201
|
Abstract
MOTIVATION Cancer develops through a process of clonal evolution in which an initially healthy cell gives rise to progeny gradually differentiating through the accumulation of genetic and epigenetic mutations. These mutations can take various forms, including single-nucleotide variants (SNVs), copy number alterations (CNAs) or structural variations (SVs), with each variant type providing complementary insights into tumor evolution as well as offering distinct challenges to phylogenetic inference. RESULTS In this work, we develop a tumor phylogeny method, TUSV-ext, which incorporates SNVs, CNAs and SVs into a single inference framework. We demonstrate on simulated data that the method produces accurate tree inferences in the presence of all three variant types. We further demonstrate the method through application to real prostate tumor data, showing how our approach to coordinated phylogeny inference and clonal construction with all three variant types can reveal a more complicated clonal structure than is suggested by prior work, consistent with extensive polyclonal seeding or migration. AVAILABILITY AND IMPLEMENTATION https://github.com/CMUSchwartzLab/TUSV-ext. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
|
202
|
Laganà A. The Architecture of a Precision Oncology Platform. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1361:1-22. [DOI: 10.1007/978-3-030-91836-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
203
|
Kang M, Na HY, Ahn S, Kim JW, Lee S, Ahn S, Lee JH, Youk J, Kim HT, Kim KJ, Suh KJ, Lee JS, Kim SH, Kim JW, Kim YJ, Lee KW, Yoon YS, Kim JH, Chung JH, Han HS, Lee JS. Gallbladder adenocarcinomas undergo subclonal diversification and selection from precancerous lesions to metastatic tumors. eLife 2022; 11:78636. [PMID: 36476508 PMCID: PMC9771369 DOI: 10.7554/elife.78636] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
We aimed to elucidate the evolutionary trajectories of gallbladder adenocarcinoma (GBAC) using multi-regional and longitudinal tumor samples. Using whole-exome sequencing data, we constructed phylogenetic trees in each patient and analyzed mutational signatures. A total of 11 patients including 2 rapid autopsy cases were enrolled. The most frequently altered gene in primary tumors was ERBB2 and TP53 (54.5%), followed by FBXW7 (27.3%). Most mutations in frequently altered genes in primary tumors were detectable in concurrent precancerous lesions (biliary intraepithelial neoplasia [BilIN]), but a substantial proportion was subclonal. Subclonal diversity was common in BilIN (n=4). However, among subclones in BilIN, a certain subclone commonly shrank in concurrent primary tumors. In addition, selected subclones underwent linear and branching evolution, maintaining subclonal diversity. Combined analysis with metastatic tumors (n=11) identified branching evolution in nine patients (81.8%). Of these, eight patients (88.9%) had a total of 11 subclones expanded at least sevenfold during metastasis. These subclones harbored putative metastasis-driving mutations in cancer-related genes such as SMAD4, ROBO1, and DICER1. In mutational signature analysis, six mutational signatures were identified: 1, 3, 7, 13, 22, and 24 (cosine similarity >0.9). Signatures 1 (age) and 13 (APOBEC) decreased during metastasis while signatures 22 (aristolochic acid) and 24 (aflatoxin) were relatively highlighted. Subclonal diversity arose early in precancerous lesions and clonal selection was a common event during malignant transformation in GBAC. However, selected cancer clones continued to evolve and thus maintained subclonal diversity in metastatic tumors.
Collapse
Affiliation(s)
- Minsu Kang
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Hee Young Na
- Department of Pathology, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Soomin Ahn
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of MedicineSeoulRepublic of Korea
| | - Ji-Won Kim
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnamRepublic of Korea,Genealogy IncSeoulRepublic of Korea
| | - Sejoon Lee
- Center for Precision Medicine, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Soyeon Ahn
- Medical Research Collaboration Center, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Ju Hyun Lee
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Jeonghwan Youk
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Haesook T Kim
- Department of Data Science, Dana Farber Cancer Institute, Harvard T.H. Chan School of Public HealthBostonUnited States
| | - Kui-Jin Kim
- Biomedical Research Institute, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Koung Jin Suh
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Jun Suh Lee
- Department of Surgery, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Se Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Jin Won Kim
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Yu Jung Kim
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Yoo-Seok Yoon
- Department of Surgery, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Jin-Haeng Chung
- Department of Pathology, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Ho-Seong Han
- Department of Surgery, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Jong Seok Lee
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnamRepublic of Korea
| |
Collapse
|
204
|
Lee M, Untch BR, Xu B, Ghossein R, Han C, Kuo F, Valero C, Nadeem Z, Patel N, Makarov V, Dogan S, Wong RJ, Sherman EJ, Ho AL, Chan TA, Fagin JA, Morris LGT. Genomic and Transcriptomic Correlates of Thyroid Carcinoma Evolution after BRAF Inhibitor Therapy. Mol Cancer Res 2022; 20:45-55. [PMID: 34635506 PMCID: PMC8738128 DOI: 10.1158/1541-7786.mcr-21-0442] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/12/2021] [Accepted: 10/04/2021] [Indexed: 12/16/2022]
Abstract
Targeted inhibition of BRAF V600E achieves tumor control in a subset of advanced thyroid tumors. Nearly all tumors develop resistance, and some have been observed to subsequently undergo dedifferentiation. The molecular alterations associated with thyroid cancer dedifferentiation in the setting of BRAF inhibition are unknown. We analyzed targeted next-generation sequencing data from 639 advanced, recurrent and/or metastatic thyroid carcinomas, including 15 tumors that were treated with BRAF inhibitor drugs and had tissue sampled during or posttreatment, 8 of which had matched pretherapy samples. Pre- and posttherapy tissues from one additional patient were profiled with whole-exome sequencing and RNA expression profiling. Mutations in genes comprising the SWI/SNF chromatin remodeling complex and the PI3K-AKT-mTOR, MAPK, and JAK-STAT pathways all increased in prevalence across more dedifferentiated thyroid cancer histologies. Of 7 thyroid cancers that dedifferentiated after BRAF inhibition, 6 had mutations in these pathways. These mutations were mostly absent from matched pretreatment samples and were rarely detected in tumors that did not dedifferentiate. Additional analyses in one of the vemurafenib-treated tumors before and after anaplastic transformation revealed the emergence of an oncogenic PIK3CA mutation, activation of ERK signaling, dedifferentiation, and development of an immunosuppressive tumor microenvironment. These findings validate earlier preclinical data implicating these genetic pathways in resistance to BRAF inhibitors, and suggest that genetic alterations mediating acquired drug resistance may also promote thyroid tumor dedifferentiation. IMPLICATIONS: The possibility that thyroid cancer dedifferentiation may be attributed to selective pressure applied by BRAF inhibitor-targeted therapy should be investigated further.
Collapse
Affiliation(s)
- Mark Lee
- Weill Cornell Medicine, New York, New York
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brian R Untch
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bin Xu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronald Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Catherine Han
- Weill Cornell Medicine, New York, New York
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fengshen Kuo
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cristina Valero
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zaineb Nadeem
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neal Patel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vladimir Makarov
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio
| | - Snjezana Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard J Wong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric J Sherman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alan L Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Timothy A Chan
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio
| | - James A Fagin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Luc G T Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
205
|
Liu X, Wang W, Liu X, Zhang Z, Yu L, Li R, Guo D, Cai W, Quan X, Wu H, Dai M, Liang Z. Multi-omics analysis of intra-tumoural and inter-tumoural heterogeneity in pancreatic ductal adenocarcinoma. Clin Transl Med 2022; 12:e670. [PMID: 35061935 PMCID: PMC8782496 DOI: 10.1002/ctm2.670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022] Open
Abstract
The poor prognosis of pancreatic ductal adenocarcinoma (PDAC) is associated with the tumour heterogeneity. To explore intra- and inter-tumoural heterogeneity in PDAC, we analysed the multi-omics profiles of 61 PDAC lesion samples, along with the matched pancreatic normal tissue samples, from 19 PDAC patients. Haematoxylin and Eosin (H&E) staining revealed that diversely differentiated lesions coexisted both within and across individual tumours. Whole exome sequencing (WES) of samples from multi-region revealed diverse types of mutations in diverse genes between cancer cells within a tumour and between tumours from different individuals. The copy number variation (CNV) analysis also showed that PDAC exhibited intra- and inter-tumoural heterogeneity in CNV and that high average CNV burden was associated poor prognosis of the patients. Phylogenetic tree analysis and clonality/timing analysis of mutations displayed diverse evolutionary pathways and spatiotemporal characteristics of genomic alterations between different lesions from the same or different tumours. Hierarchical clustering analysis illustrated higher inter-tumoural heterogeneity than intra-tumoural heterogeneity of PDAC at the transcriptional levels as lesions from the same patients are grouped into a single cluster. Immune marker genes are differentially expressed in different regions and tumour samples as shown by tumour microenvironment (TME) analysis. TME appeared to be more heterogeneous than tumour cells in the same patient. Lesion-specific differentially methylated regions (DMRs) were identified by methylated DNA immunoprecipitation sequencing (MeDIP-seq). Furthermore, the integration analysis of multi-omics data showed that the mRNA levels of some genes, such as PLCB4, were significantly correlated with the gene copy numbers. The mRNA expressions of potential PDAC biomarkers ZNF521 and KDM6A were correlated with copy number alteration and methylation, respectively. Taken together, our results provide a comprehensive view of molecular heterogeneity and evolutionary trajectories of PDAC and may guide personalised treatment strategies in PDAC therapy.
Collapse
Affiliation(s)
- Xiaoqian Liu
- Department of PathologyState Key Laboratory of Complex Severe and Rare DiseasesMolecular Pathology Research CenterPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of PathologyQilu Hospital (Qingdao)Cheeloo College of MedicineShandong UniversityQingdaoShandongChina
| | - Wenqian Wang
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaoding Liu
- Department of PathologyState Key Laboratory of Complex Severe and Rare DiseasesMolecular Pathology Research CenterPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhiwen Zhang
- Department of PathologyState Key Laboratory of Complex Severe and Rare DiseasesMolecular Pathology Research CenterPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lianyuan Yu
- Department of PathologyState Key Laboratory of Complex Severe and Rare DiseasesMolecular Pathology Research CenterPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ruiyu Li
- Department of PathologyState Key Laboratory of Complex Severe and Rare DiseasesMolecular Pathology Research CenterPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Dan Guo
- Clinical BiobankMedical Research CentrePeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Weijing Cai
- Shanghai Tongshu Biotechnology Co., LtdShanghaiChina
| | - Xueping Quan
- Shanghai Tongshu Biotechnology Co., LtdShanghaiChina
| | - Huanwen Wu
- Department of PathologyState Key Laboratory of Complex Severe and Rare DiseasesMolecular Pathology Research CenterPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Menghua Dai
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhiyong Liang
- Department of PathologyState Key Laboratory of Complex Severe and Rare DiseasesMolecular Pathology Research CenterPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
206
|
Park S, Lee D, Kim Y, Lim S, Chae H, Kim S. BioVLAB-Cancer-Pharmacogenomics: tumor heterogeneity and pharmacogenomics analysis of multi-omics data from tumor on the cloud. Bioinformatics 2021; 38:275-277. [PMID: 34185062 DOI: 10.1093/bioinformatics/btab478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/12/2021] [Accepted: 06/28/2021] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION Multi-omics data in molecular biology has accumulated rapidly over the years. Such data contains valuable information for research in medicine and drug discovery. Unfortunately, data-driven research in medicine and drug discovery is challenging for a majority of small research labs due to the large volume of data and the complexity of analysis pipeline. RESULTS We present BioVLAB-Cancer-Pharmacogenomics, a bioinformatics system that facilitates analysis of multi-omics data from breast cancer to analyze and investigate intratumor heterogeneity and pharmacogenomics on Amazon Web Services. Our system takes multi-omics data as input to perform tumor heterogeneity analysis in terms of TCGA data and deconvolve-and-match the tumor gene expression to cell line data in CCLE using DNA methylation profiles. We believe that our system can help small research labs perform analysis of tumor multi-omics without worrying about computational infrastructure and maintenance of databases and tools. AVAILABILITY AND IMPLEMENTATION http://biohealth.snu.ac.kr/software/biovlab_cancer_pharmacogenomics. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Sungjoon Park
- Department of Computer Science and Engineering, Seoul National University, Seoul 08840, Republic of Korea
| | - Dohoon Lee
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08840, Republic of Korea
| | - Youngkuk Kim
- Department of Computer Science and Engineering, Seoul National University, Seoul 08840, Republic of Korea
| | - Sangsoo Lim
- Bioinformatics Institute, Seoul National University, Seoul 08840, Republic of Korea
| | - Heejoon Chae
- Division of Computer Science, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Sun Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul 08840, Republic of Korea.,Bioinformatics Institute, Seoul National University, Seoul 08840, Republic of Korea.,Institute of Engineering Research, Seoul National University, Seoul 08840, Republic of Korea
| |
Collapse
|
207
|
Jin Y, Chen Y, Tang H, Hu X, Hubert SM, Li Q, Su D, Xu H, Fan Y, Yu X, Chen Q, Liu J, Hong W, Xu Y, Deng H, Zhu D, Li P, Gong Y, Xia X, Gay CM, Zhang J, Chen M. Activation of PI3K/AKT pathway is a potential mechanism of treatment resistance in small cell lung cancer. Clin Cancer Res 2021; 28:526-539. [PMID: 34921019 DOI: 10.1158/1078-0432.ccr-21-1943] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/30/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022]
Affiliation(s)
- Ying Jin
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, China
| | - Yamei Chen
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, China
| | - Huarong Tang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, China
| | - Xiao Hu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, China
| | - Shawna M Hubert
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qian Li
- Geneplus-Beijing Institute, Beijing, China
| | - Dan Su
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Haimiao Xu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Yun Fan
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xinmin Yu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Qixun Chen
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jinshi Liu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wei Hong
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Yujin Xu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, China
| | - Huan Deng
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, China
| | - Dapeng Zhu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Pansong Li
- Geneplus-Beijing Institute, Beijing, China
| | - Yuhua Gong
- Geneplus-Beijing Institute, Beijing, China
| | | | - Carl M Gay
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas.
- Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ming Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, China
| |
Collapse
|
208
|
Benard BA, Leak LB, Azizi A, Thomas D, Gentles AJ, Majeti R. Clonal architecture predicts clinical outcomes and drug sensitivity in acute myeloid leukemia. Nat Commun 2021; 12:7244. [PMID: 34903734 PMCID: PMC8669028 DOI: 10.1038/s41467-021-27472-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/17/2021] [Indexed: 12/17/2022] Open
Abstract
The impact of clonal heterogeneity on disease behavior or drug response in acute myeloid leukemia remains poorly understood. Using a cohort of 2,829 patients, we identify features of clonality associated with clinical features and drug sensitivities. High variant allele frequency for 7 mutations (including NRAS and TET2) associate with dismal prognosis; elevated GATA2 variant allele frequency correlates with better outcomes. Clinical features such as white blood cell count and blast percentage correlate with the subclonal abundance of mutations such as TP53 and IDH1. Furthermore, patients with cohesin mutations occurring before NPM1, or transcription factor mutations occurring before splicing factor mutations, show shorter survival. Surprisingly, a branched pattern of clonal evolution is associated with superior clinical outcomes. Finally, several mutations (including NRAS and IDH1) predict drug sensitivity based on their subclonal abundance. Together, these results demonstrate the importance of assessing clonal heterogeneity with implications for prognosis and actionable biomarkers for therapy.
Collapse
Affiliation(s)
- Brooks A Benard
- Department of Medicine, Division of Hematology, Cancer Institute, Stanford University, Stanford, CA, USA
- Cancer Biology Program, Stanford University, Stanford, CA, USA
| | - Logan B Leak
- Cancer Biology Program, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Armon Azizi
- Department of Medicine, Division of Hematology, Cancer Institute, Stanford University, Stanford, CA, USA
| | - Daniel Thomas
- Department of Medicine, Division of Hematology, Cancer Institute, Stanford University, Stanford, CA, USA
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Andrew J Gentles
- Department of Medicine (Biomedical Informatics/Quantitative Sciences unit), Stanford University, Stanford, CA, USA
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Cancer Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
209
|
Zhang W, Wang T, Wang Y, Zhu F, Shi H, Zhang J, Wang Z, Qu M, Zhang H, Wang T, Qian Y, Yang J, Gao X, Li J. Intratumor heterogeneity and clonal evolution revealed in castration-resistant prostate cancer by longitudinal genomic analysis. Transl Oncol 2021; 16:101311. [PMID: 34902740 PMCID: PMC8681025 DOI: 10.1016/j.tranon.2021.101311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/25/2021] [Accepted: 12/03/2021] [Indexed: 11/29/2022] Open
Abstract
Intratumor heterogeneity is a key driver for local relapse and treatment failure. Thus, using multifocal prostate cancer as a model to investigate tumor inter-clonal relationships and tumor evolution could aid in our understanding of drug resistance. Previous studies discovered genomic alterations by comparing hormone-sensitive prostate cancer (HSPC) with castration-resistant prostate cancer (CRPC) in large cohorts. However, most studies did not sequentially sample tumors from the same patient. In our study, we performed whole-exome sequencing (WES) on 14 specimens from five locally relapsed patients before and after androgen-deprivation therapy. We described the landscape of genomic alterations before and after treatment and identified critical driver events that could have contributed to the evolution of CRPC. In addition to confirming known cancer genes such as TP53 and CDK12, we also identified new candidate genes that may play a role in the progression of prostate cancer, including MYO15A, CHD6 and LZTR1. At copy number alteration (CNA) level, gain of 8q24.13-8q24.3 was observed in 60% of patients and was the most commonly altered locus in both HSPC and CRPC tumors. Finally, utilizing phylogenetic reconstruction, we explored the clonal progression pattern from HSPC to CRPC in each patient. Our findings highlight the complex and heterogeneous mechanisms underlying the development of drug resistance, and underscore the potential value of monitoring tumor clonal architectures during disease progression in a clinical setting.
Collapse
Affiliation(s)
- Wenhui Zhang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Tao Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yan Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Feng Zhu
- Department of Urology, Tianyou Hospital, Tongji University, Shanghai 200333, China
| | - Haoqing Shi
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jili Zhang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Ziwei Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Min Qu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Huaru Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Tianyi Wang
- Department of Nuclear Medicine, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yuping Qian
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jinjian Yang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Xu Gao
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| | - Jing Li
- Department of Bioinformatics, Center for Translational Medicine, Second Military Medical University, Shanghai 200433, China; Shanghai Key Laboratory of Cell Engineering, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
210
|
Tang M, Abbas HA, Negrao MV, Ramineni M, Hu X, Hubert SM, Fujimoto J, Reuben A, Varghese S, Zhang J, Li J, Chow CW, Mao X, Song X, Lee WC, Wu J, Little L, Gumbs C, Behrens C, Moran C, Weissferdt A, Lee JJ, Sepesi B, Swisher S, Cheng C, Kurie J, Gibbons D, Heymach JV, Wistuba II, Futreal PA, Kalhor N, Zhang J. The histologic phenotype of lung cancers is associated with transcriptomic features rather than genomic characteristics. Nat Commun 2021; 12:7081. [PMID: 34873156 PMCID: PMC8648877 DOI: 10.1038/s41467-021-27341-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 11/16/2021] [Indexed: 12/31/2022] Open
Abstract
Histology plays an essential role in therapeutic decision-making for lung cancer patients. However, the molecular determinants of lung cancer histology are largely unknown. We conduct whole-exome sequencing and microarray profiling on 19 micro-dissected tumor regions of different histologic subtypes from 9 patients with lung cancers of mixed histology. A median of 68.9% of point mutations and 83% of copy number aberrations are shared between different histologic components within the same tumors. Furthermore, different histologic components within the tumors demonstrate similar subclonal architecture. On the other hand, transcriptomic profiling reveals shared pathways between the same histologic subtypes from different patients, which is supported by the analyses of the transcriptomic data from 141 cell lines and 343 lung cancers of different histologic subtypes. These data derived from mixed histologic subtypes in the setting of identical genetic background and exposure history support that the histologic fate of lung cancer cells is associated with transcriptomic features rather than the genomic profiles in most tumors. The molecular determinants of lung cancer histologic subtypes are not well understood. Here the authors analyze lung cancers of mixed histology and find that histologic subtypes are associated with transcriptomic features rather than genomic profiles in most tumors.
Collapse
Affiliation(s)
- Ming Tang
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hussein A Abbas
- Medical Oncology Fellowship, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Marcelo V Negrao
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Maheshwari Ramineni
- Department of Pathology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xin Hu
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Shawna Marie Hubert
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Junya Fujimoto
- Department of Translational Molecular Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Susan Varghese
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jun Li
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Chi-Wan Chow
- Department of Translational Molecular Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xizeng Mao
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xingzhi Song
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Won-Chul Lee
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jia Wu
- Department of Imaging Physics, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Latasha Little
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Curtis Gumbs
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Carmen Behrens
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Cesar Moran
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Annikka Weissferdt
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - J Jack Lee
- Department of Biostatistics, Division of Basic Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Boris Sepesi
- Department of Thoracic Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Stephen Swisher
- Department of Thoracic Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Chao Cheng
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jonathan Kurie
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Don Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ignacio I Wistuba
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.,Department of Translational Molecular Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - P Andrew Futreal
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Neda Kalhor
- Department of Pathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Jianjun Zhang
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
211
|
Harnessing Mitochondrial Mutations to ATAC Clonal Evolution in CLL. Cancer Discov 2021; 11:2965-2967. [DOI: 10.1158/2159-8290.cd-21-1225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
212
|
Matsutani T, Hamada M. Clone decomposition based on mutation signatures provides novel insights into mutational processes. NAR Genom Bioinform 2021; 3:lqab093. [PMID: 34734181 PMCID: PMC8559167 DOI: 10.1093/nargab/lqab093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/17/2021] [Accepted: 10/19/2021] [Indexed: 11/24/2022] Open
Abstract
Intra-tumor heterogeneity is a phenomenon in which mutation profiles differ from cell to cell within the same tumor and is observed in almost all tumors. Understanding intra-tumor heterogeneity is essential from the clinical perspective. Numerous methods have been developed to predict this phenomenon based on variant allele frequency. Among the methods, CloneSig models the variant allele frequency and mutation signatures simultaneously and provides an accurate clone decomposition. However, this method has limitations in terms of clone number selection and modeling. We propose SigTracer, a novel hierarchical Bayesian approach for analyzing intra-tumor heterogeneity based on mutation signatures to tackle these issues. We show that SigTracer predicts more reasonable clone decompositions than the existing methods against artificial data that mimic cancer genomes. We applied SigTracer to whole-genome sequences of blood cancer samples. The results were consistent with past findings that single base substitutions caused by a specific signature (previously reported as SBS9) related to the activation-induced cytidine deaminase intensively lie within immunoglobulin-coding regions for chronic lymphocytic leukemia samples. Furthermore, we showed that this signature mutates regions responsible for cell-cell adhesion. Accurate assignments of mutations to signatures by SigTracer can provide novel insights into signature origins and mutational processes.
Collapse
Affiliation(s)
- Taro Matsutani
- Graduate School of Advanced Science and Engineering, Waseda University, 55N-06-10, 3-4-1, Okubo Shinjuku-ku, Tokyo 169–8555, Japan
- Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo 169–8555, Japan
| | - Michiaki Hamada
- Graduate School of Advanced Science and Engineering, Waseda University, 55N-06-10, 3-4-1, Okubo Shinjuku-ku, Tokyo 169–8555, Japan
- Computational Bio Big-Data Open Innovation Laboratory (CBBD-OIL), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo 169–8555, Japan
- Graduate School of Medicine, Nippon Medical School, Sendagi, Bunkyo, Tokyo 113-8602, Japan
| |
Collapse
|
213
|
Kristensen NP, Heeke C, Tvingsholm SA, Borch A, Draghi A, Crowther MD, Carri I, Munk KK, Holm JS, Bjerregaard AM, Bentzen AK, Marquard AM, Szallasi Z, McGranahan N, Andersen R, Nielsen M, Jönsson GB, Donia M, Svane IM, Hadrup SR. Neoantigen-reactive CD8+ T cells affect clinical outcome of adoptive transfer with tumor-infiltrating lymphocytes in melanoma. J Clin Invest 2021; 132:150535. [PMID: 34813506 PMCID: PMC8759789 DOI: 10.1172/jci150535] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 11/18/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Neoantigen-driven recognition and T cell-mediated killing contribute to tumor clearance following adoptive cell therapy (ACT) with Tumor-Infiltrating Lymphocytes (TILs). Yet, how diversity, frequency, and persistence of expanded neoepitope-specific CD8+ T cells derived from TIL infusion products affect patient outcome is not fully determined. METHODS Using barcoded pMHC multimers, we provide a comprehensive mapping of CD8+ T cells recognizing neoepitopes in TIL infusion products and blood samples from 26 metastatic mela-noma patients who received ACT. RESULTS We identified 106 neoepitopes within TIL infusion products corresponding to 1.8% of all predicted neoepitopes. We observed neoepitope-specific recognition to be virtually devoid in TIL infusion products given to patients with progressive disease outcome. Moreover, we found that the frequency of neoepitope-specific CD8+ T cells in TIL infusion products correlated with in-creased survival, and that detection of engrafted CD8+ T cells in post-treatment (i.e. originating from the TIL infusion product) were unique to responders of TIL-ACT. Finally, we found that a transcriptional signature for lymphocyte activity within the tumor microenvironment was associated with a higher frequency of neoepitope-specific CD8+ T cells in the infusion product. CONCLUSIONS These data support previous case studies of neoepitope-specific CD8+ T cells in melanoma, and indicate that successful TIL-ACT is associated with an expansion of neoepitope-specific CD8+ T cells. FUNDING NEYE Foundation; European Research Council; Lundbeck Foundation Fellowship; Carlsberg Foundation.
Collapse
Affiliation(s)
- Nikolaj Pagh Kristensen
- Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, Denmark
| | - Christina Heeke
- Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, Denmark
| | - Siri A Tvingsholm
- Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, Denmark
| | - Annie Borch
- Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, Denmark
| | - Arianna Draghi
- Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | | | - Ibel Carri
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, Buenos Aires, Argentina
| | - Kamilla K Munk
- Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, Denmark
| | - Jeppe Sejerø Holm
- Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, Denmark
| | - Anne-Mette Bjerregaard
- Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, Denmark
| | - Amalie Kai Bentzen
- Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, Denmark
| | - Andrea M Marquard
- Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, Denmark
| | - Zoltan Szallasi
- Danish Cancer Society Research Center, Danish Cancer Society, Copenhagen, Denmark
| | | | - Rikke Andersen
- Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Morten Nielsen
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, Denmark
| | - Göran B Jönsson
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Marco Donia
- Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Inge Marie Svane
- Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Sine Reker Hadrup
- Department of Health Technology, Technical University of Denmark (DTU), Kgs. Lyngby, Denmark
| |
Collapse
|
214
|
RDAClone: Deciphering Tumor Heterozygosity through Single-Cell Genomics Data Analysis with Robust Deep Autoencoder. Genes (Basel) 2021; 12:genes12121847. [PMID: 34946794 PMCID: PMC8701080 DOI: 10.3390/genes12121847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/27/2022] Open
Abstract
Rapid advances in single-cell genomics sequencing (SCGS) have allowed researchers to characterize tumor heterozygosity with unprecedented resolution and reveal the phylogenetic relationships between tumor cells or clones. However, high sequencing error rates of current SCGS data, i.e., false positives, false negatives, and missing bases, severely limit its application. Here, we present a deep learning framework, RDAClone, to recover genotype matrices from noisy data with an extended robust deep autoencoder, cluster cells into subclones by the Louvain-Jaccard method, and further infer evolutionary relationships between subclones by the minimum spanning tree. Studies on both simulated and real datasets demonstrate its robustness and superiority in data denoising, cell clustering, and evolutionary tree reconstruction, particularly for large datasets.
Collapse
|
215
|
Chen M, Chen R, Jin Y, Li J, Hu X, Zhang J, Fujimoto J, Hubert SM, Gay CM, Zhu B, Tian Y, McGranahan N, Lee WC, George J, Hu X, Chen Y, Wu M, Behrens C, Chow CW, Pham HHN, Fukuoka J, Wu J, Parra ER, Little LD, Gumbs C, Song X, Wu CJ, Diao L, Wang Q, Cardnell R, Zhang J, Wang J, Le X, Gibbons DL, Heymach JV, Jack Lee J, William WN, Cheng C, Glisson B, Wistuba I, Andrew Futreal P, Thomas RK, Reuben A, Byers LA, Zhang J. Cold and heterogeneous T cell repertoire is associated with copy number aberrations and loss of immune genes in small-cell lung cancer. Nat Commun 2021; 12:6655. [PMID: 34789716 PMCID: PMC8599854 DOI: 10.1038/s41467-021-26821-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/25/2021] [Indexed: 02/03/2023] Open
Abstract
Small-cell lung cancer (SCLC) is speculated to harbor complex genomic intratumor heterogeneity (ITH) associated with high recurrence rate and suboptimal response to immunotherapy. Here, using multi-region whole exome/T cell receptor (TCR) sequencing as well as immunohistochemistry, we reveal a rather homogeneous mutational landscape but extremely cold and heterogeneous TCR repertoire in limited-stage SCLC tumors (LS-SCLCs). Compared to localized non-small cell lung cancers, LS-SCLCs have similar predicted neoantigen burden and genomic ITH, but significantly colder and more heterogeneous TCR repertoire associated with higher chromosomal copy number aberration (CNA) burden. Furthermore, copy number loss of IFN-γ pathway genes is frequently observed and positively correlates with CNA burden. Higher mutational burden, higher T cell infiltration and positive PD-L1 expression are associated with longer overall survival (OS), while higher CNA burden is associated with shorter OS in patients with LS-SCLC.
Collapse
Affiliation(s)
- Ming Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510060, China. .,The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, 310022, China. .,Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China. .,Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China.
| | - Runzhe Chen
- grid.12981.330000 0001 2360 039XDepartment of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510060 China ,grid.240145.60000 0001 2291 4776Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA ,grid.240145.60000 0001 2291 4776Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Ying Jin
- grid.410726.60000 0004 1797 8419The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022 China ,grid.9227.e0000000119573309Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022 China ,Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022 China
| | - Jun Li
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Xin Hu
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Jiexin Zhang
- grid.240145.60000 0001 2291 4776Department of Bioinformatics and Computational Biology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Junya Fujimoto
- grid.240145.60000 0001 2291 4776Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Shawna M. Hubert
- grid.240145.60000 0001 2291 4776Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA ,grid.240145.60000 0001 2291 4776Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Carl M. Gay
- grid.240145.60000 0001 2291 4776Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Bo Zhu
- grid.240145.60000 0001 2291 4776Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA ,grid.240145.60000 0001 2291 4776Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Yanhua Tian
- grid.240145.60000 0001 2291 4776Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA ,grid.240145.60000 0001 2291 4776Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Nicholas McGranahan
- grid.11485.390000 0004 0422 0975Cancer Research United Kingdom-University College London Lung Cancer Centre of Excellence, London, WC1E6BT UK
| | - Won-Chul Lee
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Julie George
- grid.6190.e0000 0000 8580 3777Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931 Germany ,grid.411097.a0000 0000 8852 305XDepartment of Otorhinolaryngology, Head and Neck Surgery, University Hospital Cologne, 50937 Cologne, Germany
| | - Xiao Hu
- grid.410726.60000 0004 1797 8419The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022 China ,grid.9227.e0000000119573309Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022 China ,Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022 China
| | - Yamei Chen
- grid.410726.60000 0004 1797 8419The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022 China ,grid.9227.e0000000119573309Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022 China ,Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022 China
| | - Meijuan Wu
- grid.410726.60000 0004 1797 8419The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022 China ,grid.9227.e0000000119573309Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022 China ,Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang 310022 China
| | - Carmen Behrens
- grid.240145.60000 0001 2291 4776Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Chi-Wan Chow
- grid.240145.60000 0001 2291 4776Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Hoa H. N. Pham
- grid.174567.60000 0000 8902 2273Department of Pathology, Nagasaki University Graduate school of Biomedical Sciences, Nagasaki, Japan
| | - Junya Fukuoka
- grid.174567.60000 0000 8902 2273Department of Pathology, Nagasaki University Graduate school of Biomedical Sciences, Nagasaki, Japan
| | - Jia Wu
- grid.240145.60000 0001 2291 4776Department of Image Physics, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Edwin Roger Parra
- grid.240145.60000 0001 2291 4776Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Latasha D. Little
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Curtis Gumbs
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Xingzhi Song
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Chang-Jiun Wu
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Lixia Diao
- grid.240145.60000 0001 2291 4776Department of Bioinformatics and Computational Biology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Qi Wang
- grid.240145.60000 0001 2291 4776Department of Bioinformatics and Computational Biology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Robert Cardnell
- grid.240145.60000 0001 2291 4776Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Jianhua Zhang
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Jing Wang
- grid.240145.60000 0001 2291 4776Department of Bioinformatics and Computational Biology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Xiuning Le
- grid.240145.60000 0001 2291 4776Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Don L. Gibbons
- grid.240145.60000 0001 2291 4776Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - John V. Heymach
- grid.240145.60000 0001 2291 4776Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - J. Jack Lee
- grid.240145.60000 0001 2291 4776Department of Biostatistics, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - William N. William
- grid.240145.60000 0001 2291 4776Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Chao Cheng
- grid.39382.330000 0001 2160 926XInstitute for Clinical and Translational Research, Baylor College of Medicine, Houston, Texas 77030 USA
| | - Bonnie Glisson
- grid.240145.60000 0001 2291 4776Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Ignacio Wistuba
- grid.240145.60000 0001 2291 4776Department of Translational Molecular Pathology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - P. Andrew Futreal
- grid.240145.60000 0001 2291 4776Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Roman K. Thomas
- grid.6190.e0000 0000 8580 3777Department of Translational Genomics, Medical Faculty, University of Cologne, Cologne, 50931 Germany ,grid.411097.a0000 0000 8852 305XDepartment of Pathology, Medical Faculty, University Hospital Cologne, Cologne, 50931 Germany ,grid.7497.d0000 0004 0492 0584DKFZ, German Cancer Research Center and German Cancer Consortium (DKTK), Heidelberg, 69115 Germany
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA.
| | - Lauren A. Byers
- grid.240145.60000 0001 2291 4776Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030 USA
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA. .,Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas, 77030, USA.
| |
Collapse
|
216
|
Utro F, Levovitz C, Rhrissorrakrai K, Parida L. A common methodological phylogenomics framework for intra-patient heteroplasmies to infer SARS-CoV-2 sublineages and tumor clones. BMC Genomics 2021; 22:518. [PMID: 34789161 PMCID: PMC8596094 DOI: 10.1186/s12864-021-07660-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 04/28/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND All diseases containing genetic material undergo genetic evolution and give rise to heterogeneity including cancer and infection. Although these illnesses are biologically very different, the ability for phylogenetic retrodiction based on the genomic reads is common between them and thus tree-based principles and assumptions are shared. Just as the different frequencies of tumor genomic variants presupposes the existence of multiple tumor clones and provides a handle to computationally infer them, we postulate that the different variant frequencies in viral reads offers the means to infer multiple co-infecting sublineages. RESULTS We present a common methodological framework to infer the phylogenomics from genomic data, be it reads of SARS-CoV-2 of multiple COVID-19 patients or bulk DNAseq of the tumor of a cancer patient. We describe the Concerti computational framework for inferring phylogenies in each of the two scenarios.To demonstrate the accuracy of the method, we reproduce some known results in both scenarios. We also make some additional discoveries. CONCLUSIONS Concerti successfully extracts and integrates information from multi-point samples, enabling the discovery of clinically plausible phylogenetic trees that capture the heterogeneity known to exist both spatially and temporally. These models can have direct therapeutic implications by highlighting "birth" of clones that may harbor resistance mechanisms to treatment, "death" of subclones with drug targets, and acquisition of functionally pertinent mutations in clones that may have seemed clinically irrelevant. Specifically in this paper we uncover new potential parallel mutations in the evolution of the SARS-CoV-2 virus. In the context of cancer, we identify new clones harboring resistant mutations to therapy.
Collapse
Affiliation(s)
- Filippo Utro
- IBM Research, T.J. Watson Research Center, Yorktown Heights, USA
| | - Chaya Levovitz
- IBM Research, T.J. Watson Research Center, Yorktown Heights, USA
| | | | - Laxmi Parida
- IBM Research, T.J. Watson Research Center, Yorktown Heights, USA
| |
Collapse
|
217
|
Chattopadhyay S, Karlsson J, Valind A, Andersson N, Gisselsson D. Tracing the evolution of aneuploid cancers by multiregional sequencing with CRUST. Brief Bioinform 2021; 22:bbab292. [PMID: 34343239 PMCID: PMC8981300 DOI: 10.1093/bib/bbab292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/16/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022] Open
Abstract
Clonal deconvolution of mutational landscapes is crucial to understand the evolutionary dynamics of cancer. Two limiting factors for clonal deconvolution that have remained unresolved are variation in purity and chromosomal copy number across different samples of the same tumor. We developed a semi-supervised algorithm that tracks variant calls through multi-sample spatiotemporal tumor data. While normalizing allele frequencies based on purity, it also adjusts for copy number changes at clonal deconvolution. Absent à priori copy number data, it renders in silico copy number estimations from bulk sequences. Using published and simulated tumor sequences, we reliably segregated clonal/subclonal variants even at a low sequencing depth (~50×). Given at least one pure tumor sample (>70% purity), we could normalize and deconvolve paired samples down to a purity of 40%. This renders a reliable clonal reconstruction well adapted to multi-regionally sampled solid tumors, which are often aneuploid and contaminated by non-cancer cells.
Collapse
Affiliation(s)
- Subhayan Chattopadhyay
- Division of Clinical Genetics, Department of Laboratory
Medicine, Lund University, Lund, Sweden
| | - Jenny Karlsson
- Division of Clinical Genetics, Department of Laboratory
Medicine, Lund University, Lund, Sweden
| | - Anders Valind
- Division of Clinical Genetics, Department of Laboratory
Medicine, Lund University, Lund, Sweden
- Department of Pediatrics, Skåne University
Hospital, Lund, Sweden
| | - Natalie Andersson
- Division of Clinical Genetics, Department of Laboratory
Medicine, Lund University, Lund, Sweden
| | - David Gisselsson
- Division of Clinical Genetics, Department of Laboratory
Medicine, Lund University, Lund, Sweden
- Division of Oncology and Pathology, Department of Clinical
Sciences, Lund University, Lund, Sweden
- Clinical Genetics and Pathology, Laboratory Medicine,
Lund University Hospital, Lund, Sweden
| |
Collapse
|
218
|
Benchmarking pipelines for subclonal deconvolution of bulk tumour sequencing data. Nat Commun 2021; 12:6396. [PMID: 34737285 PMCID: PMC8569188 DOI: 10.1038/s41467-021-26698-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 10/20/2021] [Indexed: 11/09/2022] Open
Abstract
Intratumour heterogeneity provides tumours with the ability to adapt and acquire treatment resistance. The development of more effective and personalised treatments for cancers, therefore, requires accurate characterisation of the clonal architecture of tumours, enabling evolutionary dynamics to be tracked. Many methods exist for achieving this from bulk tumour sequencing data, involving identifying mutations and performing subclonal deconvolution, but there is a lack of systematic benchmarking to inform researchers on which are most accurate, and how dataset characteristics impact performance. To address this, we use the most comprehensive tumour genome simulation tool available for such purposes to create 80 bulk tumour whole exome sequencing datasets of differing depths, tumour complexities, and purities, and use these to benchmark subclonal deconvolution pipelines. We conclude that i) tumour complexity does not impact accuracy, ii) increasing either purity or purity-corrected sequencing depth improves accuracy, and iii) the optimal pipeline consists of Mutect2, FACETS and PyClone-VI. We have made our benchmarking datasets publicly available for future use.
Collapse
|
219
|
Ogundijo OE, Zhu K, Wang X, Anastassiou D. Characterizing Intra-Tumor Heterogeneity From Somatic Mutations Without Copy-Neutral Assumption. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:2271-2280. [PMID: 32070995 DOI: 10.1109/tcbb.2020.2973635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Bulk samples of the same patient are heterogeneous in nature, comprising of different subpopulations (subclones) of cancer cells. Cells in a tumor subclone are characterized by unique mutational genotype profile. Resolving tumor heterogeneity by estimating the genotypes, cellular proportions and the number of subclones present in the tumor can help in understanding cancer progression and treatment. We present a novel method, ChaClone2, to efficiently deconvolve the observed variant allele fractions (VAFs), with consideration for possible effects from copy number aberrations at the mutation loci. Our method describes a state-space formulation of the feature allocation model, deconvolving the observed VAFs from samples of the same patient into three matrices: subclonal total and variant copy numbers for mutated genes, and proportions of subclones in each sample. We describe an efficient sequential Monte Carlo (SMC) algorithm to estimate these matrices. Extensive simulation shows that the ChaClone2 yields better accuracy when compared with other state-of-the-art methods for addressing similar problem and it offers scalability to large datasets. Also, ChaClone2 features that the model parameter estimates can be refined whenever new mutation data of freshly sequenced genomic locations are available. MATLAB code and datasets are available to download at: https://github.com/moyanre/method2.
Collapse
|
220
|
Zhai W, Lai H, Kaya NA, Chen J, Yang H, Lu B, Lim JQ, Ma S, Chew SC, Chua KP, Alvarez JJS, Chen PJ, Chang MM, Wu L, Goh BKP, Chung AYF, Chan CY, Cheow PC, Lee SY, Kam JH, Kow AWC, Ganpathi IS, Chanwat R, Thammasiri J, Yoong BK, Ong DBL, de Villa VH, Dela Cruz RD, Loh TJ, Wan WK, Zeng Z, Skanderup AJ, Pang YH, Madhavan K, Lim TKH, Bonney G, Leow WQ, Chew V, Dan YY, Tam WL, Toh HC, Foo RSY, Chow PKH. Dynamic phenotypic heterogeneity and the evolution of multiple RNA subtypes in hepatocellular carcinoma: the PLANET study. Natl Sci Rev 2021; 9:nwab192. [PMID: 35382356 PMCID: PMC8973408 DOI: 10.1093/nsr/nwab192] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
Intra-tumor heterogeneity (ITH) is a key challenge in cancer treatment, but previous studies have focused mainly on the genomic alterations without exploring phenotypic (transcriptomic and immune) heterogeneity. Using one of the largest prospective surgical cohorts for hepatocellular carcinoma (HCC) with multi-region sampling, we sequenced whole genomes and paired transcriptomes from 67 HCC patients (331 samples). We found that while genomic ITH was rather constant across stages, phenotypic ITH had a very different trajectory and quickly diversified in stage II patients. Most strikingly, 30% of patients were found to contain more than one transcriptomic subtype within a single tumor. Such phenotypic ITH was found to be much more informative in predicting patient survival than genomic ITH and explains the poor efficacy of single-target systemic therapies in HCC. Taken together, we not only revealed an unprecedentedly dynamic landscape of phenotypic heterogeneity in HCC, but also highlighted the importance of studying phenotypic evolution across cancer types.
Collapse
Affiliation(s)
- Weiwei Zhai
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Hannah Lai
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Neslihan Arife Kaya
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Jianbin Chen
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Hechuan Yang
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Bingxin Lu
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Jia Qi Lim
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Siming Ma
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Sin Chi Chew
- Division of Surgery and Surgical Oncology, National Cancer Centre, Singapore 169610, Singapore
| | - Khi Pin Chua
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | | | - Pauline Jieqi Chen
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Mei Mei Chang
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Lingyan Wu
- Division of Surgery and Surgical Oncology, National Cancer Centre, Singapore 169610, Singapore
| | - Brian K P Goh
- Department of Hepato-Pancreato-Biliary and Transplant Surgery, Singapore General Hospital, Singapore 169608, Singapore
| | - Alexander Yaw-Fui Chung
- Department of Hepato-Pancreato-Biliary and Transplant Surgery, Singapore General Hospital, Singapore 169608, Singapore
| | - Chung Yip Chan
- Department of Hepato-Pancreato-Biliary and Transplant Surgery, Singapore General Hospital, Singapore 169608, Singapore
| | - Peng Chung Cheow
- Department of Hepato-Pancreato-Biliary and Transplant Surgery, Singapore General Hospital, Singapore 169608, Singapore
| | - Ser Yee Lee
- Department of Hepato-Pancreato-Biliary and Transplant Surgery, Singapore General Hospital, Singapore 169608, Singapore
| | - Juinn Huar Kam
- Department of Hepato-Pancreato-Biliary and Transplant Surgery, Singapore General Hospital, Singapore 169608, Singapore
| | - Alfred Wei-Chieh Kow
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore 119228, Singapore
| | - Iyer Shridhar Ganpathi
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore 119228, Singapore
| | - Rawisak Chanwat
- Hepato-Pancreato-Biliary Surgery Unit, Department of Surgery, National Cancer Institute, Bangkok 10310, Thailand
| | - Jidapa Thammasiri
- Division of Pathology, National Cancer Institute, Bangkok 10400, Thailand
| | - Boon Koon Yoong
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 59100, Malaysia
| | - Diana Bee-Lan Ong
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 59100, Malaysia
| | - Vanessa H de Villa
- Department of Surgery and Center for Liver Disease Management and Transplantation, The Medical City, Pasig City, Metro Manila, Philippines
| | | | - Tracy Jiezhen Loh
- Department of Pathology, Singapore General Hospital, Singapore 169608, Singapore
| | - Wei Keat Wan
- Department of Pathology, Singapore General Hospital, Singapore 169608, Singapore
| | - Zeng Zeng
- Institute for Infocomm Research, ASTAR, Singapore 138632, Singapore
| | - Anders Jacobsen Skanderup
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Yin Huei Pang
- Department of Pathology, National University Health System, Singapore 119228, Singapore
| | - Krishnakumar Madhavan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore 119228, Singapore
| | - Tony Kiat-Hon Lim
- Department of Pathology, Singapore General Hospital, Singapore 169608, Singapore
| | - Glenn Bonney
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, University Surgical Cluster, National University Health System, Singapore 119228, Singapore
| | - Wei Qiang Leow
- Department of Pathology, Singapore General Hospital, Singapore 169608, Singapore
| | - Valerie Chew
- Translational Immunology Institute (TII), SingHealth Duke-NUS Academic Medical Centre, Singapore 168753, Singapore
| | - Yock Young Dan
- Division of Gastroenterology and Hepatology, University Medicine Cluster, National University Hospital, Singapore 119228, Singapore
| | - Wai Leong Tam
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Center Singapore, Singapore 169610, Singapore
| | - Roger Sik-Yin Foo
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| | - Pierce Kah-Hoe Chow
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore 138672, Singapore
| |
Collapse
|
221
|
Rosendahl Huber A, Van Hoeck A, Van Boxtel R. The Mutagenic Impact of Environmental Exposures in Human Cells and Cancer: Imprints Through Time. Front Genet 2021; 12:760039. [PMID: 34745228 PMCID: PMC8565797 DOI: 10.3389/fgene.2021.760039] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/05/2021] [Indexed: 12/25/2022] Open
Abstract
During life, the DNA of our cells is continuously exposed to external damaging processes. Despite the activity of various repair mechanisms, DNA damage eventually results in the accumulation of mutations in the genomes of our cells. Oncogenic mutations are at the root of carcinogenesis, and carcinogenic agents are often highly mutagenic. Over the past decade, whole genome sequencing data of healthy and tumor tissues have revealed how cells in our body gradually accumulate mutations because of exposure to various mutagenic processes. Dissection of mutation profiles based on the type and context specificities of the altered bases has revealed a variety of signatures that reflect past exposure to environmental mutagens, ranging from chemotherapeutic drugs to genotoxic gut bacteria. In this review, we discuss the latest knowledge on somatic mutation accumulation in human cells, and how environmental mutagenic factors further shape the mutation landscapes of tissues. In addition, not all carcinogenic agents induce mutations, which may point to alternative tumor-promoting mechanisms, such as altered clonal selection dynamics. In short, we provide an overview of how environmental factors induce mutations in the DNA of our healthy cells and how this contributes to carcinogenesis. A better understanding of how environmental mutagens shape the genomes of our cells can help to identify potential preventable causes of cancer.
Collapse
Affiliation(s)
- Axel Rosendahl Huber
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - Arne Van Hoeck
- Oncode Institute, Utrecht, Netherlands
- Center for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Ruben Van Boxtel
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
222
|
Yan H, Tarabichi M, McGranahan N, Van Loo P. DeCiFering the subclonal composition of tumors. Cell Syst 2021; 12:955-957. [PMID: 34672959 DOI: 10.1016/j.cels.2021.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Accurately identifying the subclones that make up tumors is critical for understanding cancer biology. In an article in this issue of Cell Systems, Satas et al. examine mutations with an evolutionary perspective to decipher the composition of tumors.
Collapse
Affiliation(s)
- Haixi Yan
- The Francis Crick Institute, London, UK
| | - Maxime Tarabichi
- The Francis Crick Institute, London, UK; Institute for Interdisciplinary Research, Université Libre de Bruxelles, Brussels, Belgium
| | | | | |
Collapse
|
223
|
Satas G, Zaccaria S, El-Kebir M, Raphael BJ. DeCiFering the elusive cancer cell fraction in tumor heterogeneity and evolution. Cell Syst 2021; 12:1004-1018.e10. [PMID: 34416171 DOI: 10.1016/j.cels.2021.07.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/09/2021] [Accepted: 07/21/2021] [Indexed: 12/22/2022]
Abstract
The cancer cell fraction (CCF), or proportion of cancerous cells in a tumor containing a single-nucleotide variant (SNV), is a fundamental statistic used to quantify tumor heterogeneity and evolution. Existing CCF estimation methods from bulk DNA sequencing data assume that every cell with an SNV contains the same number of copies of the SNV. This assumption is unrealistic in tumors with copy-number aberrations that alter SNV multiplicities. Furthermore, the CCF does not account for SNV losses due to copy-number aberrations, confounding downstream phylogenetic analyses. We introduce DeCiFer, an algorithm that overcomes these limitations by clustering SNVs using a novel statistic, the descendant cell fraction (DCF). The DCF quantifies both the prevalence of an SNV at the present time and its past evolutionary history using an evolutionary model that allows mutation losses. We show that DeCiFer yields more parsimonious reconstructions of tumor evolution than previously reported for 49 prostate cancer samples.
Collapse
Affiliation(s)
- Gryte Satas
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Simone Zaccaria
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Mohammed El-Kebir
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Benjamin J Raphael
- Department of Computer Science, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
224
|
Chen L, Qing Y, Li R, Li C, Li H, Feng X, Li SC. Somatic variant analysis suite: copy number variation clonal visualization online platform for large-scale single-cell genomics. Brief Bioinform 2021; 23:6406714. [PMID: 34671807 DOI: 10.1093/bib/bbab452] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 11/15/2022] Open
Abstract
The recent advance of single-cell copy number variation (CNV) analysis plays an essential role in addressing intratumor heterogeneity, identifying tumor subgroups and restoring tumor-evolving trajectories at single-cell scale. Informative visualization of copy number analysis results boosts productive scientific exploration, validation and sharing. Several single-cell analysis figures have the effectiveness of visualizations for understanding single-cell genomics in published articles and software packages. However, they almost lack real-time interaction, and it is hard to reproduce them. Moreover, existing tools are time-consuming and memory-intensive when they reach large-scale single-cell throughputs. We present an online visualization platform, single-cell Somatic Variant Analysis Suite (scSVAS), for real-time interactive single-cell genomics data visualization. scSVAS is specifically designed for large-scale single-cell genomic analysis that provides an arsenal of unique functionalities. After uploading the specified input files, scSVAS deploys the online interactive visualization automatically. Users may conduct scientific discoveries, share interactive visualizations and download high-quality publication-ready figures. scSVAS provides versatile utilities for managing, investigating, sharing and publishing single-cell CNV profiles. We envision this online platform will expedite the biological understanding of cancer clonal evolution in single-cell resolution. All visualizations are publicly hosted at https://sc.deepomics.org.
Collapse
Affiliation(s)
- Lingxi Chen
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| | - Yuhao Qing
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| | - Ruikang Li
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| | - Chaohui Li
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| | - Hechen Li
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong, China.,School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta GA 30332, USA
| | - Xikang Feng
- School of Software, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Shuai Cheng Li
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| |
Collapse
|
225
|
Luttermann T, Rückert C, Wibberg D, Busche T, Schwarzhans JP, Friehs K, Kalinowski J. Establishment of a near-contiguous genome sequence of the citric acid producing yeast Yarrowia lipolytica DSM 3286 with resolution of rDNA clusters and telomeres. NAR Genom Bioinform 2021; 3:lqab085. [PMID: 34661101 PMCID: PMC8515841 DOI: 10.1093/nargab/lqab085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 09/01/2021] [Accepted: 10/13/2021] [Indexed: 11/29/2022] Open
Abstract
Yarrowia lipolytica is an oleaginous yeast that is particularly suitable for the sustainable production of secondary metabolites. The genome of this yeast is characterized by its relatively large size and its high number of different rDNA clusters located in its telomeric regions. However, due to the presence of long repetitive elements in the sub-telomeric regions, rDNA clusters and telomeres are missing in current genome assemblies of Y. lipolytica. Here, we present the near-contiguous genome sequence of the biotechnologically relevant strain DSM 3286. We employed a hybrid assembly strategy combining Illumina and nanopore sequencing reads to integrate all six rDNA clusters as well as telomeric repeats into the genome sequence. By fine-tuning of DNA isolation and library preparation protocols, we were able to create ultra-long reads that not only contained multiples of mitochondrial genomes but also shed light on the inter- and intra-chromosomal diversity of rDNA cluster types. We show that there are ten different rDNA units present in this strain that additionally appear in a predefined order in a cluster. Based on single reads, we also demonstrate that the number of rDNA repeats in a specific cluster varies from cell to cell within a population.
Collapse
Affiliation(s)
- Tobias Luttermann
- Microbial Genomics and Biotechnology, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, NRW 33615, Germany
| | - Christian Rückert
- Microbial Genomics and Biotechnology, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, NRW 33615, Germany
| | - Daniel Wibberg
- Genome Research of Industrial Microorganisms, Bielefeld University, Bielefeld, NRW 33615, Germany
| | - Tobias Busche
- Microbial Genomics and Biotechnology, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, NRW 33615, Germany
| | | | - Karl Friehs
- Fermentation Engineering, Bielefeld University, Bielefeld, NRW 33615, Germany
| | - Jörn Kalinowski
- Microbial Genomics and Biotechnology, Center for Biotechnology (CeBiTec), Bielefeld University, Bielefeld, NRW 33615, Germany
| |
Collapse
|
226
|
Lu X, Gao W, Zhang Y, Wang T, Gao H, Chen Q, Shi X, Lian B, Zhang W, Gao X, Li J. Case Report: Systemic Treatment and Serial Genomic Sequencing of Metastatic Prostate Adenocarcinoma Progressing to Small Cell Carcinoma. Front Oncol 2021; 11:732071. [PMID: 34646773 PMCID: PMC8503647 DOI: 10.3389/fonc.2021.732071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/06/2021] [Indexed: 12/18/2022] Open
Abstract
Small cell carcinoma (SCC)/neuroendocrine prostate cancer (NEPC) is a rare and highly aggressive subtype of prostate cancer associated with an AR(androgen receptor)-null phenotype and visceral metastases. This study presents a 44-year-old man originally diagnosed with metastatic hormone-sensitive prostatic adenocarcinoma. After 6-month androgen deprivation therapy (ADT) combined with docetaxel, the patient developed paraplegia. Laminectomy was performed, and a thoracic vertebral biopsy revealed neuroendocrine differentiation and mixed adenocarcinoma. The patient developed liver metastases and experienced stable disease for 4 months following etoposide combined with cisplatin and pembrolizumab. Seminal vesicle biopsy after chemotherapy revealed small-cell cancer. The prostate biopsy specimen also indicated pure SCC. We witnessed the dynamic evolution from pure adenocarcinoma to fully differentiated SCC, leading to obstruction and death. In addition, whole-exome sequencing was performed on both biopsy specimens of the thoracic vertebra at the beginning of castration resistance and that of seminal vesicle after multiple lines of treatment failure. Utilizing phylogenetic reconstruction, we observed that both samples shared a common ancestor clone harboring aberrations in the TP53, RB1, and NF2 genes. We also discovered that driver events in the private subclones of both samples, such as alterations in CDC27 and RUNX1, might have played a significant role in tumor progression or even neuroendocrine differentiation. Tumor biopsy and IHC assessment must be repeated at different stages of progression, because of intrapatient spatial and temporal heterogeneity of adenocarcinoma versus SCC/NEPC. Although, typical treatments including ADT, docetaxel, etoposide, cisplatin, and pembrolizumab provided temporary response, the patient still had a poor prognosis.
Collapse
Affiliation(s)
- XiaoJun Lu
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Wenwen Gao
- Department of Oncology, Shidong Hospital, Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Yu Zhang
- Department of Bioinformatics, Center for Translational Medicine, Second Military Medical University, Shanghai, China
| | - Tao Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongliang Gao
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qing Chen
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiaolei Shi
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Bijun Lian
- Department of Urology, The 903th PLA Hospital, Hangzhou, China
| | - Wenhui Zhang
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xu Gao
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jing Li
- Department of Bioinformatics, Center for Translational Medicine, Second Military Medical University, Shanghai, China
| |
Collapse
|
227
|
Genomic comparison between cerebrospinal fluid and primary tumor revealed the genetic events associated with brain metastasis in lung adenocarcinoma. Cell Death Dis 2021; 12:935. [PMID: 34642306 PMCID: PMC8511004 DOI: 10.1038/s41419-021-04223-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 12/26/2022]
Abstract
Lung adenocarcinoma (LUAD) is most common pathological type of lung cancer. LUAD with brain metastases (BMs) usually have poor prognosis. To identify the potential genetic factors associated with BM, a genomic comparison for BM cerebrospinal fluid (CSF) and primary lung tumor samples obtained from 1082 early- and late-stage LUAD patients was performed. We found that single nucleotide variation (SNV) of EGFR was highly enriched in CSF (87% of samples). Compared with the other primary lung tissues, copy number gain of EGFR (27%), CDK4 (11%), PMS2 (11%), MET (10%), IL7R (8%), RICTOR (7%), FLT4 (5%), and FGFR4 (4%), and copy number loss of CDKN2A (28%) and CDKN2B (18%) were remarkably more frequent in CSF samples. CSF had significantly lower tumor mutation burden (TMB) level but more abundant copy number variant. It was also found that the relationships among co-occurrent and mutually exclusive genes were dynamically changing with LUAD development. Additionally, CSF (97% of samples) harbored more abundant targeted drugs related driver and fusion genes. The signature 15 associated with defective DNA mismatch repair (dMMR) was only identified in the CSF group. Cancer associated pathway analysis further revealed that ErbB (95%) and cell cycle (84%) were unique pathways in CSF samples. The tumor evolution analysis showed that CSF carried significantly fewer clusters, but subclonal proportion of EGFR was remarkably increased with tumor progression. Collectively, CSF sequencing showed unique genomic characteristics and the intense copy number instability associated with cell cycle disorder and dMMR might be the crucial genetic factors in BM of LUAD.
Collapse
|
228
|
Zhou YF, Xiao Y, Jin X, Di GH, Jiang YZ, Shao ZM. Integrated analysis reveals prognostic value of HLA-I LOH in triple-negative breast cancer. J Immunother Cancer 2021; 9:jitc-2021-003371. [PMID: 34615706 PMCID: PMC8496394 DOI: 10.1136/jitc-2021-003371] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2021] [Indexed: 12/20/2022] Open
Abstract
Background Triple-negative breast cancers (TNBCs), especially those non-immune-inflamed tumors, have a poor prognosis and limited therapies. Human leukocyte antigen (HLA)-I not only contributes to antitumor immune response and the phenotype of the tumor microenvironment, but also is a negative predictor of outcomes after immunotherapy. However, the importance of HLA functional status in TNBCs remains poorly understood. Methods Using the largest original multiomics datasets on TNBCs, we systematically characterized the HLA-Ⅰ status of TNBCs from the perspective of HLA-Ⅰ homogeneity and loss of heterozygosity (LOH). The prognostic significance of HLA-I status was measured. To explain the potential mechanism of prognostic value in HLA-Ⅰ status, the mutational signature, copy number alteration, neoantigen and intratumoral heterogeneity were measured. Furthermore, the correlation between HLA-Ⅰ functional status and the tumor immune microenvironment was analyzed. Results LOH and homogeneity in HLA-I accounted for 18% and 21% of TNBCs, respectively. HLA-I LOH instead of HLA-I homogeneity was an independent prognostic biomarker in TNBCs. In particular, for patients with non-immune-inflamed tumors, HLA-I LOH indicated a worse prognosis than HLA-I non-LOH. Furthermore, integrated genomic and transcriptomic analysis showed that HLA-I LOH was accompanied by upregulated scores of mutational signature 3 and homologous recombination deficiency scores, which implied the failure of DNA double-strand break repair. Moreover, HLA-I LOH had higher mutation and neoantigen loads and more subclones than HLA-I non-LOH. These results indicated that although HLA-I LOH tumors with failure of DNA double-strand break repair were prone to produce neoantigens, their limited capacity for antigen presentation finally contributed to poor immune selection pressure. Conclusion Our study illustrates the genomic landscape of HLA-I functional status and stresses the prognostic significance of HLA-I LOH in TNBCs. For “cold” tumors in TNBCs, HLA-I LOH indicated a worse prognosis than HLA-I non-LOH.
Collapse
Affiliation(s)
- Yi-Fan Zhou
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Xi Jin
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Gen-Hong Di
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Institutes of Biomedical Sciences, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
229
|
Feng Y, Liu Y, Yuan M, Dong G, Zhang H, Zhang T, Chang L, Xia X, Li L, Zhu H, Xing P, Wang H, Shi Y, Wang Z, Hu X. The Feasibility of Using Biomarkers Derived from Circulating Tumor DNA Sequencing as Predictive Classifiers in Patients with Small-Cell Lung Cancer. Cancer Res Treat 2021; 54:753-766. [PMID: 34645133 PMCID: PMC9296939 DOI: 10.4143/crt.2021.905] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/28/2021] [Indexed: 11/21/2022] Open
Abstract
Purpose To investigate the feasibility of biomarkers based on dynamic circulating tumor DNA (ctDNA) to classify small cell lung cancer (SCLC) into different subtypes. Materials and Methods Tumor and longitudinal plasma ctDNA samples were analyzed by next-generation sequencing of 1,021 genes. PyClone was used to infer the molecular tumor burden index (mTBI). Pre-treatment tumor tissues [T1] and serial plasma samples were collected (pre-treatment [B1], after two [B2], six [B3] cycles of chemotherapy and at progression [B4]). Results Overall concordance between T1 and B1 sequencing (n=30) was 66.5%, and 89.5% in the gene of RB1. A classification method was designed according to the changes of RB1 mutation, named as subtype Ⅰ (both positive at B1 and B2), subtype Ⅱ (positive at B1 but negative at B2), and subtype Ⅲ (both negative at B1 and B2). The median progressive-free survival for subtype Ⅰ patients (4.5 months [95%CI: 2.6-5.8]) was inferior to subtype Ⅱ (not reached, p<0.0001) and subtype Ⅲ (10.8 months [95%CI: 6.0-14.4], p=0.002). The median overall survival for subtype Ⅰ patients (16.3 months [95%CI: 5.3-22.9]) was inferior to subtype Ⅱ (not reached, p=0.01) and subtype Ⅲ (not reached, p=0.02). Patients with a mTBI dropped to zero at B2 had longer median overall survival (not reached vs. 19.5 months, p=0.01). The changes of mTBI from B4 to B1 were sensitive to predict new metastases, with a sensitivity of 100% and a specificity of 85.7%. Conclusion Monitoring ctDNA based RB1 mutation and mTBI provided a feasible tool to predict the prognosis of SCLC.
Collapse
Affiliation(s)
- Yu Feng
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | | | - Guilan Dong
- Department of Medical Oncology, The People's Hospital of Tangshan city, Tangshan, China
| | - Hongxia Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Tongmei Zhang
- Department of General Medicine, Beijing Chest Hospital, Capital Medical University & Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | | | - Xuefeng Xia
- Medical Center, Geneplus-Beijing, Beijing, China
| | - Lifeng Li
- Medical Center, Geneplus-Beijing, Beijing, China
| | - Haohua Zhu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Puyuan Xing
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hongyu Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhijie Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xingsheng Hu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
230
|
Lo AA, Wallace A, Oreper D, Lounsbury N, Havnar C, Pechuan-Jorge X, Wu TD, Bourgon R, Jones R, Krogh K, Yang GY, Zill OA. Indication-specific tumor evolution and its impact on neoantigen targeting and biomarkers for individualized cancer immunotherapies. J Immunother Cancer 2021; 9:jitc-2021-003001. [PMID: 34599029 PMCID: PMC8488717 DOI: 10.1136/jitc-2021-003001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2021] [Indexed: 12/12/2022] Open
Abstract
Background Individualized neoantigen-specific immunotherapy (iNeST) requires robustly expressed clonal neoantigens for efficacy, but tumor mutational heterogeneity, loss of neoantigen expression, and variable tissue sampling present challenges. It is assumed that clonal neoantigens are preferred targets for immunotherapy, but the distributions of clonal neoantigens are not well characterized across cancer types. Methods We combined multiregion sequencing (MR-seq) analysis of five untreated, synchronously sampled metastatic solid tumors with re-analysis of published MR-seq data from 103 patients in order to characterize their globally clonal neoantigen content and factors that would impact neoantigen targeting. Results Branching evolution in colorectal cancer and renal cell carcinoma led to fewer clonal neoantigens and to clade-specific neoantigens (those shared across a subset of tumor regions but not fully clonal), with the latter not being readily distinguishable in single tumor samples. In colorectal, renal, and bladder cancer, most tumors had few globally clonal neoantigens. Prioritizing mutations with higher purity-adjusted and ploidy-adjusted variant allele frequency enriched for globally clonal neoantigens (those found in all tumor regions), whereas estimated cancer cell fraction derived from clustering-based tools, surprisingly, did not. Neoantigen quality was associated with loss of neoantigen expression in the bladder cancer case, and HLA-allele loss was observed in the renal and non-small cell lung cancer cases. Conclusions We show that tumor type, multilesion sampling, neoantigen expression, and HLA allele retention are important factors for iNeST targeting and patient selection, and may also be important factors to consider in the development of biomarker strategies.
Collapse
Affiliation(s)
- Amy A Lo
- Department of Research Pathology, Genentech Inc, South San Francisco, California, USA
| | - Andrew Wallace
- Department of Oncology Bioinformatics, Genentech Inc, South San Francisco, California, USA
| | - Daniel Oreper
- Department of Oncology Bioinformatics, Genentech Inc, South San Francisco, California, USA
| | - Nicolas Lounsbury
- Department of Oncology Bioinformatics, Genentech Inc, South San Francisco, California, USA
| | - Charles Havnar
- Department of Research Pathology, Genentech Inc, South San Francisco, California, USA
| | - Ximo Pechuan-Jorge
- Department of Cancer Immunology, Genentech Inc, South San Francisco, California, USA
| | - Thomas D Wu
- Department of Oncology Bioinformatics, Genentech Inc, South San Francisco, California, USA
| | - Richard Bourgon
- Department of Oncology Bioinformatics, Genentech Inc, South San Francisco, California, USA
| | - Ryan Jones
- Department of Pathology, Northwestern University, Chicago, Illinois, USA
| | - Katrina Krogh
- Department of Pathology, Northwestern University, Chicago, Illinois, USA
| | - Guang-Yu Yang
- Department of Pathology, Northwestern University, Chicago, Illinois, USA
| | - Oliver A Zill
- Department of Oncology Bioinformatics, Genentech Inc, South San Francisco, California, USA .,Current affiliation, init.bio, Inc, San Mateo, CA, USA
| |
Collapse
|
231
|
Bortolomeazzi M, Keddar MR, Montorsi L, Acha-Sagredo A, Benedetti L, Temelkovski D, Choi S, Petrov N, Todd K, Wai P, Kohl J, Denner T, Nye E, Goldstone R, Ward S, Wilson GA, Al Bakir M, Swanton C, John S, Miles J, Larijani B, Kunene V, Fontana E, Arkenau HT, Parker PJ, Rodriguez-Justo M, Shiu KK, Spencer J, Ciccarelli FD. Immunogenomics of Colorectal Cancer Response to Checkpoint Blockade: Analysis of the KEYNOTE 177 Trial and Validation Cohorts. Gastroenterology 2021; 161:1179-1193. [PMID: 34197832 PMCID: PMC8527923 DOI: 10.1053/j.gastro.2021.06.064] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/18/2021] [Accepted: 06/22/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND & AIMS Colorectal cancer (CRC) shows variable response to immune checkpoint blockade, which can only partially be explained by high tumor mutational burden (TMB). We conducted an integrated study of the cancer tissue and associated tumor microenvironment (TME) from patients treated with pembrolizumab (KEYNOTE 177 clinical trial) or nivolumab to dissect the cellular and molecular determinants of response to anti- programmed cell death 1 (PD1) immunotherapy. METHODS We selected multiple regions per tumor showing variable T-cell infiltration for a total of 738 regions from 29 patients, divided into discovery and validation cohorts. We performed multiregional whole-exome and RNA sequencing of the tumor cells and integrated these with T-cell receptor sequencing, high-dimensional imaging mass cytometry, detection of programmed death-ligand 1 (PDL1) interaction in situ, multiplexed immunofluorescence, and computational spatial analysis of the TME. RESULTS In hypermutated CRCs, response to anti-PD1 immunotherapy was not associated with TMB but with high clonality of immunogenic mutations, clonally expanded T cells, low activation of Wnt signaling, deregulation of the interferon gamma pathway, and active immune escape mechanisms. Responsive hypermutated CRCs were also rich in cytotoxic and proliferating PD1+CD8 T cells interacting with PDL1+ antigen-presenting macrophages. CONCLUSIONS Our study clarified the limits of TMB as a predictor of response of CRC to anti-PD1 immunotherapy. It identified a population of antigen-presenting macrophages interacting with CD8 T cells that consistently segregate with response. We therefore concluded that anti-PD1 agents release the PD1-PDL1 interaction between CD8 T cells and macrophages to promote cytotoxic antitumor activity.
Collapse
Affiliation(s)
- Michele Bortolomeazzi
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, United Kingdom; School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Mohamed Reda Keddar
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, United Kingdom; School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Lucia Montorsi
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, United Kingdom; School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Amelia Acha-Sagredo
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, United Kingdom; School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Lorena Benedetti
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, United Kingdom; School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Damjan Temelkovski
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, United Kingdom; School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Subin Choi
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, United Kingdom; School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| | - Nedyalko Petrov
- Biomedical Research Centre, Guy's and St. Thomas' National Health Service Trust, London, United Kingdom
| | - Katrina Todd
- Biomedical Research Centre, Guy's and St. Thomas' National Health Service Trust, London, United Kingdom
| | - Patty Wai
- State-Dependent Neural Processing Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Johannes Kohl
- State-Dependent Neural Processing Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Tamara Denner
- Experimental Histopathology, The Francis Crick Institute, London, United Kingdom
| | - Emma Nye
- Experimental Histopathology, The Francis Crick Institute, London, United Kingdom
| | - Robert Goldstone
- Advanced Sequencing Facility, The Francis Crick Institute, London, United Kingdom
| | - Sophia Ward
- Advanced Sequencing Facility, The Francis Crick Institute, London, United Kingdom
| | - Gareth A Wilson
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Maise Al Bakir
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Susan John
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom
| | | | - Banafshe Larijani
- FASTBASE Solutions S.L, Derio, Spain; Cell Biophysics Laboratory, Ikerbasque, Basque Foundation for Science, Research Centre for Experimental Marine Biology and Biotechnology & Biophysics Institute, University of the Basque Country, Leioa, Bizkaia, Spain; Centre for Therapeutic Innovation, Cell Biophysics Laboratory, Department of Pharmacy and Pharmacology & Department of Physics, University of Bath, Bath, United Kingdom
| | - Victoria Kunene
- Medical Oncology, University Hospitals Birmingham National Health Service Foundation Trust, Birmingham, United Kingdom
| | - Elisa Fontana
- Drug Development Unit, Sarah Cannon Research Institute UK, London, United Kingdom
| | - Hendrik-Tobias Arkenau
- Drug Development Unit, Sarah Cannon Research Institute UK, London, United Kingdom; Department of Oncology, University College Hospital, London, United Kingdom
| | - Peter J Parker
- School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom; Protein Phosphorylation Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Manuel Rodriguez-Justo
- Department of Histopathology, University College London Cancer Institute, London, United Kingdom
| | - Kai-Keen Shiu
- Department of Gastrointestinal Oncology, University College London Hospital National Health Service Foundation Trust, London, United Kingdom
| | - Jo Spencer
- School of Immunology and Microbial Sciences, King's College London, London, United Kingdom.
| | - Francesca D Ciccarelli
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, United Kingdom; School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom.
| |
Collapse
|
232
|
In-depth cell-free DNA sequencing reveals genomic landscape of Hodgkin’s lymphoma and facilitates ultrasensitive residual disease detection. MED 2021; 2:1171-1193.e11. [DOI: 10.1016/j.medj.2021.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/12/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022]
|
233
|
Duchmann M, Laplane L, Itzykson R. Clonal Architecture and Evolutionary Dynamics in Acute Myeloid Leukemias. Cancers (Basel) 2021; 13:4887. [PMID: 34638371 PMCID: PMC8507870 DOI: 10.3390/cancers13194887] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/19/2022] Open
Abstract
Acute myeloid leukemias (AML) results from the accumulation of genetic and epigenetic alterations, often in the context of an aging hematopoietic environment. The development of high-throughput sequencing-and more recently, of single-cell technologies-has shed light on the intratumoral diversity of leukemic cells. Taking AML as a model disease, we review the multiple sources of genetic, epigenetic, and functional heterogeneity of leukemic cells and discuss the definition of a leukemic clone extending its definition beyond genetics. After introducing the two dimensions contributing to clonal diversity, namely, richness (number of leukemic clones) and evenness (distribution of clone sizes), we discuss the mechanisms at the origin of clonal emergence (mutation rate, number of generations, and effective size of the leukemic population) and the causes of clonal dynamics. We discuss the possible role of neutral drift, but also of cell-intrinsic and -extrinsic influences on clonal fitness. After reviewing available data on the prognostic role of genetic and epigenetic diversity of leukemic cells on patients' outcome, we discuss how a better understanding of AML as an evolutionary process could lead to the design of novel therapeutic strategies in this disease.
Collapse
Affiliation(s)
- Matthieu Duchmann
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université de Paris, 75010 Paris, France;
- Laboratoire d’Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, 75010 Paris, France
| | - Lucie Laplane
- Institut d’Histoire et Philosophie des Sciences et des Techniques UMR 8590, CNRS, Université Paris 1 Panthéon-Sorbonne, 75010 Paris, France;
- Gustave Roussy Cancer Center, UMR1287, 94805 Villejuif, France
| | - Raphael Itzykson
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université de Paris, 75010 Paris, France;
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, 75010 Paris, France
| |
Collapse
|
234
|
Zhang M, Wang D, Su L, Ma J, Wang S, Cui M, Hong S, Guan B, Ma X. Activity of Wnt/PCP Regulation Pathway Classifies Patients of Low-Grade Glioma Into Molecularly Distinct Subgroups With Prognostic Difference. Front Oncol 2021; 11:726034. [PMID: 34540693 PMCID: PMC8440981 DOI: 10.3389/fonc.2021.726034] [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: 06/16/2021] [Accepted: 08/09/2021] [Indexed: 11/13/2022] Open
Abstract
Wingless/Int-1 (Wnt) signaling is one of the most well-known oncogenic pathways. Numerous studies have uncovered an aberrant expression of Wnt in cancer and its association with multiple oncogenic processes, such as cell proliferation, epithelial–mesenchymal transition (EMT), and invasiveness. Most previous studies mainly focused on the canonical branch of Wnt signaling pathway, i.e., Wnt/β-catenin signaling. The Wnt/planar cell polarity (PCP) signaling pathway, as the most recently described branch of Wnt signaling, was much less investigated in oncology research. In this study, we thoroughly characterized the activity of the Wnt/PCP regulation pathway in low-grade glioma (LGG) patients. Subtyping based on the expression pattern of the Wnt/PCP regulation pathway revealed three (C1–C3) subgroups with significant survival differences. Each group displayed distinct genomic characteristics. For instance, C1 was enriched with capicua transcriptional repressor (CIC) truncating mutations and 1p19q codel. C2 was characterized with tumor protein p53 (TP53) and ATRX chromatin remodeler (ATRX) inactivating mutations but depletion of telomerase reverse transcriptase (TERT) promoter mutations. C3 showed elevated malignancy reflected from several oncogenic characteristics, such as tumor heterogeneity and cell stemness, and demonstrated the worst survival outcome. In addition, C3 showed elevated macrophage segregation via induction of cytokines that are able to enhance the permeability of the brain–blood barrier (BBB). Lastly, we developed a prognostic model based on the risk score system. Validation indicated that our model can independently predict the prognosis of LGG patients.
Collapse
Affiliation(s)
- Meng Zhang
- Medical School of Chinese People's Liberation Army, Beijing, China.,Department of Neurosurgery, The First Medical Centre, Chinese People's Liberation Army General Hospital, Beijing, China.,Department of Neurosurgery, The Second Hospital of Southern District of Chinese People's Liberation Army Navy, Sanya, China
| | - Dan Wang
- Genetron Health (Beijing) Co. Ltd., Beijing, China
| | - Lan Su
- Genetron Health (Beijing) Co. Ltd., Beijing, China
| | - Jingjiao Ma
- Genetron Health (Beijing) Co. Ltd., Beijing, China
| | - Sizhen Wang
- Genetron Health (Beijing) Co. Ltd., Beijing, China
| | - Meng Cui
- Department of Neurosurgery, The First Medical Centre, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Shunming Hong
- Department of Neurosurgery, The Third Medical Centre, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Bing Guan
- Health Economics Department, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaodong Ma
- Department of Neurosurgery, The First Medical Centre, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
235
|
Andersson N, Chattopadhyay S, Valind A, Karlsson J, Gisselsson D. DEVOLUTION-A method for phylogenetic reconstruction of aneuploid cancers based on multiregional genotyping data. Commun Biol 2021; 4:1103. [PMID: 34545199 PMCID: PMC8452746 DOI: 10.1038/s42003-021-02637-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 09/03/2021] [Indexed: 02/05/2023] Open
Abstract
Phylogenetic reconstruction of cancer cell populations remains challenging. There is a particular lack of tools that deconvolve clones based on copy number aberration analyses of multiple tumor biopsies separated in time and space from the same patient. This has hampered investigations of tumors rich in aneuploidy but few point mutations, as in many childhood cancers and high-risk adult cancer. Here, we present DEVOLUTION, an algorithm for subclonal deconvolution followed by phylogenetic reconstruction from bulk genotyping data. It integrates copy number and sequencing information across multiple tumor regions throughout the inference process, provided that the mutated clone fraction for each mutation is known. We validate DEVOLUTION on data from 56 pediatric tumors comprising 253 tumor biopsies and show a robust performance on simulations of bulk genotyping data. We also benchmark DEVOLUTION to similar bioinformatic tools using an external dataset. DEVOLUTION holds the potential to facilitate insights into the development, progression, and response to treatment, particularly in tumors with high burden of chromosomal copy number alterations.
Collapse
Affiliation(s)
- Natalie Andersson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| | - Subhayan Chattopadhyay
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Anders Valind
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Department of Pediatrics, Skåne University Hospital, Lund, Sweden
| | - Jenny Karlsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - David Gisselsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Division of Oncology-Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
- Clinical Genetics and Pathology, Laboratory Medicine, Lund University Hospital, Skåne Healthcare Region, Lund, Sweden
| |
Collapse
|
236
|
Abécassis J, Reyal F, Vert JP. CloneSig can jointly infer intra-tumor heterogeneity and mutational signature activity in bulk tumor sequencing data. Nat Commun 2021; 12:5352. [PMID: 34504064 PMCID: PMC8429716 DOI: 10.1038/s41467-021-24992-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 07/12/2021] [Indexed: 02/07/2023] Open
Abstract
Systematic DNA sequencing of cancer samples has highlighted the importance of two aspects of cancer genomics: intra-tumor heterogeneity (ITH) and mutational processes. These two aspects may not always be independent, as different mutational processes could be involved in different stages or regions of the tumor, but existing computational approaches to study them largely ignore this potential dependency. Here, we present CloneSig, a computational method to jointly infer ITH and mutational processes in a tumor from bulk-sequencing data. Extensive simulations show that CloneSig outperforms current methods for ITH inference and detection of mutational processes when the distribution of mutational signatures changes between clones. Applied to a large cohort of 8,951 tumors with whole-exome sequencing data from The Cancer Genome Atlas, and on a pan-cancer dataset of 2,632 whole-genome sequencing tumor samples from the Pan-Cancer Analysis of Whole Genomes initiative, CloneSig obtains results overall coherent with previous studies.
Collapse
Affiliation(s)
- Judith Abécassis
- Institut Curie, PSL Research University, Translational Research Department, INSERM, U932 Immunity and Cancer, Residual Tumor & Response to Treatment Laboratory (RT2Lab), Paris, France
- MINES ParisTech, PSL University, CBIO - Centre for Computational Biology, Paris, France
- Institut Curie, PSL Research University, Paris, France
| | - Fabien Reyal
- Institut Curie, PSL Research University, Translational Research Department, INSERM, U932 Immunity and Cancer, Residual Tumor & Response to Treatment Laboratory (RT2Lab), Paris, France
- Department of Surgery, Institut Curie, Paris, France
| | - Jean-Philippe Vert
- MINES ParisTech, PSL University, CBIO - Centre for Computational Biology, Paris, France.
- Google Research, Brain team, Paris, France.
| |
Collapse
|
237
|
Min Q, Wang Y, Wu Q, Li X, Teng H, Fan J, Cao Y, Fan P, Zhan Q. Genomic and epigenomic evolution of acquired resistance to combination therapy in esophageal squamous cell carcinoma. JCI Insight 2021; 6:150203. [PMID: 34494553 PMCID: PMC8492345 DOI: 10.1172/jci.insight.150203] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/21/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUNDTargeted arterial infusion of verapamil combined with chemotherapy (TVCC) is an effective clinical interventional therapy for esophageal squamous cell carcinoma (ESCC), but multidrug resistance (MDR) remains the major cause of relapse or poor prognosis, and the underlying molecular mechanisms of MDR, temporal intratumoral heterogeneity, and clonal evolutionary processes of resistance have not been determined.METHODSTo elucidate the roles of genetic and epigenetic alterations in the evolution of acquired resistance during therapies, we performed whole-exome sequencing on 16 serial specimens from 7 patients with ESCC at every cycle of therapeutic intervention from 3 groups, complete response, partial response, and progressive disease, and we performed whole-genome bisulfite sequencing for 3 of these 7 patients, 1 patient from each group.RESULTSPatients with progressive disease exhibited a substantially higher genomic and epigenomic temporal heterogeneity. Subclonal expansions driven by the beneficial new mutations were observed during combined therapies, which explained the emergence of MDR. Notably, SLC7A8 was identified as a potentially novel MDR gene, and functional assays demonstrated that mutant SLC7A8 promoted the resistance phenotypes of ESCC cell lines. Promoter methylation dynamics during treatments revealed 8 drug resistance protein-coding genes characterized by hypomethylation in promoter regions. Intriguingly, promoter hypomethylation of SLC8A3 and mutant SLC7A8 were enriched in an identical pathway, protein digestion and absorption, indicating a potentially novel MDR mechanism during treatments.CONCLUSIONOur integrated multiomics investigations revealed the dynamics of temporal genetic and epigenetic inter- and intratumoral heterogeneity, clonal evolutionary processes, and epigenomic changes, providing potential MDR therapeutic targets in treatment-resistant patients with ESCC during combined therapies.FUNDINGNational Natural Science Foundation of China, Science Foundation of Peking University Cancer Hospital, CAMS Innovation Fund for Medical Sciences, Major Program of Shenzhen Bay Laboratory, Guangdong Basic and Applied Basic Research Foundation, and the third round of public welfare development and reform pilot projects of Beijing Municipal Medical Research Institutes.
Collapse
Affiliation(s)
- Qingjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qingnan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xianfeng Li
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Huajing Teng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiawen Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yiren Cao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Pingsheng Fan
- Department of Medical Oncology, Anhui Provincial Cancer Hospital, Hefei, China
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
238
|
Chen ZH, Yan SM, Chen XX, Zhang Q, Liu SX, Liu Y, Luo YL, Zhang C, Xu M, Zhao YF, Huang LY, Liu BL, Xia TL, Xu DZ, Liang Y, Chen YM, Wang W, Yuan SQ, Zhang HZ, Yun JP, Zhai WW, Zeng MS, Bai F, Zhong Q. The genomic architecture of EBV and infected gastric tissue from precursor lesions to carcinoma. Genome Med 2021; 13:146. [PMID: 34493320 PMCID: PMC8422682 DOI: 10.1186/s13073-021-00963-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 08/29/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Epstein-Barr virus (EBV)-associated gastric carcinomas (EBVaGCs) present unique molecular signatures, but the tumorigenesis of EBVaGCs and the role EBV plays during this process remain poorly understood. METHODS We applied whole-exome sequencing, EBV genome sequencing, and whole-genome bisulfite sequencing to multiple samples (n = 123) derived from the same patients (n = 25), which covered saliva samples and different histological stages from morphologically normal epithelial tissues to dysplasia and EBVaGCs. We compared the genomic landscape between EBVaGCs and their precursor lesions and traced the clonal evolution for each patient. We also analyzed genome sequences of EBV from samples of different histological types. Finally, the key molecular events promoting the tumor evolution were demonstrated by MTT, IC50, and colony formation assay in vitro experiments and in vivo xenograft experiments. RESULTS Our analysis revealed increasing mutational burden and EBV load from normal tissues and low-grade dysplasia (LD) to high-grade dysplasia (HD) and EBVaGCs, and oncogenic amplifications occurred late in EBVaGCs. Interestingly, within each patient, EBVaGCs and HDs were monoclonal and harbored single-strain-originated EBV, but saliva or normal tissues/LDs had different EBV strains from that in EBVaGCs. Compared with precursor lesions, tumor cells showed incremental methylation in promotor regions, whereas EBV presented consistent hypermethylation. Dominant alterations targeting the PI3K-Akt and Wnt pathways were found in EBV-infected cells. The combinational inhibition of these two pathways in EBV-positive tumor cells confirmed their synergistic function. CONCLUSIONS We portrayed the (epi) genomic evolution process of EBVaGCs, revealed the extensive genomic diversity of EBV between tumors and normal tissue sites, and demonstrated the synergistic activation of the PI3K and Wnt pathways in EBVaGCs, offering a new potential treatment strategy for this disease.
Collapse
Affiliation(s)
- Zhang-Hua Chen
- Biomedical Pioneering Innovation Center (BIOPIC), Integrated Research Building Room 330, School of Life Sciences, Peking University, Yiheyuan Road No.5, Haidian District, Beijing, 100871, China
| | - Shu-Mei Yan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Xi-Xi Chen
- Biomedical Pioneering Innovation Center (BIOPIC), Integrated Research Building Room 330, School of Life Sciences, Peking University, Yiheyuan Road No.5, Haidian District, Beijing, 100871, China
| | - Qi Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Ultrasound, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
- Department of Oncology, Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shang-Xin Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yang Liu
- Biomedical Pioneering Innovation Center (BIOPIC), Integrated Research Building Room 330, School of Life Sciences, Peking University, Yiheyuan Road No.5, Haidian District, Beijing, 100871, China
| | - Yi-Ling Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Chao Zhang
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, USA
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, USA
| | - Miao Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yi-Fan Zhao
- Biomedical Pioneering Innovation Center (BIOPIC), Integrated Research Building Room 330, School of Life Sciences, Peking University, Yiheyuan Road No.5, Haidian District, Beijing, 100871, China
| | - Li-Yun Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Bin-Liu Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Tian-Liang Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Da-Zhi Xu
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yao Liang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yong-Ming Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei Wang
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shu-Qiang Yuan
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hui-Zhong Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Jing-Ping Yun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Centre, Guangzhou, China
| | - Wei-Wei Zhai
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Fan Bai
- Biomedical Pioneering Innovation Center (BIOPIC), Integrated Research Building Room 330, School of Life Sciences, Peking University, Yiheyuan Road No.5, Haidian District, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China.
| | - Qian Zhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
239
|
Shin G, Greer SU, Hopmans E, Grimes SM, Lee H, Zhao L, Miotke L, Suarez C, Almeda AF, Haraldsdottir S, Ji HP. Profiling diverse sequence tandem repeats in colorectal cancer reveals co-occurrence of microsatellite and chromosomal instability involving Chromosome 8. Genome Med 2021; 13:145. [PMID: 34488871 PMCID: PMC8420050 DOI: 10.1186/s13073-021-00958-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/23/2021] [Indexed: 11/10/2022] Open
Abstract
We developed a sensitive sequencing approach that simultaneously profiles microsatellite instability, chromosomal instability, and subclonal structure in cancer. We assessed diverse repeat motifs across 225 microsatellites on colorectal carcinomas. Our study identified elevated alterations at both selected tetranucleotide and conventional mononucleotide repeats. Many colorectal carcinomas had a mix of genomic instability states that are normally considered exclusive. An MSH3 mutation may have contributed to the mixed states. Increased copy number of chromosome arm 8q was most prevalent among tumors with microsatellite instability, including a case of translocation involving 8q. Subclonal analysis identified co-occurring driver mutations previously known to be exclusive.
Collapse
Affiliation(s)
- GiWon Shin
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA, 94305-5151, USA
| | - Stephanie U Greer
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA, 94305-5151, USA
| | - Erik Hopmans
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA, 94305-5151, USA
| | - Susan M Grimes
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA, 94305-5151, USA
| | - HoJoon Lee
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA, 94305-5151, USA
| | - Lan Zhao
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA, 94305-5151, USA
| | - Laura Miotke
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA, 94305-5151, USA
| | - Carlos Suarez
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Alison F Almeda
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA, 94305-5151, USA
| | - Sigurdis Haraldsdottir
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA, 94305-5151, USA
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA, 94305-5151, USA. .,Stanford Genome Technology Center, Stanford University, Palo Alto, CA, 94304, USA.
| |
Collapse
|
240
|
Fridland S, Choi J, Nam M, Schellenberg SJ, Kim E, Lee G, Yoon N, Chae YK. Assessing tumor heterogeneity: integrating tissue and circulating tumor DNA (ctDNA) analysis in the era of immuno-oncology - blood TMB is not the same as tissue TMB. J Immunother Cancer 2021; 9:jitc-2021-002551. [PMID: 34462324 PMCID: PMC8407207 DOI: 10.1136/jitc-2021-002551] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2021] [Indexed: 12/29/2022] Open
Abstract
Tissue tumor mutational burden (tTMB) is calculated to aid in cancer treatment selection. High tTMB predicts a favorable response to immunotherapy in patients with non-small cell lung cancer. Blood TMB (bTMB) from circulating tumor DNA is reported to have similar predictive power and has been proposed as an alternative to tTMB. Across many studies not only are tTMB and bTMB not concordant but also as reported previously by our group predict conflicting outcomes. This implies that bTMB is not a substitute for tTMB, but rather a composite index that may encompass tumor heterogeneity. Here, we provide a thorough overview of the predictive power of TMB, discuss the use of tumor heterogeneity alongside TMB to predict treatment response and review several methods of tumor heterogeneity assessment. Furthermore, we propose a hypothetical method of estimating tumor heterogeneity and touch on its clinical implications.
Collapse
Affiliation(s)
- Stanislav Fridland
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jaeyoun Choi
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Myungwoo Nam
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | - Eugene Kim
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Grace Lee
- Northwestern University, Evanston, Illinois, USA
| | | | - Young Kwang Chae
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA .,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
241
|
Xiang X, Liu Z, Zhang C, Li Z, Gao J, Zhang C, Cao Q, Cheng J, Liu H, Chen D, Cheng Q, Zhang N, Xue R, Bai F, Zhu J. IDH Mutation Subgroup Status Associates with Intratumor Heterogeneity and the Tumor Microenvironment in Intrahepatic Cholangiocarcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101230. [PMID: 34250753 PMCID: PMC8425914 DOI: 10.1002/advs.202101230] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/01/2021] [Indexed: 05/03/2023]
Abstract
Intrahepatic cholangiocarcinoma (ICC) is highly heterogeneous. Here, the authors perform exome sequencing and bulk RNA sequencing on 73 tumor regions from 14 ICC patients to portray the multi-faceted intratumor heterogeneity (ITH) landscape of ICC. The authors show that ITH is highly concordant across genomic, transcriptomic, and immune levels. Comparison of these data to 8 published datasets reveals significantly higher degrees of ITH in ICC than hepatocellular carcinoma. Remarkably, the authors find that high-ITH tumors highly overlap with the IDH (isocitrate dehydrogenase)-mutant subgroup (IDH-SG), comprising of IDH-mutated tumors and IDH-like tumors, that is, those IDH-wildtype tumors that exhibit similar molecular profiles to the IDH-mutated ones. Furthermore, IDH-SG exhibits less T cell infiltration and lower T cell cytotoxicity, indicating a colder tumor microenvironment (TME). The higher ITH and colder TME of IDH-SG are successfully validated by single-cell RNA sequencing on 17 503 cells from 4 patients. Collectively, the study shows that IDH mutant subgroup status, rather than IDH mutation alone, is associated with ITH and the TME of ICC tumors. The results highlight that IDH-like patients may also benefit from IDH targeted therapies and provide important implications for the diagnosis and treatment of ICC.
Collapse
Affiliation(s)
- Xiao Xiang
- Department of Hepatobiliary Surgery, Peking University People's HospitalBeijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Ziyang Liu
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life SciencesPeking UniversityBeijing100871China
- Beijing Advanced Innovation Center for Genomics (ICG)Peking UniversityBeijing100871China
| | - Chong Zhang
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life SciencesPeking UniversityBeijing100871China
- Beijing Advanced Innovation Center for Genomics (ICG)Peking UniversityBeijing100871China
| | - Zhao Li
- Department of Hepatobiliary Surgery, Peking University People's HospitalBeijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Jie Gao
- Department of Hepatobiliary Surgery, Peking University People's HospitalBeijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Changkun Zhang
- Department of Hepatobiliary Surgery, Peking University People's HospitalBeijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Qi Cao
- Translational Cancer Research CenterPeking University First HospitalBeijing100034China
| | - Jinghui Cheng
- Translational Cancer Research CenterPeking University First HospitalBeijing100034China
| | - Hengkang Liu
- Translational Cancer Research CenterPeking University First HospitalBeijing100034China
| | - Dingbao Chen
- Department of Hepatobiliary Surgery, Peking University People's HospitalBeijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Qian Cheng
- Department of Hepatobiliary Surgery, Peking University People's HospitalBeijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| | - Ning Zhang
- Translational Cancer Research CenterPeking University First HospitalBeijing100034China
| | - Ruidong Xue
- Translational Cancer Research CenterPeking University First HospitalBeijing100034China
| | - Fan Bai
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life SciencesPeking UniversityBeijing100871China
- Beijing Advanced Innovation Center for Genomics (ICG)Peking UniversityBeijing100871China
| | - Jiye Zhu
- Department of Hepatobiliary Surgery, Peking University People's HospitalBeijing Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver CancerBeijing100044China
| |
Collapse
|
242
|
Giunta S. Decoding human cancer with whole genome sequencing: a review of PCAWG Project studies published in February 2020. Cancer Metastasis Rev 2021; 40:909-924. [PMID: 34097189 PMCID: PMC8180541 DOI: 10.1007/s10555-021-09969-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/21/2021] [Indexed: 12/15/2022]
Abstract
Cancer is underlined by genetic changes. In an unprecedented international effort, the Pan-Cancer Analysis of Whole Genomes (PCAWG) of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA) sequenced the tumors of over two thousand five hundred patients across 38 different cancer types, as well as the corresponding healthy tissue, with the aim of identifying genome-wide mutations exclusively found in cancer and uncovering new genetic changes that drive tumor formation. What set this project apart from earlier efforts is the use of whole genome sequencing (WGS) that enabled to explore alterations beyond the coding DNA, into cancer's non-coding genome. WGS of the entire cohort allowed to tease apart driving mutations that initiate and support carcinogenesis from passenger mutations that do not play an overt role in the disease. At least one causative mutation was found in 95% of all cancers, with many tumors showing an average of 5 driver mutations. The PCAWG Project also assessed the transcriptional output altered in cancer and rebuilt the evolutionary history of each tumor showing that initial driver mutations can occur years if not decades prior to a diagnosis. Here, I provide a concise review of the Pan-Cancer Project papers published on February 2020, along with key computational tools and the digital framework generated as part of the project. This represents an historic effort by hundreds of international collaborators, which provides a comprehensive understanding of cancer genetics, with publicly available data and resources representing a treasure trove of information to advance cancer research for years to come.
Collapse
Affiliation(s)
- Simona Giunta
- Laboratory of Genome Evolution, Department of Biology & Biotechnology "Charles Darwin", University of Rome Sapienza, Rome, Italy.
- The Rockefeller University, 1230 York Avenue, New York, NY, USA.
| |
Collapse
|
243
|
Baghaarabani L, Goliaei S, Foroughmand-Araabi MH, Shariatpanahi SP, Goliaei B. Conifer: clonal tree inference for tumor heterogeneity with single-cell and bulk sequencing data. BMC Bioinformatics 2021; 22:416. [PMID: 34461827 PMCID: PMC8404257 DOI: 10.1186/s12859-021-04338-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 08/16/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Genetic heterogeneity of a cancer tumor that develops during clonal evolution is one of the reasons for cancer treatment failure, by increasing the chance of drug resistance. Clones are cell populations with different genotypes, resulting from differences in somatic mutations that occur and accumulate during cancer development. An appropriate approach for identifying clones is determining the variant allele frequency of mutations that occurred in the tumor. Although bulk sequencing data can be used to provide that information, the frequencies are not informative enough for identifying different clones with the same prevalence and their evolutionary relationships. On the other hand, single-cell sequencing data provides valuable information about branching events in the evolution of a cancerous tumor. However, the temporal order of mutations may be determined with ambiguities using only single-cell data, while variant allele frequencies from bulk sequencing data can provide beneficial information for inferring the temporal order of mutations with fewer ambiguities. RESULT In this study, a new method called Conifer (ClONal tree Inference For hEterogeneity of tumoR) is proposed which combines aggregated variant allele frequency from bulk sequencing data with branching event information from single-cell sequencing data to more accurately identify clones and their evolutionary relationships. It is proven that the accuracy of clone identification and clonal tree inference is increased by using Conifer compared to other existing methods on various sets of simulated data. In addition, it is discussed that the evolutionary tree provided by Conifer on real cancer data sets is highly consistent with information in both bulk and single-cell data. CONCLUSIONS In this study, we have provided an accurate and robust method to identify clones of tumor heterogeneity and their evolutionary history by combining single-cell and bulk sequencing data.
Collapse
Affiliation(s)
- Leila Baghaarabani
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Sama Goliaei
- Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | | | | | - Bahram Goliaei
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
244
|
Whole exome and transcriptome sequencing reveal clonal evolution and exhibit immune-related features in metastatic colorectal tumors. Cell Death Discov 2021; 7:222. [PMID: 34453042 PMCID: PMC8397721 DOI: 10.1038/s41420-021-00607-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/14/2021] [Accepted: 07/29/2021] [Indexed: 01/05/2023] Open
Abstract
Liver is the most common site where metastatic lesions of colorectal cancer (CRC) arise. Although researches have shown mutations in driver genes, copy number variations (CNV) and alterations in relevant signaling pathways promoted the tumor evolution and immune escape during colorectal liver metastasis (CLM), the underlying mechanism remains largely elusive. Tumor and matched metastatic tissues were collected from 16 patients diagnosed with colorectal cancer and subjected to whole-exome sequencing (WES) and RNA sequencing (RNA-seq) for studying colorectal cancer clonal evolution and immune escape during CLM. Shared somatic mutations between primary and metastatic tissues with a commonly observed subclonal-clonal (S-C) changing pattern indicated a common clonal origin between two lesions. The recurrent mutations with S-C changing pattern included those in KRAS, SYNE1, CACNA1H, PCLO, FBXL2, and DNAH11. The main CNV events underwent clonal-clonal evolution (20q amplification (amp), 17p deletion (del), 18q del and 8p del), subclonal-clonal evolution (8q amp, 13q amp, 8p del) and metastasis-specific evolution (8q amp) during the process of CLM. In addition, we revealed a potential mechanism of tumor cell immune escape by analyzing human leukocytes antigens (HLA) related clonal neoantigens and immune cell components in CLM. Our study proposed a novel liver metastasis-related evolutionary process in colorectal cancer and emphasized the theory of neo-immune escape in colorectal liver metastasis.
Collapse
|
245
|
Hu H, Zhang Q, Huang R, Gao Z, Yuan Z, Tang Q, Gao F, Wang M, Zhang W, Ma T, Qiao T, Jin Y, Wang G. Genomic Analysis Reveals Heterogeneity Between Lesions in Synchronous Primary Right-Sided and Left-Sided Colon Cancer. Front Mol Biosci 2021; 8:689466. [PMID: 34422903 PMCID: PMC8371635 DOI: 10.3389/fmolb.2021.689466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/07/2021] [Indexed: 01/05/2023] Open
Abstract
Background: The synchronous primary right-sided and left-sided colon cancer (sRL-CC) is a peculiar subtype of colorectal cancer. However, the genomic landscape of sRL-CC remains elusive. Methods: Twenty-eight paired tumor samples and their corresponding normal mucosa samples from 14 patients were collected from the Second Affiliated Hospital of Harbin Medical University from 2011 to 2018. The clinical-pathological data were obtained, and whole-exome sequencing was performed based on formalin-fixed and paraffin-embedded samples of these patients, and then, comprehensive bioinformatic analyses were conducted. Results: Both the lesions of sRL-CC presented dissimilar histological grade and differentiation. Based on sequencing data, few overlapping SNV signatures, onco-driver gene mutations, and SMGs were identified. Moreover, the paired lesions harbored a different distribution of copy number variants (CNVs) and loss of heterozygosity. The clonal architecture analysis demonstrated the polyclonal origin of sRL-CC and inter-cancerous heterogeneity between two lesions. Conclusion: Our work provides evidence that lesions of sRL-CC share few overlapping mutational signatures and CNVs, and may originate from different clones.
Collapse
Affiliation(s)
- Hanqing Hu
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qian Zhang
- Colorectal Cancer Surgery Department, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Rui Huang
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhifeng Gao
- Department of Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ziming Yuan
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qingchao Tang
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Feng Gao
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meng Wang
- Colorectal Cancer Surgery Department, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Weiyuan Zhang
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianyi Ma
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianyu Qiao
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yinghu Jin
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guiyu Wang
- Colorectal Cancer Surgery Department, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| |
Collapse
|
246
|
Stiehl T, Marciniak-Czochra A. Computational Reconstruction of Clonal Hierarchies From Bulk Sequencing Data of Acute Myeloid Leukemia Samples. Front Physiol 2021; 12:596194. [PMID: 34497529 PMCID: PMC8419336 DOI: 10.3389/fphys.2021.596194] [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: 08/18/2020] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
Acute myeloid leukemia is an aggressive cancer of the blood forming system. The malignant cell population is composed of multiple clones that evolve over time. Clonal data reflect the mechanisms governing treatment response and relapse. Single cell sequencing provides most direct insights into the clonal composition of the leukemic cells, however it is still not routinely available in clinical practice. In this work we develop a computational algorithm that allows identifying all clonal hierarchies that are compatible with bulk variant allele frequencies measured in a patient sample. The clonal hierarchies represent descendance relations between the different clones and reveal the order in which mutations have been acquired. The proposed computational approach is tested using single cell sequencing data that allow comparing the outcome of the algorithm with the true structure of the clonal hierarchy. We investigate which problems occur during reconstruction of clonal hierarchies from bulk sequencing data. Our results suggest that in many cases only a small number of possible hierarchies fits the bulk data. This implies that bulk sequencing data can be used to obtain insights in clonal evolution.
Collapse
Affiliation(s)
- Thomas Stiehl
- Institute for Computational Biomedicine – Disease Modeling, RWTH Aachen University, Aachen, Germany
- Institute of Applied Mathematics, Interdisciplinary Center for Scientific Computing and Bioquant Center, Heidelberg University, Heidelberg, Germany
| | - Anna Marciniak-Czochra
- Institute of Applied Mathematics, Interdisciplinary Center for Scientific Computing and Bioquant Center, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
247
|
Na D, Chae J, Cho SY, Kang W, Lee A, Min S, Kang J, Kim MJ, Choi J, Lee W, Shin D, Min A, Kim YJ, Lee KH, Kim TY, Suh YS, Kong SH, Lee HJ, Kim WH, Park H, Im SA, Yang HK, Lee C, Kim JI. Predictive biomarkers for 5-fluorouracil and oxaliplatin-based chemotherapy in gastric cancers via profiling of patient-derived xenografts. Nat Commun 2021; 12:4840. [PMID: 34376661 PMCID: PMC8355375 DOI: 10.1038/s41467-021-25122-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 07/16/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is commonly treated by chemotherapy using 5-fluorouracil (5-FU) derivatives and platinum combination, but predictive biomarker remains lacking. We develop patient-derived xenografts (PDXs) from 31 GC patients and treat with a combination of 5-FU and oxaliplatin, to determine biomarkers associated with responsiveness. When the PDXs are defined as either responders or non-responders according to tumor volume change after treatment, the responsiveness of PDXs is significantly consistent with the respective clinical outcomes of the patients. An integrative genomic and transcriptomic analysis of PDXs reveals that pathways associated with cell-to-cell and cell-to-extracellular matrix interactions enriched among the non-responders in both cancer cells and the tumor microenvironment (TME). We develop a 30-gene prediction model to determine the responsiveness to 5-FU and oxaliplatin-based chemotherapy and confirm the significant poor survival outcomes among cases classified as non-responder-like in three independent GC cohorts. Our study may inform clinical decision-making when designing treatment strategies.
Collapse
Affiliation(s)
- Deukchae Na
- Ewha Institute of Convergence Medicine, Ewha Womans University Mokdong Hospital, Seoul, Korea
| | - Jeesoo Chae
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Evolution Research Center, The Catholic University of Korea, Seoul, Korea
| | - Sung-Yup Cho
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea
| | - Wonyoung Kang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Ahra Lee
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Seoyeon Min
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Jinjoo Kang
- Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Min Jung Kim
- Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea
| | - Jaeyong Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Woochan Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Dongjin Shin
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Ahrum Min
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yu-Jin Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea
| | - Kyung-Hun Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Tae-Yong Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
- Department of Surgery, Seoul National University Bundang Hospital, Seoul, Korea
| | - Seong-Ho Kong
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Hyuk-Joon Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Woo-Ho Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Hansoo Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Korea
| | - Seock-Ah Im
- Cancer Research Institute, Seoul National University, Seoul, Korea.
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
| | - Han-Kwang Yang
- Cancer Research Institute, Seoul National University, Seoul, Korea.
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea.
| | - Charles Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
- Department of Life Science, Ewha Womans University, Seoul, Korea.
- Precision Medicine Center, The First Affiliated Hospital of Xiu'an Jiaotong University, Shaanxi, People's Republic of China.
| | - Jong-Il Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.
- Cancer Research Institute, Seoul National University, Seoul, Korea.
- Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea.
| |
Collapse
|
248
|
Chronological genome and single-cell transcriptome integration characterizes the evolutionary process of adult T cell leukemia-lymphoma. Nat Commun 2021; 12:4821. [PMID: 34376672 PMCID: PMC8355240 DOI: 10.1038/s41467-021-25101-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 07/23/2021] [Indexed: 02/05/2023] Open
Abstract
Subclonal genetic heterogeneity and their diverse gene expression impose serious problems in understanding the behavior of cancers and contemplating therapeutic strategies. Here we develop and utilize a capture-based sequencing panel, which covers host hotspot genes and the full-length genome of human T-cell leukemia virus type-1 (HTLV-1), to investigate the clonal architecture of adult T-cell leukemia-lymphoma (ATL). For chronologically collected specimens from patients with ATL or pre-onset individuals, we integrate deep DNA sequencing and single-cell RNA sequencing to detect the somatic mutations and virus directly and characterize the transcriptional readouts in respective subclones. Characteristic genomic and transcriptomic patterns are associated with subclonal expansion and switches during the clinical timeline. Multistep mutations in the T-cell receptor (TCR), STAT3, and NOTCH pathways establish clone-specific transcriptomic abnormalities and further accelerate their proliferative potential to develop highly malignant clones, leading to disease onset and progression. Early detection and characterization of newly expanded subclones through the integrative analytical platform will be valuable for the development of an in-depth understanding of this disease. Characterising the clonal architecture of Adult T-cell leukemia-lymphoma (ATL) remains crucial. Here, the authors develop a capture-based sequencing panel and use deep DNA and single cell RNA sequencing and report distinct genomic and transcriptomic features associated with subclonal evolution.
Collapse
|
249
|
Sayyab S, Lundmark A, Larsson M, Ringnér M, Nystedt S, Marincevic-Zuniga Y, Tamm KP, Abrahamsson J, Fogelstrand L, Heyman M, Norén-Nyström U, Lönnerholm G, Harila-Saari A, Berglund EC, Nordlund J, Syvänen AC. Mutational patterns and clonal evolution from diagnosis to relapse in pediatric acute lymphoblastic leukemia. Sci Rep 2021; 11:15988. [PMID: 34362951 PMCID: PMC8346595 DOI: 10.1038/s41598-021-95109-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
The mechanisms driving clonal heterogeneity and evolution in relapsed pediatric acute lymphoblastic leukemia (ALL) are not fully understood. We performed whole genome sequencing of samples collected at diagnosis, relapse(s) and remission from 29 Nordic patients. Somatic point mutations and large-scale structural variants were called using individually matched remission samples as controls, and allelic expression of the mutations was assessed in ALL cells using RNA-sequencing. We observed an increased burden of somatic mutations at relapse, compared to diagnosis, and at second relapse compared to first relapse. In addition to 29 known ALL driver genes, of which nine genes carried recurrent protein-coding mutations in our sample set, we identified putative non-protein coding mutations in regulatory regions of seven additional genes that have not previously been described in ALL. Cluster analysis of hundreds of somatic mutations per sample revealed three distinct evolutionary trajectories during ALL progression from diagnosis to relapse. The evolutionary trajectories provide insight into the mutational mechanisms leading relapse in ALL and could offer biomarkers for improved risk prediction in individual patients.
Collapse
Affiliation(s)
- Shumaila Sayyab
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Box 1432, 75144, Uppsala, Sweden.
| | - Anders Lundmark
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Box 1432, 75144, Uppsala, Sweden
| | - Malin Larsson
- Department of Physics, Chemistry and Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Linköping University, Linköping, Sweden
| | - Markus Ringnér
- Department of Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Lund University, Lund, Sweden
| | - Sara Nystedt
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Box 1432, 75144, Uppsala, Sweden
| | - Yanara Marincevic-Zuniga
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Box 1432, 75144, Uppsala, Sweden
| | | | - Jonas Abrahamsson
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,For the Nordic Society of Pediatric Hematology and Oncology, Stockholm, Sweden
| | - Linda Fogelstrand
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden.,For the Nordic Society of Pediatric Hematology and Oncology, Stockholm, Sweden
| | - Mats Heyman
- Childhood Cancer Research Unit, Karolinska University Hospital, Stockholm, Sweden.,For the Nordic Society of Pediatric Hematology and Oncology, Stockholm, Sweden
| | - Ulrika Norén-Nyström
- Department of Clinical Sciences and Pediatrics, University of Umeå, Umeå, Sweden.,For the Nordic Society of Pediatric Hematology and Oncology, Stockholm, Sweden
| | - Gudmar Lönnerholm
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Arja Harila-Saari
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden.,For the Nordic Society of Pediatric Hematology and Oncology, Stockholm, Sweden
| | - Eva C Berglund
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Box 1432, 75144, Uppsala, Sweden
| | - Jessica Nordlund
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Box 1432, 75144, Uppsala, Sweden
| | - Ann-Christine Syvänen
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Box 1432, 75144, Uppsala, Sweden.
| |
Collapse
|
250
|
Genomic characterization of co-existing neoplasia and carcinoma lesions reveals distinct evolutionary paths of gallbladder cancer. Nat Commun 2021; 12:4753. [PMID: 34362903 PMCID: PMC8346570 DOI: 10.1038/s41467-021-25012-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 07/16/2021] [Indexed: 12/30/2022] Open
Abstract
Gallbladder carcinoma is the most common cancer of the biliary tract with dismal survival largely due to delayed diagnosis. Biliary tract intraepithelial neoplasia (BilIN) is the common benign tumor that is suspected to be precancerous lesions. However, the genetic and evolutionary relationships between BilIN and carcinoma remain unclear. Here we perform whole-exome sequencing of coexisting low-grade BilIN (adenoma), high-grade BilIN, and carcinoma lesions, and normal tissues from the same patients. We identify aging as a major factor contributing to accumulated mutations and a critical role of CTNNB1 mutations in these tumors. We reveal two distinct carcinoma evolutionary paths: carcinoma can either diverge earlier and evolve more independently or form through the classic adenoma/dysplasia-carcinoma sequence model. Our analysis suggests that extensive loss-of-heterozygosity and mutation events in the initial stage tend to result in a cancerous niche, leading to the subsequent BilIN-independent path. These results reframes our understanding of tumor transformation and the evolutionary trajectory of carcinogenesis in the gallbladder, laying a foundation for the early diagnosis and effective treatment of gallbladder cancer. The progression from biliary tract intraepithelial neoplasia (BilIN) to gallbladder carcinoma (GBC) remains unclear. Here the authors use genomics to analyze coexisting GBC lesions, low-grade and high-grade BilINs, revealing two distinct evolutionary paths for GBC development.
Collapse
|