1
|
Byrne ME, Selenica P, Dessources K, Da Cruz Paula A, Gordhandas S, Wu M, Pareja F, Roche KL, Mueller JJ, Sonoda Y, Abu-Rustum NR, Weigelt B. Peritoneal washings analysis in endometrial cancer: Comparison of somatic mutation detection with panel sequencing and traditional cytology. Gynecol Oncol 2025; 197:155-162. [PMID: 40347837 DOI: 10.1016/j.ygyno.2025.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
OBJECTIVES The prognostic significance of positive pelvic washings in endometrial cancer (EC) remains unknown, and little data exist regarding washings as a source of genetic information in relation to a patient's tumor. We sought to assess the feasibility of identifying EC mutations in peritoneal washings. METHODS Peritoneal washings from 21 biopsy-confirmed newly diagnosed patients with EC across disease stages between 09/2018 and 07/2019 were collected. Peritoneal washings, primary EC, and normal DNA samples were subjected to next-generation sequencing targeting 468 cancer-related genes. Sequencing results were compared to cytological analysis. RESULTS For the 21 EC cases included, cytology found 8 (38 %) of the peritoneal washings as positive, 7 (33 %) as negative, and 6 (29 %) as suspicious or rare-atypical cells. Based on molecular analysis, tumor mutations (TMs) were detected in 18/21 (86 %) of peritoneal washings. Overall, 11/21 (52 %) samples demonstrated concordant results between cytologic and molecular analysis, and all positive cytologic results were confirmed with molecular analysis. However, of cases with negative or suspicious cytology results, 77 % (10/13) were found to have TMs in washings. Five patients with negative cytology were positive on molecular analysis (5/7, 71 %), and 5 patients with suspicious washings demonstrated TMs (5/6, 83 %). Of the 10 EC patients who developed recurrences, regardless of stage, 5/10 (50 %) patients had positive cytology, whereas 9/10 (90 %) had TMs based on molecular analysis. CONCLUSIONS Mutational analysis of peritoneal washings using panel sequencing in EC is feasible. A substantial subset of patients with cytology-negative or suspicious washings had TMs detected.
Collapse
Affiliation(s)
- Maureen E Byrne
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pier Selenica
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kimberly Dessources
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Arnaud Da Cruz Paula
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sushmita Gordhandas
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michelle Wu
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fresia Pareja
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kara Long Roche
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of OB/GYN, Weill Cornell Medical College, New York, NY, USA
| | - Jennifer J Mueller
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of OB/GYN, Weill Cornell Medical College, New York, NY, USA
| | - Yukio Sonoda
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of OB/GYN, Weill Cornell Medical College, New York, NY, USA
| | - Nadeem R Abu-Rustum
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of OB/GYN, Weill Cornell Medical College, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
2
|
Zheng Y, Ahmad K, Henikoff S. Total whole-arm chromosome losses predict malignancy in human cancer. Proc Natl Acad Sci U S A 2025; 122:e2505385122. [PMID: 40314975 PMCID: PMC12067283 DOI: 10.1073/pnas.2505385122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Aneuploidy is observed as gains or losses of whole chromosomes or chromosome arms and is a common hallmark of cancer. Whereas models for the generation of aneuploidy in cancer invoke mitotic chromosome segregation errors, whole-arm losses might occur simply as a result of centromere breakage. We recently showed that elevated RNA Polymerase II level over the S-phase-dependent histone genes predicts rapid recurrence of human meningioma and is correlated with total whole-arm losses relative to gains. To explain this imbalance in arm losses over gains, we have proposed that histone overexpression at S-phase competes with the histone H3 variant CENP-A, resulting in centromere breaks and whole-arm losses. To test whether centromere breaks alone can drive aneuploidy, we ask whether total whole-arm aneuploids can predict outcomes across different cancer types in large RNA and whole-genome sequencing databanks. We find that total whole-arm losses generally predict outcome, suggesting that centromere breakage is a major initiating factor leading to aneuploidy and the resulting changes in the selective landscape that drive most cancers. We also present evidence that centromere breakage alone is sufficient to account for whole-arm losses and gains, contrary to mitotic spindle error models for the generation of aneuploidy. Our results suggest that therapeutic intervention targeting histone overexpression has the potential to reduce aneuploidy and slow cancer progression.
Collapse
Affiliation(s)
- Ye Zheng
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA98109
| | - Kami Ahmad
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA98109
| | - Steven Henikoff
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA98109
- HHMI, Chevy Chase, MD20815
| |
Collapse
|
3
|
Zhang D, Segerstolpe Å, Slyper M, Waldman J, Murray E, Strasser R, Watter J, Cohen O, Ashenberg O, Abravanel D, Jané-Valbuena J, Mages S, Lako A, Helvie K, Rozenblatt-Rosen O, Rodig S, Chen F, Wagle N, Regev A, Klughammer J. SlideCNA: spatial copy number alteration detection from Slide-seq-like spatial transcriptomics data. Genome Biol 2025; 26:112. [PMID: 40317049 PMCID: PMC12046676 DOI: 10.1186/s13059-025-03573-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/08/2025] [Indexed: 05/04/2025] Open
Abstract
Solid tumors are spatially heterogeneous in their genetic, molecular, and cellular composition, but recent spatial profiling studies have mostly charted genetic and RNA variation in tumors separately. To leverage the potential of RNA to identify copy number alterations (CNAs), we develop SlideCNA, a computational tool to extract CNA signals from sparse spatial transcriptomics data with near single cellular resolution. SlideCNA uses expression-aware spatial binning to overcome sparsity limitations while maintaining spatial signal to recover CNA patterns. We test SlideCNA on simulated and real Slide-seq data of (metastatic) breast cancer and demonstrate its potential for spatial subclone detection.
Collapse
Affiliation(s)
- Diane Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Åsa Segerstolpe
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michal Slyper
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - Julia Waldman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Evan Murray
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robert Strasser
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jan Watter
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ofir Cohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University, Beersheba, Israel
| | - Orr Ashenberg
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel Abravanel
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Judit Jané-Valbuena
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - Simon Mages
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ana Lako
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Karla Helvie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Orit Rozenblatt-Rosen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - Scott Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Fei Chen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nikhil Wagle
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Genentech, 1 DNA Way, South San Francisco, CA, USA.
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Massachusetts Institute of Technology, Cambridge, MA, USA.
- Genentech, 1 DNA Way, South San Francisco, CA, USA.
| | - Johanna Klughammer
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
4
|
Dong M, Chen J, Lu N, Wang S, Wei W, Wang Z, Wang J, Zhang J, Han X, Wang F, Ou Q, Bao H, Ma X, Shan B, Pan Y. Unraveling breast cancer response to neoadjuvant chemotherapy through integrated genomic, transcriptomic, and circulating tumor DNA analysis. Breast Cancer Res 2025; 27:64. [PMID: 40312292 PMCID: PMC12044986 DOI: 10.1186/s13058-025-02026-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/16/2025] [Indexed: 05/03/2025] Open
Abstract
INTRODUCTION Neoadjuvant chemotherapy (NAC) is a standard treatment for breast cancer (BC) to shrink tumors and facilitate surgery. However, the molecular underpinnings of response to NAC and prognosis have not been well characterized. METHODS We enrolled 73 stage II/III BC patients who received NAC followed by surgery. Tumor tissue samples were available from 36 patients at baseline and 38 at the time of surgery. Plasma circulating tumor DNA (ctDNA) was collected at three time points: before NAC (n = 63), during NAC (n = 42), and after NAC (n = 40). Comprehensive genomic, transcriptomic, and ctDNA analyses were performed to identify biomarkers associated with pathological complete response (pCR) and survival outcomes. RESULTS Nine baseline mutations, including DNHD1 and PLEC, along with HIPPO pathway alterations, were associated with pCR. Responsive tumors exhibited immune activation and downregulated PI3K-Akt and AGE-RAGE pathways, while non-pCR tumors showed reduced cytokine and immune receptor activity. Undetectable ctDNA during and after NAC was predictive of treatment efficacy and correlated with improved survival. Baseline mutations in USH2A were associated with shorter disease-free survival (hazard ratio: 11.9; 95% confidence interval: 2.8-50.8; P < 0.001), with a consistent trend observed for overall survival. Elevated NHSL1 expression in baseline tumors indicated an initial treatment response but was later associated with tumor relapse and poor overall survival (P = 0.026 and P = 0.023, respectively), findings that were validated in an independent clinical cohort (N = 30) through immunohistochemistry staining. CONCLUSION Our comprehensive multi-omics analysis identified promising biomarkers predictive of treatment response and survival in BC patients receiving NAC followed by surgery. These findings underscore the importance of early tumor assessment for improved patient stratification and prognostication.
Collapse
Affiliation(s)
- Menghao Dong
- Department of Clinical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 17 Lujiang Road, Luyang District, Hefei, Anhui, 230001, China
| | - Jian Chen
- Department of Clinical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 17 Lujiang Road, Luyang District, Hefei, Anhui, 230001, China
| | - Nannan Lu
- Department of Clinical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 17 Lujiang Road, Luyang District, Hefei, Anhui, 230001, China
| | - Song Wang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, 210032, China
| | - Wenhui Wei
- Department of Clinical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 17 Lujiang Road, Luyang District, Hefei, Anhui, 230001, China
| | - Ziming Wang
- Department of Clinical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 17 Lujiang Road, Luyang District, Hefei, Anhui, 230001, China
| | - Jinnan Wang
- Department of Clinical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 17 Lujiang Road, Luyang District, Hefei, Anhui, 230001, China
| | - Jinguo Zhang
- Department of Clinical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 17 Lujiang Road, Luyang District, Hefei, Anhui, 230001, China
| | - Xinghua Han
- Department of Clinical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 17 Lujiang Road, Luyang District, Hefei, Anhui, 230001, China
| | - Fufeng Wang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, 210032, China
| | - Qiuxiang Ou
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, 210032, China
| | - Hua Bao
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, 210032, China
| | - Xiaopeng Ma
- Department of Breast Surgery, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 17 Lujiang Road, Luyang District, Hefei, Anhui, 230001, China.
| | - Benjie Shan
- Department of Clinical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 17 Lujiang Road, Luyang District, Hefei, Anhui, 230001, China.
| | - Yueyin Pan
- Department of Clinical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No. 17 Lujiang Road, Luyang District, Hefei, Anhui, 230001, China.
| |
Collapse
|
5
|
Tao T, Liu S, He M, Zhao M, Chen C, Peng J, Wang Y, Cai J, Xiong J, Lai C, Gu W, Ying M, Mao J, Li L, Jia X, Wu X, Peng W, Zhang X, Li Y, Li T, Wang J, Shu Q. Synchronous bilateral Wilms tumors are prone to develop independently and respond differently to preoperative chemotherapy. Int J Cancer 2025; 156:1746-1755. [PMID: 39723643 DOI: 10.1002/ijc.35297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/20/2024] [Accepted: 11/28/2024] [Indexed: 12/28/2024]
Abstract
Wilms tumor (WT) is the most common kidney cancer in infants and young children. The determination of the clonality of bilateral WTs is critical to the treatment, because lineage-independent and metastatic tumors may require different treatment strategies. Here we found synchronous bilateral WT (n = 24 tumors from 12 patients) responded differently to preoperative chemotherapy. Transcriptome, whole-exome and whole-genome analysis (n = 12 tumors from 6 patients) demonstrated that each side of bilateral WT was clonally independent in terms of somatic driver mutations, copy number variations and transcriptomic profile. Molecular timing analysis revealed distinct timing and patterns of chromosomal evolution and mutational processes between the two sides of WT. Mutations in WT1, CTNNB1 and copy-neutral loss of heterozygosity of 11p15.5 provide possible genetic predisposition for the early initiation of bilateral WT. Our results provide comprehensive evidence and new insights regarding the separate initiation and early embryonic development of bilateral WT, which may benefit clinical practices in treating metastatic or refractory bilateral WT.
Collapse
Affiliation(s)
- Ting Tao
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Shuangai Liu
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- The First Clinical Institute, Zunyi Medical University, Zunyi, China
| | - Min He
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Manli Zhao
- Department of Pathology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chen Chen
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jinkai Peng
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yilong Wang
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Neurology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jiabin Cai
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jieni Xiong
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Can Lai
- Department of Radiology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Weizhong Gu
- Department of Pathology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Meidan Ying
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
- Nanhu Brain-computer Interface Institute, Hangzhou, China
| | - Junqing Mao
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Linjie Li
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xuan Jia
- Department of Radiology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xuan Wu
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Wanxin Peng
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Xiang Zhang
- The Affiliated Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yong Li
- Hunan Children's Hospital, Changsha, China
| | - Tao Li
- Department of Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jinhu Wang
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Qiang Shu
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
6
|
Sia TY, Allison DHR, Da Cruz Paula A, da Silva EM, Ye Q, Selenica P, Pareja F, Green H, Abu-Rustum NR, Weigelt B, Ellenson LH. Clinicopathologic and Genomic Analysis of Uterine Serous Carcinomas Arising From Endometrial Hyperplasia. Am J Surg Pathol 2025:00000478-990000000-00514. [PMID: 40298247 DOI: 10.1097/pas.0000000000002401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Uterine serous carcinoma (USC) typically arises from atrophic endometrium but may be associated with hyperplasia in 5% to 10% of cases. We sought to identify USC with concurrent hyperplasia and (i) define if these are clonally related, and (ii) determine if USC associated with hyperplasia is genetically distinct from USC without hyperplasia. Patients diagnosed with USC and hyperplasia from their hysterectomy specimen between January 1, 2014 and February 29, 2022 were identified. Hyperplasia and carcinoma were separately subjected to tumor-normal panel sequencing. Their repertoire of genetic alterations was compared with that of a separate cohort of atrophy-associated USCs. Of 267 USCs with clinical sequencing and slides available for review, 8 with concurrent carcinoma and hyperplasia had sufficient tissue for molecular studies. In 7 (87.5%) of these 8 cases, USC and hyperplasia were clonally related and shared multiple mutations, including TP53 in 4 cases (57%). In 1 case (USC4), USC and hyperplasia were unrelated at the genetic level, and the hyperplasia was TP53 wild-type. In another case (USC5), USC and TP53 wild-type hyperplasia shared 1 of 11 mutations while being distinct at the copy number level. The prevalence of ARID1A mutations was higher in hyperplasia-associated USC compared with atrophy-associated USC (43% vs. 0%, respectively; P=0.02). USC and co-occurring hyperplasia were clonally related in most cases, commonly harboring TP53 hotspot mutations in both components. These results suggest an alternative origin of tumorigenesis in this rare subset of endometrial cancers.
Collapse
Affiliation(s)
| | | | - Arnaud Da Cruz Paula
- Department of Pathology, Memorial Sloan Kettering Cancer Center
- i3S Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | | | - Qiqi Ye
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | - Hunter Green
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | - Nadeem R Abu-Rustum
- Department of Surgery, Gynecology Service
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| | - Lora H Ellenson
- Department of Pathology, Memorial Sloan Kettering Cancer Center
| |
Collapse
|
7
|
Wong J, Tian Y, Patel MS, Avasthi K, Hanson C, Larsen M, Ampaw E, Fadlullah MZH, Finklestein J, Tan AC, Park J, Manley BJ, Huang CC, Kohli M, Wang L. Plasma Cell-Free DNA Methylation-Based Prognosis in Metastatic Castrate-Resistant Prostate Cancer. RESEARCH SQUARE 2025:rs.3.rs-6331572. [PMID: 40313768 PMCID: PMC12045346 DOI: 10.21203/rs.3.rs-6331572/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Molecular prognostication in metastatic castration prostate cancer (mCRPC) remains challenging due to the lack of validated biomarkers. This study developed a plasma cell-free DNA (cfDNA) methylation-based prognostic model in mCRPC. Targeted cfDNA methylation sequencing in 96 prostate cancer patients in different states of cancer progression revealed 78 methylation haplotype blocks (MHBs) differentially methylated from organ-confined prostate cancer to mCRPC states. Among these 78 MHBs, the top 20 MHBs were associated with mCRPC overall survival and most MHB methylation levels positively correlated with predicted circulating tumor DNA (ctDNA) fraction. By integrating the MHB-based risk score with currently available prognostic clinical variables and ctDNA fraction a prognostic nomogram was developed which showed high predictive performance for mCRPC survival (AUC = 0.99 for 6 months, AUC = 0.90 for 1 year, and AUC = 0.87 for 2 years). These findings demonstrate potential of cfDNA methylation as a molecular biology-driven biomarker for mCRPC prognosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Matt Larsen
- University of Utah, Huntsman Cancer Institute
| | - Enos Ampaw
- University of Utah, Huntsman Cancer Institute
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Fiore D, Cappelli LV, Zhaoqi L, Kotlov N, Sorokina M, Phillip J, Zumbo P, Yoffe L, Ghione P, Wang A, Han X, Taylor A, Chiu W, Fragliasso V, Tabbo F, Zamponi N, Di Siervi N, Kayembe C, Medico G, Patel RP, Gaudiano M, Machiorlatti R, Astone G, Cacciapuoti MT, Zanetti G, Pignataro C, Eric RA, Patel S, Zammarchi F, Zanettini C, Queiroz L, Nikitina A, Kudryashova O, Karelin A, Nikitin D, Tychinin D, Postovalova E, Bagaev A, Svekolkin V, Belova E, Tikhonova K, Degryse S, Xu C, Novero D, Ponzoni M, Tiacci E, Falini B, Song J, Khodos I, De Stanchina E, Macari G, Cafforio L, Gardini S, Piva R, Medico E, Ng SY, Moskowitz A, Epstein Z, Intlekofer A, Ahmed D, Chan WC, Martin P, Ruan J, Bertoni F, Foà R, Brody JD, Weinstock DM, Osan J, Santambrogio L, Elemento O, Betel D, Tam W, Ruella M, Cerchietti L, Rabadan R, Horwitz S, Inghirami G. A patient-derived T cell lymphoma biorepository uncovers pathogenetic mechanisms and host-related therapeutic vulnerabilities. Cell Rep Med 2025; 6:102029. [PMID: 40147445 PMCID: PMC12047492 DOI: 10.1016/j.xcrm.2025.102029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 04/24/2024] [Accepted: 02/21/2025] [Indexed: 03/29/2025]
Abstract
Peripheral T cell lymphomas (PTCLs) comprise heterogeneous malignancies with limited therapeutic options. To uncover targetable vulnerabilities, we generate a collection of PTCL patient-derived tumor xenografts (PDXs) retaining histomorphology and molecular donor-tumor features over serial xenografting. PDX demonstrates remarkable heterogeneity, complex intratumor architecture, and stepwise trajectories mimicking primary evolutions. Combining functional transcriptional stratification and multiparametric imaging, we identify four distinct PTCL microenvironment subtypes with prognostic value. Mechanistically, we discover a subset of PTCLs expressing Epstein-Barr virus-specific T cell receptors and uncover the capacity of cancer-associated fibroblasts of counteracting treatments. PDXs' pre-clinical testing captures individual vulnerabilities, mirrors donor patients' clinical responses, and defines effective patient-tailored treatments. Ultimately, we assess the efficacy of CD5KO- and CD30- Chimeric Antigen Receptor T Cells (CD5KO-CART and CD30_CART, respectively), demonstrating their therapeutic potential and the synergistic role of immune checkpoint inhibitors for PTCL treatment. This repository represents a resource for discovering and validating intrinsic and extrinsic factors and improving the selection of drugs/combinations and immune-based therapies.
Collapse
Affiliation(s)
- Danilo Fiore
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; Institute for Experimental Endocrinology and Oncology, "G.Salvatore" IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Luca Vincenzo Cappelli
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Liu Zhaoqi
- Program for Mathematical Genomics, Department of Systems Biology, Department of Biomedical Informatics, Columbia University, New York, NY 10027 USA; China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | | | | | - Jude Phillip
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US; Chemical and Biomolecular Engineering, Oncology, Sidney Kimmel Comprehensive Cancer Center, Core Member, Institute for Nanobiotechnology (INBT), Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Paul Zumbo
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, US
| | - Liron Yoffe
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Paola Ghione
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anqi Wang
- Program for Mathematical Genomics, Department of Systems Biology, Department of Biomedical Informatics, Columbia University, New York, NY 10027 USA
| | - Xueshuai Han
- Program for Mathematical Genomics, Department of Systems Biology, Department of Biomedical Informatics, Columbia University, New York, NY 10027 USA; China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Abigail Taylor
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - William Chiu
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Valentina Fragliasso
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Laboratory of translational research, Azienda USL - IRCCS di Reggio Emilia, 42122 Reggio Emila, Italy
| | - Fabrizio Tabbo
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; SC Oncologia ASL CN2 Alba Bra Ospedale Michele e Pietro Ferrero, 12060 Verduno, (CN), Italy
| | - Nahuel Zamponi
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US
| | - Nicolás Di Siervi
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US
| | - Clarisse Kayembe
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Giovanni Medico
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ruchi P Patel
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Perelman Center for Advanced Medicine, SPE 8-112, Philadelphia, PA 19104, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marcello Gaudiano
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Rodolfo Machiorlatti
- Department of Pathology, Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy
| | - Giuseppina Astone
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Maria Teresa Cacciapuoti
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Giorgia Zanetti
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Claudia Pignataro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Ruiz Arvin Eric
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sanjay Patel
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Claudio Zanettini
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lucio Queiroz
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Chengqi Xu
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Domenico Novero
- Division of Pathological Anatomy, Quality and Safety of Diagnosis and Treatment, Città della Salute e della Scienza, 10126 Turin, Italy
| | - Maurilio Ponzoni
- Pathology Unit, San Raffaele Scientific Institute, Milan, Italy; Unit of Lymphoid Malignancies, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Enrico Tiacci
- Institute of Hematology, University of Perugia, Ospedale S. Maria della Misericordia, S. Andrea delle Fratte, 06156 Perugia Italy
| | - Brunangelo Falini
- Institute of Hematology, University of Perugia, Ospedale S. Maria della Misericordia, S. Andrea delle Fratte, 06156 Perugia Italy
| | - Joo Song
- Department of Pathology, City of Hope Medical Center, Duarte, CA 91010, US
| | - Inna Khodos
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, US
| | - Elisa De Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, US
| | | | | | | | - Roberto Piva
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; Medical Genetics Unit, Città della Salute e della Scienza University Hospital, 10126 Turin, Italy
| | - Enzo Medico
- Department of Oncology, University of Torino, Candiolo, TO, Italy; Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy
| | - Samuel Y Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; National Cancer Institute, Bethesda, MD 20892, USA
| | - Allison Moskowitz
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zachary Epstein
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrew Intlekofer
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dogan Ahmed
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Wing C Chan
- Department of Pathology, City of Hope Medical Center, Duarte, CA 91010, US
| | - Peter Martin
- Lymphoma Service, Weill Cornell Medical Center, New York, NY 10065, USA
| | - Jia Ruan
- Lymphoma Service, Weill Cornell Medical Center, New York, NY 10065, USA
| | - Francesco Bertoni
- Lymphoma Genomics, Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; Oncology Institute of Southern Switzerland, EOC,6500 Bellinzona, Switzerland
| | - Robin Foà
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Joshua D Brody
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, US; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Jaspreet Osan
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Oliver Elemento
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Doron Betel
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US; Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, US
| | - Wayne Tam
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Division of Hematopathology, Northwell Health, New York, NY 11740, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Perelman Center for Advanced Medicine, SPE 8-112, Philadelphia, PA 19104, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Leandro Cerchietti
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US
| | - Raul Rabadan
- Program for Mathematical Genomics, Department of Systems Biology, Department of Biomedical Informatics, Columbia University, New York, NY 10027 USA
| | - Steven Horwitz
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Giorgio Inghirami
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
9
|
Kus ME, Sahin C, Kilic E, Askin A, Ozgur MM, Karahanogullari G, Aksit A, O'Connell RM, Ekiz HA. TCGEx: a powerful visual interface for exploring and analyzing cancer gene expression data. EMBO Rep 2025; 26:1863-1890. [PMID: 40033050 PMCID: PMC11976970 DOI: 10.1038/s44319-025-00407-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
Analyzing gene expression data from the Cancer Genome Atlas (TCGA) and similar repositories often requires advanced coding skills, creating a barrier for many researchers. To address this challenge, we developed The Cancer Genome Explorer (TCGEx), a user-friendly, web-based platform for conducting sophisticated analyses such as survival modeling, gene set enrichment analysis, unsupervised clustering, and linear regression-based machine learning. TCGEx provides access to preprocessed TCGA data and immune checkpoint inhibition studies while allowing integration of user-uploaded data sets. Using TCGEx, we explore molecular subsets of human melanoma and identify microRNAs associated with intratumoral immunity. These findings are validated with independent clinical trial data on immune checkpoint inhibitors for melanoma and other cancers. In addition, we identify cytokine genes that can be used to predict treatment responses to various immune checkpoint inhibitors prior to treatment. Built on the R/Shiny framework, TCGEx offers customizable features to adapt analyses for diverse research contexts and generate publication-ready visualizations. TCGEx is freely available at https://tcgex.iyte.edu.tr , providing an accessible tool to extract insights from cancer transcriptomics data.
Collapse
Affiliation(s)
- M Emre Kus
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Cagatay Sahin
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Emre Kilic
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Arda Askin
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - M Mert Ozgur
- The Department of Molecular Biology and Genetics, Bilkent University, 06800, Cankaya, Ankara, Turkey
| | - Gokhan Karahanogullari
- The Department of Mathematics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Ahmet Aksit
- The Department of Information Technologies, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey
| | - Ryan M O'Connell
- The Department of Pathology, University of Utah, Salt Lake City, UT, 84112, USA
| | - H Atakan Ekiz
- The Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Gulbahce, Izmir, Turkey.
| |
Collapse
|
10
|
Zhong Y, Zhang W, Zheng C, Wu H, Luo J, Yuan Z, Zhang H, Wang C, Feng H, Wang M, Zhang Q, Ju H, Wang G. Multi-omic analyses reveal PTPN6's impact on tumor immunity across various cancers. Sci Rep 2025; 15:11025. [PMID: 40164665 PMCID: PMC11958644 DOI: 10.1038/s41598-025-96302-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/27/2025] [Indexed: 04/02/2025] Open
Abstract
Protein Tyrosine Phosphatase Non-Receptor Type 6 (PTPN6) plays a crucial regulatory role in cellular processes and has been implicated in oncogenesis. This pan-cancer analysis aimed to elucidate PTPN6's involvement across various cancer types, with a particular emphasis on its association with tumor immunity. We analyzed PTPN6 expression data from open access databases using various statistical techniques, including survival analysis, genetic heterogeneity analysis, immune profiling, single-cell analysis, drug sensitivity analysis, and protein interaction analysis. We also conducted in vitro experiments utilizing colorectal cancer cell lines to validate PTPN6's functional role. PTPN6 exhibited distinct expression patterns across cancers, and its prognostic significance was apparent in several cancer types, particularly in glioblastoma, sarcoma, and melanoma. We observed correlations between PTPN6 and immune genes/cell infiltration in these cancers, suggesting a potential role in modulating the tumor immune microenvironment. Single-cell analysis revealed that PTPN6 is predominantly localized in macrophages, B cells, and dendritic cells within the tumor microenvironment, implying its involvement in regulating immune cell function. Enrichment analysis highlighted PTPN6's role in immune-related pathways. Drug sensitivity analysis identified specific drugs, including PAC-1, SNX-2112, BELINOSTAT, VORINOSTAT, TPCA-1, and PHA-893,888, whose efficacy may be influenced by PTPN6 expression. Knocking down PTPN6 expression inhibited the proliferation and migration of colorectal cancer cells in vitro, confirming its oncogenic role in this cancer type. This pan-cancer analysis establishes PTPN6's multifaceted influence on tumor immunity and its potential as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Yuchen Zhong
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China
| | - Weiyuan Zhang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Chaojing Zheng
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Hongyu Wu
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Jun Luo
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Ziming Yuan
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Hao Zhang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Chunlin Wang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Haiyang Feng
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Meng Wang
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Qian Zhang
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China.
| | - Haixing Ju
- Department of Colorectal Cancer Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China.
| | - Guiyu Wang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, Heilongjiang, People's Republic of China.
| |
Collapse
|
11
|
Zheng Y, Ahmad K, Henikoff S. Total whole-arm chromosome losses predict malignancy in human cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.09.642243. [PMID: 40236246 PMCID: PMC11996446 DOI: 10.1101/2025.03.09.642243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Aneuploidy is observed as gains or losses of whole chromosomes or chromosome arms and is a common hallmark of cancer. Whereas models for the generation of aneuploidy in cancer invoke mitotic chromosome segregation errors, whole-arm losses might occur simply as a result of centromere breakage. We recently showed that elevated RNA Polymerase II (RNAPII) level over S-phase-dependent histone genes predicts rapid recurrence of human meningioma and is correlated with total whole-arm losses relative to gains. To explain this imbalance in arm losses over gains, we have proposed that histone overexpression at S-phase competes with the histone H3 variant CENP-A, resulting in centromere breaks and whole-arm losses. To test whether centromere breaks alone can drive aneuploidy, we ask whether total whole-arm aneuploids can predict outcome across different cancer types in large RNA and whole-genome sequencing databanks. We find that total whole-arm losses generally predict outcome, suggesting that centromere breakage is a major initiating factor leading to aneuploidy and the resulting changes in the selective landscape that drive most cancers. We also present evidence that centromere breakage alone is sufficient to account for whole-arm losses and gains, contrary to mitotic spindle error models for generation of aneuploidy. Our results suggest that therapeutic intervention targeting histone overexpression has the potential of reducing aneuploidy and slowing cancer progression. Significance Statement Gain or loss of whole chromosome arms following centromere breaks is frequent in cancer, but whether or not there is a common initiating event is unknown. Here we show that the total number of whole-arm losses predicts patient outcomes across cancer types, suggesting a causal relationship. This general excess of losses over gains is not predicted by mitotic error models of aneuploidy but rather suggests that centromere breaks themselves initiate whole-arm aneuploidies. Insofar as aneuploidy reshapes the selective landscapes that drive most cancers, our results have potential clinical implications.
Collapse
|
12
|
Sitthirak S, Wangwiwatsin A, Jusakul A, Namwat N, Klanrit P, Dokduang H, Sa-Ngiamwibool P, Titapun A, Jareanrat A, Thanasukarn V, Khuntikeo N, Teh BT, Boulter L, Murakami Y, Loilome W. Whole exome sequencing of multi-regions reveals tumor heterogeneity in Opisthorchis viverrini-associated cholangiocarcinoma. Sci Rep 2025; 15:10886. [PMID: 40157958 PMCID: PMC11954897 DOI: 10.1038/s41598-025-95142-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 03/19/2025] [Indexed: 04/01/2025] Open
Abstract
The study examines Opisthorchis viverrini (OV)-related cholangiocarcinoma (CCA), a serious malignancy common in Southeast Asia. Through multi-regional whole-exome sequencing of 52 tumor samples and 13 adjacent tissues from 13 patients, significant intratumoral heterogeneity (ITH) and inter-patient heterogeneity are shown. Chronic liver fluke infection induces a distinct mutational landscape, with 48-90% of mutations concentrated in each region of the tumor. The average mutation burden is 95 non-synonymous mutations per area, exceeding previous CCA investigations. Critical driver mutations in TP53, SMAD4, and other genes underscore their significance in pathogenesis. Mutational markers elucidate mechanisms including spontaneous deamination and impaired DNA repair. Unique mutation patterns distinguish OV-associated CCA from other variants. Chromosomal instability in patient K110 signifies aggressive tumor behavior and unfavorable prognosis. Targetable mutations such as ERBB2 underscore the possibility for personalized therapeutics. These findings underscore the necessity for personalized strategies for treatment that target both trunk and branch mutations in endemic areas.
Collapse
Affiliation(s)
- Sirinya Sitthirak
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Arporn Wangwiwatsin
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Apinya Jusakul
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
- Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Nisana Namwat
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Poramate Klanrit
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Hasaya Dokduang
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
- Faculty of Medicine, Mahasarakham University, Kantharawichai District, Mahasarakham, 44000, Thailand
| | - Prakasit Sa-Ngiamwibool
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Attapol Titapun
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Apiwat Jareanrat
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Vasin Thanasukarn
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Natcha Khuntikeo
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Bin Tean Teh
- National Cancer Centre Singapore, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh, EH4 2XU, Scotland, UK
| | - Yoshinori Murakami
- Department of Molecular Biology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Watcharin Loilome
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
13
|
Zhan S, Chen F, Huang L, Chen L, Jia H, Ma S, Tang M, Zhou C, Chen Y, Yang Y. The Clinical Pathological Characteristics and Prognostic Relevance of Homologous Recombination Repair Gene Mutations in Ovarian Cancer Patients: A Prospective Cohort Study. Obstet Gynecol Int 2025; 2025:5578247. [PMID: 40166687 PMCID: PMC11957853 DOI: 10.1155/ogi/5578247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 02/21/2025] [Indexed: 04/02/2025] Open
Abstract
Backgrouds: Whether homologous recombination repair (HRR) mutation has a differential effect on the prognosis has not been confirmed by current studies. The purpose of this study was to explore the clinical importance, prognostic value, and frequency of pathogenic changes in HRR genes in patients with ovarian cancer (OC). Methods: We analyze information including HRR mutation and clinical prognosis of OC patients both in our cohort and in the TCGA-OV database. Blood and/or tumor samples from 98 women admitted to Shanghai General Hospital between January 2021 and May 2024, and DNA sequencing was performed on these samples for all patients included in this retrospective study. Testing was performed for HRR mutations, including germline BRCA1/2 mutations, and defects in HRR were defined as detrimental mutations within relevant genes. Comprehensive medical records were gathered for all patients, with a follow-up period recorded for 74 of them. Results: HRR pathway genes, including BRCA1/2, CDK12, RAD54L, RAD51, ATM, MRE11, and BRIP2, are highly expressed in FIGO Stages I-II OCs among 482 patients in the TCGA-OV database, and 95.06% samples presented mutations. The alignment diagram analyzed by logistic and Cox regression was derived to investigate HRR genes on overall survival (OS < 763 days) of OC patients. A total of 98 patients were enrolled in our study, with 70 harboring HRR mutations (HRRmt) and 28 having the HRR wild-type (HRRwt). The predominant pathological type across all four patient groups was high-grade serous adenocarcinoma, with similar prevalence in HRRmt (84.30%) versus HRRwt (75%, p=0.360) and BRCAmt (94.20%) versus BRCAwt (74.60%, p=0.151) groups. Survival prediction data were collected from 74 patients, and the HRRmt group (n = 50) exhibited a numerically longer PFS compared to the HRRwt group (n = 24), with 23 months versus 17 months, respectively. A significant disparity was noted in the percentage of patients administered PARPi medication between the HRRmt and HRRwt groups (58.00% vs. 20.20%; p=0.003). Patients in both the HRRmt group (p=0.049) and the BRCAwt group (p=0.046) receiving PARPi treatment have extended PFS. Significant differences were identified between HRRmt and HRRwt groups in the size of the initial debulking surgery achieving R0 status (p=0.005), low CA125 levels (< 1000 U/mL) at diagnosis (p=0.015), and the use of PARP inhibitors (PARPi) (p=0.024) and antiangiogenic drugs (p < 0.001). For patients with HRR mutations, the use of PARPi significantly impacted PFS (p=0.049), and achieving R0 status (p=0.005) significantly influenced PFS. Conclusions: This study indicates that mutations in the HRR gene possess significant potential as a prognostic marker in OC. Our aim was to comprehensively explore how HRR gene mutations, including but not limited to BRCA, might influence the clinical course and survival of patients, shedding light on potential new avenues for personalized treatment strategies.
Collapse
Affiliation(s)
- Shitong Zhan
- Obstetrics and Gynecology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Hongkou, Shanghai 200080, China
| | - Feng Chen
- Obstetrics and Gynecology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Hongkou, Shanghai 200080, China
| | - Lijuan Huang
- Obstetrics and Gynecology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Hongkou, Shanghai 200080, China
| | - Lin Chen
- Obstetrics and Gynecology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Hongkou, Shanghai 200080, China
| | - Haoyi Jia
- Obstetrics and Gynecology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Hongkou, Shanghai 200080, China
| | - Shaofei Ma
- Pathology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Hongkou, Shanghai 200080, China
| | - Min Tang
- Surgery Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Hongkou, Shanghai 200080, China
| | - Chongzhi Zhou
- Educational Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Hongkou, Shanghai 200080, China
| | - Yanmin Chen
- Educational Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Hongkou, Shanghai 200080, China
| | - Ye Yang
- Obstetrics and Gynecology Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 85 Wujin Road, Hongkou, Shanghai 200080, China
| |
Collapse
|
14
|
Xian W, Wang S, Xie J, Yamamoto Y, Khorrami M, Zhang Y, Montes RC, Desales C, Khorrami M, Mory Z, Hoffman A, Su A, Nguyen C, Davies PJA, Stephan C, Pan S, Wu W, Liu Y, Siegelman J, Waters RE, Ross WA, Song S, Metersky M, Beer DG, Crum CP, Stewart AJ, Vincent M, Russell R, Izard RA, Ho KY, Hung-Sen Lai J, Bachovchin WW, Ajani JA, McKeon FD. Evolution of Esophageal Adenocarcinoma From Precursor Lesion Stem Cells. Gastroenterology 2025:S0016-5085(25)00521-9. [PMID: 40090599 DOI: 10.1053/j.gastro.2025.02.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND AND AIMS Metastatic cancers arise from a decades-long succession of increasingly virulent precursor lesions, each of which represents prospective targets for therapeutic intervention. This evolutionary process has been particularly vivid in esophageal adenocarcinoma (EAC), as this cancer and associated precursor lesions, including Barrett's esophagus (BE), low-grade dysplasia (LGD), and high-grade dysplasia (HGD), coexist in an accessible, 2-dimensional pattern in esophageal mucosa. Given the durability of these precursor lesions, it is likely that they, like EAC, rely on stem cells for their regenerative growth. To assess the role of stem cells in the evolution of EAC, we apply technology that selectively clones stem cells from the gastrointestinal tract to patient-matched endoscopic biopsies from each of the precursor lesions implicated in EAC. METHODS Histologically validated, endoscopic biopsy series including EAC, HGD, LGD, BE, and normal esophageal mucosa were obtained from patients presenting with EAC. Rare (1:1000) cells from each of these lesions proved clonogenic and were assessed by in vitro differentiation, tumorigenicity in mice, and by molecular genetics. RESULTS Each of the lesions in the evolution of EAC possesses a discrete set of clonogenic cells marked by immaturity, enormous proliferative potential, and lesion-specific differentiation fate. DNA sequencing of these clones reveals intralesional heterogeneity and clonal resolution of the mutation progression within a given patient from BE, LGD, HGD, and EAC. High-throughput chemical screens against BE stem cells reveal drug combinations that are similarly effective against stem cells of LGD, HGD, and EAC. CONCLUSIONS All lesions in the evolution of EAC possess discrete populations of stem cells that are potential therapeutic targets.
Collapse
Affiliation(s)
- Wa Xian
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Shan Wang
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Jingzhong Xie
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Yusuke Yamamoto
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Melina Khorrami
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Yanting Zhang
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | | | - Caycel Desales
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Melika Khorrami
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Zaal Mory
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Ashley Hoffman
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Amber Su
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Crystal Nguyen
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | | | | | - Shuang Pan
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts
| | - Wengen Wu
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts
| | - Yuxin Liu
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts
| | - Jeremy Siegelman
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Rebecca E Waters
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - William A Ross
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark Metersky
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | - David G Beer
- Departments of Thoracic Surgery and Radiation Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Christopher P Crum
- Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Alexander J Stewart
- School of Mathematics and Statistics, University of St. Andrews, North Haugh, UK
| | | | | | | | - Khek Yu Ho
- Department of Medicine, National University of Singapore, Singapore
| | - Jack Hung-Sen Lai
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts; Department of Developmental, Molecular and Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, Massachusetts
| | - William W Bachovchin
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts; Department of Developmental, Molecular and Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, Massachusetts
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frank D McKeon
- Department of Biology and Biochemistry, University of Houston, Houston, Texas.
| |
Collapse
|
15
|
Pitolli C, Marini A, Sette C, Pagliarini V. Physiological and pathological roles of the transcriptional kinases CDK12 and CDK13 in the central nervous system. Cell Death Differ 2025; 32:371-381. [PMID: 39533070 PMCID: PMC11893892 DOI: 10.1038/s41418-024-01413-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
The cyclin-dependent kinases 12 (CDK12) and 13 (CDK13) govern several steps of gene expression, including transcription, RNA processing and translation. The main target of CDK12/13 is the serine 2 residue of the carboxy-terminal domain of RNA polymerase II (RNAPII), thus influencing the directionality, elongation rate and processivity of the enzyme. The CDK12/13-dependent regulation of RNAPII activity influences the expression of selected target genes with important functional roles in the proliferation and viability of all eukaryotic cells. Neuronal cells are particularly affected by the loss of CDK12/13, as result of the high dependency of neuronal genes on RNAPII processivity for their expression. Deregulation of CDK12/13 activity strongly affects brain physiology by influencing the stemness potential and differentiation properties of neuronal precursor cells. Moreover, mounting evidence also suggest the involvement of CDK12/13 in brain tumours. Herein, we discuss the functional role(s) of CDK12 and CDK13 in gene expression regulation and highlight similarities and differences between these highly homologous kinases, with particular attention to their impact on brain physiology and pathology. Lastly, we provide an overview of CDK12/13 inhibitors and of their efficacy in brain tumours and other neoplastic diseases.
Collapse
Affiliation(s)
- Consuelo Pitolli
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, 00168, Rome, Italy
| | - Alberto Marini
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, 00168, Rome, Italy
- GSTEP-Organoids Research Core Facility, IRCCS Fondazione Policlinico Universitario Agostino Gemelli, 00168, Rome, Italy
- Saint Camillus International University of Health and Medical Sciences, 00131, Rome, Italy
| | - Claudio Sette
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, 00168, Rome, Italy.
- GSTEP-Organoids Research Core Facility, IRCCS Fondazione Policlinico Universitario Agostino Gemelli, 00168, Rome, Italy.
| | - Vittoria Pagliarini
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, 00168, Rome, Italy.
- GSTEP-Organoids Research Core Facility, IRCCS Fondazione Policlinico Universitario Agostino Gemelli, 00168, Rome, Italy.
| |
Collapse
|
16
|
Sasagawa S, Honma Y, Peng X, Maejima K, Nagaoka K, Kobayashi Y, Oosawa A, Johnson TA, Okawa Y, Liang H, Kakimi K, Yamada Y, Nakagawa H. Predicting chemotherapy responsiveness in gastric cancer through machine learning analysis of genome, immune, and neutrophil signatures. Gastric Cancer 2025; 28:228-244. [PMID: 39621213 PMCID: PMC11842519 DOI: 10.1007/s10120-024-01569-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/11/2024] [Indexed: 02/21/2025]
Abstract
BACKGROUND Gastric cancer is a major oncological challenge, ranking highly among causes of cancer-related mortality worldwide. This study was initiated to address the variability in patient responses to combination chemotherapy, highlighting the need for personalized treatment strategies based on genomic data. METHODS We analyzed whole-genome and RNA sequences from biopsy specimens of 65 advanced gastric cancer patients before their chemotherapy treatment. Using machine learning techniques, we developed a model with 123 omics features, such as immune signatures and copy number variations, to predict their chemotherapy outcomes. RESULTS The model demonstrated a prediction accuracy of 70-80% in forecasting chemotherapy responses in both test and validation cohorts. Notably, tumor-associated neutrophils emerged as significant predictors of treatment efficacy. Further single-cell analyses from cancer tissues revealed different neutrophil subgroups with potential antitumor activities suggesting their usefulness as biomarkers for treatment decisions. CONCLUSIONS This study confirms the utility of machine learning in advancing personalized medicine for gastric cancer by identifying tumor-associated neutrophils and their subgroups as key indicators of chemotherapy response. These findings could lead to more tailored and effective treatment plans for patients.
Collapse
Affiliation(s)
- Shota Sasagawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Yoshitaka Honma
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Xinxin Peng
- Precision Scientific (Beijing) Ltd, Beijing, 100085, China
| | - Kazuhiro Maejima
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Koji Nagaoka
- Department of Immunotherapeutics, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Immunology, Faculty of Medicine, Kindai University, Sayama, Osaka, 589-8511, Japan
| | - Yukari Kobayashi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Immunology, Faculty of Medicine, Kindai University, Sayama, Osaka, 589-8511, Japan
| | - Ayako Oosawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Todd A Johnson
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Yuki Okawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Han Liang
- Department of Bioinformatics and Computational Biology, Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Immunology, Faculty of Medicine, Kindai University, Sayama, Osaka, 589-8511, Japan
| | - Yasuhide Yamada
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
- Department of Medical Research, National Center for Global Health and Medicine, Tokyo, 162-8655, Japan
| | - Hidewaki Nakagawa
- Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan.
| |
Collapse
|
17
|
Duan W, Hosea R, Wang L, Ruan C, Zhao F, Liu J, Zhao H, Miyagishi M, Wu S, Kasim V. Chromosome Missegregation Triggers Tumor Cell Pyroptosis and Enhances Anti-Tumor Immunotherapy in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409769. [PMID: 39903759 PMCID: PMC11948012 DOI: 10.1002/advs.202409769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Immune checkpoint inhibitor (ICI) therapy is a promising anti-tumor therapeutic strategy; however, its efficacy in solid tumors is limited. Chromosome missegregation is common in various solid tumors; however, its role in tumor progression remains poorly understood, and its correlation with ICI is yet to be explored. Here, it is found that increased chromosome missegregation promotes tumor immune microenvironment, and eventually immunotherapeutic efficacy, by triggering pyroptosis. yin yang 2 (YY2) is identified as a mitotic checkpoint regulator, which promotes chromosome missegregation by upregulating BUB1B transcription. Increased chromosome missegregation promoted the formation of micronuclei and release of double-stranded DNA (dsDNA) into the cytosol, triggering an AIM2-mediated cytosolic dsDNA response. The subsequent pyroptosis strengthened the tumor immune microenvironment, thereby enhancing immunoinfiltration and cytotoxicity of CD8+ T cells, while preventing their exhaustion. Finally, through in vitro and in vivo experiments, it is demonstrated that combining YY2 overexpression-induced chromosome missegregation/cytosolic dsDNA response and PD-1 inhibitor significantly enhanced the efficacy of ICI immunotherapy in microsatellite instable and microsatellite stable colorectal cancer cells. Together, these findings provide new insights on the role of chromosome missegregation in triggering cytosolic dsDNA response-mediated pyroptosis and modulating the tumor immune microenvironment, suggesting a novel strategy for improving ICI therapeutic efficacy in colorectal cancer.
Collapse
Affiliation(s)
- Wei Duan
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
| | - Rendy Hosea
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
| | - Lingxian Wang
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
| | - Cao Ruan
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
| | - Fuqiang Zhao
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
| | - Jingyi Liu
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
| | - Hezhao Zhao
- Department of Gastrointestinal SurgeryChongqing University Cancer HospitalChongqing UniversityChongqing400030China
| | - Makoto Miyagishi
- Life Science InnovationSchool of Integrative and Global MajorsUniversity of TsukubaTsukubaIbaraki305‐0006Japan
| | - Shourong Wu
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized TreatmentChongqing University Cancer HospitalChongqing UniversityChongqing400030China
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized TreatmentChongqing University Cancer HospitalChongqing UniversityChongqing400030China
| |
Collapse
|
18
|
Braun DA, Moranzoni G, Chea V, McGregor BA, Blass E, Tu CR, Vanasse AP, Forman C, Forman J, Afeyan AB, Schindler NR, Liu Y, Li S, Southard J, Chang SL, Hirsch MS, LeBoeuf NR, Olive O, Mehndiratta A, Greenslade H, Shetty K, Klaeger S, Sarkizova S, Pedersen CB, Mossanen M, Carulli I, Tarren A, Duke-Cohan J, Howard AA, Iorgulescu JB, Shim B, Simon JM, Signoretti S, Aster JC, Elagina L, Carr SA, Leshchiner I, Getz G, Gabriel S, Hacohen N, Olsen LR, Oliveira G, Neuberg DS, Livak KJ, Shukla SA, Fritsch EF, Wu CJ, Keskin DB, Ott PA, Choueiri TK. A neoantigen vaccine generates antitumour immunity in renal cell carcinoma. Nature 2025; 639:474-482. [PMID: 39910301 PMCID: PMC11903305 DOI: 10.1038/s41586-024-08507-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 12/10/2024] [Indexed: 02/07/2025]
Abstract
Personalized cancer vaccines (PCVs) can generate circulating immune responses against predicted neoantigens1-6. However, whether such responses can target cancer driver mutations, lead to immune recognition of a patient's tumour and result in clinical activity are largely unknown. These questions are of particular interest for patients who have tumours with a low mutational burden. Here we conducted a phase I trial (ClinicalTrials.gov identifier NCT02950766) to test a neoantigen-targeting PCV in patients with high-risk, fully resected clear cell renal cell carcinoma (RCC; stage III or IV) with or without ipilimumab administered adjacent to the vaccine. At a median follow-up of 40.2 months after surgery, none of the 9 participants enrolled in the study had a recurrence of RCC. No dose-limiting toxicities were observed. All patients generated T cell immune responses against the PCV antigens, including to RCC driver mutations in VHL, PBRM1, BAP1, KDM5C and PIK3CA. Following vaccination, there was a durable expansion of peripheral T cell clones. Moreover, T cell reactivity against autologous tumours was detected in seven out of nine patients. Our results demonstrate that neoantigen-targeting PCVs in high-risk RCC are highly immunogenic, capable of targeting key driver mutations and can induce antitumour immunity. These observations, in conjunction with the absence of recurrence in all nine vaccinated patients, highlights the promise of PCVs as effective adjuvant therapy in RCC.
Collapse
Affiliation(s)
- David A Braun
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Giorgia Moranzoni
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Vipheaviny Chea
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Bradley A McGregor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Eryn Blass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Chloe R Tu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Allison P Vanasse
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Cleo Forman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Juliet Forman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alexander B Afeyan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nicholas R Schindler
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Yiwen Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shuqiang Li
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jackson Southard
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Steven L Chang
- Harvard Medical School, Boston, MA, USA
- Department of Urology, Brigham and Women's Hospital, Boston, MA, USA
| | - Michelle S Hirsch
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Nicole R LeBoeuf
- Harvard Medical School, Boston, MA, USA
- Center for Cutaneous Oncology, Dana-Farber Brigham and Women's Cancer Center, Boston, MA, USA
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA, USA
| | - Oriol Olive
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ambica Mehndiratta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Haley Greenslade
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Keerthi Shetty
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Susan Klaeger
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Christina B Pedersen
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- Center for Genomic Medicine, Rigshospitalet-Copenhagen University Hospital, Copenhagen, Denmark
| | - Matthew Mossanen
- Harvard Medical School, Boston, MA, USA
- Department of Urology, Brigham and Women's Hospital, Boston, MA, USA
| | - Isabel Carulli
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Anna Tarren
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Joseph Duke-Cohan
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alexis A Howard
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - J Bryan Iorgulescu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Molecular Diagnostics Laboratory, Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bohoon Shim
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jeremy M Simon
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Sabina Signoretti
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jon C Aster
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ignaty Leshchiner
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, USA
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | | | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Lars R Olsen
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Donna S Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kenneth J Livak
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sachet A Shukla
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edward F Fritsch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Derin B Keskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Computer Science, Metropolitan College, Boston University, Boston, MA, USA
| | - Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Bökenkamp JE, Keuper K, Redel S, Barthel K, Johnson L, Becker A, Wieland A, Räschle M, Storchová Z. Proteogenomic analysis reveals adaptive strategies for alleviating the consequences of aneuploidy in cancer. EMBO J 2025; 44:1829-1865. [PMID: 39930267 PMCID: PMC11914506 DOI: 10.1038/s44318-025-00372-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 03/19/2025] Open
Abstract
Aneuploidy is prevalent in cancer and associates with fitness advantage and poor patient prognosis. Yet, experimentally induced aneuploidy initially leads to adverse effects and impaired proliferation, suggesting that cancer cells must adapt to aneuploidy. We performed in vitro evolution of cells with extra chromosomes and obtained cell lines with improved proliferation and gene expression changes congruent with changes in aneuploid cancers. Integrated analysis of cancer multi-omics data and model cells revealed increased expression of DNA replicative and repair factors, reduced genomic instability, and reduced lysosomal degradation. We identified E2F4 and FOXM1 as transcription factors strongly associated with adaptation to aneuploidy in vitro and in cancers and validated this finding. The adaptation to aneuploidy also coincided with specific copy number aberrations that correlate with poor patient prognosis. Chromosomal engineering mimicking these aberrations improved aneuploid cell proliferation, while loss of previously present extra chromosomes impaired it. The identified common adaptation strategies suggest replication stress, genomic instability, and lysosomal stress as common liabilities of aneuploid cancers.
Collapse
Affiliation(s)
- Jan-Eric Bökenkamp
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Kristina Keuper
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
- Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Stefan Redel
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Karen Barthel
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Leah Johnson
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Amelie Becker
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Angela Wieland
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Markus Räschle
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Zuzana Storchová
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany.
| |
Collapse
|
20
|
Wu Y, Chen M, Qin Y. Anticancer drug response prediction integrating multi-omics pathway-based difference features and multiple deep learning techniques. PLoS Comput Biol 2025; 21:e1012905. [PMID: 40163555 PMCID: PMC11978092 DOI: 10.1371/journal.pcbi.1012905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 04/08/2025] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Individualized prediction of cancer drug sensitivity is of vital importance in precision medicine. While numerous predictive methodologies for cancer drug response have been proposed, the precise prediction of an individual patient's response to drug and a thorough understanding of differences in drug responses among individuals continue to pose significant challenges. This study introduced a deep learning model PASO, which integrated transformer encoder, multi-scale convolutional networks and attention mechanisms to predict the sensitivity of cell lines to anticancer drugs, based on the omics data of cell lines and the SMILES representations of drug molecules. First, we use statistical methods to compute the differences in gene expression, gene mutation, and gene copy number variations between within and outside biological pathways, and utilized these pathway difference values as cell line features, combined with the drugs' SMILES chemical structure information as inputs to the model. Then the model integrates various deep learning technologies multi-scale convolutional networks and transformer encoder to extract the properties of drug molecules from different perspectives, while an attention network is devoted to learning complex interactions between the omics features of cell lines and the aforementioned properties of drug molecules. Finally, a multilayer perceptron (MLP) outputs the final predictions of drug response. Our model exhibits higher accuracy in predicting the sensitivity to anticancer drugs comparing with other methods proposed recently. It is found that PARP inhibitors, and Topoisomerase I inhibitors were particularly sensitive to SCLC when analyzing the drug response predictions for lung cancer cell lines. Additionally, the model is capable of highlighting biological pathways related to cancer and accurately capturing critical parts of the drug's chemical structure. We also validated the model's clinical utility using clinical data from The Cancer Genome Atlas. In summary, the PASO model suggests potential as a robust support in individualized cancer treatment. Our methods are implemented in Python and are freely available from GitHub (https://github.com/queryang/PASO).
Collapse
Affiliation(s)
- Yang Wu
- College of Information Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Fisheries Information Ministry of Agriculture, Shanghai, China
| | - Ming Chen
- College of Information Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Fisheries Information Ministry of Agriculture, Shanghai, China
| | - Yufang Qin
- College of Information Technology, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Fisheries Information Ministry of Agriculture, Shanghai, China
| |
Collapse
|
21
|
Blohmer M, Cheek DM, Hung WT, Kessler M, Chatzidimitriou F, Wang J, Hung W, Lee IH, Gorelick AN, Wassenaar EC, Yang CY, Yeh YC, Ho HL, Speiser D, Karsten MM, Lanuti M, Pai SI, Kranenburg O, Lennerz JK, Chou TY, Kloor M, Naxerova K. Quantifying cell divisions along evolutionary lineages in cancer. Nat Genet 2025; 57:706-717. [PMID: 39905260 DOI: 10.1038/s41588-025-02078-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 01/07/2025] [Indexed: 02/06/2025]
Abstract
Cell division drives somatic evolution but is challenging to quantify. We developed a framework to count cell divisions with DNA replication-related mutations in polyguanine homopolymers. Analyzing 505 samples from 37 patients, we studied the milestones of colorectal cancer evolution. Primary tumors diversify at ~250 divisions from the founder cell, while distant metastasis divergence occurs significantly later, at ~500 divisions. Notably, distant but not lymph node metastases originate from primary tumor regions that have undergone surplus divisions, tying subclonal expansion to metastatic capacity. Then, we analyzed a cohort of 73 multifocal lung cancers and showed that the cell division burden of the tumors' common ancestor distinguishes independent primary tumors from intrapulmonary metastases and correlates with patient survival. In lung cancer too, metastatic capacity is tied to more extensive proliferation. The cell division history of human cancers is easily accessible using our simple framework and contains valuable biological and clinical information.
Collapse
Affiliation(s)
- Martin Blohmer
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
- Department of Gynecology with Breast Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - David M Cheek
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Wei-Ting Hung
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, Taiwan
| | - Maria Kessler
- Department of Applied Tumor Biology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Foivos Chatzidimitriou
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Jiahe Wang
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - William Hung
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - I-Hsiu Lee
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Alexander N Gorelick
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
| | - Emma Ce Wassenaar
- Department of Surgery, St. Antonius Hospital, Nieuwegein, the Netherlands
- Department of Surgical Oncology, Laboratory Translational Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ching-Yeuh Yang
- Department of Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yi-Chen Yeh
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiang-Ling Ho
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Dorothee Speiser
- Department of Gynecology with Breast Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Maria M Karsten
- Department of Gynecology with Breast Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Lanuti
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Sara I Pai
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Onno Kranenburg
- Department of Surgical Oncology, Laboratory Translational Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jochen K Lennerz
- Department of Pathology, Center for Integrated Diagnostics, Massachusetts General Hospital, Boston, MA, USA
| | - Teh-Ying Chou
- Department of Pathology and Precision Medicine Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Kamila Naxerova
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Witz A, Dardare J, Betz M, Michel C, Husson M, Gilson P, Merlin JL, Harlé A. Homologous recombination deficiency (HRD) testing landscape: clinical applications and technical validation for routine diagnostics. Biomark Res 2025; 13:31. [PMID: 39985088 PMCID: PMC11846297 DOI: 10.1186/s40364-025-00740-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 02/04/2025] [Indexed: 02/24/2025] Open
Abstract
The use of poly(ADP-ribose) polymerase inhibitors (PARPi) revolutionized the treatment of BRCA-mutated cancers. Identifying patients exhibiting homologous recombination deficiency (HRD) has been proved useful to predict PARPi efficacy. However, obtaining HRD status remains an arduous task due to its evolution over the time. This causes HRD status to become obsolete when obtained from genomic scars, rendering PARPi ineffective for these patients. Only two HRD tests are currently FDA-approved, both based on genomic scars detection and BRCA mutations testing. Nevertheless, new technologies for obtaining an increasingly reliable HRD status continue to evolve. Application of these tests in clinical practice is an additional challenge due to the need for lower costs and shorter time to results delay.In this review, we describe the currently available methods for HRD testing, including the methodologies and corresponding tests for assessing HRD status, and discuss the clinical routine application of these tests and their technical validation.
Collapse
Affiliation(s)
- Andréa Witz
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France.
| | - Julie Dardare
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Margaux Betz
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Cassandra Michel
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Marie Husson
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Pauline Gilson
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Jean-Louis Merlin
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Alexandre Harlé
- Département de Biopathologie, Institut de Cancérologie de Lorraine, CNRS UMR 7039 CRAN - Université de Lorraine, Vandoeuvre-lès-Nancy, France
| |
Collapse
|
23
|
Turova P, Kushnarev V, Baranov O, Butusova A, Menshikova S, Yong ST, Nadiryan A, Antysheva Z, Khorkova S, Guryleva MV, Bagaev A, Lennerz JK, Chernyshov K, Kotlov N. The Breast Cancer Classifier refines molecular breast cancer classification to delineate the HER2-low subtype. NPJ Breast Cancer 2025; 11:19. [PMID: 39979291 PMCID: PMC11842814 DOI: 10.1038/s41523-025-00723-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 01/19/2025] [Indexed: 02/22/2025] Open
Abstract
Current breast cancer classification methods, particularly immunohistochemistry and PAM50, face challenges in accurately characterizing the HER2-low subtype, a therapeutically relevant entity with distinct biological features. This notable gap can lead to misclassification, resulting in inappropriate treatment decisions and suboptimal patient outcomes. Leveraging RNA-seq and machine-learning algorithms, we developed the Breast Cancer Classifier (BCC), a unique transcriptomic classifier for more precise breast cancer subtyping, specifically by delineating and incorporating HER2-low as a distinct subtype. BCC also redefined the PAM50 Normal subtype into other subtypes, disputing its classification as a unique molecular group. Our statistical analysis not only confirmed the reproducibility and accuracy of BCC, but also revealed similarities in prognostic characteristics between the HER2-low and Basal subtypes. Addressing this gap in breast cancer classification is clinically significant because it not only improves treatment stratification, but also uncovers novel molecular and immunohistochemical features associated with the HER2-low and HER2-high subtypes, thereby advancing our understanding of breast cancer heterogeneity and providing guidance in precision oncology.
Collapse
|
24
|
Roma L, Lorber T, Rau S, Barrett MT, Ercan C, Panebianco F, Piscuoglio S, Glatz K, Bubendorf L, Ruiz C. Tracking the Evolution of Cutaneous Melanoma by Multiparameter Flow Sorting and Genomic Profiling. Int J Mol Sci 2025; 26:1758. [PMID: 40004220 PMCID: PMC11855598 DOI: 10.3390/ijms26041758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Intratumoral heterogeneity and clonal evolution are pivotal in the progression and metastasis of melanoma. However, when combined with variable tumor cellularity, intratumoral heterogeneity limits the sensitivity and accuracy of uncovering a cancer's clonal evolution. In this study, we combined fluorescence-activated cell sorting (FACS) with whole-exome sequencing (WES) to investigate the clonal composition and evolutionary patterns in seven melanoma biopsies obtained from three patients, each having both primary site and metastatic samples. We employed a multiparameter ploidy sorting approach to isolate tumor populations based on DNA ploidy and melanoma biomarkers (SOX10 or S100), enabling us to investigate clonal evolution with high resolution. Our approach increased the mean tumor purity from 70% (range 19-88%) in unsorted material to 91% (range 87-96%) post-sorting. Our findings revealed significant inter- and intratumor heterogeneity, with one patient exhibiting two genomically distinct clonal tumor populations within a single primary site biopsy, each giving rise to different metastases. Our findings highlight the critical role of intratumoral heterogeneity and clonal evolution in melanoma, especially when analyzing tumor trajectories. The unique combination of multiparameter FACS and WES provides a powerful method for identifying clonal populations and reconstructing clonal evolution. This study provides valuable insights into the clonal architecture of melanoma and lays the groundwork for future research with larger patient groups.
Collapse
Affiliation(s)
- Luca Roma
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, CH-4031 Basel, Switzerland
| | - Thomas Lorber
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, CH-4031 Basel, Switzerland
| | - Sabrina Rau
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, CH-4031 Basel, Switzerland
| | - Michael T. Barrett
- Department of Research, Mayo Clinic in Arizona, Scottsdale, AZ 85259, USA
| | - Caner Ercan
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, CH-4031 Basel, Switzerland
| | - Federica Panebianco
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, CH-4031 Basel, Switzerland
| | | | - Katharina Glatz
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, CH-4031 Basel, Switzerland
| | - Lukas Bubendorf
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, CH-4031 Basel, Switzerland
| | - Christian Ruiz
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, CH-4031 Basel, Switzerland
| |
Collapse
|
25
|
Liu F, Zhang T, Yang Y, Wang K, Wei J, Shi JH, Zhang D, Sheng X, Zhang Y, Zhou J, Zhao F. Integrated analysis of single-cell and bulk transcriptomics reveals cellular subtypes and molecular features associated with osteosarcoma prognosis. BMC Cancer 2025; 25:280. [PMID: 39962461 PMCID: PMC11834279 DOI: 10.1186/s12885-025-13714-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Osteosarcoma (OS) is the most common primary bone malignancy with variable molecular biology and prognosis. However, our understanding of the association between cell types and OS progression remains poor. METHODS We generated a human OS cell atlas by integrating over 110,000 single cells from 17 samples. Multiple machine learning algorithms were applied to develop tumor purity prediction models based on transcriptomic profile of OS. The Scissor algorithm and gene enrichment analyses were conducted to delve into cell-intrinsic molecular characteristics linked to OS prognosis. Moreover, the study investigated the impact of ATF6α in OS aggressiveness through genetic and pharmacological loss of function analyses. Lastly, the CellChat algorithm was employed to investigate cell-cell communications. RESULTS Utilizing the high-quality human OS cell atlas, we identified tumor purity as a prognostic indicator and developed a robust tumor purity prediction model. We respectively delineated cancer cell- and immune cell-intrinsic molecular characteristics associated with OS prognosis at single-cell resolution. Interestingly, tumor cells with activated unfolded protein response (UPR) pathway were significantly associated with disease aggressiveness. Notably, ATF6α emerged as the top-activated transcription factor for this tumor subcluster. Subsequently, we confirmed that ATF6α was markedly associated with OS progression, while both genetic and pharmacological inhibition of ATF6α impaired the survival of HOS cells. Lastly, we depicted the landscape of signal crosstalk between the UPR-related subcluster and other cell types within the tumor microenvironment. CONCLUSION In summary, our work provides novel insights into the molecular biology of OS, and offers valuable resource for OS biomarker discovery and treatment strategy development.
Collapse
Affiliation(s)
- Feng Liu
- Department of Hand/Foot/Ankle Surgery, Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655099, China
| | - Tingting Zhang
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Yongqiang Yang
- Department of Hand/Foot/Ankle Surgery, Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655099, China
| | - Kailun Wang
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Jinlan Wei
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Ji-Hua Shi
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Dong Zhang
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250021, China
| | - Xia Sheng
- Department of Hand/Foot/Ankle Surgery, Qujing Affiliated Hospital of Kunming Medical University, Qujing, 655099, China
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Yi Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Jing Zhou
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Faming Zhao
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China.
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
26
|
Schiantarelli J, Benamar M, Park J, Sax HE, Oliveira G, Bosma-Moody A, Campbell KM, Liu D, Johnson DB, Rodig S, Wu CJ, Hodi FS, Ribas A, Van Allen E, Haq R. Genomic mediators of acquired resistance to immunotherapy in metastatic melanoma. Cancer Cell 2025; 43:308-316.e6. [PMID: 39933900 DOI: 10.1016/j.ccell.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 10/21/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025]
Abstract
Although some patients with metastatic melanoma experience durable responses to immune checkpoint inhibitors (ICIs), most exhibit intrinsic or acquired resistance to these therapies. Here, we compare somatic genomic profiles from matched pre-treatment and post-resistance tumor biopsies in patients (n = 25) with metastatic melanoma who exhibited heterogeneous ICI responses to nominate additional mediators of acquired resistance. We find that several acquired resistance tumors exhibit defects in B2M or JAK1/2, consistent with prior findings. We also discover resistance-associated mutations in SEC24C and SEC24D in 3 patients. SEC24 has an essential role in the trafficking of the dsDNA sensor STING and has been linked to interferonopathies. Melanoma cells engineered to express the SEC24C mutations observed in patients exhibit diminished STING signaling, including decreased type I interferon production, antigen presentation, and a reduced capacity to activate cytotoxic T cells. This study nominates a role for aberrant STING trafficking in acquired resistance to ICIs.
Collapse
Affiliation(s)
- Julia Schiantarelli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Mouadh Benamar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Jihye Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Haley E Sax
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Alice Bosma-Moody
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Katie M Campbell
- Division of Hematology/Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - David Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | | | - Scott Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Antoni Ribas
- Division of Hematology/Oncology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Eliezer Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| | - Rizwan Haq
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
27
|
Chih YC, Dietsch AC, Koopmann P, Ma X, Agardy DA, Zhao B, De Roia A, Kourtesakis A, Kilian M, Krämer C, Suwala AK, Stenzinger M, Boenig H, Blum A, Pienkowski VM, Aman K, Becker JP, Feldmann H, Bunse T, Harbottle R, Riemer AB, Liu HK, Etminan N, Sahm F, Ratliff M, Wick W, Platten M, Green EW, Bunse L. Vaccine-induced T cell receptor T cell therapy targeting a glioblastoma stemness antigen. Nat Commun 2025; 16:1262. [PMID: 39893177 PMCID: PMC11787355 DOI: 10.1038/s41467-025-56547-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025] Open
Abstract
T cell receptor-engineered T cells (TCR-T) could be advantageous in glioblastoma by allowing safe and ubiquitous targeting of the glioblastoma-derived peptidome. Protein tyrosine phosphatase receptor type Z1 (PTPRZ1), is a clinically targetable glioblastoma antigen associated with glioblastoma cell stemness. Here, we identify a therapeutic HLA-A*02-restricted PTPRZ1-reactive TCR retrieved from a vaccinated glioblastoma patient. Single-cell sequencing of primary brain tumors shows PTPRZ1 overexpression in malignant cells, especially in glioblastoma stem cells (GSCs) and astrocyte-like cells. The validated vaccine-induced TCR recognizes the endogenously processed antigen without off-target cross-reactivity. PTPRZ1-specific TCR-T (PTPRZ1-TCR-T) kill target cells antigen-specifically, and in murine experimental brain tumors, their combined intravenous and intracerebroventricular administration is efficacious. PTPRZ1-TCR-T maintain stem cell memory phenotype in vitro and in vivo and lyse all examined HLA-A*02+ primary glioblastoma cell lines with a preference for GSCs and astrocyte-like cells. In summary, we demonstrate the proof of principle to employ TCR-T to treat glioblastoma.
Collapse
MESH Headings
- Glioblastoma/immunology
- Glioblastoma/therapy
- Glioblastoma/pathology
- Glioblastoma/genetics
- Humans
- Animals
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Mice
- Brain Neoplasms/immunology
- Brain Neoplasms/therapy
- Brain Neoplasms/pathology
- Cell Line, Tumor
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
- Cancer Vaccines/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Antigens, Neoplasm/immunology
- Receptor-Like Protein Tyrosine Phosphatases, Class 5/immunology
- Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics
- Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism
- HLA-A2 Antigen/immunology
- Immunotherapy, Adoptive/methods
- Xenograft Model Antitumor Assays
- Female
Collapse
Grants
- Swiss Cancer Foundation (Swiss Bridge Award), the Else Kröner Fresenius Foundation (2019_EKMS.49), the University Heidelberg Foundation (Hella Buühler Award), the DFG (German Research Foundation), project 404521405 (SFB1389 UNITE Glioblastoma B03), the DKFZ Hector institute (T-SIRE), the Hertie Foundation, the University of Heidelberg, ExploreTech! the DKTK Joint Funding AMI2GO, the Rolf Schwiete Foundation (2021-009), the HI-TRON strategy project PACESSETTING, the DKTK Joint Funding Program INNOVATION INVENT4GB.
- The DFG, project 404521405 (SFB1389 UNITE Glioblastoma B01) the DKTK Joint Funding AMI2GO, the Rolf Schwiete Foundation (2021-009), the HI-TRON strategy project PACESSETTING, the DKTK Joint Funding Program INNOVATION INVENT4GB.
Collapse
Affiliation(s)
- Yu-Chan Chih
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
| | - Amelie C Dietsch
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
| | - Philipp Koopmann
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
| | - Xiujian Ma
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Division of Molecular Neurogenetics, DKFZ, DKFZ-ZMBH alliance, Heidelberg, Germany
| | - Dennis A Agardy
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
| | - Binghao Zhao
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
| | - Alice De Roia
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
- DNA Vector Laboratory, DKFZ, Heidelberg, Germany
| | - Alexandros Kourtesakis
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany
- CCU Neurooncology, DKFZ, Heidelberg, Germany
| | - Michael Kilian
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christopher Krämer
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
| | - Abigail K Suwala
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Institute for Pathology, Department of Neuropathology, Heidelberg University, Heidelberg, Germany
- CCU Neuropathology, DKFZ, Heidelberg, Germany
| | - Miriam Stenzinger
- Institute for Clinical Transfusion Medicine and Cell Therapy, Heidelberg, Germany
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Halvard Boenig
- Faculty of Medicine, Goethe University, Frankfurt a.M., Frankfurt, Germany
- Institute for Transfusion Medicine and Immunohematology, German Red Cross Blood Service Baden-Württemberg-Hessen, Frankfurt a.M., Frankfurt, Germany
| | | | | | - Kuralay Aman
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
| | - Jonas P Becker
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Division of Immunotherapy and Immunoprevention, DKFZ, Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), partner site Heidelberg, Heidelberg, Germany
| | - Henrike Feldmann
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
| | - Theresa Bunse
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
| | - Richard Harbottle
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- DNA Vector Laboratory, DKFZ, Heidelberg, Germany
| | - Angelika B Riemer
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Division of Immunotherapy and Immunoprevention, DKFZ, Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), partner site Heidelberg, Heidelberg, Germany
| | - Hai-Kun Liu
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Division of Molecular Neurogenetics, DKFZ, DKFZ-ZMBH alliance, Heidelberg, Germany
| | - Nima Etminan
- Department of Neurosurgery, University Hospital Mannheim, Mannheim, Germany
| | - Felix Sahm
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Institute for Pathology, Department of Neuropathology, Heidelberg University, Heidelberg, Germany
- CCU Neuropathology, DKFZ, Heidelberg, Germany
| | - Miriam Ratliff
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- CCU Neurooncology, DKFZ, Heidelberg, Germany
- Department of Neurosurgery, University Hospital Mannheim, Mannheim, Germany
| | - Wolfgang Wick
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Neurology Clinic, Heidelberg University Hospital, Heidelberg, Germany
- CCU Neurooncology, DKFZ, Heidelberg, Germany
| | - Michael Platten
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany
- Immune Monitoring Unit, National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Helmholtz Institute for Translational Oncology Mainz (HI-TRON Mainz) - A Helmholtz Institute of the DKFZ, Mainz, Germany
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Edward W Green
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany
| | - Lukas Bunse
- Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Cancer Consortium (DKTK), DKFZ, core center Heidelberg, Heidelberg, Germany.
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translation Neuroscience (MCTN), Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
| |
Collapse
|
28
|
Jammihal T, Saliby RM, Labaki C, Soulati H, Gallegos J, Peris A, McCurry D, Yu C, Shah V, Poduval D, El Zarif T, El Ahmar N, Laimon YN, Eid M, Sheshdeh AB, Krajewski KM, Büttner FA, Schwab M, Heng D, Casellas RC, Rai K, Zacharias Millward NM, Msaouel P, Karam J, Signoretti S, Van Allen E, Choueiri TK, Braun DA, Shukla SA. Immunogenomic determinants of exceptional response to immune checkpoint inhibition in renal cell carcinoma. NATURE CANCER 2025; 6:372-384. [PMID: 39789182 DOI: 10.1038/s43018-024-00896-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 12/10/2024] [Indexed: 01/12/2025]
Abstract
Immune checkpoint inhibitors can lead to 'exceptional', durable responses in a subset of persons. However, the molecular basis of exceptional response (ER) to immunotherapy in metastatic clear cell renal cell carcinoma (mccRCC) has not been well characterized. Here we analyzed pretherapy genomic and transcriptomic data in treatment-naive persons with mccRCC treated with standard-of-care immunotherapies: (1) combination of programmed cell death protein and ligand 1 (PD1/PDL1) and cytotoxic T lymphocyte-associated protein 4 inhibitors (IO/IO) or (2) combination of PD1/PDL1 and vascular endothelial growth factor (VEGF) receptor inhibitors (IO/VEGF). In the IO/IO cohort, clonal neoantigen load was significantly higher in persons with ER. In the IO/VEGF cohort, ER participants displayed strong enrichment of B cell receptor signaling-related pathways, tertiary lymphoid structure (TLS) signatures and evidence of increased metabolic activity. Our results suggest that ER may be related to clonal neoantigen-driven cytotoxic T cell responses and TLS formation in tumor microenvironments. Therapeutic combinations that elicit both T cell-directed and B cell-directed antitumor immunity may be important to achieve exceptional benefit to IO-based treatment in ccRCC.
Collapse
Affiliation(s)
- Tejas Jammihal
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Renee Maria Saliby
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Yale Center of Cellular and Molecular Oncology, Yale School of Medicine, New Haven, CT, USA
| | - Chris Labaki
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hanna Soulati
- Yale Center of Cellular and Molecular Oncology, Yale School of Medicine, New Haven, CT, USA
| | - Juan Gallegos
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Arnau Peris
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dustin McCurry
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chunlei Yu
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Valisha Shah
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Deepak Poduval
- Yale Center of Cellular and Molecular Oncology, Yale School of Medicine, New Haven, CT, USA
| | - Talal El Zarif
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Nourhan El Ahmar
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yasmin Nabil Laimon
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marc Eid
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Aseman Bagheri Sheshdeh
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Katherine M Krajewski
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Florian A Büttner
- Dr. Margarete Fischer-Bosch-Institut of Clinical Pharmacology, Stuttgart, Germany
- Departments of Clinical Pharmacology, and of Biochemistry and Pharmacy, University Tübingen, Tübingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institut of Clinical Pharmacology, Stuttgart, Germany
- Departments of Clinical Pharmacology, and of Biochemistry and Pharmacy, University Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) 'Image-Guided and Functionally Instructed Tumor Therapies', University Tübingen, Tübingen, Germany
| | - Daniel Heng
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | - Rafael C Casellas
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kunal Rai
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Niki M Zacharias Millward
- Department of Urology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jose Karam
- Department of Urology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Eliezer Van Allen
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Toni K Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - David A Braun
- Yale Center of Cellular and Molecular Oncology, Yale School of Medicine, New Haven, CT, USA.
| | - Sachet A Shukla
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
29
|
Schoenpflug LA, Chatzipli A, Sirinukunwattana K, Richman S, Blake A, Robineau J, Mertz KD, Verrill C, Leedham SJ, Hardy C, Whalley C, Redmond K, Dunne P, Walker S, Beggs AD, McDermott U, Murray GI, Samuel LM, Seymour M, Tomlinson I, Quirke P, Rittscher J, Maughan T, Domingo E, Koelzer VH. Tumour purity assessment with deep learning in colorectal cancer and impact on molecular analysis. J Pathol 2025; 265:184-197. [PMID: 39710952 PMCID: PMC11717495 DOI: 10.1002/path.6376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/11/2024] [Accepted: 10/29/2024] [Indexed: 12/24/2024]
Abstract
Tumour content plays a pivotal role in directing the bioinformatic analysis of molecular profiles such as copy number variation (CNV). In clinical application, tumour purity estimation (TPE) is achieved either through visual pathological review [conventional pathology (CP)] or the deconvolution of molecular data. While CP provides a direct measurement, it demonstrates modest reproducibility and lacks standardisation. Conversely, deconvolution methods offer an indirect assessment with uncertain accuracy, underscoring the necessity for innovative approaches. SoftCTM is an open-source, multiorgan deep-learning (DL) model for the detection of tumour and non-tumour cells in H&E-stained slides, developed within the Overlapped Cell on Tissue Dataset for Histopathology (OCELOT) Challenge 2023. Here, using three large multicentre colorectal cancer (CRC) cohorts (N = 1,097 patients) with digital pathology and multi-omic data, we compare the utility and accuracy of TPE with SoftCTM versus CP and bioinformatic deconvolution methods (RNA expression, DNA methylation) for downstream molecular analysis, including CNV profiling. SoftCTM showed technical repeatability when applied twice on the same slide (r = 1.0) and excellent correlations in paired H&E slides (r > 0.9). TPEs profiled by SoftCTM correlated highly with RNA expression (r = 0.59) and DNA methylation (r = 0.40), while TPEs by CP showed a lower correlation with RNA expression (r = 0.41) and DNA methylation (r = 0.29). We show that CP and deconvolution methods respectively underestimate and overestimate tumour content compared to SoftCTM, resulting in 6-13% differing CNV calls. In summary, TPE with SoftCTM enables reproducibility, automation, and standardisation at single-cell resolution. SoftCTM estimates (M = 58.9%, SD ±16.3%) reconcile the overestimation by molecular data extrapolation (RNA expression: M = 79.2%, SD ±10.5, DNA methylation: M = 62.7%, SD ±11.8%) and underestimation by CP (M = 35.9%, SD ±13.1%), providing a more reliable middle ground. A fully integrated computational pathology solution could therefore be used to improve downstream molecular analyses for research and clinics. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Lydia A Schoenpflug
- Department of Pathology and Molecular PathologyUniversity Hospital and University of ZurichZurichSwitzerland
| | | | - Korsuk Sirinukunwattana
- Institute of Biomedical Engineering (IBME), Department of Engineering Science, Old Road Campus Research BuildingUniversity of OxfordOxfordUK
- Li Ka Shing Centre for Health Information and DiscoveryBig Data Institute, University of OxfordOxfordUK
- Oxford NIHR Biomedical Research CentreOxford University Hospitals TrustOxfordUK
- Ground Truth Labs LtdOxfordUK
| | - Susan Richman
- Department of Pathology and Tumour BiologyLeeds Institute of Cancer and PathologyLeedsUK
| | - Andrew Blake
- Department of OncologyUniversity of OxfordOxfordUK
| | | | - Kirsten D Mertz
- Cantonal Hospital BasellandInstitute of PathologyLiestalSwitzerland
- Institute of Medical Genetics and PathologyUniversity Hospital BaselBaselSwitzerland
| | - Clare Verrill
- Li Ka Shing Centre for Health Information and DiscoveryBig Data Institute, University of OxfordOxfordUK
- Department of Cellular PathologyOxford University Hospitals NHS Foundation TrustOxfordUK
- Nuffield Department of Surgical Sciences and NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
| | - Simon J Leedham
- Gastrointestinal Stem‐cell Biology Laboratory, Oxford Centre for Cancer Gene Research, Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUK
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Clinical MedicineJohn Radcliffe HospitalOxfordUK
| | | | - Celina Whalley
- Institute of Cancer and Genomic ScienceUniversity of BirminghamBirminghamUK
| | - Keara Redmond
- The Patrick G Johnston Centre for Cancer ResearchQueens UniversityBelfastUK
| | - Philip Dunne
- The Patrick G Johnston Centre for Cancer ResearchQueens UniversityBelfastUK
| | - Steven Walker
- The Patrick G Johnston Centre for Cancer ResearchQueens UniversityBelfastUK
- Almac DiagnosticsCraigavonUK
| | - Andrew D Beggs
- Institute of Cancer and Genomic ScienceUniversity of BirminghamBirminghamUK
| | | | - Graeme I Murray
- Department of Pathology, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenAberdeenUK
| | - Leslie M Samuel
- Department of Clinical OncologyAberdeen Royal Infirmary, NHS GRAMPIANAberdeenUK
| | - Matthew Seymour
- Department of Pathology and Tumour BiologyLeeds Institute of Cancer and PathologyLeedsUK
| | | | - Philip Quirke
- Department of Pathology and Tumour BiologyLeeds Institute of Cancer and PathologyLeedsUK
| | - Jens Rittscher
- Institute of Biomedical Engineering (IBME), Department of Engineering Science, Old Road Campus Research BuildingUniversity of OxfordOxfordUK
- Li Ka Shing Centre for Health Information and DiscoveryBig Data Institute, University of OxfordOxfordUK
- Oxford NIHR Biomedical Research CentreOxford University Hospitals TrustOxfordUK
- Ground Truth Labs LtdOxfordUK
- Nuffield Department of MedicineLudwig Institute for Cancer Research, University of OxfordOxfordUK
| | - Tim Maughan
- Department of OncologyUniversity of OxfordOxfordUK
- University of LiverpoolLiverpoolUK
| | | | - Viktor H Koelzer
- Department of Pathology and Molecular PathologyUniversity Hospital and University of ZurichZurichSwitzerland
- Department of OncologyUniversity of OxfordOxfordUK
- Institute of Medical Genetics and PathologyUniversity Hospital BaselBaselSwitzerland
- Nuffield Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
30
|
Gardner AL, Zheng L, Howland K, Saunders A, Ramirez A, Parker P, Iloegbunam C, Morgan D, Jost TA, Brock A. Mapping cell-cell fusion at single-cell resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.11.627873. [PMID: 39896473 PMCID: PMC11785005 DOI: 10.1101/2024.12.11.627873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Cell-cell fusion is a tightly controlled process in the human body known to be involved in fertilization, placental development, muscle growth, bone remodeling, and viral response. Fusion between cancer cells results first in a whole-genome doubled state, which may be followed by the generation of aneuploidies; these genomic alterations are known drivers of tumor evolution. The role of cell-cell fusion in cancer progression and treatment response has been understudied due to limited experimental systems for tracking and analyzing individual fusion events. To meet this need, we developed a molecular toolkit to map the origins and outcomes of individual cell fusion events within a tumor cell population. This platform, ClonMapper Duo ('CMDuo'), identifies cells that have undergone cell-cell fusion through a combination of reporter expression and engineered fluorescence-associated index sequences paired to randomly generated nucleotide barcodes. scRNA-seq of the indexed barcodes enables the mapping of each set of parental cells and fusion progeny throughout the cell population. In triple-negative breast cancer cells CMDuo uncovered subclonal transcriptomic hybridization and unveiled distinct cell-states which arise in direct consequence of homotypic cell-cell fusion. CMDuo is a platform that enables mapping of cell-cell fusion events in high-throughput single cell data and enables the study of cell fusion in disease progression and therapeutic response.
Collapse
Affiliation(s)
- Andrea L Gardner
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Lan Zheng
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Kennedy Howland
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Andrew Saunders
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Andrea Ramirez
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Patrik Parker
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Chisom Iloegbunam
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Daylin Morgan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Tyler A Jost
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Amy Brock
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
31
|
Liu B, Xie Y, Zhang Y, Tang G, Lin J, Yuan Z, Liu X, Wang X, Huang M, Luo Y, Yu H. Spatial deconvolution from bulk DNA methylation profiles determines intratumoral epigenetic heterogeneity. Cell Biosci 2025; 15:7. [PMID: 39844296 PMCID: PMC11756021 DOI: 10.1186/s13578-024-01337-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Intratumoral heterogeneity emerges from accumulating genetic and epigenetic changes during tumorigenesis, which may contribute to therapeutic failure and drug resistance. However, the lack of a quick and convenient approach to determine the intratumoral epigenetic heterogeneity (eITH) limit the application of eITH in clinical settings. Here, we aimed to develop a tool that can evaluate the eITH using the DNA methylation profiles from bulk tumors. METHODS Genomic DNA of three laser micro-dissected tumor regions, including digestive tract surface, central bulk, and invasive front, was extracted from formalin-fixed paraffin-embedded sections of colorectal cancer patients. The genome-wide methylation profiles were generated with methylation array. The most variable methylated probes were selected to construct a DNA methylation-based heterogeneity (MeHEG) estimation tool that can deconvolve the proportion of each reference tumor region with the support vector machine model-based method. A PCR-based assay for quantitative analysis of DNA methylation (QASM) was developed to specifically determine the methylation status of each CpG in MeHEG assay at single-base resolution to realize fast evaluation of epigenetic heterogeneity. RESULTS In the discovery set with 79 patients, the differentially methylated CpGs among the three tumor regions were found. The 7 most representative CpGs were identified and subsequently selected to develop the MeHEG algorithm. We validated its performance of deconvolution of tumor regions in an independent cohort. In addition, we showed the significant association of MeHEG-based epigenetic heterogeneity with the genomic heterogeneity in mutation and copy number variation in our in-house and TCGA cohorts. Besides, we found that the patients with higher MeHEG score had worse disease-free and overall survival outcomes. Finally, we found dynamic change of epigenetic heterogeneity based on MeHEG score in cancer cells under the treatment of therapeutic drugs. CONCLUSION By developing a 7-loci panel using a machine learning approach combined with the QASM assay for PCR-based application, we present a valuable method for evaluating intratumoral heterogeneity. The MeHEG algorithm offers novel insights into tumor heterogeneity from an epigenetic perspective, potentially enriching current knowledge of tumor complexity and providing a new tool for clinical and research applications in cancer biology.
Collapse
Affiliation(s)
- Binbin Liu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
| | - Yumo Xie
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
| | - Yu Zhang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
| | - Guannan Tang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
| | - Jinxin Lin
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
| | - Ze Yuan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
| | - Xiaoxia Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Guangzhou, Guangdong, China
- Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaolin Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Guangzhou, Guangdong, China
- Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Meijin Huang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Guangzhou, Guangdong, China
- Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanxin Luo
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Guangzhou, Guangdong, China
- Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huichuan Yu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, Guangdong, China.
- Guangdong Institute of Gastroenterology, Guangzhou, 510655, Guangdong, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, Guangdong, China.
- Ministry of Education, Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Guangzhou, Guangdong, China.
- Innovation Center of the Sixth Affiliated Hospital, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
32
|
Zhang Q, Xu X, Jiang D, Wang Y, Wang H, Zhu J, Tang S, Wang R, Zhao S, Li K, Feng J, Xiang H, Yao Z, Xu N, Fang R, Guo W, Liu Y, Hou Y, Ding C. Integrated proteogenomic characterization of ampullary adenocarcinoma. Cell Discov 2025; 11:2. [PMID: 39762212 PMCID: PMC11704194 DOI: 10.1038/s41421-024-00742-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 09/29/2024] [Indexed: 01/11/2025] Open
Abstract
Ampullary adenocarcinoma (AMPAC) is a rare and heterogeneous malignancy. Here we performed a comprehensive proteogenomic analysis of 198 samples from Chinese AMPAC patients and duodenum patients. Genomic data illustrate that 4q loss causes fatty acid accumulation and cell proliferation. Proteomic analysis has revealed three distinct clusters (C-FAM, C-AD, C-CC), among which the most aggressive cluster, C-AD, is associated with the poorest prognosis and is characterized by focal adhesion. Immune clustering identifies three immune clusters and reveals that immune cluster M1 (macrophage infiltration cluster) and M3 (DC cell infiltration cluster), which exhibit a higher immune score compared to cluster M2 (CD4+ T-cell infiltration cluster), are associated with a poor prognosis due to the potential secretion of IL-6 by tumor cells and its consequential influence. This study provides a comprehensive proteogenomic analysis for seeking for better understanding and potential treatment of AMPAC.
Collapse
Affiliation(s)
- Qiao Zhang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Xiaomeng Xu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Dongxian Jiang
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yunzhi Wang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Haixing Wang
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Jiajun Zhu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Shaoshuai Tang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Ronghua Wang
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of Hematology & Oncology, Shanghai Jiao Tong University, Shanghai, China
| | - Shuang Zhao
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of Hematology & Oncology, Shanghai Jiao Tong University, Shanghai, China
| | - Kai Li
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Jinwen Feng
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Hang Xiang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Zhenmei Yao
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Ning Xu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Rundong Fang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Wenjia Guo
- Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Translational Biomedical Engineering, Urumqi, Xinjiang, China
| | - Yu Liu
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of Hematology & Oncology, Shanghai Jiao Tong University, Shanghai, China.
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital Fudan University, Shanghai, China.
| | - Chen Ding
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200433, China.
- Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Translational Biomedical Engineering, Urumqi, Xinjiang, China.
| |
Collapse
|
33
|
Nadeem O, Aranha MP, Redd R, Timonian M, Magidson S, Lightbody ED, Alberge JB, Bertamini L, Dutta AK, El-Khoury H, Bustoros M, Laubach JP, Bianchi G, O'Donnell E, Wu T, Tsuji J, Anderson KC, Getz G, Trippa L, Richardson PG, Sklavenitis-Pistofidis R, Ghobrial IM. Deeper response predicts better outcomes in high-risk-smoldering-myeloma: results of the I-PRISM phase II clinical trial. Nat Commun 2025; 16:358. [PMID: 39753553 PMCID: PMC11698957 DOI: 10.1038/s41467-024-55308-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/06/2024] [Indexed: 01/06/2025] Open
Abstract
Early therapeutic intervention in high-risk smoldering multiple myeloma (HR-SMM) has shown benefits, however, no studies have assessed whether biochemical progression or response depth predicts long-term outcomes. The single-arm I-PRISM phase II trial (NCT02916771) evaluated ixazomib, lenalidomide, and dexamethasone in 55 patients with HR-SMM. The primary endpoint, median progression-free survival (PFS), was not reached (NR) (95% CI: 57.7-NR, median follow-up 50 months). The secondary endpoint, biochemical PFS, was 48.6 months (95% CI: 39.9-NR) and coincided with or preceded SLiM-CRAB in eight patients. For additional secondary objectives, the overall response rate was 93% with 31% achieving complete response (CR) and 45% very good partial response (VGPR) or better. CR correlated strongly with the absence of SLiM-CRAB and biochemical progression. MRD-negativity (10-5 sensitivity) predicted a 5-year biochemical PFS of 100% versus 40% in MRD-positive patients (p = 0.051), demonstrating that deep responses significantly improve time to progression. Exploratory single-cell RNA sequencing linked tumor MHC class I expression to proteasome inhibitor response, and a lower proportion of GZMB+ T cells within clonally expanded CD8+ T cells associated with suboptimal outcomes.
Collapse
Affiliation(s)
- Omar Nadeem
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Michelle P Aranha
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Robert Redd
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael Timonian
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Sophie Magidson
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Elizabeth D Lightbody
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Jean-Baptiste Alberge
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Luca Bertamini
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Ankit K Dutta
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Habib El-Khoury
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Mark Bustoros
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Hematology and Medical Oncology, Meyer Cancer Center, New York-Presbyterian Hospital, New York, NY, USA
| | - Jacob P Laubach
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Giada Bianchi
- Amyloidosis Program, Division of Hematology, Brigham and Women's Hospital and Dana Farber Cancer Institute, Boston, MA, USA
| | - Elizabeth O'Donnell
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ting Wu
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Junko Tsuji
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Kenneth C Anderson
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gad Getz
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
- Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Lorenzo Trippa
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Paul G Richardson
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Romanos Sklavenitis-Pistofidis
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Irene M Ghobrial
- Center for Early Detection and Interception of Blood Cancers, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Boston, MA, USA.
| |
Collapse
|
34
|
Voutsadakis IA. Molecular alterations in claudin 18 suppressed and non-suppressed gastric adenocarcinomas to guide targeted therapies. Tissue Barriers 2025; 13:2348852. [PMID: 38713052 PMCID: PMC11970779 DOI: 10.1080/21688370.2024.2348852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Gastric adenocarcinoma represents an aggressive type of cancer and an important cause of cancer mortality. Progress in gastric cancer therapeutics has resulted from a better understanding of the molecular pathogenesis of the disease and introduction of targeted therapies, but most gastric cancer patients still rely on non-targeted chemotherapy as the mainstay of treatment for advanced disease. METHODS An analysis of publicly available series from The Cancer Genome Atlas (TCGA) gastric cancer cohort was undertaken to delineate the clinical and genomic landscape of gastric cancers with suppressed expression of claudin 18 compared with cancers with non-suppressed claudin 18. Claudin 18 suppressed cancers were defined as having an mRNA expression z-score relative to normal samples (log RNA Seq V2) of less than -1. Claudin 18 non-suppressed cancers were defined as having an mRNA expression z-score relative to normal samples (log RNA Seq V2) above 0.5. RESULTS Gastric cancers with claudin 18 mRNA suppression represented 7.7% of the gastric adenocarcinomas of TCGA cohort, while non-suppressed cancers represented 46.6% of the cases. The two groups did not differ in clinical and genomic characteristics, such as mean age, histology, grade, and stage. The mutation landscape of claudin 18 suppressed cases included high mutation rates of TP53, of genes of the WNT/β-catenin pathway and of ubiquitin ligase FBXW7. Moreover, a subset of both claudin 18 suppressed and non-suppressed cancers displayed mutations in Mismatch Repair (MMR) associated genes or a high tumor mutation burden (TMB). At the mRNA expression level, claudin 18 suppressed gastric cancers showed up-regulation of EMT core transcription factor Snail 2 and down-regulation of genes of HLA cluster. The survival of gastric cancer patients with claudin 18 mRNA suppression was not significantly different compared with patients with non-suppressed claudin 18. CONCLUSION Sub-sets of gastric cancers with claudin 18 mRNA suppression displayed characteristics of potential therapeutic interest, such as mutations in WNT and PI3K pathways and MMR defects. These may guide the development of alternative targeted therapies, in this sub-set of gastric cancers which are not candidates for claudin 18 targeting therapies.
Collapse
Affiliation(s)
- Ioannis A. Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste Marie, Ontario, Canada
- Division of Clinical Sciences, Section of Internal Medicine, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
35
|
Fathi Kazerooni A, Kraya A, Rathi KS, Kim MC, Vossough A, Khalili N, Familiar AM, Gandhi D, Khalili N, Kesherwani V, Haldar D, Anderson H, Jin R, Mahtabfar A, Bagheri S, Guo Y, Li Q, Huang X, Zhu Y, Sickler A, Lueder MR, Phul S, Koptyra M, Storm PB, Ware JB, Song Y, Davatzikos C, Foster JB, Mueller S, Fisher MJ, Resnick AC, Nabavizadeh A. Multiparametric MRI along with machine learning predicts prognosis and treatment response in pediatric low-grade glioma. Nat Commun 2025; 16:340. [PMID: 39747214 PMCID: PMC11697432 DOI: 10.1038/s41467-024-55659-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
Pediatric low-grade gliomas (pLGGs) exhibit heterogeneous prognoses and variable responses to treatment, leading to tumor progression and adverse outcomes in cases where complete resection is unachievable. Early prediction of treatment responsiveness and suitability for immunotherapy has the potential to improve clinical management and outcomes. Here, we present a radiogenomic analysis of pLGGs, integrating MRI and RNA sequencing data. We identify three immunologically distinct clusters, with one group characterized by increased immune activity and poorer prognosis, indicating potential benefit from immunotherapies. We develop a radiomic signature that predicts these immune profiles with over 80% accuracy. Furthermore, our clinicoradiomic model predicts progression-free survival and correlates with treatment response. We also identify genetic variants and transcriptomic pathways associated with progression risk, highlighting links to tumor growth and immune response. This radiogenomic study in pLGGs provides a framework for the identification of high-risk patients who may benefit from targeted therapies.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Adam Kraya
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Komal S Rathi
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Meen Chul Kim
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Arastoo Vossough
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Radiology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nastaran Khalili
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ariana M Familiar
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Deep Gandhi
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Neda Khalili
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Varun Kesherwani
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Debanjan Haldar
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Hannah Anderson
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Run Jin
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Aria Mahtabfar
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sina Bagheri
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yiran Guo
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Qi Li
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Xiaoyan Huang
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yuankun Zhu
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alex Sickler
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Matthew R Lueder
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Saksham Phul
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mateusz Koptyra
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Phillip B Storm
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jeffrey B Ware
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuanquan Song
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica B Foster
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sabine Mueller
- Department of Neurology and Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Michael J Fisher
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adam C Resnick
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ali Nabavizadeh
- Center for Data-Driven Discovery in Biomedicine (D3b), The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
36
|
Mertens F, Hofvander J, Mandahl N, Mitelman F. Aneuploidy in neoplasia: Single-cell data on 83,862 tumors. Int J Cancer 2025; 156:34-39. [PMID: 39222304 DOI: 10.1002/ijc.35163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/05/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Chromosomal aneuploidy, that is, numerical chromosome aberrations, is one of the molecular hallmarks of cancer. However, when neoplasms are studied with sequencing- and array-based approaches, chromosome numbers and ploidy states are typically inferred from bulk DNA data. Furthermore, published molecular estimates of neoplasia-associated aneuploidy often also include genomic imbalances resulting from various types of structural rearrangement, which likely result from other mechanisms than numerical chromosome aberrations. We thus analyzed chromosome numbers using single-cell cytogenetic data from 83,862 tumors, and show that both benign and malignant tumors are highly heterogeneous with regard to deviations from the normal, diploid state. Focusing on the chromosome numbers in 112 specific tumor types, defined by both exact morphologic diagnosis and organ location and from which data from ≥50 cases were available, we found two major clusters: one predominated by near-diploid neoplasms and one by neoplasms with extensive aneuploidy and one or more whole genome doublings. The former cluster included most benign solid tumors, myeloid neoplasms, and malignant gene fusion-associated solid tumors, whereas the latter was predominated by malignant solid tumors and lymphomas. For 16 malignant tumor types, the distribution of chromosome numbers could be compared to TCGA ploidy level data. Cytogenetic and molecular data correlated well, but the former indicates a higher level of clonal heterogeneity. The results presented here suggest shared pathogenetic mechanisms in certain tumor types and provide a reference for molecular analyses.
Collapse
Affiliation(s)
- Fredrik Mertens
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Department of Clinical Genetics, Pathology, and Molecular Diagnostics, Division of Laboratory Medicine, Lund, Sweden
| | - Jakob Hofvander
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Nils Mandahl
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Felix Mitelman
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
37
|
Darzi M, Shokrollahi-Barough M, Nazeri E, Majidzadeh-A K, Esmaeili R. Gene co-expression network analysis reveals relationship between leukocyte fraction and genomic instability in dedifferentiated liposarcoma. MOLECULAR BIOLOGY RESEARCH COMMUNICATIONS 2025; 14:203-218. [PMID: 40321702 PMCID: PMC12046367 DOI: 10.22099/mbrc.2025.51329.2050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Dedifferentiated Liposarcoma (DDLPS) is one of the common subtypes of liposarcoma that is considered a highly malignant category. This study aims to investigate DDLPS through a system biology approach. The gene expression profiles and clinical traits of the DDLPS were acquired from The Cancer Genome Atlas (TCGA). The identification of co-expressed modules was conducted using the weighted gene co-expression network analysis. The immune cell-related gene function was studied by a web-based tool, TIMER, and, the survival analysis was performed at both the module and single-gene levels through Cox Regression analysis. Gene enrichment analysis was also conducted using the DAVID tool. One of the nine co-expressed DDLPS modules was significantly correlated with leukocyte fraction, hyper/hypo methylation, tumor purity, and chromosome instability (CIN). Based on the biological processes used to classify genes, the hub genes in a particular module play important roles in DNA repair, microtubule organizing clusters, mitotic checkpoint dysregulation, and cell proliferation signaling pathways. After screening the genes based on intra-module connectivity, module membership, and gene significance RAD54L was selected as one of the important hub genes. RAD54L showed poor prognosis to the overall survival (OS) analysis (HR=1.6, 95% CI=1.1-2.4, p=0.02). No co-expressed modules had relationship with OS. Through DDLPS traits, CIN and hyper/hypo methylation had significant negative relationship with OS. Our achievement confirmed the inverse association between tumor purity for DDLPS gene profiles and leukocyte fraction and negative leukocyte fraction (LF) gene significance in some genes was justified according to the sub-population analyses of immune cells in TIMER.
Collapse
Affiliation(s)
- Mohammad Darzi
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Medical Informatics Research Group, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mahdieh Shokrollahi-Barough
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Elahe Nazeri
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Keivan Majidzadeh-A
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Rezvan Esmaeili
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
38
|
Pan H, Zhang H, Zhang Y, Chen X, Liu Z, Wu Y, Bai N, Shi Y, Zhao M, Zhu L. Genetic profile in primary tumor tissue of advanced lung adenocarcinoma patients with adrenal metastasis. Cancer Genet 2025; 290-291:36-43. [PMID: 39673828 DOI: 10.1016/j.cancergen.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/07/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
The aim of this study was to examine the genomic characteristics and explore the molecular mechanisms underlying adrenal metastases in lung adenocarcinoma. 57 patients diagnosed with lung adenocarcinoma (LUAD) and adrenal metastases (AM) were enrolled, alongside 33 controls diagnosed with non-adrenal metastases (non-AM) at the time of diagnosis. The primary lung cancer tissue sample were analyzed using next-generation sequencing. The molecular and clinical features were correlated with clinical outcomes. TP53, EGFR and KRAS were the most frequently mutated gene in both groups. EGFR mutations, especially rare variants (G724A, L747P, Q701 L, G719C, V769 L and S768I), exhibited significant enrichment in the non-AM group (P<0.001). An elevated age-related signature in the group of patients with AM, whereas the non-AM group exhibited a higher BRCA signature. Potential prognostic biomarkers such as KEAP1, LRP1B, NOTCH1 and RET mutations were detected in the non-AM group, while ALK mutations in the AM group correlated with shorter overall survival (P<0.001). KRAS mutations in the early synchronous adrenal metastases group were also associated with shorter OS (P<0.001). The analysis of 425 tumor genes in 29 patients with adrenal metastases showed significant enrichment in pathways associated with invasion and metastasis, including TNF signaling pathway and TGF-β signaling pathway. Patients without EGFR mutations in LUAD need to be more concerned about adrenal metastases. Meanwhile, patients with adrenal metastases harboring ALK or KRAS mutations have a poor prognosis and require more aggressive treatment. The TNF and TGF-β pathways might be associated with adrenal metastasis.
Collapse
Affiliation(s)
- Haiqiao Pan
- Chengde Medical University, Chengde, Hebei 067000, PR China; Department of Oncology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei 050000, PR China
| | - Hongbin Zhang
- Department of Oncology, Hebei Chest Hospital, Shijiazhuang, Hebei 050000, PR China
| | - Yongqian Zhang
- Department of Oncology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei 050000, PR China
| | - Xiaojing Chen
- Department of Oncology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei 050000, PR China; Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Zhai Liu
- Department of Radiotherapy, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Yajing Wu
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Na Bai
- Geneseeq Research Institute, Geneseeq Technology Inc., Nanjing, Jiangsu 210000, PR China
| | - Yan Shi
- Geneseeq Research Institute, Geneseeq Technology Inc., Nanjing, Jiangsu 210000, PR China
| | - Min Zhao
- Department of Oncology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei 050000, PR China.
| | - Lingling Zhu
- Department of Oncology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei 050000, PR China.
| |
Collapse
|
39
|
Lu WT, Zalmas LP, Bailey C, Black JRM, Martinez-Ruiz C, Pich O, Gimeno-Valiente F, Usaite I, Magness A, Thol K, Webber TA, Jiang M, Saunders RE, Liu YH, Biswas D, Ige EO, Aerne B, Grönroos E, Venkatesan S, Stavrou G, Karasaki T, Al Bakir M, Renshaw M, Xu H, Schneider-Luftman D, Sharma N, Tovini L, Jamal-Hanjani M, McClelland SE, Litchfield K, Birkbak NJ, Howell M, Tapon N, Fugger K, McGranahan N, Bartek J, Kanu N, Swanton C. TRACERx analysis identifies a role for FAT1 in regulating chromosomal instability and whole-genome doubling via Hippo signalling. Nat Cell Biol 2025; 27:154-168. [PMID: 39738653 PMCID: PMC11735399 DOI: 10.1038/s41556-024-01558-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/16/2024] [Indexed: 01/02/2025]
Abstract
Chromosomal instability (CIN) is common in solid tumours and fuels evolutionary adaptation and poor prognosis by increasing intratumour heterogeneity. Systematic characterization of driver events in the TRACERx non-small-cell lung cancer (NSCLC) cohort identified that genetic alterations in six genes, including FAT1, result in homologous recombination (HR) repair deficiencies and CIN. Using orthogonal genetic and experimental approaches, we demonstrate that FAT1 alterations are positively selected before genome doubling and associated with HR deficiency. FAT1 ablation causes persistent replication stress, an elevated mitotic failure rate, nuclear deformation and elevated structural CIN, including chromosome translocations and radial chromosomes. FAT1 loss contributes to whole-genome doubling (a form of numerical CIN) through the dysregulation of YAP1. Co-depletion of YAP1 partially rescues numerical CIN caused by FAT1 loss but does not relieve HR deficiencies, nor structural CIN. Importantly, overexpression of constitutively active YAP15SA is sufficient to induce numerical CIN. Taken together, we show that FAT1 loss in NSCLC attenuates HR and exacerbates CIN through two distinct downstream mechanisms, leading to increased tumour heterogeneity.
Collapse
Affiliation(s)
| | | | | | - James R M Black
- The Francis Crick Institute, London, UK
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Carlos Martinez-Ruiz
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | | | - Francisco Gimeno-Valiente
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Ieva Usaite
- The Francis Crick Institute, London, UK
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | | | - Kerstin Thol
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | | | | | | | - Yun-Hsin Liu
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Dhruva Biswas
- The Francis Crick Institute, London, UK
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | | | | | | | - Subramanian Venkatesan
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Georgia Stavrou
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Takahiro Karasaki
- The Francis Crick Institute, London, UK
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
- Department of Thoracic Surgery, Respiratory Center, Toranomon Hospital, Tokyo, Japan
| | - Maise Al Bakir
- The Francis Crick Institute, London, UK
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | | | - Hang Xu
- The Francis Crick Institute, London, UK
| | | | - Natasha Sharma
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Laura Tovini
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Mariam Jamal-Hanjani
- The Francis Crick Institute, London, UK
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | | | - Kevin Litchfield
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Nicolai J Birkbak
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | | | - Kasper Fugger
- University College London Cancer Institute, London, UK
| | - Nicholas McGranahan
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Jiri Bartek
- Danish Cancer Society Research Centre, Copenhagen, Denmark.
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Laboratory, Karolinska Institutet, Solna, Sweden.
| | - Nnennaya Kanu
- CRUK Lung Cancer Centre of Excellence, London, UK.
- University College London Cancer Institute, London, UK.
| | - Charles Swanton
- The Francis Crick Institute, London, UK.
- CRUK Lung Cancer Centre of Excellence, London, UK.
- University College London Cancer Institute, London, UK.
| |
Collapse
|
40
|
Voutsadakis IA. Gastric Adenocarcinomas with CDX2 Induction Show Higher Frequency of TP53 and KMT2B Mutations and MYC Amplifications but Similar Survival Compared with Cancers with No CDX2 Induction. J Clin Med 2024; 13:7635. [PMID: 39768557 PMCID: PMC11727917 DOI: 10.3390/jcm13247635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Background: Gastric cancer is one of the most prevalent gastrointestinal cancers. Mortality is high, and improved treatments are needed. A better understanding of the pathophysiology of the disease and discovery of biomarkers for targeted therapies are paramount for therapeutic progress. CDX2, a transcription factor of hindgut specification, is induced in several gastric cancers, especially with intestinal differentiation, and could be helpful for defining sub-types with particular characteristics. Methods: Gastric cancers with induced CDX2 mRNA expression were identified from the gastric cohort of The Cancer Genome Atlas (TCGA) and were compared with cancers that had no CDX2 mRNA induction. Induced CDX2 mRNA expression was defined as mRNA expression z-score relative to all samples above 0, and non-induced CDX2 mRNA expression was defined as mRNA expression z-score relative to all samples below -1. Results: Patients with gastric cancers with CDX2 mRNA induction were older, had less frequently diffuse histology, and more often had mutations in TP53 and KMT2B and amplifications in MYC. CDX2 induction was correlated with HNF4α induction and was reversely correlated with SOX2. Gastric cancers with CDX2 mRNA induction showed lower PD-L1 expression than cancers with lower CDX2 expression but did not differ in CLDN18 mRNA expression. Progression-free and overall survival of the two groups was also not significantly different. Conclusion: Gastric cancers with CDX2 mRNA induction displayed specific characteristics that differentiate them from cancers with no CDX2 induction and could be of interest for optimizing current and future therapies.
Collapse
Affiliation(s)
- Ioannis A. Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, 750 Great Northern Road, Sault Ste Marie, ON P6B 0A8, Canada; or
- Division of Clinical Sciences, Section of Internal Medicine, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
41
|
Wu Q, Hu C, Feng L, Yang X, Cui Y, Zhao H, Xiao T, Guo H. Comprehensive genomic profiling of infiltrative follicular variant of papillary thyroid carcinoma. Cancer 2024; 130:4241-4256. [PMID: 39141684 DOI: 10.1002/cncr.35517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/11/2024] [Accepted: 07/28/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Infiltrative follicular variant of papillary thyroid carcinoma (IFVPTC) exhibits nuclear characteristics typical of papillary thyroid carcinoma (PTC) but demonstrates a follicular growth pattern. The diagnosis of IFVPTC presenting with atypical nuclear features of PTC poses challenges for both preoperative cytopathology and postoperative histopathology. In such cases, molecular markers are needed to serve as diagnostic aids. Given the limited knowledge of IFVPTC's genomic features, this study aimed to characterize its genetic alterations and identify clinically relevant molecular markers. METHODS Whole-exome sequencing of 50 IFVPTC tumor-normal pairs identified single-nucleotide variants, somatic copy number alterations (sCNAs), and subclonal architecture. Key mutations were verified via polymerase chain reaction and Sanger sequencing, whereas valuable biomarkers were validated via immunohistochemistry (IHC). RESULTS This study found that endogenous processes rather than exogenous mutagens dominated the shaping of the genome of IFVPTC during tumorigenesis. BRAF V600E was the only common trunk mutation and significantly mutated gene in IFVPTC. Subcloning analysis found that most IFVPTC samples harbored two or more coexisting clones. sCNA analysis revealed that human leukocyte antigen C (HLA-C) and HLA-A were significantly amplified. Subsequent IHC investigations indicated that HLA-C shows promise in averting the misclassification of challenging-to-interpret IFVPTC and invasive encapsulated follicular variant of PTC (I-EFVPTC) as noninvasive follicular thyroid neoplasm with papillary-like nuclear features (NIFTP). Although there were several similarities between classic PTC and IFVPTC, they differed significantly in their sCNA patterns. CONCLUSIONS This study provides valuable insights into IFVPTC's genetic alterations and highlights the potential of HLA-C IHC to distinguish challenging-to-interpret IFVPTC and I-EFVPTC from NIFTP, which will enhance the understanding of its molecular features for improved diagnosis and management.
Collapse
Affiliation(s)
- Quanyou Wu
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunfang Hu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Yang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Cui
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huan Zhao
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Xiao
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Huiqin Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Hebei Cancer Hospital, Chinese Academy of Medical Sciences, Langfang, China
| |
Collapse
|
42
|
Fu S, Xu J, Wang C, Zhang C, Li C, Xie W, Wang G, Zhu X, Xu Y, Wen Y, Pei J, Yang J, Tang M, Tan H, Cai S, Cai L, Pan M. Cancer specific up-regulated lactate genes associated with immunotherapy resistance in a pan-cancer analysis. Heliyon 2024; 10:e39491. [PMID: 39669156 PMCID: PMC11636123 DOI: 10.1016/j.heliyon.2024.e39491] [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: 02/27/2024] [Revised: 09/10/2024] [Accepted: 10/15/2024] [Indexed: 12/14/2024] Open
Abstract
Background Although the lactate pathway has been reported to lead to immune escape through the inhibition of effector T cells, the cancer-intrinsic lactate signature has not been identified, and the immunotherapeutic efficacy and potential mechanism of the lactate signature are still unclear. Methods We defined a pan-cancer up-lactate score by comparing malignant tissues and normal tissues in the TCGA cohort. The immunotherapeutic efficacy was evaluated in non-small cell lung cancer (NSCLC), metastatic renal cancer (mRCC), bladder cancer (BLCA) and melanoma cohorts. The cancer cell-intrinsic mechanism to immune checkpoint inhibitors (ICIs) resistance was measured using single cell sequencing (scRNA-seq) data. Pathway activation was evaluated in the TCGA cohort and CPTAC cohort with transcriptomics and proteomics. The co-occurrence of up-lactate signature and mTOR signaling was determined by spatial transcriptomics of the tissue samples. Immunotherapy resistance and pathway regulation were validated in the in-house NSCLC cohort. Results Patients with the high up-lactate scores had significantly short overall survival (OS) than those with the low up-lactate scores (p < 0.001) across multiple types of cancers. The up-regulated lactate signature exhibited higher expression in the malignant cells compared with stromal cells and immune cells in multiple scRNA-seq datasets. A high up-lactate score was associated with poor OS in NSCLC, mRCC, BLCA and melanoma patients who received anti-PD(L)1 antibody. The up-lactate score was higher in the responders of cancer cells, but not in immune cells and stromal cells compared with the non-responders (p < 0.05). Moreover, up-lactate score was positively correlated with mTOR signaling across multiple cancers. In patients with NSCLC who received anti-PD-1 antibody, higher up-lactate scores were associated with significantly shorter PFS compared to lower up-lactate scores (p < 0.001). Additionally, the up-lactate score was associated with cold tumor, and was positively correlated with mTOR signaling. Conclusion Collectively, we defined a pan-cancer up-lactate signature, which is a feature of malignant cells and is associated with ICIs resistance. This reveals a coherent program with prognostic and predictive value that may be therapeutically targeted.
Collapse
Affiliation(s)
- Shuiting Fu
- Department of Oral & Maxillofacial - Head & Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Jiachen Xu
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Guangdong Provincial People's Hospital/Guangdong Provincial Academy of Medical Sciences, Guangdong Provincial Key Lab of Translational Medicine in Lung Cancer, China
| | - Chunming Wang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Cheng Zhang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | | | | | | | - Xin Zhu
- Burning Rock Biotech, Guangzhou, 510300, China
| | - Yuyan Xu
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yaohong Wen
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jingyuan Pei
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jun Yang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Mingyang Tang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Hongkun Tan
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Shangli Cai
- Burning Rock Biotech, Guangzhou, 510300, China
| | - Lei Cai
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Mingxin Pan
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| |
Collapse
|
43
|
Moufarrij S, Gazzo A, Rana S, Selenica P, Abu-Rustum NR, Ellenson LH, Liu YL, Weigelt B, Momeni-Boroujeni A. Concurrent POLE hotspot mutations and mismatch repair deficiency/microsatellite instability in endometrial cancer: A challenge in molecular classification. Gynecol Oncol 2024; 191:1-9. [PMID: 39276497 DOI: 10.1016/j.ygyno.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 09/17/2024]
Abstract
OBJECTIVE Endometrial carcinoma (EC) has different molecular subtypes associated with varied prognosis. We sought to characterize the molecular features of ECs with POLE hotspot mutations and concurrent mismatch repair (MMR) deficiency/high microsatellite instability (MSI). METHODS We identified POLE-mutated (POLEmut), MMR-deficient (MMRd)/MSI-high (MSI-H), or combined POLEmut/MMRd ECs subjected to clinical tumor-normal panel sequencing between 2014 and 2023. Clonality of somatic mutations, MSI scoring, tumor mutational burden (TMB), proportion of somatic insertions and deletions (indels), and single base substitution (SBS) mutational signatures were extracted. RESULTS We identified 41 ECs harboring POLE exonuclease domain hotspot mutations, 138 MMRd and/or MSI-H ECs, and 14 POLEmut/MMRd ECs. Among the 14 POLEmut/MMRd ECs, 11 (79 %) exhibited clonal POLE hotspot mutations; 4 (29 %) had a dominant POLE-related mutational signature, 4 (29 %) displayed dominant MMRd-related signatures, and 6 (43 %) had mixtures of POLE, aging/clock, MMRd, and POLEmut/MMRd-related SBS mutational signatures. The number of single nucleotide variants was higher in POLEmut/MMR-proficient (MMRp) and in POLEmut/MMRd ECs compared to POLE wild-type (wt)/MMRd EC (both p < 0.001). Small indels were enriched in POLEwt/MMRd ECs (p < 0.001). TMB was highest in POLEmut/MMRd EC compared to POLEmut/MMRp and POLEwt/MMRd ECs (both p < 0.001). Of 14 patients with POLEmut/MMRd EC, 21 % had a recurrence, versus 10 % of those with POLEmut/MMRp EC. Similar findings were noted in 3 POLEmut ECs in patients with Lynch syndrome; akin to somatic POLEmut ECs, these tumors had high TMB. CONCLUSION POLEmut/MMRd ECs may be genetically distinct. Further studies are needed to assess the impact on outcomes and treatment response within this population.
Collapse
Affiliation(s)
- Sara Moufarrij
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea Gazzo
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Satshil Rana
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pier Selenica
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadeem R Abu-Rustum
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Lora H Ellenson
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ying L Liu
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amir Momeni-Boroujeni
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
44
|
Schmidt H, Raphael BJ. A regression based approach to phylogenetic reconstruction from multi-sample bulk DNA sequencing of tumors. PLoS Comput Biol 2024; 20:e1012631. [PMID: 39630782 DOI: 10.1371/journal.pcbi.1012631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/20/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024] Open
Abstract
MOTIVATION DNA sequencing of multiple bulk samples from a tumor provides the opportunity to investigate tumor heterogeneity and reconstruct a phylogeny of a patient's cancer. However, since bulk DNA sequencing of tumor tissue measures thousands of cells from a heterogeneous mixture of distinct sub-populations, accurate reconstruction of the tumor phylogeny requires simultaneous deconvolution of cancer clones and inference of ancestral relationships, leading to a challenging computational problem. Many existing methods for phylogenetic reconstruction from bulk sequencing data do not scale to large datasets, such as recent datasets containing upwards of ninety samples with dozens of distinct sub-populations. RESULTS We develop an approach to reconstruct phylogenetic trees from multi-sample bulk DNA sequencing data by separating the reconstruction problem into two parts: a structured regression problem for a fixed tree [Formula: see text], and an optimization over tree space. We derive an algorithm for the regression sub-problem by exploiting the unique, combinatorial structure of the matrices appearing within the problem. This algorithm has both asymptotic and empirical improvements over linear programming (LP) approaches to the problem. Using our algorithm for this regression sub-problem, we develop fastBE, a simple method for phylogenetic inference from multi-sample bulk DNA sequencing data. We demonstrate on simulated data with hundreds of samples and upwards of a thousand distinct sub-populations that fastBE outperforms existing approaches in terms of reconstruction accuracy, sample efficiency, and runtime. Owing to its scalability, fastBE enables both phylogenetic reconstruction directly from indvidual mutations without requiring the clustering of mutations into clones, as well as a new phylogeny constrained mutation clustering algorithm. On real data from fourteen B-progenitor acute lymphoblastic leukemia patients, fastBE infers mutation phylogenies with fewer violations of a widely used evolutionary constraint and better agreement to the observed mutational frequencies. Using our phylogeny constrained mutation clustering algorithm, we also find mutation clusters with lower distortion compared to state-of-the-art approaches. Finally, we show that on two patient-derived colorectal cancer models, fastBE infers mutation phylogenies with less violation of a widely used evolutionary constraint compared to existing methods.
Collapse
Affiliation(s)
- Henri Schmidt
- Department of Computer Science, Princeton University, New Jersey, United States of America
| | - Benjamin J Raphael
- Department of Computer Science, Princeton University, New Jersey, United States of America
| |
Collapse
|
45
|
Weng Z, Mai Z, Yuan J, Liu Q, Deng F, Yang H, Ling Y, Xie X, Lin X, Lin T, Chen J, Wei X, Luo K, Fu J, Wen J. Evolution of genome and immunogenome in esophageal squamous cell carcinomas driven by neoadjuvant chemoradiotherapy. Int J Cancer 2024; 155:2021-2035. [PMID: 39081132 DOI: 10.1002/ijc.35118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/26/2024] [Accepted: 07/05/2024] [Indexed: 10/04/2024]
Abstract
Neoadjuvant chemoradiotherapy (NCRT) followed by surgery is a standard treatment for locally advanced esophageal squamous cell carcinomas (ESCCs). However, the evolution of genome and immunogenome in ESCCs driven by NCRT remains incompletely elucidated. We performed whole-exome sequencing of 51 ESCC tumors collected before and after NCRT, 36 of which were subjected to transcriptome sequencing. Clonal analysis identified clonal extinction in 13 ESCC patients wherein all pre-NCRT clones disappeared after NCRT, and clonal persistence in 9 patients wherein clones endured following NCRT. The clone-persistent patients showed higher pre-NCRT genomic intratumoral heterogeneity and worse prognosis than the clone-extinct ones. In contrast to the clone-extinct patients, the clone-persistent patients demonstrated a high proportion of subclonal neoantigens within pre-treatment specimens. Transcriptome analysis revealed increased immune infiltrations and up-regulated immune-related pathways after NCRT, especially in the clone-extinct patients. The number of T cell receptor-neoantigen interactions was higher in the clone-extinct patients than in the clone-persistent ones. The decrease in T cell repertoire evenness positively correlated to the decreased number of clonal neoantigens after NCRT, especially in the clone-extinct patients. In conclusion, we identified two prognosis-related clonal dynamic modes driven by NCRT in ESCCs. This study extended our knowledge of the ESCC genome and immunogenome evolutions driven by NCRT.
Collapse
Affiliation(s)
- Zelin Weng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zihang Mai
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianye Yuan
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Qianwen Liu
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fangqi Deng
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hong Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yihong Ling
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiuying Xie
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaodan Lin
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ting Lin
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiyang Chen
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaoli Wei
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kongjia Luo
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianhua Fu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing Wen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
46
|
Sturgill I, Raab J, Hoadley K. Expanded detection and impact of BAP1 alterations in cancer. NAR Cancer 2024; 6:zcae045. [PMID: 39554490 PMCID: PMC11567159 DOI: 10.1093/narcan/zcae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/15/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024] Open
Abstract
Aberrant expression of the BAP1 (BRCA associated protein 1) tumor suppressor gene is a prominent risk factor for several tumor types and is important in tumor evolution and progression. Here we performed integrated multi-omics analyses using data from The Cancer Genome Atlas for 33 cancer types and over 10 000 individuals to identify alterations leading to BAP1 disruption. We combined existing variant calls and new calls derived from a de novo local realignment pipeline across multiple independent variant callers, increasing somatic variant detection by 41% from 182 to 257, including 11 indels ≥40 bp. The expanded detection of mutations highlights the power of new tools to uncover longer indels and impactful mutations. We developed an expression-based BAP1 activity score and identified a transcriptional profile associated with BAP1 disruption in cancer. BAP1 has been proposed to play a critical role in controlling tumor plasticity and normal cell fate. Leveraging human and mouse liver datasets, BAP1 loss in normal cells resulted in lower BAP1 activity scores and lower scores were associated with a less-differentiated phenotype in embryonic cells. Together, our expanded BAP1 mutant samples revealed a transcriptional signature in cancer cells, supporting BAP1's influences on cellular plasticity and cell identity maintenance.
Collapse
Affiliation(s)
- Ian R Sturgill
- Bioinformatics and Computational Biology Curriculum, Department of Genetics, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA
| | - Jesse R Raab
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA
| | - Katherine A Hoadley
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA
| |
Collapse
|
47
|
Tarantino G, Ricker CA, Wang A, Ge W, Aprati TJ, Huang AY, Madha S, Chen J, Shi Y, Glettig M, Feng CH, Frederick DT, Freeman S, Holovatska MM, Manos MP, Zimmer L, Rösch A, Zaremba A, Livingstone E, Jameson JC, Saghafian S, Lee A, Zhao K, Morris LG, Reardon B, Park J, Elmarakeby HA, Schilling B, Giobbie-Hurder A, Vokes NI, Buchbinder EI, Flaherty KT, Haq R, Wu CJ, Boland GM, Hodi FS, Van Allen EM, Schadendorf D, Liu D. Genomic heterogeneity and ploidy identify patients with intrinsic resistance to PD-1 blockade in metastatic melanoma. SCIENCE ADVANCES 2024; 10:eadp4670. [PMID: 39602539 PMCID: PMC11601251 DOI: 10.1126/sciadv.adp4670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024]
Abstract
The introduction of immune checkpoint blockade (ICB) has markedly improved outcomes for advanced melanoma. However, many patients develop resistance through unknown mechanisms. While combination ICB has improved response rate and progression-free survival, it substantially increases toxicity. Biomarkers to distinguish patients who would benefit from combination therapy versus aPD-1 remain elusive. We analyzed whole-exome sequencing of pretreatment tumors from four cohorts (n = 140) of ICB-naïve patients treated with aPD-1. High genomic heterogeneity and low ploidy robustly identified patients intrinsically resistant to aPD-1. To establish clinically actionable predictions, we optimized and validated a predictive model using ploidy and heterogeneity to confidently identify (90% PPV) patients with intrinsic resistance to and worse survival on aPD-1. We further observed that three of seven (43%) patients predicted to be intrinsically resistant to single-agent PD-1 ICB responded to combination ICB, suggesting that these patients may benefit disproportionately from combination ICB. These findings highlight the importance of heterogeneity and ploidy, nominating an approach toward clinical actionability.
Collapse
Affiliation(s)
- Giuseppe Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Cora A. Ricker
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Tyler J. Aprati
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amy Y. Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shariq Madha
- Worcester Polytechnic Institute, Worcester, MA, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Jiajia Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yingxiao Shi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marc Glettig
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Catherine H. Feng
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Marta M. Holovatska
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
| | - Michael P. Manos
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
| | - Lisa Zimmer
- Department of Dermatology, University HospitalEssen, Essen, Germany
| | - Alexander Rösch
- Department of Dermatology, University HospitalEssen, Essen, Germany
| | - Anne Zaremba
- Department of Dermatology, University HospitalEssen, Essen, Germany
| | | | - Jacob C. Jameson
- Interfaculty Initiative in Health Policy, Harvard University, Cambridge, MA, USA
| | | | - Andrew Lee
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karena Zhao
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luc G.T. Morris
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brendan Reardon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jihye Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Haitham A. Elmarakeby
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Al-Azhar University, Cairo, Egypt
| | - Bastian Schilling
- Department of Dermatology, University HospitalEssen, Essen, Germany
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | | | - Natalie I. Vokes
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Rizwan Haq
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | | | - F. Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
| | - Eliezer M. Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dirk Schadendorf
- Department of Dermatology, University HospitalEssen, Essen, Germany
| | - David Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
48
|
van der Sluis K, van Sandick JW, Koemans WJ, van den Bosch T, Broeks A, Peters D, Seignette IM, Rausch CR, van Dijk E, Snaebjornsson P, van den Berg JG, van Grieken NCT, Ylstra B, Carvalho B, Miedema DM, Kodach LL. Karyotype evolution in response to chemoradiotherapy and upon recurrence of esophageal adenocarcinomas. Cell Rep 2024; 43:114981. [PMID: 39535918 DOI: 10.1016/j.celrep.2024.114981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 09/06/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The genome of esophageal adenocarcinoma (EAC) is highly unstable and might evolve over time. Here, we track karyotype evolution in EACs in response to treatment and upon recurrence through multi-region and longitudinal analysis. To this end, we introduce L-PAC (low-purity inference of absolute copy-number alterations [CNAs]), a bio-informatics technique that allows inference of absolute CNAs of low-purity samples by leveraging the information of high-purity samples from the same cancer. Quantitative analysis of matched absolute CNAs reveals that the amount of karyotype evolution induced by chemoradiotherapy (CRT) is predictive for early recurrence and depends on the initial level of karyotype intra-tumor heterogeneity. We observe that CNAs acquired in response to CRT are partially reversed back to the initial state upon recurrence. Hence, CRT alters the fitness landscape to which tumors can adjust by adapting their karyotype. Together, our results indicate that karyotype plasticity contributes to the therapy resistance of EACs.
Collapse
Affiliation(s)
- Karen van der Sluis
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Johanna W van Sandick
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Willem J Koemans
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Tom van den Bosch
- Amsterdam UMC Location University of Amsterdam, Cancer Center Amsterdam & Amsterdam Gastroenterology Endocrinology Metabolism, Center for Experimental and Molecular Medicine, Amsterdam, the Netherlands; Oncode Institute, Amsterdam, the Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Dennis Peters
- Core Facility Molecular Pathology and Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Iris M Seignette
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Christian R Rausch
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Erik van Dijk
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Petur Snaebjornsson
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - José G van den Berg
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Nicole C T van Grieken
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Bauke Ylstra
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Beatriz Carvalho
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Daniël M Miedema
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Liudmila L Kodach
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
49
|
Yu J, Gui X, Zou Y, Liu Q, Yang Z, An J, Guo X, Wang K, Guo J, Huang M, Zhou S, Zuo J, Chen Y, Deng L, Yuan G, Li N, Song Y, Jia J, Zeng J, Zhao Y, Liu X, Du X, Liu Y, Wang P, Zhang B, Ding L, Robles AI, Rodriguez H, Zhou H, Shao Z, Wu L, Gao D. A proteogenomic analysis of cervical cancer reveals therapeutic and biological insights. Nat Commun 2024; 15:10114. [PMID: 39578447 PMCID: PMC11584810 DOI: 10.1038/s41467-024-53830-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 10/21/2024] [Indexed: 11/24/2024] Open
Abstract
Although the incidence of cervical cancer (CC) has been reduced in high-income countries due to human papillomavirus (HPV) vaccination and screening strategies, it remains a significant public health issue that poses a threat to women's health in low-income countries. Here, we perform a comprehensive proteogenomic profiling of CC tumors obtained from 139 Chinese women. Integrated proteogenomic analysis links genetic aberrations to downstream pathogenesis-related pathways and reveals the landscape of HPV-associated multi-omic changes. EP300 is found to enhance the acetylation of FOSL2-K222, consequently accelerating the malignant proliferation of CC cells. Proteomic stratification identifies three patient subgroups with distinct features in prognosis, genetic alterations, immune infiltration, and post-translational modification regulations. PRKCB is further identified as a potential radioresponse-related biomarker of CC patients. This study provides a valuable public resource for researchers and clinicians to delve into the molecular basis of CC, to identify potential treatments and to ultimately advance clinical practice.
Collapse
Affiliation(s)
- Jing Yu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiuqi Gui
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Yunhao Zou
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qian Liu
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Zhicheng Yang
- University of Chinese Academy of Sciences, Beijing, China
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jusheng An
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuan Guo
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kaihua Wang
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiaming Guo
- University of Chinese Academy of Sciences, Beijing, China
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Manni Huang
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuhan Zhou
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jing Zuo
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yimin Chen
- University of Chinese Academy of Sciences, Beijing, China
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Lu Deng
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guangwen Yuan
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Li
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Song
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia Jia
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia Zeng
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuxi Zhao
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianming Liu
- Bruker (Beijing) Scientific Technology Co., Ltd, Shanghai, China
| | - Xiaoxian Du
- Bruker (Beijing) Scientific Technology Co., Ltd, Shanghai, China
| | - Yansheng Liu
- Department of Pharmacology, Cancer Biology Institute, Yale University School of Medicine, West Haven, CT, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bing Zhang
- Department of Molecular and Human Genetics, Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Li Ding
- Department of Medicine, McDonnell Genome Institute, Siteman Cancer Center, Washington University, St. Louis, MI, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Hu Zhou
- University of Chinese Academy of Sciences, Beijing, China.
- Analytical Research Center for Organic and Biological Molecules, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| | - Zhen Shao
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Lingying Wu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Daming Gao
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
50
|
Lu Q, Liu Z, Wang X. Inferring tumor purity using multi-omics data based on a uniform machine learning framework MoTP. Brief Bioinform 2024; 26:bbaf056. [PMID: 39950745 PMCID: PMC11826339 DOI: 10.1093/bib/bbaf056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/24/2024] [Accepted: 01/27/2025] [Indexed: 02/17/2025] Open
Abstract
Existing algorithms for assessing tumor purity are limited to a single omics data, such as gene expression, somatic copy number variations, somatic mutations, and DNA methylation. Here we proposed the machine learning Multi-omics Tumor Purity prediction (MoTP) algorithm to estimate tumor purity based on multiple types of omics data. MoTP utilizes the Bayesian Regularized Neural Networks as the prediction algorithm, and Consensus Tumor Purity Estimates as labels. We trained MoTP using multi-omics data (mRNA, microRNA, long non-coding RNA, and DNA methylation) across 21 TCGA solid cancer types. By testing MoTP in TCGA validation sets, TCGA test sets, and eight datasets outside the TCGA cancer cohorts, we showed that although MoTP could achieve excellent performance in predicting tumor purity based on a single omics data type, the integration of multiple single omics data-based predictions can enhance the prediction performance. Moreover, we demonstrated the robustness of MoTP by testing it in datasets with Gaussian noise and feature missing. Benchmark analysis showed that MoTP outperformed most established tumor purity prediction algorithms, and that it required less running time and computational resource to fulfill the predictive task. Thus, MoTP would be an attractive option for computational tumor purity inference.
Collapse
Affiliation(s)
- Qiqi Lu
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, China
| | - Zhixian Liu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, China
| |
Collapse
|