1
|
Nussinov R, Yavuz BR, Jang H. Tumors and their microenvironments: Learning from pediatric brain pathologies. Biochim Biophys Acta Rev Cancer 2025; 1880:189328. [PMID: 40254040 DOI: 10.1016/j.bbcan.2025.189328] [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: 12/12/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
Early clues to tumors and their microenvironments come from embryonic development. Here we review the literature and consider whether the embryonic brain and its pathologies can serve as a better model. Among embryonic organs, the brain is the most heterogenous and complex, with multiple lineages leading to wide spectrum of cell states and types. Its dysregulation promotes neurodevelopmental brain pathologies and pediatric tumors. Embryonic brain pathologies point to the crucial importance of spatial heterogeneity over time, akin to the tumor microenvironment. Tumors dedifferentiate through genetic mutations and epigenetic modulations; embryonic brains differentiate through epigenetic modulations. Our innovative review proposes learning developmental brain pathologies to target tumor evolution-and vice versa. We describe ways through which tumor pharmacology can learn from embryonic brains and their pathologies, and how learning tumor, and its microenvironment, can benefit targeting neurodevelopmental pathologies. Examples include pediatric low-grade versus high-grade brain tumors as in rhabdomyosarcomas and gliomas.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
2
|
Gu Y, Ye Q, Huang X, Cao Y, Chaiswing L, She QB. Glycosaminoglycan modification of NRP1 exon 4-skipping variant drives colorectal cancer metastasis via endosomal-exosomal trafficking. Cancer Lett 2025; 620:217683. [PMID: 40157493 PMCID: PMC12014352 DOI: 10.1016/j.canlet.2025.217683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/15/2025] [Accepted: 03/27/2025] [Indexed: 04/01/2025]
Abstract
Neuropilin-1 (NRP1) is a transmembrane glycoprotein that functions as a co-receptor with various cellular functions. Our previous studies identified the NRP1 exon 4-skipping (NRP1-ΔE4) splice variant as an aggressive metastasis driver by activating endosomal signals. Here, we demonstrate the critical role of glycosaminoglycan (GAG) modification in regulating NRP1-ΔE4's cellular trafficking and oncogenic activity. NRP1-ΔE4 undergoes constitutive internalization into endosomes and subsequent exosomal release from colorectal cancer (CRC) cells. Exosomal NRP1-ΔE4 enhances the migration and invasion of both donor and recipient CRC cells. Genetic or pharmacological inhibition of exosome secretion, or immunodepletion of exosomal NRP1-ΔE4, markedly reduces its metastatic potential. Notably, GAG modification at the O-glycosylation site Ser612 is essential for NRP1-ΔE4's endosomal trafficking and exosomal release. This modification also promotes the formation of a trimeric complex with Met and β1-integrin, leading to their co-internalization and accumulation in endosomes, which activates FAK signaling and drives CRC metastasis. These findings reveal GAG modification as a key regulatory process that governs the endosomal-exosomal trafficking of NRP1-ΔE4 to facilitate CRC cell dissemination.
Collapse
Affiliation(s)
- Yiwei Gu
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40506, USA; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Qing Ye
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Xiuping Huang
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40506, USA; Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yanan Cao
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, Lexington, Lexington, KY, 40506, USA
| | - Qing-Bai She
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40506, USA.
| |
Collapse
|
3
|
Xie Y, Wang F, Wei J, Shen Z, Song X, Wang Y, Chen H, Tao L, Zheng J, Lin L, Niu Z, Guan X, Zhou T, Xu Z, Liu Y, Du D, Pan H, Li S, Ji W, Zhou W, Yang Y, Tian J, Xu J, Hu H, Liang X. Noninvasive prognostic classification of ITH in HCC with multi-omics insights and therapeutic implications. SCIENCE ADVANCES 2025; 11:eads8323. [PMID: 40315307 PMCID: PMC12047409 DOI: 10.1126/sciadv.ads8323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 03/31/2025] [Indexed: 05/04/2025]
Abstract
Intratumoral heterogeneity (ITH) is a critical factor associated with treatment failure and disease relapse in hepatocellular carcinoma (HCC). However, decoding ITH in a noninvasive and comprehensive manner remains a notable challenge. In this study involving 851 patients from five centers, we developed a noninvasive prognostic classification for ITH using radiomics based on multisequence MRI, termed radiomics ITH (RITH) phenotypes. The RITH phenotypes highly correlated with prognosis and pathological ITH. In addition, through an integrated multi-omics analysis, we uncovered the molecular mechanisms underlying RITH, notably enhancing its biological interpretability. Specifically, high-RITH tumors demonstrated an enrichment of cancer-associated fibroblasts and activation of extracellular matrix remodeling. Our approach facilitates the noninvasive refined classification of ITH using radiomics and multi-omics, paving the way for tailored treatment strategies in HCC. Extracellular matrix-receptor interaction could be a potential therapeutic target in patients with high-RITH tumors. Given the routine use of radiologic imaging in oncology, our methodology ignites versatile framework for broader application to other solid tumors.
Collapse
Affiliation(s)
- Yangyang Xie
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Fang Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, 430022 Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, 430022 Wuhan, China
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
| | - Jingwei Wei
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 100190 Beijing, China
- Beijing Key Laboratory of Molecular Imaging, 100190 Beijing, China
| | - Zefeng Shen
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Xue Song
- Department of Respiratory and Critical Care Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 310007 Hangzhou, China
| | - Yali Wang
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Hongjun Chen
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Liye Tao
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Junhao Zheng
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Lanfen Lin
- The College of Computer Science and Technology, Zhejiang University, 310027 Hangzhou, China
| | - Ziwei Niu
- The College of Computer Science and Technology, Zhejiang University, 310027 Hangzhou, China
| | - Xiaojun Guan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009 Hangzhou, China
| | - Tianhan Zhou
- Department of General Surgery, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 310007 Hangzhou, China
| | - Zhengao Xu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Yang Liu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Danwei Du
- Department of Anorectal, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 310000 Hangzhou, China
| | - Haoyu Pan
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Shihao Li
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Wenbin Ji
- Department of Radiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 317000 Taizhou, China
| | - Wei Zhou
- Department of Radiology, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, 313000 Huzhou, China
| | - Yunjun Yang
- Department of Radiology, The First Affiliated Hospital, Wenzhou Medical University, 325006 Wenzhou, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 100190 Beijing, China
- Beijing Key Laboratory of Molecular Imaging, 100190 Beijing, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 100191 Beijing, China
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, 710126 Xi’an, China
| | - Junjie Xu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Hongjie Hu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
| | - Xiao Liang
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- School of Medicine, Shaoxing University, 312000 Shaoxing, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, 310000 Hangzhou, China
| |
Collapse
|
4
|
Li C, Liao J, Chen B, Wang Q. Heterogeneity of the tumor immune cell microenvironment revealed by single-cell sequencing in head and neck cancer. Crit Rev Oncol Hematol 2025; 209:104677. [PMID: 40023465 DOI: 10.1016/j.critrevonc.2025.104677] [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: 12/05/2024] [Revised: 02/16/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025] Open
Abstract
Head and neck cancer (HNC) is the sixth most common disease in the world. The recurrence rate of patients is relatively high, and the heterogeneity of tumor immune microenvironment (TIME) cells may be an important reason for this. Single-cell sequencing (SCS) is currently the most promising and mature application in cancer research. It can identify unique genes expressed in cells and study tumor heterogeneity. According to current research, the heterogeneity of immune cells has become an important factor affecting the occurrence and development of HNC. SCSs can provide effective therapeutic targets and prognostic factors for HNC patients through analyses of gene expression levels and cell heterogeneity. Therefore, this study analyzes the basic theory of HNC and the development of SCS technology, elaborating on the application of SCS technology in HNC and its potential value in identifying HNC therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Jia Liao
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Bo Chen
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Qiang Wang
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan 629000, China.
| |
Collapse
|
5
|
Liu Z, Gu S, Peng Z, Wang Y, Li H, Zeng X, Wang H, Lv P, Wu Y, Zhou Y, Zhang Y, Jiang X, Fu P. Fusion of glioma-associated mesenchymal stem/stromal cells with glioma cells promotes macrophage recruitment and M2 polarization via m 6A modification of CSF1. Cell Death Dis 2025; 16:345. [PMID: 40287444 PMCID: PMC12033374 DOI: 10.1038/s41419-025-07678-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 04/10/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
Malignant glioma is the most common primary malignant tumor of the brain in adults, with glioblastoma (GBM) being the most aggressive subtype. Mesenchymal stem/stromal cells (MSCs) have been shown to fuse with tumor cells in various cancers including glioma, thereby regulating tumor progression. However, there has been no systematic research on the fusion of glioma-associated MSCs (GA-MSCs) with glioma cells. Here, it is shown that GA-MSCs are able to spontaneously fuse with glioma cells both in vitro and in vivo. The hybrid cells display significantly lower levels of N6-methyladenosine (m6A) modification and can modulate the glioma microenvironment by attracting and inducing M2-like polarization of macrophages. Mechanistically, the demethylase fat mass and obesity-associated protein (FTO) mediates demethylation in hybrids and promotes macrophage colony-stimulating factor (CSF1) secretion by increasing its RNA stability in an m6A-YTH domain family 2 (YTHDF2)-dependent manner. Our study reveals a novel crosstalk mechanism between glioma cells, GA-MSCs, and macrophages in glioma microenvironment, offering potential new approaches for glioma therapy.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sujie Gu
- Department of Neurosurgery, Henan Provincial People's Hospital, Zhengzhou, 450000, China
| | - Zesheng Peng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yihao Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Li
- Department of Cataract, Nanyang Eye Hospital, Nanyang, 473000, China
| | - Xiaoqing Zeng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Haofei Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Peng Lv
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuyi Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yan Zhou
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yanbin Zhang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Peng Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
6
|
Cao L, Bi W. METTL16/IGF2BP2 axis enhances malignant progression and DDP resistance through up-regulating COL4A1 by mediating the m6A methylation modification of LAMA4 in hepatocellular carcinoma. Cell Div 2025; 20:9. [PMID: 40251670 PMCID: PMC12008873 DOI: 10.1186/s13008-025-00152-2] [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: 12/31/2024] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the third most common malignant tumor after gastric cancer and esophageal cancer, which is a serious threat to human health. Methyltransferase-like protein 16 (METTL16) regulates the occurrence and development of various cancers, but its molecular mechanism in HCC has not been fully investigated. METHODS A series of databases were used to predict gene expression, methylation sites, correlation analysis, and protein interaction analysis. Gene expression levels were detected by quantitative real-time polymerase chain reaction (qRT-PCR), western blot, and immunohistochemistry (IHC). What's more, drug-resistant cell lines were established for drug resistance analysis. Cell proliferation was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and 5-ethynyl-2'-deoxyuridine (EdU) staining. Flow cytometry, transwell and wound healing assays were used for apoptosis, invasion and migration, respectively. In addition, the regulatory mechanism of METTL16 in HCC was investigated by methylated RNA immunoprecipitation (MeRIP), RNA immunoprecipitation (RIP) and co-immunoprecipitation (Co-IP). Finally, constructing subcutaneous transplanted tumor in nude mice confirmed the effect of METTL16 in vivo. RESULTS METTL16 was up-regulated in HCC drug-resistant tissues and cells. Knockdown of METTL16 inhibited Cisplatin (DDP) resistance, proliferation, invasion and migration of HCC cells, but promoted apoptosis. Besides, laminin subunit alpha 4 (LAMA4), which was overexpressed in HCC drug-resistant tissues and cells, was selected as the target of METTL16. Mechanistically, METTL16 and m6A reader insulin like growth factor 2 mRNA binding protein 2 (IGF2BP2) co-regulated the m6A modification and mRNA stability of LAMA4, and LAMA4 weakened the effects of METTL16 knockdown on HCC drug-resistance. Meanwhile, LAMA4 bound to collagen type IV alpha 1 chain (COL4A1) and facilitated DDP resistance and HCC progression via COL4A1. Similarly, in vivo, METTL16 induced tumor growth, as well as LAMA4 and COL4A1 expression, and increased DDP resistance. CONCLUSION METTL16 and IGF2BP2 jointly mediated the m6A methylation modification of LAMA4, thereby promoting DDP resistance and malignant progression of HCC through regulation of COL4A1.
Collapse
Affiliation(s)
- Liming Cao
- Department of General Surgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, China
| | - Wei Bi
- Department of General Surgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, China.
| |
Collapse
|
7
|
Tang C, Tamura-Nakano M, Tachibana K. Allogenic grafting induces PI3K-mediated tissue overgrowth in hydrozoan jellyfish Cladonema radiatum medusae. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025:105367. [PMID: 40185348 DOI: 10.1016/j.dci.2025.105367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/05/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Allorecognition, which is the ability of an organism to discriminate between self and non-self, protects multicellular animals from somatic cell/germline parasitism. We reported and characterised allorecognition in the stolons of colonies of the hydrozoan jellyfish Cladonema radiatum (C. radiatum) in our previous publication. C. radiatum has a free-swimming medusa form besides the sessile colonial form. In this study, we investigated the allorecognition responses in the medusa form of C. radiatum. By using grafting experiments, we observed that while C. radiatum medusae show tolerance to both isogenic and allogenic chimerism, allogenic grafting induces the formation of a form of circular scars-we refer to as "ring-shaped scars"-around the grafts on the host umbrella. Within the scars, overgrowth of tissues occurs with additional gastrovascular canal development. By pharmaceutical experiments, we found that tissue overgrowth is dependent on phosphoinositide 3-kinase (PI3K) and vascular endothelial growth factor (VEGF), showing a resemblance to mammalian neoplasia.
Collapse
Affiliation(s)
- Crystal Tang
- Laboratory of Chronobiology, School of Life Science and Technology, Tokyo Institute of Technology, 4259, Nagatsuta, Midori-ku, Yokohama, 226-8501, Japan.
| | - Miwa Tamura-Nakano
- Japan Institute for Health Security, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan.
| | - Kazunori Tachibana
- Laboratory of Chronobiology, School of Life Science and Technology, Tokyo Institute of Technology, 4259, Nagatsuta, Midori-ku, Yokohama, 226-8501, Japan.
| |
Collapse
|
8
|
Nesse RM, Labov JB, Madhavan G. Explanations for failures in designed and evolved systems. PNAS NEXUS 2025; 4:pgaf086. [PMID: 40309464 PMCID: PMC12041745 DOI: 10.1093/pnasnexus/pgaf086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 02/28/2025] [Indexed: 05/02/2025]
Abstract
Engineers have long studied the origins of design features that make machines prone to failure, but biologists have only recently begun investigating why organisms have traits that make them susceptible to disease. This article compares explanations for vulnerability to failure in machines with explanations for traits that make bodies vulnerable to disease. Some global explanations are relevant for both: design deficiencies, corrupted plans, assembly variations, incorrect operating environment, and trade-offs. These similarities suggest that a common framework for failure analysis could be valuable. However, a closer look at each of the 10 global categories reveals fundamental differences: machines are built to match an ideal blueprint, while species have no perfect genome or form. Design trade-offs in machines involve balancing multiple factors such as performance, robustness, and costs, while biological trade-offs maximize only gene transmission, often at the expense of health and lifespan. Detailed consideration of these and other differences reveals how the metaphor of body as a designed machine fosters tacit creationism that misrepresents the nature of organically complex systems.
Collapse
Affiliation(s)
- Randolph M Nesse
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ 85287, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jay B Labov
- National Academy of Engineering, Washington, DC 20001, USA
| | - Guru Madhavan
- National Academy of Engineering, Washington, DC 20001, USA
| |
Collapse
|
9
|
Deyra M, Gay C, Gerbaud L, Berland P, Pizon F. Perceptions of the determinants of health and cancer: trends in discourse and level of argumentation between girls and boys aged 6 to 18. Front Public Health 2025; 13:1390084. [PMID: 40034175 PMCID: PMC11872708 DOI: 10.3389/fpubh.2025.1390084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Aims To characterize the discourse trends and level of argumentation of girls and boys by analysing the differences in the conceptions and systems of conceptions of children and adolescents aged between 6 and 18. Materials and methods This is a multi-centre qualitative study in the human and social sciences, based on data collected in two different phases using two different tools: e.Photoexpression©, which deals with health in general, and Photonarration, which deals with cancer. The aim of this open, exploratory method, which uses photographs, is to gather data on the experiences and knowledge specific to each child and adolescent. The informative value of data from qualitative collection tools, combined with a mixed analysis methodology, enabled us to characterize the differences in perceptions of health determinants and cancer between girls and boys aged between 6 and 18. Results 4,174 productions were collected from 1,068 children aged 6 to 18, identifying 30 determinants of health and cancer. For all of these results, there were significant gender differences from a very early age: boys focused on leisure activities and physical activity, while girls took a more global view, focusing on the environment, food, emotional aspects of social relationships, hygiene, care, prevention, etc. As they got older, the focus shifted to health determinants. As we get older, we see a change in discourse trends and in the level of argumentation, with girls becoming richer and boys poorer. Discussion and outlook The trends in girls' and boys' discourse on what, in their view, determines health demonstrate the interest and relevance of adapting the prevention methods used as closely as possible to the conceptions of children and adolescents. The differences observed between boys and girls are a crucial lever that takes into account the specific characteristics of a population. They offer the possibility of taking more effective action, both in the context of interventions aimed at teenagers and in support of decision-making in the context of prevention policies.
Collapse
Affiliation(s)
- Maéliane Deyra
- Université Clermont Auvergne CNRS, SIGMA Clermont, Institut Pascal, Clermont-Ferrand, France
| | | | | | | | | |
Collapse
|
10
|
Mellon WJ, Sterner B, Ågren JA, Vincze O, Marx MT, Kapsetaki SE, Huang PH, Yavari B, McCollum HW, Natterson-Horowitz B, Human H, Baciu C, Richker H, Mallo D, Maley C, Harmon LJ, Compton ZT. Leveraging Comparative Phylogenetics for Evolutionary Medicine: Applications to Comparative Oncology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637459. [PMID: 39990350 PMCID: PMC11844554 DOI: 10.1101/2025.02.11.637459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Comparative phylogenetics provides a wealth of computational tools to understand evolutionary processes and their outcomes. Advances in these methodologies have occurred in parallel with a surge in cross-species genomic and phenotypic data. To date, however, the majority of published studies have focused on classical questions in evolutionary biology, such as speciation and the ecological drivers of trait evolution. Here, we argue that evolutionary medicine in general, and our understanding of the origin and diversification of disease traits in particular, would be greatly expanded by a wider integration of phylogenetic comparative methods (PCMs). We use comparative oncology - the study of cancer across the tree of life - as a case study to demonstrate the power of the approach and show that implementing PCMs can highlight the mode and tempo of the evolutionary changes in intrinsic, species-level disease vulnerabilities.
Collapse
Affiliation(s)
- Walker J Mellon
- Arizona Cancer Evolution Center, The Biodesign Institute, Tempe, AZ
| | - Beckett Sterner
- School of Life Sciences, Arizona State University, Tempe, AZ
| | - J Arvid Ågren
- Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - Orsolya Vincze
- Institute of Aquatic Ecology, Centre for Ecological Research, 4026 Debrecen, Hungary
- Evolutionary Ecology Group, Hungarian Department of Biology and Ecology, Babeş-Bolyai
| | - Matthew T Marx
- Arizona Cancer Evolution Center, The Biodesign Institute, Tempe, AZ
| | - Stefania E Kapsetaki
- Arizona Cancer Evolution Center, The Biodesign Institute, Tempe, AZ
- Frederick University, School of Health Sciences, Department of Pharmacology, Nicosia, Cyprus
- Hellenic Open University, Patras, Greece
| | - Ping-Han Huang
- School of Mathematical and Statistical Sciences, Arizona State University
| | - Bryan Yavari
- Arizona Cancer Evolution Center, The Biodesign Institute, Tempe, AZ
| | | | | | - Hannah Human
- Arizona Cancer Evolution Center, The Biodesign Institute, Tempe, AZ
| | - Cristina Baciu
- Arizona Cancer Evolution Center, The Biodesign Institute, Tempe, AZ
| | - Harley Richker
- Arizona Cancer Evolution Center, The Biodesign Institute, Tempe, AZ
| | - Diego Mallo
- Arizona Cancer Evolution Center, The Biodesign Institute, Tempe, AZ
| | - Carlo Maley
- Arizona Cancer Evolution Center, The Biodesign Institute, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe, AZ
| | - Luke J Harmon
- Department of Biological Sciences, University of Idaho, Moscow, Idaho
| | - Zachary T Compton
- Arizona Cancer Evolution Center, The Biodesign Institute, Tempe, AZ
- University of Arizona Cancer Center, Tucson, AZ
- University of Arizona College of Medicine, Tucson, AZ
| |
Collapse
|
11
|
Korsavidou Hult N, Tarai S, Hammarström K, Kullberg J, Lundström E, Bjerner T, Glimelius B, Ahlström H. Inclusion of tumor periphery in radiomics analysis of magnetic resonance images does not improve predictions of preoperative therapy response in patients with rectal cancer. Abdom Radiol (NY) 2025:10.1007/s00261-025-04815-0. [PMID: 39907722 DOI: 10.1007/s00261-025-04815-0] [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: 11/25/2024] [Revised: 01/10/2025] [Accepted: 01/17/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND/PURPOSE To evaluate the advantages of including versus excluding the tumor periphery and combining diffusion-weighted imaging (DWI) with T2-weighted imaging (T2w) for outcome predictions of preoperative radio(chemo)therapy in rectal cancer. METHODS Four analysis strategies, based on two segmentation methods and two magnetic resonance imaging (MRI) sequences, were evaluated in 106 patients examined with pretreatment MRI. One segmentation method included the tumor periphery in the region of interest (ROI) encompassing the whole tumor (wROI), considered as the reference segmentation approach, and one included only the central part (cROI). Relevant radiomics imaging features were extracted from either T2w alone or from both T2w and DWI and used by a machine learning algorithm for the prediction of pathologic complete response (pCR), neoadjuvant rectal (NAR) score, and disease recurrence. The area under the curve (AUC) was the performance measure. AUCs were compared with a bootstrapping method based on 104 bootstraps. RESULTS cROI applied to both T2w and DWI provided the highest numerical prediction of pCR (AUC 0.76), however, not significantly superior to the other strategies (p ≥ 0.138). cROI applied to both T2w and DWI also yielded the highest numerical prediction of NAR score (AUC 0.84), showing advantages over wROI-based analysis strategies (AUC 0.66 and 0.69; p ≤ 0.008). When compared to cROI applied to T2w alone (AUC 0.73), the benefit was borderline statistically significant (p = 0.053). For prediction of disease recurrence, no differences were found between the analysis strategies. CONCLUSIONS Inclusion of the tumor periphery in radiomic analysis of magnetic resonance images does not improve predictions of the preoperative therapy response in patients with rectal cancer. Excluding tumor periphery while adding DWI to T2w improves prediction of the NAR score, although it does not affect pCR or recurrence prediction.
Collapse
Affiliation(s)
- Nafsika Korsavidou Hult
- Radiology, Department of Surgical Sciences, Uppsala University, Akademiska Sjukhuset, Ingång 70, Uppsala, 751 85, Sweden.
| | - Sambit Tarai
- Radiology, Department of Surgical Sciences, Uppsala University, Akademiska Sjukhuset, Ingång 70, Uppsala, 751 85, Sweden
| | - Klara Hammarström
- Department of Immunology, Genetics and Pathology, Uppsala University, Dag Hammarskjölds v 20, Uppsala, 751 85, Sweden
| | - Joel Kullberg
- Radiology, Department of Surgical Sciences, Uppsala University, Akademiska Sjukhuset, Ingång 70, Uppsala, 751 85, Sweden
- Antaros Medical AB, GoCo House Entreprenörsstråket 10, Mölndal, 431 53, Sweden
| | - Elin Lundström
- Radiology, Department of Surgical Sciences, Uppsala University, Akademiska Sjukhuset, Ingång 70, Uppsala, 751 85, Sweden
| | - Tomas Bjerner
- Dept. of Health, Medicine and Caring Sciences (HMV), Division of Diagnostics and Specialist Medicine (DISP), Linköping University, Linköping, 581 83, Sweden
| | - Bengt Glimelius
- Department of Immunology, Genetics and Pathology, Uppsala University, Dag Hammarskjölds v 20, Uppsala, 751 85, Sweden
| | - Håkan Ahlström
- Radiology, Department of Surgical Sciences, Uppsala University, Akademiska Sjukhuset, Ingång 70, Uppsala, 751 85, Sweden
- Antaros Medical AB, GoCo House Entreprenörsstråket 10, Mölndal, 431 53, Sweden
| |
Collapse
|
12
|
Punzi S, Cittaro D, Gatti G, Crupi G, Botrugno OA, Cartalemi AA, Gutfreund A, Oneto C, Giansanti V, Battistini C, Santacatterina G, Patruno L, Villanti I, Palumbo M, Laverty DJ, Giannese F, Graudenzi A, Caravagna G, Antoniotti M, Nagel Z, Cavallaro U, Lanfrancone L, Yap TA, Draetta G, Balaban N, Tonon G. Early tolerance and late persistence as alternative drug responses in cancer. Nat Commun 2025; 16:1291. [PMID: 39900637 PMCID: PMC11790948 DOI: 10.1038/s41467-024-54728-7] [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: 04/11/2024] [Accepted: 11/20/2024] [Indexed: 02/05/2025] Open
Abstract
Bacteria withstand antibiotic treatment through three alternative mechanisms: resistance, persistence or tolerance. While resistance and persistence have been described, whether drug-induced tolerance exists in cancer cells remains largely unknown. Here, we show that human cancer cells elicit a tolerant response when exposed to commonly used chemotherapy regimens, propelled by the pervasive activation of autophagy, leading to the comprehensive activation of DNA damage repair pathways. After prolonged drug exposure, such tolerant responses morph into persistence, whereby the increased DNA damage repair is entirely reversed. The central regulator of mitophagy PINK1 drives this reduction in DNA repair via the cytoplasmic relocalization of the cell identity master HNF4A, thus hampering HNF4A transcriptional activation of DNA repair genes. We conclude that exposing cancer cells to relevant standard-of-care antitumour therapies induces a pervasive drug-induced tolerant response that might be broadly exploited to increase the impact of first-line, adjuvant treatments and debulking in advanced cancers.
Collapse
Affiliation(s)
- Simona Punzi
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Università Vita-Salute San Raffaele, Milan, Italy.
| | - Davide Cittaro
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Guido Gatti
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Gemma Crupi
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Oronza A Botrugno
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antonino Alex Cartalemi
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Alon Gutfreund
- The Racah Institute of Physics, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Caterina Oneto
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Giansanti
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Battistini
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - Giovanni Santacatterina
- Cancer Data Science Laboratory, Department of Mathematics and Geosciences, University of Trieste, Trieste, Italy
| | - Lucrezia Patruno
- Department of Informatics, Systems and Communication of the University of Milan-Bicocca, Milan, Italy
| | | | - Martina Palumbo
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Francesca Giannese
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alex Graudenzi
- Department of Informatics, Systems and Communication of the University of Milan-Bicocca, Milan, Italy
| | - Giulio Caravagna
- Cancer Data Science Laboratory, Department of Mathematics and Geosciences, University of Trieste, Trieste, Italy
| | - Marco Antoniotti
- Department of Informatics, Systems and Communication of the University of Milan-Bicocca, Milan, Italy
| | - Zachary Nagel
- Harvard Chan School of Public Health, Boston, MA, USA
| | - Ugo Cavallaro
- Unit of Gynaecological Oncology Research, European Institute of Oncology IRCSS, Milan, Italy
| | - Luisa Lanfrancone
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Timothy A Yap
- Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giulio Draetta
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center Houston, Houston, TX, USA
| | - Nathalie Balaban
- The Racah Institute of Physics, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Giovanni Tonon
- Functional Genomics of Cancer Unit, Division of Experimental Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Università Vita-Salute San Raffaele, Milan, Italy.
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
13
|
Liu Z, Wang Y, Peng Z, Li H, Wang H, Wu Y, Jiang X, Fu P. Fusion of tumor cells and mesenchymal stem/stroma cells: a source of tumor heterogeneity, evolution and recurrence. Med Oncol 2025; 42:52. [PMID: 39838167 DOI: 10.1007/s12032-024-02595-z] [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: 10/25/2024] [Accepted: 12/28/2024] [Indexed: 01/23/2025]
Abstract
The heterogeneity and evolution of tumors remain significant obstacles in cancer treatment, contributing to both therapy resistance and relapse. Mesenchymal stem/stromal cells (MSCs) are multipotent stromal cells within the tumor microenvironment that interact with tumor cells through various mechanisms, including cell fusion. While previous research has largely focused on the effects of MSC-tumor cell fusion on tumor proliferation, migration, and tumorigenicity, emerging evidence indicates that its role in tumor maintenance, evolution, and recurrence, particularly under stress conditions, may be even more pivotal. This review examines the connection between MSC-tumor cell fusion and several critical factors like tumor heterogeneity, cancer stem cells, and therapy resistance, highlighting the crucial role of cell fusion in tumor survival, evolution, and recurrence. Additionally, we explore potential therapeutic strategies aimed at targeting this process.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yihao Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zesheng Peng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Li
- Department of Cataract, Nanyang Eye Hospital, Nanyang, 473000, China
| | - Haofei Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuyi Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Peng Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
14
|
Derbal Y. Adaptive Treatment of Metastatic Prostate Cancer Using Generative Artificial Intelligence. Clin Med Insights Oncol 2025; 19:11795549241311408. [PMID: 39776668 PMCID: PMC11701910 DOI: 10.1177/11795549241311408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Despite the expanding therapeutic options available to cancer patients, therapeutic resistance, disease recurrence, and metastasis persist as hallmark challenges in the treatment of cancer. The rise to prominence of generative artificial intelligence (GenAI) in many realms of human activities is compelling the consideration of its capabilities as a potential lever to advance the development of effective cancer treatments. This article presents a hypothetical case study on the application of generative pre-trained transformers (GPTs) to the treatment of metastatic prostate cancer (mPC). The case explores the design of GPT-supported adaptive intermittent therapy for mPC. Testosterone and prostate-specific antigen (PSA) are assumed to be repeatedly monitored while treatment may involve a combination of androgen deprivation therapy (ADT), androgen receptor-signalling inhibitors (ARSI), chemotherapy, and radiotherapy. The analysis covers various questions relevant to the configuration, training, and inferencing of GPTs for the case of mPC treatment with a particular attention to risk mitigation regarding the hallucination problem and its implications to clinical integration of GenAI technologies. The case study provides elements of an actionable pathway to the realization of GenAI-assisted adaptive treatment of metastatic prostate cancer. As such, the study is expected to help facilitate the design of clinical trials of GenAI-supported cancer treatments.
Collapse
Affiliation(s)
- Youcef Derbal
- Ted Rogers School of Information Technology Management, Toronto Metropolitan University, Toronto, ON, Canada
| |
Collapse
|
15
|
Jakobsen MZ, Brøndum RF, Gregersen H, Due H, Dybkær K. A systematic literature review on clonal evolution events preceding relapse in multiple myeloma. Crit Rev Oncol Hematol 2025; 205:104560. [PMID: 39549892 DOI: 10.1016/j.critrevonc.2024.104560] [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: 09/19/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024] Open
Abstract
Despite considerable treatment advances, multiple myeloma (MM) remains an incurable hematological cancer due to treatment resistance. A systematic literature search was conducted to identify determinants for clonal evolution driving relapse and drug resistance in MM. A total of 631 non-duplicate publications were screened of which 28 articles were included for data extraction. Genetic alterations, mutational signatures, evolutionary trajectories, and non-genetic determinants were identified as key topics to characterize clonal evolution in relapsed MM. A variety of factors led to clonal diversification and increased tumor mutation burden, such as MAPK-Ras mutations and incremental changes related to chromosomal bands 1 and 17, while mutational signature analyses revealed that APOBEC activity and melphalan treatment leave a distinct impact on the clonal composition in MM genomes. To capture and dissect tumor heterogeneity, our review suggests combining methods or using technical approaches with high resolution to assess the impact of clonal evolution.
Collapse
Affiliation(s)
- Maja Zimmer Jakobsen
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Rasmus Froberg Brøndum
- Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Center for Clinical Data Science, Aalborg University, and Aalborg University Hospital, Aalborg, Denmark
| | - Henrik Gregersen
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Hanne Due
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Karen Dybkær
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark.
| |
Collapse
|
16
|
Li K, Huang J, Tan Y, Sun J, Zhou M. Single-cell and bulk transcriptome analysis reveals tumor cell heterogeneity and underlying molecular program in uveal melanoma. J Transl Med 2024; 22:1020. [PMID: 39533334 PMCID: PMC11555829 DOI: 10.1186/s12967-024-05831-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Uveal melanoma (UM) is a rare and deadly eye cancer with high metastatic potential. Despite the predominance of malignant cells within the tumor microenvironment, the heterogeneity and underlying molecular features remain to be fully characterized. METHODS We analyzed single-cell transcriptomic profiling of 37,660 malignant cells from 17 UM tumors. A consensus non-negative factorization algorithm was used to decipher transcriptional programs underlying tumor cell-intrinsic heterogeneity. Tumor-infiltrated immune cells were computationally estimated from bulk transcriptomes and bioinformatics methods. A gene signature was derived for subtyping and prognostic stratification and validated in multicenter cohorts. RESULTS ScRNA-seq analysis revealed the existence of diverse subpopulations and transcriptional variability among malignant cells within and between tumors. Furthermore, we observed that the heterogeneity of malignant cell states and compositions correlated with genomic, immunological, and clinical characteristics. By identifying gene expression programs associated with malignant cell heterogeneity at the single cell level, UM samples were classified into two distinct intra-tumoral subtypes (ITMHlo and ITMHhi) with different prognoses and immune microenvironments. Finally, a machine learning-derived 9-gene signature was developed to translate single-cell information into bulk tissue transcriptomes for patient stratification and was validated in multicenter cohorts. CONCLUSIONS Our study provides insight into understanding the intra-tumoral heterogeneity of UM and its potential impact on patient stratification.
Collapse
Affiliation(s)
- Ke Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jingzhe Huang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ying Tan
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jie Sun
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Meng Zhou
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- National Clinical Research Center for Ocular Diseases, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
17
|
Ahmed M, Kim DR. Life history dynamics of evolving tumors: insights into task specialization, trade-offs, and tumor heterogeneity. Cancer Cell Int 2024; 24:364. [PMID: 39506763 PMCID: PMC11539310 DOI: 10.1186/s12935-024-03538-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
The evolution of cancer cells parallels species evolution in numerous ways. Variations arise and spread under the pressure of competition between cancer cells. Current investigations of tumor evolution echo earlier debates between biologists. These include the role of non-Darwinian mechanisms, the contribution of neutral evolution, and life history dynamics. The trade-off between proliferation and metastasis is the most well-studied application of life history theory to cancer evolution. This article briefly introduces some parallels between cancer and species evolution, focusing on the life history of evolving tumors. Next, we review evidence from simulation and experimental studies supporting task specialization and trade-offs in cancer. We also cover recent work on inferring tumor tasks from data. We then turn to the implications of multi-tasking and the utility of the theory in explaining critical aspects of tumor heterogeneity. Finally, we discuss some of the criticism and future directions of this research topic.
Collapse
Affiliation(s)
- Mahmoud Ahmed
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University,College of Medicine, Jinju, South Korea
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University,College of Medicine, Jinju, South Korea.
| |
Collapse
|
18
|
Krishnamurthy K, Chai J, Liu X, Elsayad M, Goldstein DY. Intratumoral heterogeneity of oncogenic drivers in mixed histology lung adenocarcinomas: How tissue selection impacts molecular testing? Pathol Res Pract 2024; 263:155577. [PMID: 39265501 DOI: 10.1016/j.prp.2024.155577] [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: 06/23/2024] [Revised: 08/21/2024] [Accepted: 08/31/2024] [Indexed: 09/14/2024]
Abstract
Majority of the lung adenocarcinomas show a mixture of different histological patterns. The possibility of histologically heterogeneous areas of the adenocarcinoma showing genetic heterogeneity and harboring different driver mutations, with potentially significant clinical impact, has not been adequately addressed. Currently, there are no guidelines to suggest how to submit tumor tissue in adenocarcinomas with mixed histological features for molecular testing. The objective of this study is to assess intra-tumoral heterogeneity in prominent driver mutations among different morphological patterns of lung adenocarcinoma, its implications on the future of molecular testing as well as its potential impact on patient management. Twenty-three cases of mixed histology lung adenocarcinoma resected between 2018 and 2023 were retrieved from the archives. H&E slides were reviewed to identify the predominant and second most predominant histological patterns. The morphologically different tumor areas were manually macro-dissected for DNA extraction. Next-Generation Sequencing with Ion AmpliSeq™ Cancer Hotspot Panel v2 (Thermo Fisher Scientific, USA). Thirteen cases showed the same pathological variant in both histological components tested. Three cases (13 %) exhibited disparities in the variants detected across the different histological patterns tested (p=0.025). The discrepant findings had a direct therapeutic impact in 4.3 % cases. Seven cases showed no pathogenic variants detected on either of the histological components tested. This study elucidates the presence of infrequent yet significant intra-tumoral heterogeneity in the molecular profiles of mixed histology adenocarcinomas, highlighting the need for guidelines directing tissue selection for molecular testing to avoid missed therapeutic opportunities and mitigate disease relapse.
Collapse
Affiliation(s)
| | - Jiani Chai
- Montefiore medical Center, Bronx, NY, USA
| | | | | | - Doctor Y Goldstein
- Montefiore medical Center, Bronx, NY, USA; Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
19
|
Giraudeau M, Vincze O, Dupont SM, Sepp T, Baines C, Lemaitre JF, Lemberger K, Gentès S, Boddy A, Dujon AM, Bramwell G, Harris V, Ujvari B, Alix-Panabières C, Lair S, Sayag D, Conde DA, Colchero F, Harrison TM, Pavard S, Padilla-Morales B, Chevallier D, Hamede R, Roche B, Malkocs T, Aktipis AC, Maley C, DeGregori J, Loc’h GL, Thomas F. Approaches and methods to study wildlife cancer. J Anim Ecol 2024; 93:1410-1428. [PMID: 39189422 PMCID: PMC11745198 DOI: 10.1111/1365-2656.14144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/23/2024] [Indexed: 08/28/2024]
Abstract
The last few years have seen a surge of interest from field ecologists and evolutionary biologists to study neoplasia and cancer in wildlife. This contributes to the One Health Approach, which investigates health issues at the intersection of people, wild and domestic animals, together with their changing environments. Nonetheless, the emerging field of wildlife cancer is currently constrained by methodological limitations in detecting cancer using non-invasive sampling. In addition, the suspected differential susceptibility and resistance of species to cancer often make the choice of a unique model species difficult for field biologists. Here, we provide an overview of the importance of pursuing the study of cancer in non-model organisms and we review the currently available methods to detect, measure and quantify cancer in the wild, as well as the methodological limitations to be overcome to develop novel approaches inspired by diagnostic techniques used in human medicine. The methodology we propose here will help understand and hopefully fight this major disease by generating general knowledge about cancer, variation in its rates, tumour-suppressor mechanisms across species as well as its link to life history and physiological characters. Moreover, this is expected to provide key information about cancer in wildlife, which is a top priority due to the accelerated anthropogenic change in the past decades that might favour cancer progression in wild populations.
Collapse
Affiliation(s)
- Mathieu Giraudeau
- Littoral Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, La Rochelle, France
| | - Orsolya Vincze
- Littoral Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, La Rochelle, France
- ImmunoConcEpT, CNRS UMR 5164, University of Bordeaux, Bordeaux, France
- Hungarian Department of Biology and Ecology, Evolutionary Ecology Group, Babeş-Bolyai University, Cluj-Napoca, Romania
- HUN-REN-DE Conservation Biology Research Group, Debrecen, Hungary
| | - Sophie M. Dupont
- Littoral Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, La Rochelle, France
- Laboratoire de Biologie des ORganismes et Ecosystèmes Aquatiques (BOREA), FRE 2030, Muséum National d’Histoire Naturelle, CNRS, IRD, Sorbonne Université, Université de Caen Normandie, Université des Antilles, Paris, France
| | - Tuul Sepp
- Institute of Ecology and Earth Sciences, University of Tartu, Tartu, Estonia
| | - Ciara Baines
- Department of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Jean-Francois Lemaitre
- Laboratoire de Biométrie et Biologie Évolutive, CNRS, UMR5558, Université Lyon 1, Villeurbanne, France
| | | | - Sophie Gentès
- Littoral Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, La Rochelle, France
| | - Amy Boddy
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, California, USA
| | - Antoine M. Dujon
- School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Victoria, Australia
- CREEC/CANECEV, MIVEGEC, Unité Mixte de Recherches, IRD 224–CNRS5290–Université de Montpellier, Montpellier, France
| | - Georgina Bramwell
- School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Victoria, Australia
| | - Valerie Harris
- Arizona Cancer Evolution Center, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - Beata Ujvari
- School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Victoria, Australia
- Centre de Recherches Ecologiques et Evolutives sur le Cancer, Montpellier, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
| | - Stephane Lair
- Faculté de médecine vétérinaire, Canadian Wildlife Health Cooperative/Centre québécois sur la santé des animaux sauvages, Université de Montréal, Saint-Hyacinthe, Quebec, Canada
| | - David Sayag
- ONCOnseil—Unité d’expertise en oncologie vétérinaire, Toulouse, France
| | - Dalia A. Conde
- Department of Biology, University of Southern Denmark, Odense M, Denmark
- Interdisciplinary Centre on Population Dynamics, University of Southern Denmark, Odense M, Denmark
| | - Fernando Colchero
- Interdisciplinary Centre on Population Dynamics, University of Southern Denmark, Odense M, Denmark
- Department of Primate Behavior and Evolution, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Department of Mathematics and Computer Sciences, University of Southern Denmark, Odense M, Denmark
| | - Tara M. Harrison
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Samuel Pavard
- Unité Eco-Anthropologie (EA), Muséum National d’Histoire Naturelle, CNRS 7206, Université Paris Cité, Paris, France
| | - Benjamin Padilla-Morales
- Department of Biology and Biochemistry, Milner Centre for Evolution, University of Bath, Bath, UK
| | - Damien Chevallier
- Laboratoire de Biologie des ORganismes et Ecosystèmes Aquatiques (BOREA), FRE 2030, Muséum National d’Histoire Naturelle, CNRS, IRD, Sorbonne Université, Université de Caen Normandie, Université des Antilles, Paris, France
| | - Rodrigo Hamede
- Centre de Recherches Ecologiques et Evolutives sur le Cancer, Montpellier, France
- School of Natural Sciences, University of Tasmania, Hobart, Tasmania, Australia
| | - Benjamin Roche
- CREEC/CANECEV, MIVEGEC, Unité Mixte de Recherches, IRD 224–CNRS5290–Université de Montpellier, Montpellier, France
- Centre de Recherche en Écologie et Évolution de la Santé (CREES), Montpellier, France
- Departamento de Etología, Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Tamas Malkocs
- Littoral Environnement et Sociétés (LIENSs), UMR 7266 CNRS-La Rochelle Université, La Rochelle, France
- Univ Brest, CNRS, IRD, Ifremer, LEMAR, IUEM, Plouzane, France
| | - Athena C. Aktipis
- Arizona Cancer Evolution Center, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
- Department of Psychology, Arizona State University, Tempe, Arizona, USA
| | - Carlo Maley
- Arizona Cancer Evolution Center, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Frédéric Thomas
- CREEC/CANECEV, MIVEGEC, Unité Mixte de Recherches, IRD 224–CNRS5290–Université de Montpellier, Montpellier, France
- Centre de Recherche en Écologie et Évolution de la Santé (CREES), Montpellier, France
| |
Collapse
|
20
|
Cannataro VL, Glasmacher KA, Hampson CE. Mutations, substitutions, and selection: Linking mutagenic processes to cancer using evolutionary theory. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167268. [PMID: 38823460 DOI: 10.1016/j.bbadis.2024.167268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/25/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
Cancers are the product of evolutionary events, where molecular variation occurs and accumulates in tissues and tumors. Sequencing of this molecular variation informs not only which variants are driving tumorigenesis, but also the mechanisms behind what is fueling mutagenesis. Both of these details are crucial for preventing premature deaths due to cancer, whether it is by targeting the variants driving the cancer phenotype or by measures to prevent exogenous mutations from contributing to somatic evolution. Here, we review tools to determine both molecular signatures and cancer drivers, and avenues by which these metrics may be linked.
Collapse
Affiliation(s)
| | - Kira A Glasmacher
- Emmanuel College, 400 Fenway, Boston, MA 02115, United States of America
| | - Caralynn E Hampson
- Emmanuel College, 400 Fenway, Boston, MA 02115, United States of America
| |
Collapse
|
21
|
Casotti MC, Meira DD, Zetum ASS, Campanharo CV, da Silva DRC, Giacinti GM, da Silva IM, Moura JAD, Barbosa KRM, Altoé LSC, Mauricio LSR, Góes LSBDB, Alves LNR, Linhares SSG, Ventorim VDP, Guaitolini YM, dos Santos EDVW, Errera FIV, Groisman S, de Carvalho EF, de Paula F, de Sousa MVP, Fechine PBA, Louro ID. Integrating frontiers: a holistic, quantum and evolutionary approach to conquering cancer through systems biology and multidisciplinary synergy. Front Oncol 2024; 14:1419599. [PMID: 39224803 PMCID: PMC11367711 DOI: 10.3389/fonc.2024.1419599] [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: 04/30/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer therapy is facing increasingly significant challenges, marked by a wide range of techniques and research efforts centered around somatic mutations, precision oncology, and the vast amount of big data. Despite this abundance of information, the quest to cure cancer often seems more elusive, with the "war on cancer" yet to deliver a definitive victory. A particularly pressing issue is the development of tumor treatment resistance, highlighting the urgent need for innovative approaches. Evolutionary, Quantum Biology and System Biology offer a promising framework for advancing experimental cancer research. By integrating theoretical studies, translational methods, and flexible multidisciplinary clinical research, there's potential to enhance current treatment strategies and improve outcomes for cancer patients. Establishing stronger links between evolutionary, quantum, entropy and chaos principles and oncology could lead to more effective treatments that leverage an understanding of the tumor's evolutionary dynamics, paving the way for novel methods to control and mitigate cancer. Achieving these objectives necessitates a commitment to multidisciplinary and interprofessional collaboration at the heart of both research and clinical endeavors in oncology. This entails dismantling silos between disciplines, encouraging open communication and data sharing, and integrating diverse viewpoints and expertise from the outset of research projects. Being receptive to new scientific discoveries and responsive to how patients react to treatments is also crucial. Such strategies are key to keeping the field of oncology at the forefront of effective cancer management, ensuring patients receive the most personalized and effective care. Ultimately, this approach aims to push the boundaries of cancer understanding, treating it as a manageable chronic condition, aiming to extend life expectancy and enhance patient quality of life.
Collapse
Affiliation(s)
- Matheus Correia Casotti
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Débora Dummer Meira
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | | | | | - Giulia Maria Giacinti
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Iris Moreira da Silva
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - João Augusto Diniz Moura
- Laboratório de Oncologia Clínica e Experimental, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Karen Ruth Michio Barbosa
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Lorena Souza Castro Altoé
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | | | - Lyvia Neves Rebello Alves
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | - Vinícius do Prado Ventorim
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Yasmin Moreto Guaitolini
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | | | - Sonia Groisman
- Instituto de Biologia Roberto Alcântara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Elizeu Fagundes de Carvalho
- Instituto de Biologia Roberto Alcântara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Flavia de Paula
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | - Pierre Basílio Almeida Fechine
- Group of Chemistry of Advanced Materials (GQMat), Department of Analytical Chemistry and Physical-Chemistry, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
| | - Iuri Drumond Louro
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| |
Collapse
|
22
|
Liu B, Hu S, Wang X. Applications of single-cell technologies in drug discovery for tumor treatment. iScience 2024; 27:110486. [PMID: 39171294 PMCID: PMC11338156 DOI: 10.1016/j.isci.2024.110486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024] Open
Abstract
Single-cell technologies have been known as advanced and powerful tools to study tumor biological systems at the single-cell resolution and are playing increasingly critical roles in multiple stages of drug discovery and development. Specifically, single-cell technologies can promote the discovery of drug targets, help high-throughput screening at single-cell level, and contribute to pharmacokinetic studies of anti-tumor drugs. Emerging single-cell analysis technologies have been developed to further integrating multidimensional single-cell molecular features, expanding the scale of single-cell data, profiling phenotypic impact of genes in single cell, and providing full-length coverage single-cell sequencing. In this review, we systematically summarized the applications of single-cell technologies in various sections of drug discovery for tumor treatment, including target identification, high-throughput drug screening, and pharmacokinetic evaluation and highlighted emerging single-cell technologies in providing in-depth understanding of tumor biology. Single-cell-technology-based drug discovery is expected to further optimize therapeutic strategies and improve clinical outcomes of tumor patients.
Collapse
Affiliation(s)
- Bingyu Liu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Shunfeng Hu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
- Taishan Scholars Program of Shandong Province, Jinan, Shandong 250021, China
| |
Collapse
|
23
|
Chen K, Shuen TWH, Chow PKH. The association between tumour heterogeneity and immune evasion mechanisms in hepatocellular carcinoma and its clinical implications. Br J Cancer 2024; 131:420-429. [PMID: 38760445 PMCID: PMC11300599 DOI: 10.1038/s41416-024-02684-w] [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: 12/15/2023] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 05/19/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related mortality worldwide. The emergence of combination therapy, atezolizumab (anti-PDL1, immune checkpoint inhibitor) and bevacizumab (anti-VEGF) has revolutionised the management of HCC. Despite this breakthrough, the best overall response rate with first-line systemic therapy is only about 30%, owing to intra-tumoural heterogeneity, complex tumour microenvironment and the lack of predictive biomarkers. Many groups have attempted to classify HCC based on the immune microenvironment and have consistently observed better outcomes in immunologically "hot" HCC. We summarised possible mechanisms of tumour immune evasion based on the latest literature and the rationale for combination/sequential therapy to improve treatment response. Lastly, we proposed future strategies and therapies to overcome HCC immune evasion to further improve treatment outcomes of HCC.
Collapse
Affiliation(s)
- Kaina Chen
- Department of Gastroenterology & Hepatology, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Timothy W H Shuen
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Pierce K H Chow
- Duke-NUS Medical School, Singapore, Singapore.
- Department of Hepato-pancreato-biliary and Transplant Surgery, National Cancer Centre Singapore and Singapore General Hospital, Singapore, Singapore.
- Program in Translational and Clinical Liver Cancer Research, National Cancer Centre Singapore, Singapore, Singapore.
| |
Collapse
|
24
|
Zhang ZW, Zhang KX, Liao X, Quan Y, Zhang HY. Evolutionary screening of precision oncology biomarkers and its applications in prognostic model construction. iScience 2024; 27:109859. [PMID: 38799582 PMCID: PMC11126775 DOI: 10.1016/j.isci.2024.109859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/15/2024] [Accepted: 04/27/2024] [Indexed: 05/29/2024] Open
Abstract
Biomarker screening is critical for precision oncology. However, one of the main challenges in precision oncology is that the screened biomarkers often fail to achieve the expected clinical effects and are rarely approved by regulatory authorities. Considering the close association between cancer pathogenesis and the evolutionary events of organisms, we first explored the evolutionary feature underlying clinically approved biomarkers, and two evolutionary features of approved biomarkers (Ohnologs and specific evolutionary stages of genes) were identified. Subsequently, we utilized evolutionary features for screening potential prognostic biomarkers in four common cancers: head and neck squamous cell carcinoma, liver hepatocellular carcinoma, lung adenocarcinoma, and lung squamous cell carcinoma. Finally, we constructed an evolution-strengthened prognostic model (ESPM) for cancers. These models can predict cancer patients' survival time across different cancer cohorts effectively and perform better than conventional models. In summary, our study highlights the application potentials of evolutionary information in precision oncology biomarker screening.
Collapse
Affiliation(s)
- Zhi-Wen Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Ke-Xin Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Xuan Liao
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Yuan Quan
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Hong-Yu Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| |
Collapse
|
25
|
Li L, Xie W, Zhan L, Wen S, Luo X, Xu S, Cai Y, Tang W, Wang Q, Li M, Xie Z, Deng L, Zhu H, Yu G. Resolving tumor evolution: a phylogenetic approach. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:97-106. [PMID: 39282584 PMCID: PMC11390690 DOI: 10.1016/j.jncc.2024.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/28/2024] [Accepted: 03/20/2024] [Indexed: 09/19/2024] Open
Abstract
The evolutionary dynamics of cancer, characterized by its profound heterogeneity, demand sophisticated tools for a holistic understanding. This review delves into tumor phylogenetics, an essential approach bridging evolutionary biology with oncology, offering unparalleled insights into cancer's evolutionary trajectory. We provide an overview of the workflow, encompassing study design, data acquisition, and phylogeny reconstruction. Notably, the integration of diverse data sets emerges as a transformative step, enhancing the depth and breadth of evolutionary insights. With this integrated perspective, tumor phylogenetics stands poised to redefine our understanding of cancer evolution and influence therapeutic strategies.
Collapse
Affiliation(s)
- Lin Li
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wenqin Xie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Li Zhan
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shaodi Wen
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital, Nanjing, China
| | - Xiao Luo
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shuangbin Xu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Division of Laboratory Medicine, Microbiome Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yantong Cai
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Wenli Tang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Qianwen Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ming Li
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zijing Xie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Lin Deng
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hongyuan Zhu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guangchuang Yu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Thongkumkoon P, Sangphukieo A, Tongjai S, Noisagul P, Sangkhathat S, Laochareonsuk W, Kamolphiwong R, Budprom P, Teeyakasem P, Yongpitakwattana P, Thepbundit V, Sirikaew N, Klangjorhor J, Settakorn J, Moonmuang S, Suksakit P, Pasena A, Chaijaruwanich J, Yathongkhum W, Dissook S, Pruksakorn D, Chaiyawat P. Establishment, characterization, and genetic profiling of patient-derived osteosarcoma cells from a patient with retinoblastoma. Sci Rep 2024; 14:11056. [PMID: 38744935 PMCID: PMC11094034 DOI: 10.1038/s41598-024-60628-z] [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: 05/22/2023] [Accepted: 04/25/2024] [Indexed: 05/16/2024] Open
Abstract
Osteosarcoma is the most common malignant bone cancer in pediatric patients. Patients who respond poorly to chemotherapy experience worse clinical outcomes with a high mortality rate. The major challenge is the lack of effective drugs for these patients. To introduce new drugs for clinical approval, preclinical studies based on in vitro models must demonstrate the potency of the tested drugs, enabling the drugs to enter phase 1 clinical trials. Patient-derived cell culture is a promising testing platform for in vitro studies, as they more accurately recapitulate cancer states and genetic profiles compared to cell lines. In the present study, we established patient-derived osteosarcoma cells (PDC) from a patient who had previously been diagnosed with retinoblastoma. We identified a new variant of a germline mutation in the RB1 gene in the tissue of the patient. The biological effects of this PDC were studied to observe whether the cryopreserved PDC retained a feature of fresh PDC. The cryopreserved PDC preserved the key biological effects, including cell growth, invasive capability, migration, and mineralization, that define the conserved phenotypes compared to fresh PDC. From whole genome sequencing analysis of osteosarcoma tissue and patient-derived cells, we found that cryopreserved PDC was a minor population in the origin tissue and was selectively grown under the culture conditions. The cryopreserved PDC has a high resistance to conventional chemotherapy. This study demonstrated that the established cryopreserved PDC has the aggressive characteristics of osteosarcoma, in particular the chemoresistance phenotype that might be used for further investigation in the chemoresistant mechanism of osteosarcoma. In conclusion, the approach we applied for primary cell culture might be a promising method to generate in vitro models for functional testing of osteosarcoma.
Collapse
Affiliation(s)
- Patcharawadee Thongkumkoon
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Apiwat Sangphukieo
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Siripong Tongjai
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Pitiporn Noisagul
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Surasak Sangkhathat
- Division of Surgery, Faculty of Medicine, Prince of Songkla University, Hatyai, Songkhla, 90110, Thailand
- Translational Medicine Research Center, Prince of Songkla University, Hatyai, Songkhla, 90110, Thailand
| | - Wison Laochareonsuk
- Division of Surgery, Faculty of Medicine, Prince of Songkla University, Hatyai, Songkhla, 90110, Thailand
- Translational Medicine Research Center, Prince of Songkla University, Hatyai, Songkhla, 90110, Thailand
| | - Rawikant Kamolphiwong
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Piyaporn Budprom
- Faculty of Medicine, Musculoskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Pimpisa Teeyakasem
- Faculty of Medicine, Musculoskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Petlada Yongpitakwattana
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Viraporn Thepbundit
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 10 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Nutnicha Sirikaew
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 10 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Jeerawan Klangjorhor
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
- Faculty of Medicine, Musculoskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Jongkolnee Settakorn
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sutpirat Moonmuang
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Pathacha Suksakit
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Arnat Pasena
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Jeerayut Chaijaruwanich
- Department of Computer Science, Faculty of Science, Data Science Research Center, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Wilawan Yathongkhum
- Department of Computer Science, Faculty of Science, Data Science Research Center, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sivamoke Dissook
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 10 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand.
| | - Dumnoensun Pruksakorn
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand.
- Faculty of Medicine, Musculoskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Department of Orthopedics, Faculty of Medicine, Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand.
| | - Parunya Chaiyawat
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand.
- Faculty of Medicine, Musculoskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
27
|
Zhao Y, Zhang B, Ma Y, Guo M, Zhao F, Chen J, Wang B, Jin H, Zhou F, Guan J, Zhao Q, Liu Q, Wang H, Zhao F, Wang X. Distinct molecular profiles drive multifaceted characteristics of colorectal cancer metastatic seeds. J Exp Med 2024; 221:e20231359. [PMID: 38502057 PMCID: PMC10949939 DOI: 10.1084/jem.20231359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/10/2023] [Accepted: 02/08/2024] [Indexed: 03/20/2024] Open
Abstract
Metastasis of primary tumors remains a challenge for early diagnosis and prevention. The cellular properties and molecular drivers of metastatically competent clones within primary tumors remain unclear. Here, we generated 10-16 single cell-derived lines from each of three colorectal cancer (CRC) tumors to identify and characterize metastatic seeds. We found that intrinsic factors conferred clones with distinct metastatic potential and cellular communication capabilities, determining organ-specific metastasis. Poorly differentiated or highly metastatic clones, rather than drug-resistant clones, exhibited poor clinical prognostic impact. Personalized genetic alterations, instead of mutation burden, determined the occurrence of metastatic potential during clonal evolution. Additionally, we developed a gene signature for capturing metastatic potential of primary CRC tumors and demonstrated a strategy for identifying metastatic drivers using isogenic clones with distinct metastatic potential in primary tumors. This study provides insight into the origin and mechanisms of metastasis and will help develop potential anti-metastatic therapeutic targets for CRC patients.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Institute for Intelligent Healthcare, Tsinghua University, Beijing, China
| | - Bing Zhang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yiming Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengmeng Guo
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Fuqiang Zhao
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianan Chen
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bingzhi Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hua Jin
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Fulai Zhou
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Jiawei Guan
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Qian Zhao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Qian Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongying Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fangqing Zhao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Xia Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Institute for Intelligent Healthcare, Tsinghua University, Beijing, China
| |
Collapse
|
28
|
Ginno PA, Borgers H, Ernst C, Schneider A, Behm M, Aitken SJ, Taylor MS, Odom DT. Single-mitosis dissection of acute and chronic DNA mutagenesis and repair. Nat Genet 2024; 56:913-924. [PMID: 38627597 PMCID: PMC11096113 DOI: 10.1038/s41588-024-01712-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: 05/19/2023] [Accepted: 03/08/2024] [Indexed: 04/24/2024]
Abstract
How chronic mutational processes and punctuated bursts of DNA damage drive evolution of the cancer genome is poorly understood. Here, we demonstrate a strategy to disentangle and quantify distinct mechanisms underlying genome evolution in single cells, during single mitoses and at single-strand resolution. To distinguish between chronic (reactive oxygen species (ROS)) and acute (ultraviolet light (UV)) mutagenesis, we microfluidically separate pairs of sister cells from the first mitosis following burst UV damage. Strikingly, UV mutations manifest as sister-specific events, revealing mirror-image mutation phasing genome-wide. In contrast, ROS mutagenesis in transcribed regions is reduced strand agnostically. Successive rounds of genome replication over persisting UV damage drives multiallelic variation at CC dinucleotides. Finally, we show that mutation phasing can be resolved to single strands across the entire genome of liver tumors from F1 mice. This strategy can be broadly used to distinguish the contributions of overlapping cancer relevant mutational processes.
Collapse
Affiliation(s)
- Paul Adrian Ginno
- German Cancer Research Center (DKFZ), Division of Regulatory Genomics and Cancer Evolution, Heidelberg, Germany
| | - Helena Borgers
- German Cancer Research Center (DKFZ), Division of Regulatory Genomics and Cancer Evolution, Heidelberg, Germany
| | - Christina Ernst
- Cancer Research UK - Cambridge Institute, University of Cambridge, Cambridge, UK
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Anja Schneider
- German Cancer Research Center (DKFZ), Division of Regulatory Genomics and Cancer Evolution, Heidelberg, Germany
| | - Mikaela Behm
- German Cancer Research Center (DKFZ), Division of Regulatory Genomics and Cancer Evolution, Heidelberg, Germany
| | - Sarah J Aitken
- Cancer Research UK - Cambridge Institute, University of Cambridge, Cambridge, UK
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Martin S Taylor
- MRC Human Genetics Unit, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| | - Duncan T Odom
- German Cancer Research Center (DKFZ), Division of Regulatory Genomics and Cancer Evolution, Heidelberg, Germany.
- Cancer Research UK - Cambridge Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
29
|
Zhang L, Yang J, Huang J, Yu Y, Ding J, Karges J, Xiao H. Development of tumor-evolution-targeted anticancer therapeutic nanomedicineEVT. Chem 2024; 10:1337-1356. [DOI: 10.1016/j.chempr.2023.12.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
|
30
|
Xu J, Gao H, Guan X, Meng J, Ding S, Long Q, Yi W. Circulating tumor DNA: from discovery to clinical application in breast cancer. Front Immunol 2024; 15:1355887. [PMID: 38745646 PMCID: PMC11091288 DOI: 10.3389/fimmu.2024.1355887] [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: 12/14/2023] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Breast cancer (BC) stands out as the cancer with the highest incidence of morbidity and mortality among women worldwide, and its incidence rate is currently trending upwards. Improving the efficiency of breast cancer diagnosis and treatment is crucial, as it can effectively reduce the disease burden. Circulating tumor DNA (ctDNA) originates from the release of tumor cells and plays a pivotal role in the occurrence, development, and metastasis of breast cancer. In recent years, the widespread application of high-throughput analytical technology has made ctDNA a promising biomarker for early cancer detection, monitoring minimal residual disease, early recurrence monitoring, and predicting treatment outcomes. ctDNA-based approaches can effectively compensate for the shortcomings of traditional screening and monitoring methods, which fail to provide real-time information and prospective guidance for breast cancer diagnosis and treatment. This review summarizes the applications of ctDNA in various aspects of breast cancer, including screening, diagnosis, prognosis, treatment, and follow-up. It highlights the current research status in this field and emphasizes the potential for future large-scale clinical applications of ctDNA-based approaches.
Collapse
Affiliation(s)
- Jiachi Xu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, China
| | - Hongyu Gao
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, China
| | - Xinyu Guan
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, China
| | - Jiahao Meng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, China
| | - Shirong Ding
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qian Long
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, China
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center For Breast Disease In Hunan Province, Changsha, China
| |
Collapse
|
31
|
Li R, Shi F, Song L, Yu Z. scGAL: unmask tumor clonal substructure by jointly analyzing independent single-cell copy number and scRNA-seq data. BMC Genomics 2024; 25:393. [PMID: 38649804 PMCID: PMC11034052 DOI: 10.1186/s12864-024-10319-w] [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: 11/09/2023] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Accurately deciphering clonal copy number substructure can provide insights into the evolutionary mechanism of cancer, and clustering single-cell copy number profiles has become an effective means to unmask intra-tumor heterogeneity (ITH). However, copy numbers inferred from single-cell DNA sequencing (scDNA-seq) data are error-prone due to technically confounding factors such as amplification bias and allele-dropout, and this makes it difficult to precisely identify the ITH. RESULTS We introduce a hybrid model called scGAL to infer clonal copy number substructure. It combines an autoencoder with a generative adversarial network to jointly analyze independent single-cell copy number profiles and gene expression data from same cell line. Under an adversarial learning framework, scGAL exploits complementary information from gene expression data to relieve the effects of noise in copy number data, and learns latent representations of scDNA-seq cells for accurate inference of the ITH. Evaluation results on three real cancer datasets suggest scGAL is able to accurately infer clonal architecture and surpasses other similar methods. In addition, assessment of scGAL on various simulated datasets demonstrates its high robustness against the changes of data size and distribution. scGAL can be accessed at: https://github.com/zhyu-lab/scgal . CONCLUSIONS Joint analysis of independent single-cell copy number and gene expression data from a same cell line can effectively exploit complementary information from individual omics, and thus gives more refined indication of clonal copy number substructure.
Collapse
Affiliation(s)
- Ruixiang Li
- School of Information Engineering, Ningxia University, Yinchuan, 750021, China
| | - Fangyuan Shi
- School of Information Engineering, Ningxia University, Yinchuan, 750021, China
- Collaborative Innovation Center for Ningxia Big Data and Artificial Intelligence Co-founded by Ningxia Municipality and Ministry of Education, Ningxia University, Yinchuan, 750021, China
| | - Lijuan Song
- School of Information Engineering, Ningxia University, Yinchuan, 750021, China
- Collaborative Innovation Center for Ningxia Big Data and Artificial Intelligence Co-founded by Ningxia Municipality and Ministry of Education, Ningxia University, Yinchuan, 750021, China
| | - Zhenhua Yu
- School of Information Engineering, Ningxia University, Yinchuan, 750021, China.
- Collaborative Innovation Center for Ningxia Big Data and Artificial Intelligence Co-founded by Ningxia Municipality and Ministry of Education, Ningxia University, Yinchuan, 750021, China.
| |
Collapse
|
32
|
Oncel S, Safratowich BD, Lindlauf JE, Liu Z, Palmer DG, Briske-Anderson M, Zeng H. Efficacy of Butyrate to Inhibit Colonic Cancer Cell Growth Is Cell Type-Specific and Apoptosis-Dependent. Nutrients 2024; 16:529. [PMID: 38398853 PMCID: PMC10892417 DOI: 10.3390/nu16040529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Increasing dietary fiber consumption is linked to lower colon cancer incidence, and this anticancer effect is tied to elevated levels of short-chain fatty acids (e.g., butyrate) because of the fermentation of fiber by colonic bacteria. While butyrate inhibits cancer cell proliferation, the impact on cancer cell type remains largely unknown. To test the hypothesis that butyrate displays different inhibitory potentials due to cancer cell type, we determined half-maximal inhibitory concentrations (IC50) of butyrate in HCT116, HT-29, and Caco-2 human colon cancer cell proliferation at 24, 48, and 72 h. The IC50 (mM) butyrate concentrations of HCT116, HT-29, and Caco-2 cells were [24 h, 1.14; 48 h, 0.83; 72 h, 0.86], [24 h, N/D; 48 h, 2.42; 72 h, 2.15], and [24 h, N/D; 48 h, N/D; 72 h, 2.15], respectively. At the molecular level, phosphorylated ERK1/2 and c-Myc survival signals were decreased by (>30%) in HCT116, HT-29, and Caco-2 cells treated with 4 mM butyrate. Conversely, butyrate displayed a stronger potential (>1-fold) for inducing apoptosis and nuclear p21 tumor suppressor in HCT116 cells compared to HT-29 and Caco-2 cells. Moreover, survival analysis demonstrated that a cohort with high p21 gene expression in their colon tissue significantly increased survival time compared to a low-p21-expression cohort of colon cancer patients. Collectively, the inhibitory efficacy of butyrate is cell type-specific and apoptosis-dependent.
Collapse
Affiliation(s)
- Sema Oncel
- USDA-ARS Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA; (S.O.); (B.D.S.); (J.E.L.); (D.G.P.); (M.B.-A.)
| | - Bryan D. Safratowich
- USDA-ARS Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA; (S.O.); (B.D.S.); (J.E.L.); (D.G.P.); (M.B.-A.)
| | - James E. Lindlauf
- USDA-ARS Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA; (S.O.); (B.D.S.); (J.E.L.); (D.G.P.); (M.B.-A.)
| | - Zhenhua Liu
- School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA 01003, USA;
| | - Daniel G. Palmer
- USDA-ARS Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA; (S.O.); (B.D.S.); (J.E.L.); (D.G.P.); (M.B.-A.)
| | - Mary Briske-Anderson
- USDA-ARS Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA; (S.O.); (B.D.S.); (J.E.L.); (D.G.P.); (M.B.-A.)
| | - Huawei Zeng
- USDA-ARS Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA; (S.O.); (B.D.S.); (J.E.L.); (D.G.P.); (M.B.-A.)
| |
Collapse
|
33
|
Zhu Q, Dai H, Qiu F, Lou W, Wang X, Deng L, Shi C. Heterogeneity of computational pathomic signature predicts drug resistance and intra-tumor heterogeneity of ovarian cancer. Transl Oncol 2024; 40:101855. [PMID: 38185058 PMCID: PMC10808968 DOI: 10.1016/j.tranon.2023.101855] [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: 09/06/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
BACKGROUND Chemotherapy resistance is the main cause of ovarian cancer progression and even death. However, there are no clear indicators for predicting the risk of drug resistance in patients. Intra-tumor heterogeneity (ITH) is one of the characteristics of malignant tumors, which is associated with the treatment and prognosis of tumors. Accordingly, our study aims to investigate the correlation between the image features of intra-tumor heterogeneity and drug resistance of ovarian cancer based on artificial intelligence. METHODS We obtained hematoxylin and eosin staining frozen histopathological images of ovarian cancer and paracarcinoma tissues from the Cancer Genome Atlas. We extracted quantitative image features of whole-slide images based on the automatic image nuclear segmentation processing technology. After that, we used bioinformatics analysis to find the relationship between image features of intra-tumor heterogeneity and drug resistance. RESULTS Our results show that our automatic image processing process based on computer artificial intelligence can extract image features effectively, and the key image features extracted are closely related to ITH. Among them, the Perimeter.sd image feature with the most prominent ITH feature can accurately predict the risk of platinum-based chemotherapy drug resistance in ovarian cancer patients. CONCLUSION Automatic image processing and feature extraction based on artificial intelligence have excellent results. Perimeter.sd can be used as a useful image feature indicator for evaluating ITH. ITH is associated with drug resistance of ovarian cancer, so ITH characteristics can be used as an effective indicator to evaluate drug resistance in patients with ovarian cancer.
Collapse
Affiliation(s)
- Qiuli Zhu
- Department of Genetics, Gaoxin Branch of The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hua Dai
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Feng Qiu
- Department of Oncology, Gaoxin Branch of The First Affiliated Hospital of Nanchang University, No.7889 of Changdong avenue, Gaoxin District, Nanchang, Jiangxi, China
| | - Weiming Lou
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xin Wang
- Queen Mary School of Nanchang University, Nanchang University, Nanchang, China
| | - Libin Deng
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, China.
| | - Chao Shi
- Department of Oncology, Gaoxin Branch of The First Affiliated Hospital of Nanchang University, No.7889 of Changdong avenue, Gaoxin District, Nanchang, Jiangxi, China.
| |
Collapse
|
34
|
Mahgoub EO, Cho WC, Sharifi M, Falahati M, Zeinabad HA, Mare HE, Hasan A. Role of functional genomics in identifying cancer drug resistance and overcoming cancer relapse. Heliyon 2024; 10:e22095. [PMID: 38249111 PMCID: PMC10797146 DOI: 10.1016/j.heliyon.2023.e22095] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 10/28/2023] [Accepted: 11/03/2023] [Indexed: 01/23/2024] Open
Abstract
Functional genomics is an emerging field focused on elucidating the functions of genes or proteins, which can help solve challenges related to reliable cancer therapy. One of the main challenges currently faced by cancer therapy is the variations in the number of mutations in patients, leading to drug resistance and cancer relapses. Drug intrinsic or acquired resistance, is generally associated with most cancer relapses. There are advanced tools that can help identify the mutant genes in cancer tissues causing cancer drug resistance (CDR). Such tools include but are not limited to DNA and RNA sequencing as well assynthetic lethality gene screen (CRISPR)-based diagnosis. This review discusses the role of functional genomics in understanding CDR and finding tools for discovering drug target genes for cancer therapy.
Collapse
Affiliation(s)
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Majid Sharifi
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, 3614773947, Iran
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hojjat Alizadeh Zeinabad
- Apoptosis Research Centre, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Hany E. Mare
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35116, Egypt
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha, 2713, Qatar
- Biomedical Research Center, Qatar University, Doha, 2713, Qatar
| |
Collapse
|
35
|
Liu SJ, Casey-Clyde T, Cho NW, Swinderman J, Pekmezci M, Dougherty MC, Foster K, Chen WC, Villanueva-Meyer JE, Swaney DL, Vasudevan HN, Choudhury A, Pak J, Breshears JD, Lang UE, Eaton CD, Hiam-Galvez KJ, Stevenson E, Chen KH, Lien BV, Wu D, Braunstein SE, Sneed PK, Magill ST, Lim D, McDermott MW, Berger MS, Perry A, Krogan NJ, Hansen MR, Spitzer MH, Gilbert L, Theodosopoulos PV, Raleigh DR. Epigenetic reprogramming shapes the cellular landscape of schwannoma. Nat Commun 2024; 15:476. [PMID: 38216587 PMCID: PMC10786948 DOI: 10.1038/s41467-023-40408-5] [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: 02/06/2023] [Accepted: 05/25/2023] [Indexed: 01/14/2024] Open
Abstract
Mechanisms specifying cancer cell states and response to therapy are incompletely understood. Here we show epigenetic reprogramming shapes the cellular landscape of schwannomas, the most common tumors of the peripheral nervous system. We find schwannomas are comprised of 2 molecular groups that are distinguished by activation of neural crest or nerve injury pathways that specify tumor cell states and the architecture of the tumor immune microenvironment. Moreover, we find radiotherapy is sufficient for interconversion of neural crest schwannomas to immune-enriched schwannomas through epigenetic and metabolic reprogramming. To define mechanisms underlying schwannoma groups, we develop a technique for simultaneous interrogation of chromatin accessibility and gene expression coupled with genetic and therapeutic perturbations in single-nuclei. Our results elucidate a framework for understanding epigenetic drivers of tumor evolution and establish a paradigm of epigenetic and metabolic reprograming of cancer cells that shapes the immune microenvironment in response to radiotherapy.
Collapse
Affiliation(s)
- S John Liu
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
- Arc Institute, Palo Alto, CA, 94304, USA
| | - Tim Casey-Clyde
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Nam Woo Cho
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, and Departments of Otolaryngology, and Microbiology and Immunology, University of California San Francisco, San Francisco, CA, 94115, USA
| | - Jason Swinderman
- Arc Institute, Palo Alto, CA, 94304, USA
- Department of Urology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Melike Pekmezci
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Mark C Dougherty
- Departments of Otolaryngology and Neurosurgery, University of Iowa, Iowa City, IA, 52242, USA
| | - Kyla Foster
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - William C Chen
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Danielle L Swaney
- J. David Gladstone Institutes, California Institute for Quantitative Biosciences, Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Harish N Vasudevan
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Abrar Choudhury
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Joanna Pak
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
- Arc Institute, Palo Alto, CA, 94304, USA
| | - Jonathan D Breshears
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ursula E Lang
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Dermatology, University of California San Francisco, San Francisco, CA, 94115, USA
| | - Charlotte D Eaton
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Kamir J Hiam-Galvez
- Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, and Departments of Otolaryngology, and Microbiology and Immunology, University of California San Francisco, San Francisco, CA, 94115, USA
| | - Erica Stevenson
- J. David Gladstone Institutes, California Institute for Quantitative Biosciences, Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Kuei-Ho Chen
- J. David Gladstone Institutes, California Institute for Quantitative Biosciences, Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Brian V Lien
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - David Wu
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Steve E Braunstein
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Penny K Sneed
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Stephen T Magill
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - Daniel Lim
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | | | - Mitchel S Berger
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Arie Perry
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Nevan J Krogan
- J. David Gladstone Institutes, California Institute for Quantitative Biosciences, Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Marlan R Hansen
- Departments of Otolaryngology and Neurosurgery, University of Iowa, Iowa City, IA, 52242, USA
| | - Matthew H Spitzer
- Parker Institute for Cancer Immunotherapy, Chan Zuckerberg Biohub, and Departments of Otolaryngology, and Microbiology and Immunology, University of California San Francisco, San Francisco, CA, 94115, USA
| | - Luke Gilbert
- Arc Institute, Palo Alto, CA, 94304, USA
- Department of Urology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Philip V Theodosopoulos
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - David R Raleigh
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA.
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA.
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
36
|
Li K, You G, Jiang K, Wang R, Li W, Meng Y, Fang Y, Chen W, Zhu G, Song J, Wang W, Su H, Hu B, Sun F, Jia Z, Li C, Zhu J. Root extract of Hemsleya amabilis Diels suppresses renal cell carcinoma cell growth through inducing apoptosis and G 2/M phase arrest via PI3K/AKT signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:117014. [PMID: 37557938 DOI: 10.1016/j.jep.2023.117014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/16/2023] [Accepted: 08/07/2023] [Indexed: 08/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hemsleya amabilis Diels, belongs to cucurbitaceae, was traditional Chinese medicine (TCM). It is widely used to treat various diseases. However, these diseases may contribute to the development of RCC. AIM OF THE STUDY investigated the anticancer activities of root extract of Hemsleya amabilis Diels (HRE), and elucidated the underlying molecular mechanism in vivo and in vitro. MATERIALS AND METHODS Dried Hemsleya amabilis Diels roots were extracted by ethyl acetate and used to treat RCC4, OS-RC-2 and ACHN cells. UHPLC-MS was used to analyze the chemical composition of the extract. CCK-8 and colony formation assay were used to investigate proliferation. PI staining was used to detect cell cycle. Annexin-V-FITC, AO/EB and TEM were used to evaluate apoptosis. Transwell and wound healing assays were used to evaluate migration and invasion. RNA-seq, Network pharmacology, autodocking for virtual screening and molecular dynamics simulation were used to analyze potential molecular mechanisms and active components of HRE inhibiting proliferation of RCC. LY294002 and UC2288 were used to inhibit PI3K and P21 expression, respectively. IGF-1 was used to activate PI3K. Xenograft tumor model was established to evaluate its anti-tumor potential in vivo. Immunohistochemistry and Western blot were used to test protein expression levels. H&E staining was used to explore the side effects of HRE in vivo. Applying bioinformatics to analyze the effect of P21 on RCC. RESULTS HRE consists of 739 compounds. CCK-8 and colony formation assay showed that HRE significantly inhibited RCC cells proliferation. PI staining indicated that HRE caused G2/M phase arrest. Annexin-V-FITC, AO/EB and TEM experiments revealed that HRE significantly promoted apoptosis of RCC cells. Transwell and wound healing assays showed that HRE can inhibit the migration and invasion of RCC cells. RNA-seq showed that HRE induced 230 gene changes. Network pharmacology analysis found the relationship between HRE-component-target-RCC. Auto-docking found that Epitulipinolide diepoxide in HRE can stably bind to PIK3CA (-7.22 kJ/mol), and molecular dynamics simulation verified the combination between Epitulipinolide diepoxide of PIK3CA. In RCC4 cells, pretreatment with IGF-1, attenuated HRE-induced apoptosis and G2/M arrest. When pretreated with PIK3 inhibitor LY294002, the opposite result appears. Pretreatment with CDKN1A (P21) inhibitor UC2288 attenuated HRE-induced G2/M arrest. Xenograft tumor model showed that HRE inhibited tumor growth. Western blot analysis indicated that HRE can regulating Bax, Bcl-2, PARP, cleared-PARP, Caspase-9, Caspase-8, Caspase-3, Survivin, Cyclin-B1, CDK1, N-cadherin, snail, slug, E-cadherin, MMP-9. Immunohistochemical staining showed that in the treated group, expression of E-cadherin, Bax, P21 was up-regulated, while N-cadherin, PI3K, AKT and Bcl-2 were down-regulated. H&E staining showed that compared to control groups, the main organs in the HRE-treated groups showed no histological abnormalities. The overall survival rate of RCC patients in the high-expression group of P21 was higher than in the low-expression group of P21 on bioinformatics analysis. CONCLUSIONS HRE inhibited RCC migration and invasion through EMT, and inhibited proliferation in vivo and in vitro. In addition, HRE inhibited proliferation through promoting apoptosis and P21-induced G2/M phase arrest via PI3K/AKT signaling pathway. Overall, these results suggest that HRE may be a promising chemotherapy agent for RCC.
Collapse
Affiliation(s)
- Kai Li
- Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, 550025, China
| | - Ganhua You
- The Second People's Hospital of Guizhou Province, Guiyang, 550002, China
| | - Kehua Jiang
- Department of Urology, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550002, China
| | - Rongpin Wang
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550002, China
| | - Wuchao Li
- Department of Radiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550002, China
| | - Yonglu Meng
- Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, 550025, China
| | - Yinyi Fang
- Medical College of Guizhou University, Guiyang, Guizhou Province, 550025, China
| | - Weiming Chen
- Medical College of Guizhou University, Guiyang, Guizhou Province, 550025, China
| | - Guohua Zhu
- Department of Pedictric, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550002, China
| | - Jukun Song
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, China
| | - Wei Wang
- Department of Pedictric, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550002, China
| | - Hao Su
- Department of Urology, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550002, China
| | - Bin Hu
- Department of Urology, Kweichow Moutai Hospital, Renhuai, China
| | - Fa Sun
- Department of Urology, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550002, China.
| | - Zhenyu Jia
- University of California of Riverside, Riverside, CA, 92521, USA.
| | - Can Li
- Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, 550025, China.
| | - Jianguo Zhu
- Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, 550025, China; Department of Urology, Guizhou Provincial People's Hospital, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, 550002, China.
| |
Collapse
|
37
|
Luo F, Li H, Ma W, Cao J, Chen Q, Lu F, Qiu M, Zhou P, Xia Z, Zeng K, Zhan J, Zhou T, Luo Q, Pan W, Zhang L, Lin C, Huang Y, Zhang L, Yang D, Zhao H. The BCL-2 inhibitor APG-2575 resets tumor-associated macrophages toward the M1 phenotype, promoting a favorable response to anti-PD-1 therapy via NLRP3 activation. Cell Mol Immunol 2024; 21:60-79. [PMID: 38062129 PMCID: PMC10757718 DOI: 10.1038/s41423-023-01112-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 10/12/2023] [Accepted: 11/13/2023] [Indexed: 01/01/2024] Open
Abstract
The main challenges in the use of immune checkpoint inhibitors (ICIs) are ascribed to the immunosuppressive tumor microenvironment and the lack of sufficient infiltration of activated CD8+ T cells. Transforming the tumor microenvironment (TME) from "cold" to "hot" and thus more likely to potentiate the effects of ICIs is a promising strategy for cancer treatment. We found that the selective BCL-2 inhibitor APG-2575 can enhance the antitumor efficacy of anti-PD-1 therapy in syngeneic and humanized CD34+ mouse models. Using single-cell RNA sequencing, we found that APG-2575 polarized M2-like immunosuppressive macrophages toward the M1-like immunostimulatory phenotype with increased CCL5 and CXCL10 secretion, restoring T-cell function and promoting a favorable immunotherapy response. Mechanistically, we demonstrated that APG-2575 directly binds to NF-κB p65 to activate NLRP3 signaling, thereby mediating macrophage repolarization and the activation of proinflammatory caspases and subsequently increasing CCL5 and CXCL10 chemokine production. As a result, APG-2575-induced macrophage repolarization could remodel the tumor immune microenvironment, thus improving tumor immunosuppression and further enhancing antitumor T-cell immunity. Multiplex immunohistochemistry confirmed that patients with better immunotherapeutic efficacy had higher CD86, p-NF-κB p65 and NLRP3 levels, accompanied by lower CD206 expression on macrophages. Collectively, these data provide evidence that further study on APG-2575 in combination with immunotherapy for tumor treatment is required.
Collapse
Affiliation(s)
- Fan Luo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Han Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenjuan Ma
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiaxin Cao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qun Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Feiteng Lu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Miaozhen Qiu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Penghui Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zengfei Xia
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kangmei Zeng
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianhua Zhan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ting Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiuyun Luo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wentao Pan
- Ascentage Pharma (Suzhou) Co Ltd, 218 Xinghu Street, Suzhou, Jiangsu Province, China
| | - Lin Zhang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chaozhuo Lin
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan Huang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Dajun Yang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| | - Hongyun Zhao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
38
|
Wang L, Diao M, Zhang Z, Jiang M, Chen S, Zhao D, Liu Z, Zhou C. Comparison of the somatic genomic landscape between central- and peripheral-type non-small cell lung cancer. Lung Cancer 2024; 187:107439. [PMID: 38113653 DOI: 10.1016/j.lungcan.2023.107439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023]
Abstract
OBJECTIVE Lung cancer is classified into central and peripheral types based on the anatomic location. The present study aimed to explore the distinct patterns of genomic alterations between central- and peripheral-type non-small cell lung cancers (NSCLCs) with negative driver genes and identify potential driver genes and biomarkers to improve therapy strategies for NSCLC. METHODS Whole-exome sequencing (WES) was performed with 182 tumor/control pairs of samples from 145 Chinese NSCLC patients without EGFR, ALK, or ROS1 alterations. Significantly mutated genes (SMGs) and somatic copy number alterations (SCNAs) were identified. Subsequently, tumor mutation burden (TMB), weighted genome integrity index (wGII), copy number alteration (CNA) burden, Shannon diversity index (SDI), intratumor heterogeneity (ITH), neoantigen load (NAL), and clonal variations were evaluated in central- and peripheral-type NSCLCs. Furthermore, mutational signature analysis and survival analysis were performed. RESULTS TP53 was the most frequently mutated gene in NSCLC and more frequently mutated in central-type NSCLC. Higher wGII, ITH, and SDI were found in central-type lung adenocarcinoma (LUAD) than in peripheral-type LUAD. The NAL of central-type lung squamous cell carcinoma (LUSC) with stage III/IV was significantly higher than that of peripheral-type LUSC. Mutational signature analysis revealed that SBS10b, SBS24, and ID7 were significantly different in central- and peripheral-type NSCLCs. Furthermore, central-type NSCLC was found to evolve at a higher level with fewer clones and more subclones, particularly in central-type LUSC. Survival analysis revealed that TMB, CNA burden, NAL, subclonal driver mutations, and subclonal mutations were negatively related to the overall survival (OS) and the progression-free survival (PFS) of central-type LUAD. CONCLUSIONS Central-type NSCLC tended to evolve at a higher level and might suggest a favorable response to immunotherapy. Our study also identified several potential driver genes and promising biomarkers for the prognosis and prediction of chemotherapy responses in NSCLC.
Collapse
Affiliation(s)
- Lei Wang
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Meng Diao
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Zheng Zhang
- Department of Radiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Minlin Jiang
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, PR China
| | - Shifu Chen
- HaploX Biotechnology Co., Shenzhen, PR China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, PR China.
| | - Zhenguo Liu
- Department of Anesthesiology, Weifang People's Hospital, Weifang, Shandong Province, PR China.
| | - Caicun Zhou
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, PR China.
| |
Collapse
|
39
|
Vukovic Đerfi K, Vasiljevic T, Matijevic Glavan T. Recent Advances in the Targeting of Head and Neck Cancer Stem Cells. APPLIED SCIENCES 2023; 13:13293. [DOI: 10.3390/app132413293] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a very heterogeneous cancer with a poor overall response to therapy. One of the reasons for this therapy resistance could be cancer stem cells (CSCs), a small population of cancer cells with self-renewal and tumor-initiating abilities. Tumor cell heterogeneity represents hurdles for therapeutic elimination of CSCs. Different signaling pathway activations, such as Wnt, Notch, and Sonic-Hedgehog (SHh) pathways, lead to the expression of several cancer stem factors that enable the maintenance of CSC features. Identification and isolation of CSCs are based either on markers (CD133, CD44, and aldehyde dehydrogenase (ALDH)), side populations, or their sphere-forming ability. A key challenge in cancer therapy targeting CSCs is overcoming chemotherapy and radiotherapy resistance. However, in novel therapies, various approaches are being employed to address this hurdle such as targeting cell surface markers, other stem cell markers, and different signaling or metabolic pathways, but also, introducing checkpoint inhibitors and natural compounds into the therapy can be beneficial.
Collapse
Affiliation(s)
- Kristina Vukovic Đerfi
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Ruđer Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| | - Tea Vasiljevic
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Ruđer Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| | - Tanja Matijevic Glavan
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Ruđer Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| |
Collapse
|
40
|
Wrenn ED, Apfelbaum AA, Rudzinski ER, Deng X, Jiang W, Sud S, Van Noord RA, Newman EA, Garcia NM, Miyaki A, Hoglund VJ, Bhise SS, Kanaan SB, Waltner OG, Furlan SN, Lawlor ER. Cancer-Associated Fibroblast-Like Tumor Cells Remodel the Ewing Sarcoma Tumor Microenvironment. Clin Cancer Res 2023; 29:5140-5154. [PMID: 37471463 PMCID: PMC10801911 DOI: 10.1158/1078-0432.ccr-23-1111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/07/2023] [Accepted: 07/18/2023] [Indexed: 07/22/2023]
Abstract
PURPOSE Despite limited genetic and histologic heterogeneity, Ewing sarcoma (EwS) tumor cells are transcriptionally heterogeneous and display varying degrees of mesenchymal lineage specification in vitro. In this study, we investigated if and how transcriptional heterogeneity of EwS cells contributes to heterogeneity of tumor phenotypes in vivo. EXPERIMENTAL DESIGN Single-cell proteogenomic-sequencing of EwS cell lines was performed and integrated with patient tumor transcriptomic data. Cell subpopulations were isolated by FACS for assessment of gene expression and phenotype. Digital spatial profiling and human whole transcriptome analysis interrogated transcriptomic heterogeneity in EwS xenografts. Tumor cell subpopulations and matrix protein deposition were evaluated in xenografts and patient tumors using multiplex immunofluorescence staining. RESULTS We identified CD73 as a biomarker of highly mesenchymal EwS cell subpopulations in tumor models and patient biopsies. CD73+ tumor cells displayed distinct transcriptional and phenotypic properties, including selective upregulation of genes that are repressed by EWS::FLI1, and increased migratory potential. CD73+ cells were distinguished in vitro and in vivo by increased expression of matrisomal genes and abundant deposition of extracellular matrix (ECM) proteins. In epithelial-derived malignancies, ECM is largely deposited by cancer-associated fibroblasts (CAF), and we thus labeled CD73+ EwS cells, CAF-like tumor cells. Marked heterogeneity of CD73+ EwS cell frequency and distribution was detected in tumors in situ, and CAF-like tumor cells and associated ECM were observed in peri-necrotic regions and invasive foci. CONCLUSIONS EwS tumor cells can adopt CAF-like properties, and these distinct cell subpopulations contribute to tumor heterogeneity by remodeling the tumor microenvironment. See related commentary by Kuo and Amatruda, p. 5002.
Collapse
Affiliation(s)
- Emma D. Wrenn
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA
| | - April A. Apfelbaum
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA
- Cancer Biology PhD Program, University of Michigan, Ann Arbor, Michigan
| | - Erin R. Rudzinski
- Pathology Department, Seattle Children’s Hospital, Seattle, Washington
| | - Xuemei Deng
- Pathology Department, Seattle Children’s Hospital, Seattle, Washington
| | - Wei Jiang
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Sudha Sud
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | | | - Erika A. Newman
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Nicolas M. Garcia
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA
| | - Aya Miyaki
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA
| | - Virginia J. Hoglund
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA
| | - Shruti S. Bhise
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Sami B. Kanaan
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Olivia G. Waltner
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Scott N. Furlan
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Pediatrics, University of Washington, Seattle, WA
| | - Elizabeth R. Lawlor
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Pediatrics, University of Washington, Seattle, WA
| |
Collapse
|
41
|
Liu Z, Xu X, Liu H, Zhao X, Yang C, Fu R. Immune checkpoint inhibitors for multiple myeloma immunotherapy. Exp Hematol Oncol 2023; 12:99. [PMID: 38017516 PMCID: PMC10685608 DOI: 10.1186/s40164-023-00456-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/02/2023] [Indexed: 11/30/2023] Open
Abstract
Multiple myeloma (MM) is related to immune disorders, recent studys have revealed that immunotherapy can greatly benefit MM patients. Immune checkpoints can negatively modulate the immune system and are closely associated with immune escape. Immune checkpoint-related therapy has attracted much attention and research in MM. However, the efficacy of those therapies need further improvements. There need more thoughts about the immune checkpoint to translate their use in clinical work. In our review, we aggregated the currently known immune checkpoints and their corresponding ligands, further more we propose various ways of potential translation applying treatment based on immune checkpoints for MM patients.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xintong Xu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xianghong Zhao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chun Yang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
42
|
Li H, Yang Z, Tu F, Deng L, Han Y, Fu X, Wang L, Gu D, Werner B, Huang W. Mutation divergence over space in tumour expansion. J R Soc Interface 2023; 20:20230542. [PMID: 37989227 PMCID: PMC10681009 DOI: 10.1098/rsif.2023.0542] [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: 09/16/2023] [Accepted: 10/30/2023] [Indexed: 11/23/2023] Open
Abstract
Mutation accumulation in tumour evolution is one major cause of intra-tumour heterogeneity (ITH), which often leads to drug resistance during treatment. Previous studies with multi-region sequencing have shown that mutation divergence among samples within the patient is common, and the importance of spatial sampling to obtain a complete picture in tumour measurements. However, quantitative comparisons of the relationship between mutation heterogeneity and tumour expansion modes, sampling distances as well as the sampling methods are still few. Here, we investigate how mutations diverge over space by varying the sampling distance and tumour expansion modes using individual-based simulations. We measure ITH by the Jaccard index between samples and quantify how ITH increases with sampling distance, the pattern of which holds in various sampling methods and sizes. We also compare the inferred mutation rates based on the distributions of variant allele frequencies under different tumour expansion modes and sampling sizes. In exponentially fast expanding tumours, a mutation rate can always be inferred for any sampling size. However, the accuracy compared with the true value decreases when the sampling size decreases, where small sampling sizes result in a high estimate of the mutation rate. In addition, such an inference becomes unreliable when the tumour expansion is slow, such as in surface growth.
Collapse
Affiliation(s)
- Haiyang Li
- Group of Theoretical Biology, The State Key Laboratory of Bio-control, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People’s Republic of China
- Evolutionary Dynamics Group, Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Zixuan Yang
- Group of Theoretical Biology, The State Key Laboratory of Bio-control, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People’s Republic of China
| | - Fengyu Tu
- Group of Theoretical Biology, The State Key Laboratory of Bio-control, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People’s Republic of China
| | - Lijuan Deng
- Group of Theoretical Biology, The State Key Laboratory of Bio-control, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People’s Republic of China
| | - Yuqing Han
- Group of Theoretical Biology, The State Key Laboratory of Bio-control, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People’s Republic of China
| | - Xing Fu
- Group of Theoretical Biology, The State Key Laboratory of Bio-control, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People’s Republic of China
| | - Long Wang
- Group of Theoretical Biology, The State Key Laboratory of Bio-control, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People’s Republic of China
| | - Di Gu
- The first affiliated hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Benjamin Werner
- Evolutionary Dynamics Group, Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Weini Huang
- Group of Theoretical Biology, The State Key Laboratory of Bio-control, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, People’s Republic of China
- School of Mathematical Sciences, Queen Mary University of London, London, UK
| |
Collapse
|
43
|
Mansur MB, deSouza NM, Natrajan R, Abegglen LM, Schiffman JD, Greaves M. Evolutionary determinants of curability in cancer. Nat Ecol Evol 2023; 7:1761-1770. [PMID: 37620552 DOI: 10.1038/s41559-023-02159-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 07/05/2023] [Indexed: 08/26/2023]
Abstract
The emergence of drug-resistant cells, most of which have a mutated TP53 gene, prevents curative treatment in most advanced and common metastatic cancers of adults. Yet, a few, rarer malignancies, all of which are TP53 wild type, have high cure rates. In this Perspective, we discuss how common features of curable cancers offer insights into the evolutionary and developmental determinants of drug resistance. Acquired loss of TP53 protein function is the most common genetic change in cancer. This probably reflects positive selection in the context of strong ecosystem pressures including microenvironmental hypoxia. Loss of TP53's functions results in multiple fitness benefits and enhanced evolvability of cancer cells. TP53-null cells survive apoptosis, and tolerate potent oncogenic signalling, DNA damage and genetic instability. In addition, critically, they provide an expanded pool of self-renewing, or stem, cells, the primary units of evolutionary selection in cancer, making subsequent adaptation to therapeutic challenge by drug resistance highly probable. The exceptional malignancies that are curable, including the common genetic subtype of childhood acute lymphoblastic leukaemia and testicular seminoma, differ from the common adult cancers in originating prenatally from embryonic or fetal cells that are developmentally primed for TP53-dependent apoptosis. Plus, they have other genetic and phenotypic features that enable dissemination without exposure to selective pressures for TP53 loss, retaining their intrinsic drug hypersensitivity.
Collapse
Affiliation(s)
| | - Nandita M deSouza
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
- Department of Imaging, The Royal Marsden National Health Service (NHS) Foundation Trust, London, UK
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, Division of Breast Cancer, The Institute of Cancer Research, London, UK
| | - Lisa M Abegglen
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Joshua D Schiffman
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Peel Therapeutics, Inc., Salt Lake City, UT, USA
| | - Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| |
Collapse
|
44
|
Dey DK, Krause D, Rai R, Choudhary S, Dockery LE, Chandra V. The role and participation of immune cells in the endometrial tumor microenvironment. Pharmacol Ther 2023; 251:108526. [PMID: 37690483 DOI: 10.1016/j.pharmthera.2023.108526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/12/2023]
Abstract
The tumor microenvironment is surrounded by blood vessels and consists of malignant, non-malignant, and immune cells, as well as signalling molecules, which primarily affect the therapeutic response and curative effects of drugs in clinical studies. Tumor-infiltrating immune cells participate in tumor progression, impact anticancer therapy, and eventually lead to the development of immune tolerance. Immunotherapy is evolving as a promising therapeutic intervention to stimulate and activate the immune system to suppress cancer cell growth. Endometrial cancer (EC) is an immunogenic disease, and in recent years, immunotherapy has shown benefit in the treatment of recurrent and advanced EC. This review discusses the key molecular pathways associated with the intra-tumoral immune response and the involvement of circulatory signalling molecules. Specific immunologic signatures in EC which offer targets for immunomodulating agents, are also discussed. We have summarized the available literature in support of using immunotherapy in EC. Lastly, we have also discussed ongoing clinical trials that may offer additional promising immunotherapy options in the future. The manuscript also explored innovative approaches for screening and identifying effective drugs, and to reduce the financial burdens for the development of personalized treatment strategies. Collectively, we aim to provide a comprehensive review of the role of immune cells and the tumor microenvironment in the development, progression, and treatment of EC.
Collapse
Affiliation(s)
- Debasish Kumar Dey
- Gynecologic Oncology Section, Obstetrics and Gynecology Department, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Danielle Krause
- Gynecologic Oncology Section, Obstetrics and Gynecology Department, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Rajani Rai
- Gynecologic Oncology Section, Obstetrics and Gynecology Department, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Swati Choudhary
- Gynecologic Oncology Section, Obstetrics and Gynecology Department, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Lauren E Dockery
- Gynecologic Oncology Section, Obstetrics and Gynecology Department, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Vishal Chandra
- Gynecologic Oncology Section, Obstetrics and Gynecology Department, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
45
|
Su GH, Xiao Y, You C, Zheng RC, Zhao S, Sun SY, Zhou JY, Lin LY, Wang H, Shao ZM, Gu YJ, Jiang YZ. Radiogenomic-based multiomic analysis reveals imaging intratumor heterogeneity phenotypes and therapeutic targets. SCIENCE ADVANCES 2023; 9:eadf0837. [PMID: 37801493 PMCID: PMC10558123 DOI: 10.1126/sciadv.adf0837] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 09/06/2023] [Indexed: 10/08/2023]
Abstract
Intratumor heterogeneity (ITH) profoundly affects therapeutic responses and clinical outcomes. However, the widespread methods for assessing ITH based on genomic sequencing or pathological slides, which rely on limited tissue samples, may lead to inaccuracies due to potential sampling biases. Using a newly established multicenter breast cancer radio-multiomic dataset (n = 1474) encompassing radiomic features extracted from dynamic contrast-enhanced magnetic resonance images, we formulated a noninvasive radiomics methodology to effectively investigate ITH. Imaging ITH (IITH) was associated with genomic and pathological ITH, predicting poor prognosis independently in breast cancer. Through multiomic analysis, we identified activated oncogenic pathways and metabolic dysregulation in high-IITH tumors. Integrated metabolomic and transcriptomic analyses highlighted ferroptosis as a vulnerability and potential therapeutic target of high-IITH tumors. Collectively, this work emphasizes the superiority of radiomics in capturing ITH. Furthermore, we provide insights into the biological basis of IITH and propose therapeutic targets for breast cancers with elevated IITH.
Collapse
Affiliation(s)
- Guan-Hua Su
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ren-Cheng Zheng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai 201203, China
| | - Shen Zhao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shi-Yun Sun
- Department of Radiology, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jia-Yin Zhou
- Department of Radiology, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lu-Yi Lin
- Department of Radiology, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - He Wang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai 201203, China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ya-Jia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
46
|
Agten A, Blázquez-Moreno A, Crabbe M, Tuefferd M, Goehlmann H, Geys H, Peng CY, Claes J, Neyens T, Faes C. Measures of spatial heterogeneity in the liver tissue micro-environment as predictive factors for fibrosis score. Comput Biol Med 2023; 165:107382. [PMID: 37634463 DOI: 10.1016/j.compbiomed.2023.107382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/02/2023] [Accepted: 08/14/2023] [Indexed: 08/29/2023]
Abstract
The organization and interaction between hepatocytes and other hepatic non-parenchymal cells plays a pivotal role in maintaining normal liver function and structure. Although spatial heterogeneity within the tumor micro-environment has been proven to be a fundamental feature in cancer progression, the role of liver tissue topology and micro-environmental factors in the context of liver damage in chronic infection has not been widely studied yet. We obtained images from 110 core needle biopsies from a cohort of chronic hepatitis B patients with different fibrosis stages according to METAVIR score. The tissue sections were immunofluorescently stained and imaged to determine the locations of CD45 positive immune cells and HBsAg-negative and HBsAg-positive hepatocytes within the tissue. We applied several descriptive techniques adopted from ecology, including Getis-Ord, the Shannon Index and the Morisita-Horn Index, to quantify the extent to which immune cells and different types of liver cells co-localize in the tissue biopsies. Additionally, we modeled the spatial distribution of the different cell types using a joint log-Gaussian Cox process and proposed several features to quantify spatial heterogeneity. We then related these measures to the patient fibrosis stage by using a linear discriminant analysis approach. Our analysis revealed that the co-localization of HBsAg-negative hepatocytes with immune cells and the co-localization of HBsAg-positive hepatocytes with immune cells are equally important factors for explaining the METAVIR score in chronic hepatitis B patients. Moreover, we found that if we allow for an error of 1 on the METAVIR score, we are able to reach an accuracy of around 80%. With this study we demonstrate how methods adopted from ecology and applied to the liver tissue micro-environment can be used to quantify heterogeneity and how these approaches can be valuable in biomarker analyses for liver topology.
Collapse
Affiliation(s)
- Annelies Agten
- Data Science Institute, UHasselt - Hasselt University, Agoralaan 1, BE 3590 Diepenbeek, Belgium.
| | - Alfonso Blázquez-Moreno
- Discovery Statistics, Global Development, Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Marjolein Crabbe
- Discovery Statistics, Global Development, Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Marianne Tuefferd
- Translational Biomarkers, Infectious Diseases, Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Hinrich Goehlmann
- Translational Biomarkers, Infectious Diseases, Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Helena Geys
- Discovery Statistics, Global Development, Janssen Research and Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | - Jari Claes
- Data Science Institute, UHasselt - Hasselt University, Agoralaan 1, BE 3590 Diepenbeek, Belgium
| | - Thomas Neyens
- Data Science Institute, UHasselt - Hasselt University, Agoralaan 1, BE 3590 Diepenbeek, Belgium; L-BioStat, KU Leuven, Kapucijnenvoer 35, 3000 Leuven, Belgium
| | - Christel Faes
- Data Science Institute, UHasselt - Hasselt University, Agoralaan 1, BE 3590 Diepenbeek, Belgium
| |
Collapse
|
47
|
Pradeu T, Daignan-Fornier B, Ewald A, Germain PL, Okasha S, Plutynski A, Benzekry S, Bertolaso M, Bissell M, Brown JS, Chin-Yee B, Chin-Yee I, Clevers H, Cognet L, Darrason M, Farge E, Feunteun J, Galon J, Giroux E, Green S, Gross F, Jaulin F, Knight R, Laconi E, Larmonier N, Maley C, Mantovani A, Moreau V, Nassoy P, Rondeau E, Santamaria D, Sawai CM, Seluanov A, Sepich-Poore GD, Sisirak V, Solary E, Yvonnet S, Laplane L. Reuniting philosophy and science to advance cancer research. Biol Rev Camb Philos Soc 2023; 98:1668-1686. [PMID: 37157910 PMCID: PMC10869205 DOI: 10.1111/brv.12971] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
Cancers rely on multiple, heterogeneous processes at different scales, pertaining to many biomedical fields. Therefore, understanding cancer is necessarily an interdisciplinary task that requires placing specialised experimental and clinical research into a broader conceptual, theoretical, and methodological framework. Without such a framework, oncology will collect piecemeal results, with scant dialogue between the different scientific communities studying cancer. We argue that one important way forward in service of a more successful dialogue is through greater integration of applied sciences (experimental and clinical) with conceptual and theoretical approaches, informed by philosophical methods. By way of illustration, we explore six central themes: (i) the role of mutations in cancer; (ii) the clonal evolution of cancer cells; (iii) the relationship between cancer and multicellularity; (iv) the tumour microenvironment; (v) the immune system; and (vi) stem cells. In each case, we examine open questions in the scientific literature through a philosophical methodology and show the benefit of such a synergy for the scientific and medical understanding of cancer.
Collapse
Affiliation(s)
- Thomas Pradeu
- CNRS UMR5164 ImmunoConcEpT, University of Bordeaux, 146 rue Leo Saignat, Bordeaux 33076, France
- CNRS UMR8590, Institut d’Histoire et Philosophie des Sciences et des Technique, University Paris I Panthéon-Sorbonne, 13 rue du Four, Paris 75006, France
| | - Bertrand Daignan-Fornier
- CNRS UMR 5095 Institut de Biochimie et Génétique Cellulaires, University of Bordeaux, 1 rue Camille St Saens, Bordeaux 33077, France
| | - Andrew Ewald
- Departments of Cell Biology and Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Pierre-Luc Germain
- Department of Health Sciences and Technology, Institute for Neurosciences, Eidgenössische Technische Hochschule (ETH) Zürich, Universitätstrasse 2, Zürich 8092, Switzerland
- Department of Molecular Life Sciences, Laboratory of Statistical Bioinformatics, Universität Zürich, Winterthurerstrasse 190, Zurich 8057, Switzerland
| | - Samir Okasha
- Department of Philosophy, University of Bristol, Cotham House, Bristol, BS6 6JL, UK
| | - Anya Plutynski
- Department of Philosophy, Washington University in St. Louis, and Associate with Division of Biology and Biomedical Sciences, St. Louis, MO 63105, USA
| | - Sébastien Benzekry
- Computational Pharmacology and Clinical Oncology (COMPO) Unit, Inria Sophia Antipolis-Méditerranée, Cancer Research Center of Marseille, Inserm UMR1068, CNRS UMR7258, Aix Marseille University UM105, 27, bd Jean Moulin, Marseille 13005, France
| | - Marta Bertolaso
- Research Unit of Philosophy of Science and Human Development, Università Campus Bio-Medico di Roma, Via Àlvaro del Portillo, 21-00128, Rome, Italy
- Centre for Cancer Biomarkers, University of Bergen, Bergen 5007, Norway
| | - Mina Bissell
- Biological Systems & Engineering Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd, Berkeley, CA 94720, USA
| | - Joel S. Brown
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Benjamin Chin-Yee
- Division of Hematology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, 800 Commissioners Rd E, London, ON, Canada
- Rotman Institute of Philosophy, Western University, 1151 Richmond Street North, London, ON, Canada
| | - Ian Chin-Yee
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, 800 Commissioners Rd E, London, ON, Canada
| | - Hans Clevers
- Pharma, Research and Early Development (pRED) of F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Uppsalalaan 8, Utrecht 3584 CT, The Netherlands
| | - Laurent Cognet
- CNRS UMR 5298, Laboratoire Photonique Numérique et Nanosciences, University of Bordeaux, Rue François Mitterrand, Talence 33400, France
| | - Marie Darrason
- Department of Pneumology and Thoracic Oncology, University Hospital of Lyon, 165 Chem. du Grand Revoyet, 69310 Pierre Bénite, Lyon, France
- Lyon Institute of Philosophical Research, Lyon 3 Jean Moulin University, 1 Av. des Frères Lumière, Lyon 69007, France
| | - Emmanuel Farge
- Mechanics and Genetics of Embryonic and Tumor Development group, Institut Curie, CNRS, UMR168, Inserm, Centre Origines et conditions d’apparition de la vie (OCAV) Paris Sciences Lettres Research University, Sorbonne University, Institut Curie, 11 rue Pierre et Marie Curie, Paris 75005, France
| | - Jean Feunteun
- INSERM U981, Gustave Roussy, 114 Rue Edouard Vaillant, Villejuif 94800, France
| | - Jérôme Galon
- INSERM UMRS1138, Integrative Cancer Immunology, Cordelier Research Center, Sorbonne Université, Université Paris Cité, 15 rue de l’École de Médecine, Paris 75006, France
| | - Elodie Giroux
- Lyon Institute of Philosophical Research, Lyon 3 Jean Moulin University, 1 Av. des Frères Lumière, Lyon 69007, France
| | - Sara Green
- Section for History and Philosophy of Science, Department of Science Education, University of Copenhagen, Rådmandsgade 64, Copenhagen 2200, Denmark
| | - Fridolin Gross
- CNRS UMR5164 ImmunoConcEpT, University of Bordeaux, 146 rue Leo Saignat, Bordeaux 33076, France
| | - Fanny Jaulin
- INSERM U1279, Gustave Roussy, 114 Rue Edouard Vaillant, Villejuif 94800, France
| | - Rob Knight
- Department of Bioengineering, University of California San Diego, 3223 Voigt Dr, La Jolla, CA 92093, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Ezio Laconi
- Department of Biomedical Sciences, School of Medicine, University of Cagliari, Via Università 40, Cagliari 09124, Italy
| | - Nicolas Larmonier
- CNRS UMR5164 ImmunoConcEpT, University of Bordeaux, 146 rue Leo Saignat, Bordeaux 33076, France
| | - Carlo Maley
- Arizona Cancer Evolution Center, Arizona State University, 427 East Tyler Mall, Tempe, AZ 85287, USA
- School of Life Sciences, Arizona State University, 427 East Tyler Mall, Tempe, AZ 85287, USA
- Biodesign Center for Biocomputing, Security and Society, Arizona State University, 1001 S McAllister Ave, Tempe, AZ 85287, USA
- Biodesign Center for Mechanisms of Evolution, Arizona State University, 1001 S McAllister Ave, Tempe, AZ 85287, USA
- Center for Evolution and Medicine, Arizona State University, 427 East Tyler Mall, Tempe, AZ 85287, USA
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, 4 Via Rita Levi Montalcini, 20090 Pieve Emanuele, Milan, Italy
- Department of Immunology and Inflammation, Istituto Clinico Humanitas Humanitas Cancer Center (IRCCS) Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan 20089, Italy
- The William Harvey Research Institute, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Violaine Moreau
- INSERM UMR1312, Bordeaux Institute of Oncology (BRIC), University of Bordeaux, 146 Rue Léo Saignat, Bordeaux 33076, France
| | - Pierre Nassoy
- CNRS UMR 5298, Laboratoire Photonique Numérique et Nanosciences, University of Bordeaux, Rue François Mitterrand, Talence 33400, France
| | - Elena Rondeau
- INSERM U1111, ENS Lyon and Centre International de Recherche en Infectionlogie (CIRI), 46 Allée d’Italie, Lyon 69007, France
| | - David Santamaria
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, Salamanca 37007, Spain
| | - Catherine M. Sawai
- INSERM UMR1312, Bordeaux Institute of Oncology (BRIC), University of Bordeaux, 146 Rue Léo Saignat, Bordeaux 33076, France
| | - Andrei Seluanov
- Department of Biology and Medicine, University of Rochester, Rochester, NY 14627, USA
| | | | - Vanja Sisirak
- CNRS UMR5164 ImmunoConcEpT, University of Bordeaux, 146 rue Leo Saignat, Bordeaux 33076, France
| | - Eric Solary
- INSERM U1287, Gustave Roussy, 114 Rue Edouard Vaillant, Villejuif 94800, France
- Département d’hématologie, Gustave Roussy, 114 Rue Edouard Vaillant, Villejuif 94800, France
- Université Paris-Saclay, Faculté de Médecine, 63 Rue Gabriel Péri, Le Kremlin-Bicêtre 94270, France
| | - Sarah Yvonnet
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Blegdamsvej 3B, Copenhagen DK-2200, Denmark
| | - Lucie Laplane
- CNRS UMR8590, Institut d’Histoire et Philosophie des Sciences et des Technique, University Paris I Panthéon-Sorbonne, 13 rue du Four, Paris 75006, France
- INSERM U1287, Gustave Roussy, 114 Rue Edouard Vaillant, Villejuif 94800, France
- Center for Biology and Society, College of Liberal Arts and Sciences, Arizona State University, 1100 S McAllister Ave, Tempe, AZ 85281, USA
| |
Collapse
|
48
|
Mansur MB, Greaves M. Convergent TP53 loss and evolvability in cancer. BMC Ecol Evol 2023; 23:54. [PMID: 37743495 PMCID: PMC10518978 DOI: 10.1186/s12862-023-02146-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/10/2023] [Indexed: 09/26/2023] Open
Abstract
Cancer cell populations evolve by a stepwise process involving natural selection of the fittest variants within a tissue ecosystem context and as modified by therapy. Genomic scrutiny of patient samples reveals an extraordinary diversity of mutational profiles both between patients with similar cancers and within the cancer cell population of individual patients. Does this signify highly divergent evolutionary trajectories or are there repetitive and predictable patterns?Major evolutionary innovations or adaptations in different species are frequently repeated, or convergent, reflecting both common selective pressures and constraints on optimal solutions. We argue this is true of evolving cancer cells, especially with respect to the TP53 gene. Functional loss variants in TP53 are the most common genetic change in cancer. We discuss the likely microenvironmental selective pressures involved and the profound impact this has on cell fitness, evolvability and probability of subsequent drug resistance.
Collapse
Affiliation(s)
- Marcela Braga Mansur
- Centre for Evolution and Cancer, The Institute of Cancer Research, ICR, London, UK
| | - Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, ICR, London, UK.
| |
Collapse
|
49
|
Wang Y, Drum DL, Sun R, Zhang Y, Chen F, Sun F, Dal E, Yu L, Jia J, Arya S, Jia L, Fan S, Isakoff SJ, Kehlmann AM, Dotti G, Liu F, Zheng H, Ferrone CR, Taghian AG, DeLeo AB, Ventin M, Cattaneo G, Li Y, Jounaidi Y, Huang P, Maccalli C, Zhang H, Wang C, Yang J, Boland GM, Sadreyev RI, Wong L, Ferrone S, Wang X. Stressed target cancer cells drive nongenetic reprogramming of CAR T cells and solid tumor microenvironment. Nat Commun 2023; 14:5727. [PMID: 37714830 PMCID: PMC10504259 DOI: 10.1038/s41467-023-41282-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 08/29/2023] [Indexed: 09/17/2023] Open
Abstract
The poor efficacy of chimeric antigen receptor T-cell therapy (CAR T) for solid tumors is due to insufficient CAR T cell tumor infiltration, in vivo expansion, persistence, and effector function, as well as exhaustion, intrinsic target antigen heterogeneity or antigen loss of target cancer cells, and immunosuppressive tumor microenvironment (TME). Here we describe a broadly applicable nongenetic approach that simultaneously addresses the multiple challenges of CAR T as a therapy for solid tumors. The approach reprograms CAR T cells by exposing them to stressed target cancer cells which have been exposed to the cell stress inducer disulfiram (DSF) and copper (Cu)(DSF/Cu) plus ionizing irradiation (IR). The reprogrammed CAR T cells acquire early memory-like characteristics, potent cytotoxicity, enhanced in vivo expansion, persistence, and decreased exhaustion. Tumors stressed by DSF/Cu and IR also reprogram and reverse the immunosuppressive TME in humanized mice. The reprogrammed CAR T cells, derived from peripheral blood mononuclear cells of healthy donors or metastatic female breast cancer patients, induce robust, sustained memory and curative anti-solid tumor responses in multiple xenograft mouse models, establishing proof of concept for empowering CAR T by stressing tumor as a promising therapy for solid tumors.
Collapse
Affiliation(s)
- Yufeng Wang
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - David L Drum
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ruochuan Sun
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Gastrointestinal Surgery and General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yida Zhang
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Feng Chen
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Fengfei Sun
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emre Dal
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ling Yu
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jingyu Jia
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shahrzad Arya
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lin Jia
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Song Fan
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven J Isakoff
- Termeer Center for Targeted Therapies, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Allison M Kehlmann
- Termeer Center for Targeted Therapies, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Fubao Liu
- Department of Hepatobiliary & Pancreatic Surgery and Liver Transplantation, Anhui Medical University, Hefei, Anhui, China
| | - Hui Zheng
- Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina R Ferrone
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alphonse G Taghian
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Albert B DeLeo
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco Ventin
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Giulia Cattaneo
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yongxiang Li
- Department of Gastrointestinal Surgery and General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Youssef Jounaidi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peigen Huang
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Hanyu Zhang
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cheng Wang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jibing Yang
- Center for Comparative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Genevieve M Boland
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ruslan I Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - LaiPing Wong
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Soldano Ferrone
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Orthopaedics, Massachusetts General Hospital, Boston, MA, USA
| | - Xinhui Wang
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
50
|
Kok SY, Nakayama M, Morita A, Oshima H, Oshima M. Genetic and nongenetic mechanisms for colorectal cancer evolution. Cancer Sci 2023; 114:3478-3486. [PMID: 37357016 PMCID: PMC10475778 DOI: 10.1111/cas.15891] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/10/2023] [Indexed: 06/27/2023] Open
Abstract
The stepwise accumulation of key driver mutations is responsible for the development and malignant progression of colorectal cancer in primary sites. Genetic mouse model studies have revealed combinations of driver gene mutations that induce phenotypic changes in tumors toward malignancy. However, cancer evolution is regulated by not only genetic alterations but also nongenetic mechanisms. For example, certain populations of metastatic cancer cells show a loss of malignant characteristics even after the accumulation of driver mutations, and such cells are eliminated in a negative selection manner. Furthermore, a polyclonal metastasis model has recently been proposed, in which cell clusters consisting of genetically heterogeneous cells break off from the primary site, disseminate to distant organs, and develop into heterogenous metastatic tumors. Such nongenetic mechanisms for malignant progression have been elucidated using genetically engineered mouse models as well as organoid transplantation experiments. In this review article, we discuss the role of genetic alterations in the malignant progression of primary intestinal tumors and nongenetic mechanisms for negative selection and polyclonal metastasis, which we learned from model studies.
Collapse
Affiliation(s)
- Sau Yee Kok
- Division of GeneticsCancer Research Institute, Kanazawa UniversityKanazawaJapan
| | - Mizuho Nakayama
- Division of GeneticsCancer Research Institute, Kanazawa UniversityKanazawaJapan
- WPI Nano Life Science Institute (NanoLSI), Kanazawa UniversityKanazawaJapan
| | - Atsuya Morita
- Division of GeneticsCancer Research Institute, Kanazawa UniversityKanazawaJapan
| | - Hiroko Oshima
- Division of GeneticsCancer Research Institute, Kanazawa UniversityKanazawaJapan
- WPI Nano Life Science Institute (NanoLSI), Kanazawa UniversityKanazawaJapan
| | - Masanobu Oshima
- Division of GeneticsCancer Research Institute, Kanazawa UniversityKanazawaJapan
- WPI Nano Life Science Institute (NanoLSI), Kanazawa UniversityKanazawaJapan
| |
Collapse
|