - Yiwen Huo
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, Shandong, People's Republic of China
| | - Jianfang Fu
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, Shandong, People's Republic of China
| | - Yue Liu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, 261053, Shandong, People's Republic of China
| | - Qinjiang Zhou
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, Shandong, People's Republic of China
| | - Mingyue Hou
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, Shandong, People's Republic of China
| | - Xiaoxuan Duan
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, Shandong, People's Republic of China
| | - Yanna Lv
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, Shandong, People's Republic of China.
| | - Jinxing Hu
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, Shandong, People's Republic of China.
| |
Collapse
|
44
|
Kido T, Kong H, Lau YFC. The X-Linked Tumor Suppressor TSPX Regulates Genes Involved in the EGFR Signaling Pathway and Cell Viability to Suppress Lung Adenocarcinoma. Genes (Basel) 2025; 16:75. [PMID: 39858622 PMCID: PMC11764513 DOI: 10.3390/genes16010075] [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: 12/07/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/30/2025] Open
Abstract
Background: TSPX is an X-linked tumor suppressor that was initially identified in non-small cell lung cancer (NSCLC) cell lines. However, its expression patterns and downstream mechanisms in NSCLC remain unclear. This study aims to investigate the functions of TSPX in NSCLC by identifying its potential downstream targets and their correlation with clinical outcomes. Methods: RNA-seq transcriptome and pathway enrichment analyses were conducted on the TSPX-overexpressing NSCLC cell lines, A549 and SK-MES-1, originating from lung adenocarcinoma and squamous cell carcinoma subtypes, respectively. In addition, comparative analyses were performed using the data from clinical NSCLC specimens (515 lung adenocarcinomas and 502 lung squamous cell carcinomas) in the Cancer Genome Atlas (TCGA) database. Results: TCGA data analysis revealed significant downregulation of TSPX in NSCLC tumors compared to adjacent non-cancerous tissues (Wilcoxon matched pairs signed rank test p < 0.0001). Notably, the TSPX expression levels were inversely correlated with the cancer stage, and higher TSPX levels were associated with better clinical outcomes and improved survival in lung adenocarcinoma, a subtype of NSCLC (median survival extended by 510 days; log-rank test, p = 0.0025). RNA-seq analysis of the TSPX-overexpressing NSCLC cell lines revealed that TSPX regulates various genes involved in the cancer-related signaling pathways and cell viability, consistent with the suppression of cell proliferation in cell culture assays. Notably, various potential downstream targets of TSPX that correlated with patient survival (log-rank test, p = 0.016 to 4.3 × 10-10) were identified, including EGFR pathway-related genes AREG, EREG, FOSL1, and MYC, which were downregulated. Conclusions: Our results suggest that TSPX plays a critical role in suppressing NSCLC progression by downregulating pro-oncogenic genes, particularly those in the EGFR signaling pathway, and upregulating the tumor suppressors, especially in lung adenocarcinoma. These findings suggest that TSPX is a potential biomarker and therapeutic target for NSCLC management.
Collapse
Affiliation(s)
| | | | - Yun-Fai Chris Lau
- Division of Cell and Developmental Genetics, Department of Medicine, Veterans Affairs Medical Center, and the Institute for Human Genetics, University of California, San Francisco, CA 94121, USA; (T.K.); (H.K.)
| |
Collapse
|
45
|
Andrijanova A, Bugovecka L, Isajevs S, Erts D, Malinovskis U, Liepins A. Machine Learning for Lung Cancer Subtype Classification: Combining Clinical, Histopathological, and Biophysical Features. Diagnostics (Basel) 2025; 15:127. [PMID: 39857011 PMCID: PMC11764335 DOI: 10.3390/diagnostics15020127] [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: 11/11/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Despite advances in diagnostic techniques, accurate classification of lung cancer subtypes remains crucial for treatment planning. Traditional methods like genomic studies face limitations such as high cost and complexity. This study investigates whether integrating atomic force microscopy (AFM) measurements with conventional clinical and histopathological data can improve lung cancer subtype classification. Methods: We developed and analyzed a novel dataset combining clinical, histopathological, and AFM-derived biophysical characteristics from 37 lung cancer patients. Various machine learning techniques were evaluated, with a focus on Bayesian Networks due to their ability to handle complex data with missing values. Leave-One-Out Cross-Validation was employed to assess model performance. Results: The integration of biophysical features improved classification accuracy from 86.49% to 89.19% using a data-driven Bayesian Network model, though this improvement was not statistically significant (p = 1.0). Four key features were identified as highly predictive: sex, vascular invasion, perineural invasion, and ALK mutation. A simplified model using only these features achieved identical performance with significantly reduced complexity (BIC 51.931 vs. 268.586). Conclusions: While AFM-derived measurements showed promise for enhancing lung cancer subtype classification, larger datasets are needed to fully validate their impact. Our findings demonstrate the feasibility of incorporating biophysical measurements into cancer classification frameworks and identify the most predictive features for accurate diagnosis. Further research with expanded datasets is needed to validate these findings.
Collapse
Affiliation(s)
| | - Lasma Bugovecka
- Institute of Chemical Physics, Faculty of Science and Technology, University of Latvia, Jelgavas Street 1, LV-1004 Riga, Latvia; (L.B.); (D.E.); (U.M.)
| | - Sergejs Isajevs
- Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas Street 3, LV-1004 Riga, Latvia;
| | - Donats Erts
- Institute of Chemical Physics, Faculty of Science and Technology, University of Latvia, Jelgavas Street 1, LV-1004 Riga, Latvia; (L.B.); (D.E.); (U.M.)
| | - Uldis Malinovskis
- Institute of Chemical Physics, Faculty of Science and Technology, University of Latvia, Jelgavas Street 1, LV-1004 Riga, Latvia; (L.B.); (D.E.); (U.M.)
| | | |
Collapse
|
46
|
Kudelka MR, Lavin Y, Sun S, Fuchs E. Molecular and cellular dynamics of squamous cell carcinomas across tissues. Genes Dev 2025; 39:18-35. [PMID: 39455281 PMCID: PMC11789493 DOI: 10.1101/gad.351990.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
Squamous cell carcinomas (SCCs), arising from the skin, head and neck, lungs, esophagus, and cervix, are collectively among the most common cancers and a frequent cause of cancer morbidity and mortality. Despite distinct stratified epithelial tissues of origin, converging evidence points toward shared biologic pathways across SCCs. With recent breakthroughs in molecular technologies have come novel SCC treatment paradigms, including immunotherapies and targeted therapy. This review compares commonalities and differences across SCCs from different anatomical sites, including risk factors and genetics, as well as cellular and molecular programs driving tumorigenesis. We review landmark discoveries of the "cancer stem cells" (CSCs) that initiate and propagate SCCs and their gene and translational regulation programs. This has led to an appreciation that interactions between CSCs and the immune system play key roles in invasion and therapeutic resistance. Here, we review the unifying principles of SCCs that have emerged from these exciting advances in our understanding of these epithelial cancers.
Collapse
Affiliation(s)
- Matthew R Kudelka
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New York 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Yonit Lavin
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New York 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Siman Sun
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New York 10065, USA
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New York 10065, USA;
| |
Collapse
|
47
|
Zhang X, Zhang P, Ren Q, Li J, Lin H, Huang Y, Wang W. Integrative multi-omic and machine learning approach for prognostic stratification and therapeutic targeting in lung squamous cell carcinoma. Biofactors 2025; 51:e2128. [PMID: 39391958 DOI: 10.1002/biof.2128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024]
Abstract
The proliferation, metastasis, and drug resistance of cancer cells pose significant challenges to the treatment of lung squamous cell carcinoma (LUSC). However, there is a lack of optimal predictive models that can accurately forecast patient prognosis and guide the selection of targeted therapies. The extensive multi-omic data obtained from multi-level molecular biology provides a unique perspective for understanding the underlying biological characteristics of cancer, offering potential prognostic indicators and drug sensitivity biomarkers for LUSC patients. We integrated diverse datasets encompassing gene expression, DNA methylation, genomic mutations, and clinical data from LUSC patients to achieve consensus clustering using a suite of 10 multi-omics integration algorithms. Subsequently, we employed 10 commonly used machine learning algorithms, combining them into 101 unique configurations to design an optimal performing model. We then explored the characteristics of high- and low-risk LUSC patient groups in terms of the tumor microenvironment and response to immunotherapy, ultimately validating the functional roles of the model genes through in vitro experiments. Through the application of 10 clustering algorithms, we identified two prognostically relevant subtypes, with CS1 exhibiting a more favorable prognosis. We then constructed a subtype-specific machine learning model, LUSC multi-omics signature (LMS) based on seven key hub genes. Compared to previously published LUSC biomarkers, our LMS score demonstrated superior predictive performance. Patients with lower LMS scores had higher overall survival rates and better responses to immunotherapy. Notably, the high LMS group was more inclined toward "cold" tumors, characterized by immune suppression and exclusion, but drugs like dasatinib may represent promising therapeutic options for these patients. Notably, we also validated the model gene SERPINB13 through cell experiments, confirming its role as a potential oncogene influencing the progression of LUSC and as a promising therapeutic target. Our research provides new insights into refining the molecular classification of LUSC and further optimizing immunotherapy strategies.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qianhe Ren
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haoran Lin
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuming Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
48
|
Zuccato JA, Mamatjan Y, Nassiri F, Ajisebutu A, Liu JC, Muazzam A, Singh O, Zhang W, Voisin M, Mirhadi S, Suppiah S, Wybenga-Groot L, Tajik A, Simpson C, Saarela O, Tsao MS, Kislinger T, Aldape KD, Moran MF, Patil V, Zadeh G. Prediction of brain metastasis development with DNA methylation signatures. Nat Med 2025; 31:116-125. [PMID: 39379704 PMCID: PMC11750707 DOI: 10.1038/s41591-024-03286-y] [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: 07/23/2023] [Accepted: 09/05/2024] [Indexed: 10/10/2024]
Abstract
Brain metastases (BMs) are the most common and among the deadliest brain tumors. Currently, there are no reliable predictors of BM development from primary cancer, which limits early intervention. Lung adenocarcinoma (LUAD) is the most common BM source and here we obtained 402 tumor and plasma samples from a large cohort of patients with LUAD with or without BM (n = 346). LUAD DNA methylation signatures were evaluated to build and validate an accurate model predicting BM development from LUAD, which was integrated with clinical factors to provide comprehensive patient-specific BM risk probabilities in a nomogram. Additionally, immune and cell interaction gene sets were differentially methylated at promoters in BM versus paired primary LUAD and had aligning dysregulation in the proteome. Immune cells were differentially abundant in BM versus LUAD. Finally, liquid biomarkers identified from methylated cell-free DNA sequenced in plasma were used to generate and validate accurate classifiers for early BM detection. Overall, LUAD methylomes can be leveraged to predict and noninvasively identify BM, moving toward improved patient outcomes with personalized treatment.
Collapse
Affiliation(s)
- Jeffrey A Zuccato
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Yasin Mamatjan
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
- The Faculty of Science, Thompson Rivers University, Kamloops, BC, Canada
| | - Farshad Nassiri
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Ajisebutu
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey C Liu
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Ammara Muazzam
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Olivia Singh
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Wen Zhang
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Mathew Voisin
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Shideh Mirhadi
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Suganth Suppiah
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Leanne Wybenga-Groot
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- SPARC BioCentre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alireza Tajik
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
- School of Medicine, St. George's University, Grenada, Grenada
| | - Craig Simpson
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- SPARC BioCentre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Olli Saarela
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Ming S Tsao
- Department of Pathology, The Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Kenneth D Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Michael F Moran
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Vikas Patil
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada.
| | - Gelareh Zadeh
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada.
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
- The Arthur and Sonia Labatt Brain Tumour Research Centre, Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
49
|
Xiong H, Zhang X, Sun J, Xue Y, Yu W, Mou S, Hsia KJ, Wan H, Wang P. Recent advances in biosensors detecting biomarkers from exhaled breath and saliva for respiratory disease diagnosis. Biosens Bioelectron 2025; 267:116820. [PMID: 39374569 DOI: 10.1016/j.bios.2024.116820] [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: 05/28/2024] [Revised: 09/06/2024] [Accepted: 09/28/2024] [Indexed: 10/09/2024]
Abstract
The global demand for rapid and non-invasive diagnostic methods for respiratory diseases has significantly intensified due to the wide spread of respiratory infectious diseases. Recent advancements in respiratory disease diagnosis through the analysis of exhaled breath and saliva has attracted great attention all over the world. Among various analytical methods, biosensors can offer non-invasive, efficient, and cost-effective diagnostic capabilities, emerging as promising tools in this area. This review intends to provide a comprehensive overview of various biosensors for the detection of respiratory disease related biomarkers in exhaled breath and saliva. Firstly, the characteristics of exhaled breath and saliva, including their generation, composition, and relevant biomarkers are introduced. Subsequently, the design and application of various biosensors for detecting these biomarkers are presented, along with the innovative materials employed as sensitive components. Different types of biosensors are reviewed, including electrochemical, optical, piezoelectric, semiconductor, and other novel biosensors. At last, the challenges, limitations, and future trends of these biosensors are discussed. It is anticipated that biosensors will play a significant role in respiratory disease diagnosis in the future.
Collapse
Affiliation(s)
- Hangming Xiong
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China; Binjiang Institute of Zhejiang University, Hangzhou 310053, China
| | - Xiaojing Zhang
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China; Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Jiaying Sun
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yingying Xue
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China; Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Weijie Yu
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China; Binjiang Institute of Zhejiang University, Hangzhou 310053, China
| | - Shimeng Mou
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China; Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - K Jimmy Hsia
- Schools of Chemical & Biomedical Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Hao Wan
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China; Binjiang Institute of Zhejiang University, Hangzhou 310053, China.
| | - Ping Wang
- Biosensor National Special Laboratory, Key Laboratory of Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China; Cancer Center, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
50
|
Ngo HH, Yu BY, Lee JE, Kim H, Keum YS. Identification of narciclasine as a novel NRF2 inhibitor. Free Radic Res 2025; 59:102-115. [PMID: 39783823 DOI: 10.1080/10715762.2025.2451679] [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: 09/15/2024] [Revised: 12/09/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025]
Abstract
Cancer genome sequencing studies have identified somatic mutations in the KEAP1/NRF2 pathway. In an effort to identify novel NRF2 small molecule inhibitor(s), we have screened a natural compound library comprising 1330 chemicals in A549-ARE-GFP-luciferase cells and identified that narciclasine significantly inhibits NRF2-dependent luciferase activity. Narciclasine suppressed the expression of NRF2 and NRF2 target genes, caused significant oxidative stress, and sensitized cisplatin-mediated apoptosis in A549 cells. In addition, we have observed that WD Repeat Domain 43 (WDR43) serves as a direct target of narciclasine for the inhibition of NRF2 as narciclasine binds to recombinant WDR43 in vitro and silencing WDR43 attenuated the inhibition of NRF2 by narciclasine in A549 cells. Finally, we observed that administration of narciclasine significantly decreased the growth of A549 xenografts. Together, our results demonstrate that the inhibition of NRF2 by narciclasine is mediated by WDR43 and future studies are necessary to elucidate the exact mechanism of how WDR43 mediates the inhibition of NRF2 by narciclasine.
Collapse
Affiliation(s)
- Hoang Hai Ngo
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University, Goyang, South Korea
| | - Bo-Yeung Yu
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University, Goyang, South Korea
| | - Jeong-Eun Lee
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University, Goyang, South Korea
| | - Hyunwoo Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University, Goyang, South Korea
| | - Young-Sam Keum
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University, Goyang, South Korea
| |
Collapse
|