1
|
Gupta A, Gazzo A, Selenica P, Safonov A, Pareja F, da Silva EM, Brown DN, Shao H, Zhu Y, Patel J, Blanco-Heredia J, Stefanovska B, Carpenter MA, Chen Y, Vegas I, Pei X, Frosina D, Jungbluth AA, Ladanyi M, Curigliano G, Weigelt B, Riaz N, Powell SN, Razavi P, Harris RS, Reis-Filho JS, Marra A, Chandarlapaty S. APOBEC3 mutagenesis drives therapy resistance in breast cancer. Nat Genet 2025; 57:1452-1462. [PMID: 40379787 PMCID: PMC12165862 DOI: 10.1038/s41588-025-02187-1] [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: 05/08/2024] [Accepted: 04/01/2025] [Indexed: 05/19/2025]
Abstract
Acquired genetic alterations drive resistance to endocrine and targeted therapies in metastatic breast cancer; however, the underlying processes engendering these alterations are largely uncharacterized. To identify the underlying mutational processes, we utilized a clinically annotated cohort of 3,880 patient samples with tumor-normal sequencing. Mutational signatures associated with apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3 (APOBEC3) enzymes were prevalent and enriched in post-treatment hormone receptor-positive cancers. These signatures correlated with shorter progression-free survival on antiestrogen plus CDK4/6 inhibitor therapy in hormone receptor-positive metastatic breast cancer. Whole-genome sequencing of breast cancer models and paired primary-metastatic samples demonstrated that active APOBEC3 mutagenesis promoted therapy resistance through characteristic alterations such as RB1 loss. Evidence of APOBEC3 activity in pretreatment samples illustrated its pervasive role in breast cancer evolution. These studies reveal APOBEC3 mutagenesis to be a frequent mediator of therapy resistance in breast cancer and highlight its potential as a biomarker and target for overcoming resistance.
Collapse
Affiliation(s)
- Avantika Gupta
- Human Oncology and Pathogenesis Program, 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
| | - Pier Selenica
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anton Safonov
- Department of Medicine, 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
| | - Edaise M da Silva
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David N Brown
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hong Shao
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yingjie Zhu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juber Patel
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juan Blanco-Heredia
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bojana Stefanovska
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Michael A Carpenter
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yanjun Chen
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Isabella Vegas
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xin Pei
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Denise Frosina
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Achim A Jungbluth
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Giuseppe Curigliano
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCSS, Milan, Italy
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Simon N Powell
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pedram Razavi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill-Cornell Medical College, New York, NY, USA
| | - Reuben S Harris
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Jorge S Reis-Filho
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Antonio Marra
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy.
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCSS, Milan, Italy.
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill-Cornell Medical College, New York, NY, USA.
| |
Collapse
|
2
|
Matejcic M, Teer JK, Hoehn HJ, Diaz DB, Shankar K, Gong J, Nguyen NT, Loroña NC, Coppola D, Fulmer CG, Saglam O, Jiang K, Cress WD, Muñoz-Antonia T, Flores I, Gordián ER, Oliveras Torres JA, Felder SI, Sanchez J, Fleming JB, Siegel EM, Freedman JA, Dutil J, Stern MC, Fridley BL, Figueiredo JC, Schmit SL. Colorectal Tumors in Diverse Patient Populations Feature a Spectrum of Somatic Mutational Profiles. Cancer Res 2025; 85:1928-1944. [PMID: 40126181 DOI: 10.1158/0008-5472.can-24-0747] [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: 03/18/2024] [Revised: 08/21/2024] [Accepted: 02/25/2025] [Indexed: 03/25/2025]
Abstract
Admixed populations, including the Hispanic/Latino/a community, are underrepresented in cancer genetic/genomic studies. Leveraging the Latino Colorectal Cancer Consortium (LC3) and other existing datasets, we analyzed whole-exome sequencing data on tumor/normal pairs from 718 individuals with colorectal cancer to map somatic mutational features by ethnicity and genetic similarity. Global proportions of African, Asian, European, and Native American genetic ancestries were estimated using ADMIXTURE. Associations between these proportions and somatic mutational features were examined using logistic regression. APC, TP53, and KRAS were the top three mutated genes across all participants and in the subset of Latino individuals in LC3. In analyses examining recurrently mutated genes, tumors from patients of Latino ethnicity had fewer KRAS and PIK3CA mutations compared with tumors from non-Latino patients. Genetic ancestry overall was associated with CDC27 mutation status, and African genetic ancestry was associated with SMAD2 mutation status. In exome-wide analyses, African genetic ancestry was significantly associated with higher odds of mutation in KNCN and TMEM184B. Native American genetic ancestry was associated with a lower frequency of microsatellite instability-high tumors. The SBS11 mutational signature was associated with Native American genetic ancestry as well as Latino ethnicity. In an independent replication dataset, MSK-IMPACT, estimates of association were largely consistent in direction but nonsignificant. A meta-analysis of LC3 and MSK-IMPACT showed that African genetic ancestry was significantly associated with KRAS mutation status and MSI status. This work facilitates precision medicine initiatives by providing insights into the contribution of genetic ancestry to molecular features of colorectal tumors. Significance: Analysis of tumors from various populations can broadly characterize genomic landscapes and enhance precision medicine strategies.
Collapse
Affiliation(s)
- Marco Matejcic
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Hannah J Hoehn
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Non-Therapeutic Research Office, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Diana B Diaz
- Non-Therapeutic Research Office, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Kritika Shankar
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jun Gong
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Nathalie T Nguyen
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Nicole C Loroña
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Domenico Coppola
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Clifton G Fulmer
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Ozlen Saglam
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Kun Jiang
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - W Douglas Cress
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Teresita Muñoz-Antonia
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Idhaliz Flores
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Edna R Gordián
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - José A Oliveras Torres
- Department of Basic Sciences, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Seth I Felder
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Julian Sanchez
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jason B Fleming
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Erin M Siegel
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Non-Therapeutic Research Office, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jennifer A Freedman
- Division of Medical Oncology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
- Duke Cancer Institute, Durham, North Carolina
| | - Julie Dutil
- Division of Clinical and Translational Cancer Research, Comprehensive Cancer Center of the University of Puerto Rico, San Juan, Puerto Rico
| | - Mariana C Stern
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California
| | - Brooke L Fridley
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jane C Figueiredo
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Stephanie L Schmit
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
3
|
Zhang L, Gao H, Ma S, Fan X, Guo H, Sun M, Wen S, Liu T, Yu G, Yuan X, Sun X, Fan B. Functional screening of somatic mutant events in extranodal natural killer/T-cell lymphoma with adrenal involvement. Front Immunol 2025; 16:1566794. [PMID: 40433378 PMCID: PMC12106576 DOI: 10.3389/fimmu.2025.1566794] [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: 01/25/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Background Extranodal natural killer/T-cell lymphoma (ENKTL) involving the adrenal glands is extremely rare, and only a few cases have been reported. However, the genetic alterations, clinicopathological features and prognosis of these patients have not yet been fully elucidated. Methods Profiling of tumor mutations in ENKTL patients with adrenal involvement was conducted by whole-genome sequencing, and the predisposing genes and driver mutation gene variants were verified through Sanger sequencing. Immunohistochemical analysis of markers for the diagnosis and tumor microenvironment competent were performed to identify histopathological features. In addition, we searched the Surveillance, Epidemiology, and End Results (SEER), PubMed, Embase, and Scopus databases to perform a population-based study to compare the prognosis between adrenocortical carcinoma (ACC) patients and adrenal ENKTL patients using Kaplan-Meier survival curves and log-rank tests and analyzed the prognostic factors affecting the overall survival (OS) of adrenal ENKTL patients via univariate and multivariate Cox regression analyses. Results We screened 15892 somatic single-nucleotide variants (SNVs), 364 somatic insertions and deletions (INDELs), and four driver mutation genes, namely, TET2, STAT3, FAS, and TP53. In addition, immunohistochemical analysis revealed that tumor cells were positive for CD3, CD43, CD56, TIA1, granzyme B, CD2, CD4, and CD7. The immunohistochemistry for detecting components of the tumor microenvironment reveled the infiltration of tumor-associated macrophages (CD68, CD163) and tumor-associated fibroblasts (vimentin, SMA) in the tumor sample. According to our population-based analysis, Kaplan-Meier survival curves revealed that ENKTL patients with adrenal involvement had a significantly poorer prognosis than did patients with ACC (p<0.001), and chemotherapy was a significant prognostic factor for OS in ENKTL patients with adrenal involvement according to Cox multivariate analysis (hazard ratio = 0.318; p=0.027). Conclusions The metastasis of ENKTL to the adrenal gland may be due to gene mutations caused by genetic variations, which may provide new therapeutic targets for this disease. The prognosis of adrenal ENKTL patients is markedly worse than that of ACC patients, and chemotherapy may serve as an independent factor of OS in adrenal ENKTL patients. However, our findings still need to be validated in additional studies.
Collapse
Affiliation(s)
- Luxin Zhang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Haifeng Gao
- Department of Urology, Central Hospital of Dalian, Dalian, Liaoning, China
| | - Shuang Ma
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaoming Fan
- Department of Pathology, Central Hospital of Dalian, Dalian, Liaoning, China
| | - Huahang Guo
- Department of Pathology, Dalian Friendship Hospital, Dalian, Liaoning, China
| | - Man Sun
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shuang Wen
- Department of Pathology, Dalian Friendship Hospital, Dalian, Liaoning, China
| | - Tianqing Liu
- Department of Pathology, Dalian Friendship Hospital, Dalian, Liaoning, China
| | - Guanghai Yu
- Department of Urology, Central Hospital of Dalian, Dalian, Liaoning, China
| | - Xiaoying Yuan
- College of Humanities and Social Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Xiuhua Sun
- Department of Oncology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bo Fan
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
4
|
Zhang L, Li H, Tong H, Cui H, Guo H, Wen S, Song Z, Chen J, Xiang S, Liu Z, Fan B, Wang L. Clinicopathological features and genetic mutation spectrum of primary anastomosing hemangioma arising from the kidney. Front Immunol 2025; 16:1554203. [PMID: 40416971 PMCID: PMC12098607 DOI: 10.3389/fimmu.2025.1554203] [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: 01/01/2025] [Accepted: 04/14/2025] [Indexed: 05/27/2025] Open
Abstract
Background Renal anastomosing hemangioma (RAH) is a rare benign renal tumor, and its clinicopathologic characteristics and genetic mutation spectrum related to its mechanisms of pathogenesis are unclear. Methods We carried out whole-genome sequencing (WGS) on RAH samples to explore the genetic mutation spectrum and verified the results by Sanger sequencing. Immunohistochemical analysis was also performed to reveal the histopathological characteristics and the tumor microenvironment components. Moreover, a population-based study was conducted after searching the PubMed, EMBASE, and Ovid SP databases to systematically summarize the clinicopathologic features of patients with RAH. Results WGS analysis revealed 10532 somatic single-nucleotide variants (SNVs), 6705 somatic insertions and deletions (INDELs), and mutations in 32 predisposing genes and 10 driver genes, among which the mutations in 8 of the predisposing genes, CNTNAP2, NCOA2, FAT1, MET, TJP2, MAML2, SRGAP3, and CSMD3, and the mutation site in the driver gene HIP1 were confirmed by Sanger sequencing. Moreover, the immunohistochemical profile of the tumor microenvironment revealed that the expression content of tumor-associated macrophages (CD163, CD68) and fibroblasts (SMA) differs between cancerous and precancerous tissues which may regulate the disease development. On the basis of our population-based analysis, we summarized the clinicopathological features of 100 patients with RAH and identified significant differences in age (p=0.001), tumor site (p<0.001), tumor focality (p<0.001), largest tumor diameter (p=0.001) and surgical approach (p=0.010) between patients with RAH with end-stage renal disease (ESRD) and those without ESRD. Conclusions The distinct phenotypes of RAH may be associated with the different genetic mutation spectra identified in our study. The presence or absence of comorbid ESRD varies among patients with RAH. However, additional studies are required to validate our results.
Collapse
Affiliation(s)
- Luxin Zhang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian, Liaoning, China
| | - Haozhen Li
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian, Liaoning, China
| | - Heyao Tong
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian, Liaoning, China
| | - Hepeng Cui
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian, Liaoning, China
| | - Huahang Guo
- Department of Pathology, Dalian Friendship Hospital, Dalian, Liaoning, China
| | - Shuang Wen
- Department of Pathology, Dalian Friendship Hospital, Dalian, Liaoning, China
| | - Zhuwei Song
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian, Liaoning, China
| | - Jiaqiang Chen
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian, Liaoning, China
| | - Shengxiang Xiang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian, Liaoning, China
| | - Zhiyu Liu
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian, Liaoning, China
| | - Bo Fan
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian, Liaoning, China
| | - Liang Wang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian, Liaoning, China
| |
Collapse
|
5
|
Skelly DA, Graham JP, Cheng M, Furuta M, Walter A, Stoklasek TA, Yang H, Stearns TM, Poirion O, Zhang JG, Grassmann JDS, Luo D, Flynn WF, Courtois ET, Chang CH, Serreze DV, Menghi F, Reinholdt LG, Liu ET. Mapping the genetic landscape establishing a tumor immune microenvironment favorable for anti-PD-1 response. Cell Rep 2025; 44:115698. [PMID: 40343794 DOI: 10.1016/j.celrep.2025.115698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/03/2025] [Accepted: 04/23/2025] [Indexed: 05/11/2025] Open
Abstract
Identifying host genetic factors modulating immune checkpoint inhibitor (ICI) efficacy is experimentally challenging. Our approach, utilizing the Collaborative Cross mouse genetic resource, fixes the tumor genomic configuration while varying host genetics. We find that response to anti-PD-1 (aPD1) immunotherapy is significantly heritable in four distinct murine tumor models (H2: 0.18-0.40). For the MC38 colorectal carcinoma system, we map four significant ICI response quantitative trait loci (QTLs) with significant epistatic interactions. The differentially expressed genes within these QTLs that define responder genetics are highly enriched for processes involving antigen processing and presentation, allograft rejection, and graft vs. host disease (all p < 1 × 10-10). Functional blockade of two top candidate immune targets, GM-CSF and IL-2RB, completely abrogates the MC38 transcriptional response to aPD1 therapy. Thus, our in vivo experimental platform is a powerful approach for discovery of host genetic factors that establish the tumor immune microenvironment propitious for ICI response.
Collapse
Affiliation(s)
- Daniel A Skelly
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - John P Graham
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | | | - Mayuko Furuta
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Andrew Walter
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | | | | | - Timothy M Stearns
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Olivier Poirion
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Ji-Gang Zhang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Jessica D S Grassmann
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Diane Luo
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - William F Flynn
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Elise T Courtois
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; OB/Gyn Department, UConn Health, Farmington, CT 06032, USA
| | - Chih-Hao Chang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - David V Serreze
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Francesca Menghi
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Laura G Reinholdt
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Edison T Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA.
| |
Collapse
|
6
|
Wee K, Yang KC, Schaeffer DF, Zhou C, Leung E, Feng X, Laskin J, Marra MA, Loree JM, Gorski SM. Genomic and transcriptomic landscapes of metastatic neuroendocrine neoplasms from distinct primary sites and their clinical implications. Sci Rep 2025; 15:15770. [PMID: 40328872 PMCID: PMC12056210 DOI: 10.1038/s41598-025-00549-7] [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: 08/09/2024] [Accepted: 04/29/2025] [Indexed: 05/08/2025] Open
Abstract
Neuroendocrine neoplasms (NENs) encompass a highly heterogeneous group of neoplasms with varying prognoses and molecular alterations. Molecular profiling studies have furthered our understanding of NENs, but the majority of previous studies have focused on primary tumors and on mutational landscapes using DNA sequencing data. Here, we describe the genomic and transcriptomic landscapes of 28 metastatic NENs across different primary anatomical sites (PASs) and their potential clinical implications. Although our cohort is small, our analyses provide further insights on the molecular commonalities and distinctions between metastatic NENs of different PASs. Comparison to several reference transcriptome data sets revealed that despite considerable whole genome and transcriptome variability in NENs, the metastatic NENs are still more like each other than other cancer types. Our study also highlights the potential utility of NEN transcriptome data for molecular classification and clinical decision making.
Collapse
Affiliation(s)
- Kathleen Wee
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Kevin C Yang
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - David F Schaeffer
- Division of Anatomic Pathology, Vancouver General Hospital, Vancouver, BC, Canada
- Pancreas Centre BC, Vancouver, BC, Canada
| | - Chen Zhou
- Department of Pathology and Laboratory Medicine, BC Cancer, Vancouver, BC, Canada
| | - Emily Leung
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Xiaolan Feng
- Vancouver Island Centre, BC Cancer, Victoria, BC, Canada
| | - Janessa Laskin
- Division of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
- Department of Medical Genetics and Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | | | - Sharon M Gorski
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada.
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
7
|
Comino-Méndez I, Velasco-Suelto J, Pascual J, López-López E, Quirós-Ortega ME, Gaona-Romero C, Martín-Muñoz A, Losana P, Heredia Y, Alba E, Guerrero-Zotano A. Identification of minimal residual disease using the clonesight test for ultrasensitive ctDNA detection to anticipate late relapse in early breast cancer. Breast Cancer Res 2025; 27:65. [PMID: 40312346 PMCID: PMC12044774 DOI: 10.1186/s13058-025-02016-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/04/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Early-stage breast cancer (BC) diagnosis significantly reduces mortality, yet relapse remains a concern due to undetectable minimal residual disease (MRD). Liquid biopsies offer real-time insights into tumor dynamics, aiding MRD detection and therapy response evaluation. However, MRD detection is challenging due to low tumor DNA levels in circulation. METHODS This prospective study included 20 HR + BC patients who had completed at least 5 years of adjuvant endocrine therapy (ET). Plasma samples were collected every 6 months over a median follow-up period of 2 years. Tumor-specific somatic variants identified through tumor tissue sequencing served as biomarkers for a patient-informed circulating tumor DNA (ctDNA) assay (CloneSight), which utilized a multiplex PCR-based next-generation sequencing (NGS) workflow. RESULTS ctDNA was detected in patients who experienced clinical relapse, with positivity observed up to 68 months (5.7 years) prior to overt recurrence, highlighting its potential for early relapse identification. In non-relapsed patients, ctDNA remained undetectable in 93% of cases, reflecting a potential high level of specificity. The assay detected ctDNA in 50% of relapsed patients, while no ctDNA signal was identified in the majority of non-relapsed cases. CONCLUSION Our exploratory findings indicate that CloneSight could be a promising tool for MRD detection and relapse prediction, providing a cost-effective, patient-informed approach to ctDNA monitoring. The ability of this approach to detect relapse prior to clinical recurrence suggests its potential relevance in improving patient monitoring. These findings suggest that ctDNA-based MRD assays could play a role in future surveillance strategies for HR + BC, though further studies in larger cohorts are needed to confirm their clinical applicability.
Collapse
Affiliation(s)
- Iñaki Comino-Méndez
- Medical Oncology Department, Hospital Universitario Virgen de la Victoria, 29010, Malaga, Spain.
- The Biomedical Research Institute of Málaga, IBIMA-CIMES-UMA), 29010, Malaga, Spain.
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC - CB16, 12/00481), 28029, Madrid, Spain.
| | - Jesús Velasco-Suelto
- Medical Oncology Department, Hospital Universitario Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga, IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC - CB16, 12/00481), 28029, Madrid, Spain
| | - Javier Pascual
- Medical Oncology Department, Hospital Universitario Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga, IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC - CB16, 12/00481), 28029, Madrid, Spain
| | - Esperanza López-López
- Medical Oncology Department, Hospital Universitario Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga, IBIMA-CIMES-UMA), 29010, Malaga, Spain
| | - Maria Elena Quirós-Ortega
- Medical Oncology Department, Hospital Universitario Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga, IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC - CB16, 12/00481), 28029, Madrid, Spain
| | - Celia Gaona-Romero
- Medical Oncology Department, Hospital Universitario Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga, IBIMA-CIMES-UMA), 29010, Malaga, Spain
| | - Alejandro Martín-Muñoz
- Hematological Malignancies Clinical Research Unit, Centro Nacional de Investigaciones Oncologicas (CNIO), Madrid, Spain
- Altum Sequencing Co, Madrid, Spain
| | | | | | - Emilio Alba
- Medical Oncology Department, Hospital Universitario Virgen de la Victoria, 29010, Malaga, Spain
- The Biomedical Research Institute of Málaga, IBIMA-CIMES-UMA), 29010, Malaga, Spain
- Centro de Investigación Biomédica en Red de Cancer (CIBERONC - CB16, 12/00481), 28029, Madrid, Spain
- Faculty of Medicine, University of Málaga, 29010, Malaga, Spain
| | - Angel Guerrero-Zotano
- Medical Oncology Department, Fundación Instituto Valenciano de Oncología, Valencia, Spain.
| |
Collapse
|
8
|
Brumage L, Best S, Hippe DS, Grunblatt E, Chanana P, Wu F, Lee MC, Ying Z, Ibrahim A, Chung JH, Vigil A, Fatherree J, Beronja S, Paddison P, Sullivan L, Nabet B, MacPherson D. In vivo functional screens reveal KEAP1 loss as a driver of chemoresistance in small cell lung cancer. SCIENCE ADVANCES 2025; 11:eadq7084. [PMID: 40267200 PMCID: PMC12017300 DOI: 10.1126/sciadv.adq7084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 03/18/2025] [Indexed: 04/25/2025]
Abstract
Exquisitely chemosensitive initially, small cell lung cancer (SCLC) exhibits dismal outcomes owing to rapid transition to chemoresistance. Elucidating the genetic underpinnings has been challenging owing to limitations with cellular models. As SCLC patient-derived xenograft (PDX) models mimic therapeutic responses, we perform genetic screens in chemosensitive PDX models to identify drivers of chemoresistance. cDNA overexpression screens identify MYC, MYCN, and MYCL, while CRISPR deletion screens identify KEAP1 loss as driving chemoresistance. Deletion of KEAP1 switched a chemosensitive SCLC PDX model to become chemoresistant and resulted in sensitivity to inhibition of glutamine metabolism. Data from the IMpower133 clinical trial revealed ~6% of patients with extensive-stage SCLC exhibit KEAP1 genetic alterations, with activation of a KEAP1/NRF2 transcriptional signature associated with reduced survival upon chemotherapy treatment. While roles for KEAP1/NRF2 have been unappreciated in SCLC, our genetic screens revealed KEAP1 loss as a driver of chemoresistance, while patient genomic analyses demonstrate clinical importance.
Collapse
Affiliation(s)
- Lauren Brumage
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington Seattle, Seattle, WA, USA
| | - Scott Best
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington Seattle, Seattle, WA, USA
| | - Daniel S. Hippe
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Eli Grunblatt
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Pritha Chanana
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Feinan Wu
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Zhe Ying
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ali Ibrahim
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jae Heun Chung
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Anna Vigil
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jackson Fatherree
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Slobodan Beronja
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Patrick Paddison
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lucas Sullivan
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - David MacPherson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|
9
|
Abdelwahab O, Torkamaneh D. Artificial intelligence in variant calling: a review. FRONTIERS IN BIOINFORMATICS 2025; 5:1574359. [PMID: 40337525 PMCID: PMC12055765 DOI: 10.3389/fbinf.2025.1574359] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/08/2025] [Indexed: 05/09/2025] Open
Abstract
Artificial intelligence (AI) has revolutionized numerous fields, including genomics, where it has significantly impacted variant calling, a crucial process in genomic analysis. Variant calling involves the detection of genetic variants such as single nucleotide polymorphisms (SNPs), insertions/deletions (InDels), and structural variants from high-throughput sequencing data. Traditionally, statistical approaches have dominated this task, but the advent of AI led to the development of sophisticated tools that promise higher accuracy, efficiency, and scalability. This review explores the state-of-the-art AI-based variant calling tools, including DeepVariant, DNAscope, DeepTrio, Clair, Clairvoyante, Medaka, and HELLO. We discuss their underlying methodologies, strengths, limitations, and performance metrics across different sequencing technologies, alongside their computational requirements, focusing primarily on SNP and InDel detection. By comparing these AI-driven techniques with conventional methods, we highlight the transformative advancements AI has introduced and its potential to further enhance genomic research.
Collapse
Affiliation(s)
- Omar Abdelwahab
- Département de Phytologie, Université Laval, Québec City, QC, Canada
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec City, QC, Canada
- Centre de recherche et d’innovation sur les végétaux (CRIV), Université Laval, Québec City, QC, Canada
- Institut intelligence et données (IID), Université Laval, Québec City, QC, Canada
| | - Davoud Torkamaneh
- Département de Phytologie, Université Laval, Québec City, QC, Canada
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec City, QC, Canada
- Centre de recherche et d’innovation sur les végétaux (CRIV), Université Laval, Québec City, QC, Canada
- Institut intelligence et données (IID), Université Laval, Québec City, QC, Canada
| |
Collapse
|
10
|
Liu R, Roberts E, Parsons H, Stover E, Choudhury A, Rhoades J, Blewett T, Yang D, Liu J, Mayer E, Adalsteinsson V. DirectHRD enables sensitive scar-based classification of homologous recombination deficiency. Nucleic Acids Res 2025; 53:gkaf313. [PMID: 40263706 PMCID: PMC12014287 DOI: 10.1093/nar/gkaf313] [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: 01/07/2025] [Revised: 03/20/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025] Open
Abstract
Homologous recombination deficiency (HRD) is a predictive biomarker for efficacy of PARP (poly ADP-ribose polymerase) inhibition and platinum chemotherapy for cancer patients but remains challenging to detect. The discovery of patients without pathogenic mutations in known HR genes but exhibiting genomic scars indicative of HRD led to the FDA approval of the first scar-based HRD test. Despite advancements in whole genome sequencing (WGS) and integration of large training datasets with machine learning models, current methods lack the sensitivity required for detecting HRD scars in low tumor purity samples, especially in liquid biopsies. Here, we describe DirectHRD, a genomic scar-based HRD classifier based on WGS. Compared to other WGS-based methods, DirectHRD exclusively utilizes a highly specific type of HRD scar-small deletions with microhomology-and its associated signatures in a probabilistic framework. We applied DirectHRD to 501 tumor and 90 cell-free DNA (cfDNA) samples from 4 cancer types: breast, ovarian, prostate, and pancreas. Among all 501 tumor biopsies, DirectHRD achieved 100% detection of HRD with high specificity (>90%). Across all 90 cfDNA samples, the method achieved an area under the curve of 0.87 and demonstrated the ability to detect HRD at tumor fractions as low as 1%, making it 10 times more sensitive than state-of-the-art methods.
Collapse
Affiliation(s)
- Ruolin Liu
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, United States
| | - Eugenia Roberts
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, United States
| | | | | | | | - Justin Rhoades
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, United States
| | - Timothy Blewett
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, United States
| | - David D Yang
- Dana-Farber Cancer Institute, Boston, MA, 02215, United States
| | - Joyce F Liu
- Dana-Farber Cancer Institute, Boston, MA, 02215, United States
| | - Erica L Mayer
- Dana-Farber Cancer Institute, Boston, MA, 02215, United States
| | | |
Collapse
|
11
|
Fiorica PN, Golmard L, Kim J, Bao R, Lin FY, Roy A, Pribnow A, Perrino MR, Masliah-Planchon J, Michalak-Provost S, Wong J, Filser M, Stoppa-Lyonnet D, Bourdeaut F, Brahimi A, Ingster O, Saulnier Sholler G, Jackson SA, Sasaki MM, Fowler T, Ng A, Corbett RJ, Kaufman RS, Haley JS, Carey DJ, Huang KL, Diskin SJ, Rokita JL, Al-Kateb H, McGee RB, Schiffman JD, Chen KS, Stewart DR, Williams Parsons D, Plon SE, Schultz KAP, Onel K. Germline Pathogenic DROSHA Variants Are Linked to Pineoblastoma and Wilms Tumor Predisposition. Clin Cancer Res 2025; 31:1491-1503. [PMID: 39992227 PMCID: PMC11995001 DOI: 10.1158/1078-0432.ccr-24-2785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/22/2024] [Accepted: 02/13/2025] [Indexed: 02/25/2025]
Abstract
PURPOSE DROSHA, DGCR8, and DICER1 regulate miRNA biogenesis and are commonly mutated in cancer. Although DGCR8 and DICER1 germline pathogenic variants (GPV) cause autosomal dominant tumor predisposition, no association between DROSHA GPVs and clinical phenotypes has been reported. EXPERIMENTAL DESIGN After obtaining informed consent, sequencing was performed on germline and tumor samples from all patients. The occurrence of germline DROSHA GPVs was investigated in large pediatric and adult cancer datasets. The population prevalence of DROSHA GPVs was investigated in the UK Biobank and Geisinger DiscovEHR cohorts. RESULTS We describe nine children from eight families with heterozygous DROSHA GPVs and a diagnosis of pineoblastoma (n = 8) or Wilms tumor (n = 1). A somatic second hit in DROSHA was detected in all eight tumors analyzed. All pineoblastoma tumors analyzed were classified as miRNA processing-altered 1 subtype. We estimate the population prevalence of germline DROSHA loss-of-function variants to be 1:3,875 to 1:4,843 but find no evidence for increased adult cancer risk. CONCLUSIONS This is the first report of DROSHA-related tumor predisposition. As pineoblastoma and Wilms tumor are also associated with DICER1 GPVs, our results suggest that the tissues of origin for these tumors are uniquely tolerant of general miRNA loss. The miRNA processing-altered 1 pineoblastoma subtype is associated with older age of diagnosis and better outcomes than other subtypes, suggesting DROSHA GPV status may have important clinical and prognostic significance. We suggest that genetic testing for DROSHA GPVs be considered for patients with pineoblastoma, Wilms tumor, or other DICER1-/DGCR8-related conditions and propose surveillance recommendations through research studies for individuals with DROSHA GPVs.
Collapse
Affiliation(s)
- Peter N. Fiorica
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Lisa Golmard
- Department of Genetics, Institut Curie, Paris, France
- PSL Research University, Paris, France
| | - Jung Kim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Riyue Bao
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Frank Y. Lin
- Department of Pediatrics, Baylor College of Medicine, Houston Texas
- Texas Children’s Cancer and Hematology Center, Texas Children’s Hospital, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Angshumoy Roy
- Department of Pediatrics, Baylor College of Medicine, Houston Texas
- Texas Children’s Cancer and Hematology Center, Texas Children’s Hospital, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Department of Pathology, Texas Children’s Hospital, Houston, Texas
| | - Allison Pribnow
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Lucile Packard Children’s Hospital, Stanford University School of Medicine, Stanford, California
| | - Melissa R. Perrino
- Division of Cancer Predisposition, Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | | | | | - Jennifer Wong
- Department of Genetics, Institut Curie, Paris, France
- PSL Research University, Paris, France
| | - Mathilde Filser
- Department of Genetics, Institut Curie, Paris, France
- PSL Research University, Paris, France
| | - Dominique Stoppa-Lyonnet
- Department of Genetics, Institut Curie, Paris, France
- Inserm U830, Institut Curie, Paris, France
- Paris Cité University, Paris, France
| | - Franck Bourdeaut
- PSL Research University, Paris, France
- Inserm U830, Institut Curie, Paris, France
- SIREDO Oncology Center (Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer), Institut Curie, Paris, France
| | - Afane Brahimi
- Department of Clinical Genetics, Lille University Hospital, Lille, France
| | - Olivier Ingster
- Department of Genetics, Centre Angers University Hospital, Angers, France
| | | | - Sarah A. Jackson
- Department of Pediatrics, The University of Chicago, Chicago, Illinois
| | - Mark M. Sasaki
- Department of Pediatrics, The University of Chicago, Chicago, Illinois
| | - Trent Fowler
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
- Peel Therapeutics, Inc., Salt Lake City, Utah
| | - Anita Ng
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York
| | - Ryan J. Corbett
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Rebecca S. Kaufman
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | | | - Kuan-lin Huang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sharon J. Diskin
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jo Lynne Rokita
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, District of Columbia
| | - Hussam Al-Kateb
- Department of Lab Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Rose B. McGee
- Division of Cancer Predisposition, Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Joshua D. Schiffman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
- Peel Therapeutics, Inc., Salt Lake City, Utah
- Department of Pediatrics, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Kenneth S. Chen
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Douglas R. Stewart
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - D. Williams Parsons
- Department of Pediatrics, Baylor College of Medicine, Houston Texas
- Texas Children’s Cancer and Hematology Center, Texas Children’s Hospital, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Sharon E. Plon
- Department of Pediatrics, Baylor College of Medicine, Houston Texas
- Texas Children’s Cancer and Hematology Center, Texas Children’s Hospital, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Kris Ann P. Schultz
- International Pleuropulmonary Blastoma/DICER1 Registry, Minneapolis, Minnesota
| | - Kenan Onel
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Department of Clinical Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Center for Precision Oncology and Cancer Prevention, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
12
|
Chen LT, Jager M, Rebergen D, Brink GJ, van den Ende T, Vanderlinden W, Kolbeck P, Pagès-Gallego M, van der Pol Y, Besselink N, Moldovan N, Hami N, Kloosterman WP, van Laarhoven H, Mouliere F, Zweemer R, Lipfert J, Derks S, Marcozzi A, de Ridder J. Nanopore-based consensus sequencing enables accurate multimodal tumor cell-free DNA profiling. Genome Res 2025; 35:886-899. [PMID: 39805703 PMCID: PMC12047234 DOI: 10.1101/gr.279144.124] [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: 02/22/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Shallow genome-wide cell-free DNA sequencing holds great promise for noninvasive cancer monitoring by providing reliable copy number alteration (CNA) and fragmentomic profiles. Single-nucleotide variations (SNVs) are, however, much harder to identify with low sequencing depth due to sequencing errors. Here, we present Nanopore Rolling Circle Amplification (RCA)-enhanced Consensus Sequencing (NanoRCS), which leverages RCA and consensus calling based on genome-wide long-read nanopore sequencing to enable simultaneous multimodal tumor fraction (TF) estimation through SNVs, CNAs, and fragmentomics. The efficacy of NanoRCS is tested on 18 cancer patient samples and seven healthy controls, demonstrating its ability to reliably detect TFs as low as 0.24%. In vitro experiments confirm that SNV measurements are essential for detecting TFs below 3%. NanoRCS provides an opportunity for cost-effective and rapid sample processing, which aligns well with clinical needs, particularly in settings where quick and accurate cancer monitoring is essential for personalized treatment strategies.
Collapse
Affiliation(s)
- Li-Ting Chen
- Center for Molecular Medicine University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Myrthe Jager
- Center for Molecular Medicine University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | | | - Geertruid J Brink
- Department of Gynecologic Oncology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Tom van den Ende
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, 1105 AZ, Amsterdam, The Netherlands
| | - Willem Vanderlinden
- Soft Condensed Matter and Biophysics, Department of Physics and Debye Institute for Nanomaterials Science, Utrecht University, 3584 CC Utrecht, The Netherlands
- School of Physics and Astronomy, University of Edinburgh, EH9 3FD Edinburgh, United Kingdom
| | - Pauline Kolbeck
- Soft Condensed Matter and Biophysics, Department of Physics and Debye Institute for Nanomaterials Science, Utrecht University, 3584 CC Utrecht, The Netherlands
- Department of Physics and Center for NanoScience, LMU Munich, 80799 Munich, Germany
| | - Marc Pagès-Gallego
- Center for Molecular Medicine University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Ymke van der Pol
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Nicolle Besselink
- Center for Molecular Medicine University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
- Oncode Institute, 3521 AL Utrecht, The Netherlands
| | - Norbert Moldovan
- Cancer Center Amsterdam, Imaging and Biomarkers, 1105 AZ, Amsterdam, The Netherlands
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Nizar Hami
- Department of Gynecologic Oncology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | | | - Hanneke van Laarhoven
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, 1105 AZ, Amsterdam, The Netherlands
| | - Florent Mouliere
- Cancer Center Amsterdam, Imaging and Biomarkers, 1105 AZ, Amsterdam, The Netherlands
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1105 AZ, Amsterdam, The Netherlands
- Cancer Research UK National Biomarker Centre, University of Manchester, Manchester M20 4BX, United Kingdom
| | - Ronald Zweemer
- Department of Gynecologic Oncology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Jan Lipfert
- Soft Condensed Matter and Biophysics, Department of Physics and Debye Institute for Nanomaterials Science, Utrecht University, 3584 CC Utrecht, The Netherlands
| | - Sarah Derks
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | | | - Jeroen de Ridder
- Center for Molecular Medicine University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands;
- Oncode Institute, 3521 AL Utrecht, The Netherlands
- Cyclomics, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
13
|
Wang Y, Yu A, Gao Z, Yuan X, Du X, Shi P, Guan H, Wen S, Wang H, Wang L, Fan B, Liu Z. TET2 gene mutation status associated with poor prognosis of transition zone prostate cancer: a retrospective cohort study based on whole exome sequencing and machine learning models. Front Endocrinol (Lausanne) 2025; 16:1568665. [PMID: 40297177 PMCID: PMC12034557 DOI: 10.3389/fendo.2025.1568665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Background Prostate cancer (PCa) in the transition zone (TZ) is uncommon and often poses challenges for early diagnosis, but its genomic determinants and therapeutic vulnerabilities remain poorly characterized. Methods Tumor mutational landscape was characterized in nine patients with TZ PCa, identifying somatic variants through whole-exome sequencing (WES). Novel candidate variants relevant to driver gene were selected using rare-variant burden analysis. Kaplan-Meier curves with log-rank testing and Cox regression models were applied to evaluate the prognostic significance of selected mutant driver gene and clinicopathological characteristics in a cohort of 132 patients with TZ PCa. Significant prognostic determinants were integrated into a validated nomogram for individualized prediction of 3-, 4-, and 5-year biochemical recurrence-free survival (BRFS) and overall survival (OS) probabilities. Eight machine learning algorithms were employed to develop BRFS and OS prediction models in a cohort. Results A total of 5,036 somatic single nucleotide variants (SNVs) and 587 somatic insertion and deletion (INDELs) were discovered. Among eight driver gene mutations which were verified through Sanger sequencing, TET2 gene, with high mutation frequency and potential targeted drug relevance (bromodomain inhibitors and DOT1L inhibitors) was selected for further validation. Retrospective cohort study demonstrated that TET2 mutant status was significantly associated with Gleason score (p = 0.004) and distant metastasis (p = 0.002). Furthermore, TET2 mutant status was significantly correlated with inferior BRFS and OS and served as an independent predictor. Comparative evaluation of eight algorithms revealed the GBM model achieved superior discriminative ability for BRFS (AUC for 3-year: 0.752, 4-year: 0.786, 5-year: 0.796). The predictive model based on the GBM machine learning algorithm achieved the best predictive performance for OS (AUC for 3-year: 0.838, 4-year: 0.915, 5-year: 0.868). The constructed predictive nomogram provided evidence that TET2 mutant status integration conferred statistically significant improvements in model accuracy and clinical predictive value. Conclusion Our study elucidated the distinct genetic basis of prostate cancer in the transition zone and identified TET2 mutation as an independent prognostic determinant in TZ PCa. However, the limited sample size of this study necessitates cautious interpretation of these findings, and further validation in larger cohorts is warranted to confirm their generalizability.
Collapse
Affiliation(s)
- Yutong Wang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Colorectal Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning, China
| | - Ailing Yu
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- First Clinical College, Dalian Medical University, Dalian, Liaoning, China
| | - Ziping Gao
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xiaoying Yuan
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaochen Du
- College of Humanities and Social Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Peng Shi
- Second Clinical College, Dalian Medical University, Dalian, Liaoning, China
| | - Haoyun Guan
- Second Clinical College, Dalian Medical University, Dalian, Liaoning, China
| | - Shuang Wen
- Department of Pathology, Dalian Friendship Hospital, Dalian, China
| | - Honglong Wang
- Department of Pathology, Dalian Friendship Hospital, Dalian, China
| | - Liang Wang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bo Fan
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhiyu Liu
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
14
|
George MM, Brennick CA, Hagymasi AT, Shcheglova TV, Al Seesi S, Rosales TJ, Baker BM, Mandoiu II, Srivastava PK. A frameshift-generated cancer neoepitope that controls tumor burden in prophylaxis as well as therapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf016. [PMID: 40209093 DOI: 10.1093/jimmun/vkaf016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/10/2025] [Indexed: 04/12/2025]
Abstract
Insertion or deletion of one or two base pairs within a coding region causes a frameshift, which has the potential to generate neoepitopes (InDel-generated neoepitopes) that lack a self-counterpart and are entirely novel. Despite the obvious appeal of InDel-generated neoepitopes, and the demonstration of such candidate neoepitopes that can elicit a CD8 T-cell response, no InDel-generated neoepitopes that actually control tumors in vivo have been reported thus far. Here, in a mouse colon carcinoma line, we identify 11 InDels, only one of which generates a neoepitope that elicits tumor control in vivo in models of prophylaxis as well as therapy. Although this neoepitope has no self-counterpart, it has a low affinity (IC50 33,937.60 nM) for its MHC I allele. Despite its low affinity for MHC I, this neoepitope elicits antitumor activity in vivo through CD8 T cells. Furthermore, CD8 T cells elicited by this InDel-generated neoepitope, like the neoepitopes created by point mutations, show notably less exhaustion than classical immunogenic epitopes. Ironically, this InDel-generated neoepitope follows the same rules as noted for most of the tumor control-mediating neoepitopes generated by point mutations that have a poor affinity for MHC I alleles.
Collapse
Affiliation(s)
- Mariam M George
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
- Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Cory A Brennick
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
- Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Adam T Hagymasi
- Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Tatiana V Shcheglova
- Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Sahar Al Seesi
- Computer Science Department, Southern Connecticut State University, New Haven, CT, United States
| | - Tatiana J Rosales
- Harper Cancer Research Institute and the Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
| | - Brian M Baker
- Harper Cancer Research Institute and the Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, United States
| | - Ion I Mandoiu
- Department of Computer Science and Engineering, University of Connecticut, Storrs, CT, United States
| | - Pramod K Srivastava
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
- Carole and Ray Neag Comprehensive Cancer Center, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
15
|
Zhu G, Xie Y, Li L, Li R, Sun Y, Zhou T, Cun Y. Single-cell RNA sequencing reveals important role of monocytes and macrophages during mucopolysaccharidosis treatment. Sci Rep 2025; 15:12364. [PMID: 40210734 PMCID: PMC11986106 DOI: 10.1038/s41598-025-97330-7] [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: 12/31/2024] [Accepted: 04/03/2025] [Indexed: 04/12/2025] Open
Abstract
Mucopolysaccharidosis (MPS) encompasses a heterogeneous group of lysosomal storage diseases resulting from mutations in genes encoding lysosomal enzymes responsible for the degradation of mucopolysaccharides, also known as glycosaminoglycans (GAGs). Current therapeutic strategies for MPS include hematopoietic stem cell transplantation (HSCT), enzyme replacement therapy (ERT), and symptomatic therapy. This study investigated dynamic changes in MPS type II (MPS-II) through genomic and single-cell sequencing in a patient undergoing ERT. Analysis of peripheral blood mononuclear cells (PBMCs) from one MPS-II patient of 10 year old at different disease stages through scRNA-seq identified various immune cell types, including natural killer (NK) cells, NKT cells, CD4 + and CD8 + T cells, CD14 + and CD16 + monocytes, and B cells. Monocytes and macrophages were significantly reduced during the severe stage of MPS-II but increased during the recovery stage following ERT. Notably, monocyte subtype mono3 was exclusively expressed in the severe stage, while mono1_2, a subtype of mono1, was absent during the severe stage and exhibited distinct biological functions. These findings suggest that monocytes and macrophages play critical roles in the pathogenesis of MPS-II and in the response to ERT. Pseudotime, Gene Ontology, and cell-communication analyses revealed unique functions for the different cellular subtypes. Notably, key molecules mediating cellular interactions during ERT in MPS-II included CXCR3, PF4, APP, and C5AR1 in macrophages, RPS19 in T cells, HLA-DPB1 in B cells, ADRB2 in NK cells, and IL1B, C5AR1, RPS19, and TNFSF13B in monocytes. Overall, integrative analysis delineated the expression dynamics of various cell types and identified mutations in MPS-II, providing a comprehensive atlas of transcriptional programs, cellular characterizations, and genomic variation profiles in MPS-II. This dataset, along with advanced integrative analysis, represents a valuable resource for the discovery of drug targets and the improvement of therapeutic strategies for MPS-II.
Collapse
Affiliation(s)
- Gaohui Zhu
- Department of Endocrinology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yue Xie
- Pediatric Research Institute, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Li Li
- Department of Endocrinology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Rong Li
- Department of Endocrinology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yihong Sun
- Pediatric Research Institute, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Ting Zhou
- Department of Endocrinology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Yupeng Cun
- Pediatric Research Institute, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
16
|
Chen D, Lei T, Wang Y, Yu Z, Liu S, Ye L, Li W, Yang Q, Jin H, Liu F, Li Y. Isolation and Characterization of the Adamantinomatous Craniopharyngioma Primary Cells with Cancer-Associated Fibroblast Features. Biomedicines 2025; 13:912. [PMID: 40299526 PMCID: PMC12025058 DOI: 10.3390/biomedicines13040912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/31/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025] Open
Abstract
Backgrounds: Adamantinomatous craniopharyngiomas (ACPs) are benign intracranial tumors that behave aggressively due to their location, infiltration of the surrounding nervous tissue and high capacity for recurrence. In this study, we aimed to construct ACP primary cell models for further investigation of tumorigenic and recurrent mechanisms. Methods: Primary cells were isolated from primary (one case) and recurrent (one case) ACP. Short tandem repeat (STR) analysis was used to clarify the identity of the ACP primary cells we isolated. Whole exome sequencing (WES), immunofluorescence (IF) and immunohistochemistry (IHC) were performed on primary cells and corresponding ACP tissues, to determine the mutational profile and to clarify the tissue origin and phenotypic of primary cells. Transcriptome RNA-seq was performed to obtain the gene expression characteristics of ACP primary cells. Subsequently, a heterotopic ACP xenograft mouse model was established to confirm the tumorigenesis capacity of ACP primary cells. Results: ACP primary cells were successfully cultured. The genetic variants were similar to the original tumor tissue, and they owned expression of cancer-associated fibroblast (CAF) markers (FSP1/S100A4, Vimentin) and nuclear translocation β-catenin. Meanwhile, they had an high level expression of extracellular matrix components (Fibronectin). The tumor formation ability of ACP primary cells was verified. The transcriptional signatures of ACP primary cells were also explored. Conclusions: We successfully isolated and characterized ACP primary cells that acquired multiple CAF features and demonstrated stable propagation through dozens of passages. These PDC models laid the foundation for further research on ACP.
Collapse
Affiliation(s)
- Dongting Chen
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.)
| | - Ting Lei
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China; (T.L.)
| | - Yulin Wang
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.)
| | - Zaitao Yu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China; (T.L.)
| | - Siqi Liu
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.)
| | - Ling Ye
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.)
| | - Wanfang Li
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Union-Genius Pharmaceutical Technology Development Co., Ltd., Beijing 100176, China
| | - Qin Yang
- Beijing Union-Genius Pharmaceutical Technology Development Co., Ltd., Beijing 100176, China
| | - Hongtao Jin
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Union-Genius Pharmaceutical Technology Development Co., Ltd., Beijing 100176, China
| | - Fangjun Liu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China; (T.L.)
| | - Yan Li
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.)
| |
Collapse
|
17
|
Farooq AR, Zhang AX, Chan-Seng-Yue M, Topham JT, O'Kane GM, Jang GH, Fischer S, Dodd A, Holter S, Wilson J, Grant RC, Aung KL, Zogopoulos G, Elimova E, Prince R, Jang R, Moore M, Biagi J, Tang P, Goodwin R, Bathe OF, Marra M, Laskin J, Renouf DJ, Schaeffer DF, Karasinska JM, Notta F, Gallinger S, Knox JJ, Tsang ES. The tandem duplicator phenotype may be a novel targetable subgroup in pancreatic cancer. NPJ Precis Oncol 2025; 9:100. [PMID: 40185871 PMCID: PMC11971333 DOI: 10.1038/s41698-025-00888-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
Tandem duplicator phenotype (TDP) consists of distinct genomic rearrangements where tandem duplications are randomly distributed. In this study, we characterized the prevalence and outcomes of TDP in a large series of prospectively sequenced tumors from patients with pancreatic ductal adenocarcinomas (PDAC). Whole-genome sequencing (WGS) was performed in 530 PDAC cases from the PanCuRx Initiative, COMPASS and PanGen/POG trials in Canada. Of 530 cases, 52 were identified as TDP (9.8%; 13 resected, 39 advanced). Etiological subgroups of TDP included BRCA1 (n = 9), CCNE1 (n = 4), and unknown (n = 39). Presence of TDP was not prognostic in resected specimens (p = 0.77) compared with non-HRD and non-TDP cases, described as typicals. In advanced cases, when stratified for only classical subtype cases, platinum therapy was correlated with longer response in non-BRCA1 TDP vs. typicals (p = 0.0036). There was no difference in overall survival between TDP and typicals (p = 0.5).TDP represents a potential novel targetable subgroup for chemotherapy selection in PDAC.
Collapse
Affiliation(s)
| | - Amy X Zhang
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | - James T Topham
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | | | - Gun Ho Jang
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | - Anna Dodd
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Spring Holter
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Julie Wilson
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | | | | | - Elena Elimova
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | | | - Raymond Jang
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Malcolm Moore
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - James Biagi
- Cancer Centre of Southeastern Ontario/Queen's University, Kingston, ON, Canada
| | - Patricia Tang
- Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Rachel Goodwin
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital, The University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Oliver F Bathe
- Department of Oncology, Tom Baker Cancer Center, University of Calgary, 1331 29th St NW, Calgary, AB, T2N 4N2, Canada
| | - Marco Marra
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - Janessa Laskin
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - Daniel J Renouf
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - David F Schaeffer
- Department of Pathology, Vancouver General Hospital, Vancouver, BC, Canada
| | | | - Faiyaz Notta
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | | | - Erica S Tsang
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
18
|
Van Alsten SC, Love MI, Calhoun BC, Butler EN, Perou CM, Hoadley KA, Troester MA. Genomic Analysis Reveals Racial and Age-Related Differences in the Somatic Landscape of Breast Cancer and the Association with Socioeconomic Factors. Cancer Res 2025; 85:1327-1340. [PMID: 39879109 PMCID: PMC12034101 DOI: 10.1158/0008-5472.can-24-1788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/27/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025]
Abstract
Cancer genomics consortia have identified somatic drivers of breast cancer subtypes. However, these studies have predominantly included older, non-Black women, and the related socioeconomic status (SES) data are limited. Increased representation and depth of social data are crucial for understanding how health inequity is intertwined with somatic landscapes. Here, we conducted targeted sequencing on primary tumors from the Carolina Breast Cancer Study (N = 357; 52% Black; 47% <50) and compared the results with The Cancer Genome Atlas (N = 948; 18% Black; 27% <50). Race (Black vs. non-Black), age, and SES were evaluated in association with mutations, copy number alterations, and aneuploidy using generalized linear models. Pathway dysfunction was also assessed by aggregating mutation and copy number alterations. Adjusting for age, Black participants (N = 350) were significantly more likely to have TP53 and FAT1 mutations and less likely to have PIK3CA, CDH1, DDR2, and GATA3 mutations than non-Black participants. Younger participants had more GATA3 alterations and fewer KMT2C, PTEN, MAP3K1, and CDH1 alterations. Black participants had significant enrichment for MYC (8q) and PIK3CA (3q26) amplifications and higher total aneuploidy, but age was not associated with copy number variation. SES was associated with different patterns of alteration in Black versus non-Black women. Overall, Black participants showed modest differences in TP53, PIK3CA, and other alterations that further varied by SES. Race is a social construct, and varying distributions of etiologic factors across social strata may predispose Black, young, and low SES women to cancer subtypes characterized by these alterations. Significance: The collection and analysis of DNA sequencing with comprehensive socioeconomic factor associations in a large Black breast cancer patient cohort could help uncover mechanisms by which social conditions contribute to tumor biology.
Collapse
Affiliation(s)
- Sarah C. Van Alsten
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Michael I. Love
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Benjamin C. Calhoun
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Eboneé N. Butler
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Charles M. Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Katherine A. Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Melissa A. Troester
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
19
|
Noguchi R, Yanagihara K, Iino Y, Komatsu T, Kubo T, Ono T, Osaki J, Adachi Y, Iwata S, Shiota Y, Seyama T, Kondo T. Establishment and characterization of novel cancer cachexia-inducing cell line, Aku60GC, of scirrhous gastric cancer. Hum Cell 2025; 38:82. [PMID: 40178664 DOI: 10.1007/s13577-025-01208-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 03/10/2025] [Indexed: 04/05/2025]
Abstract
Cancer cachexia is a pathological state characterized by severe weight loss, skeletal muscle depletion, and adipose tissue reduction. Cancer cachexia is observed in gastric cancer (GC) with a higher incidence over 80%. Approximately 80% patients with advanced GC including scirrhous gastric cancer (SGC), which has the worst prognosis among all GC, are affected with cachexia. The exact pathophysiology in SGC cancer cachexia remains elusive, and therapeutic approaches for the cancer cachexia have not been established. Patient-derived cancer cachexia models are promising for elucidating the underlying mechanisms of disease progression and developing novel treatments, none of which originate from SGC. Therefore, we established a novel cancer cachexia-inducing cell line, designated Aku60GC, through stepwise selection of a patient-derived SGC cell line, HSC-60. Subcutaneous implantation of the Aku60GC cells into nude mice resulted in weight loss, muscle atrophy, and adipose tissue depletion with high reproducibility, accompanied by elevation of the circulating cytokines IL-8 and IL-18. Compared to parental HSC-60 cells, Aku60GC cells exhibited additional genomic changes, such as AKT2 and CCNE1 gains, a somatic mutation of RUNX1, and accelerated growth. Thus, our results demonstrate that the Aku60GC cell line is a valuable resource for research on cancer cachexia in SGC.
Collapse
Affiliation(s)
- Rei Noguchi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kazuyoshi Yanagihara
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
- Department of Life Sciences, Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima-shi, Hiroshima, 731-0153, Japan.
- Biospecimen Laboratories, Inc., 1-5-10-105 Nakamagome, Ohta-ku, Tokyo, 143-0027, Japan.
| | - Yuki Iino
- Exploratory Oncology and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
| | - Teruo Komatsu
- Exploratory Oncology and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
| | - Takanori Kubo
- Department of Life Sciences, Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima-shi, Hiroshima, 731-0153, Japan
| | - Takuya Ono
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Julia Osaki
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yuki Adachi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shuhei Iwata
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yomogi Shiota
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Toshio Seyama
- Department of Life Sciences, Faculty of Pharmacy, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami-ku, Hiroshima-shi, Hiroshima, 731-0153, Japan
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
20
|
Zhao H, Lin N, Ho VWS, Liu K, Chen X, Wu H, Chiu PK, Huang L, Dantes Z, Wong K, Chau H, Ko IC, Wong CH, Leung DK, Yuen SK, Wu D, Ding X, Ng CF, Teoh JY. Patient-Derived Bladder Cancer Organoids as a Valuable Tool for Understanding Tumor Biology and Developing Personalized Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414558. [PMID: 39921252 PMCID: PMC11967763 DOI: 10.1002/advs.202414558] [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: 11/08/2024] [Revised: 01/15/2025] [Indexed: 02/10/2025]
Abstract
Bladder cancer (BC) is a heterogeneous disease with high recurrence rates and variable treatment responses. To address these clinical challenges, the world's first bladder cancer patient-derived organoids (PDOs) biobank is established based on an Asian population. Thirty-six BC-PDOs are generated from 56 patients and demonstrated that the BC-PDOs can replicate the histological and genomic features of parental tumors. Drug screening tests are conducted with a broad spectrum of conventional chemotherapeutic and targeted therapy drugs and identified differential drug sensitivities among the BC-PDOs. These in vitro results are consistently supported by the PDO xenograft animal studies and patients' clinical treatment outcomes, thereby verifying the predictive power of PDOs for drug responses in BC patients. By analyzing the genetic profiles of the PDOs, specific driver genes that correlate with drug sensitivity to two stand-of-care chemotherapeutics, cisplatin, and gemcitabine, are identified. Additionally, the practicality of PDOs in investigating the tumor microenvironment has been demonstrated. This study underscores the utility of PDOs in advancing the understanding of bladder cancer and the development of personalized therapeutic strategies. The BC-PDOs biobank provides an ideal preclinical platform for supporting the development of personalized treatment strategies for BC patients. This study also provides insights into the potential mechanisms of drug resistance, paves the way for subsequent region-specific research, and demonstrates the possibility of using PDO-related models to direct future research in developing drugs targeting tumor microenvironments.
Collapse
Affiliation(s)
- Hongda Zhao
- S.H. Ho Urology CentreDepartment of SurgeryThe Chinese University of Hong KongHong Kong999077China
- Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Na Lin
- Department of Biomedical SciencesFaculty of Health SciencesUniversity of MacauTaipaMacaoSAR999078China
| | - Vincy Wing Sze Ho
- S.H. Ho Urology CentreDepartment of SurgeryThe Chinese University of Hong KongHong Kong999077China
- Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Kang Liu
- S.H. Ho Urology CentreDepartment of SurgeryThe Chinese University of Hong KongHong Kong999077China
- Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Xuan Chen
- S.H. Ho Urology CentreDepartment of SurgeryThe Chinese University of Hong KongHong Kong999077China
- Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Hongwei Wu
- S.H. Ho Urology CentreDepartment of SurgeryThe Chinese University of Hong KongHong Kong999077China
- Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
| | - Peter Ka‐Fung Chiu
- S.H. Ho Urology CentreDepartment of SurgeryThe Chinese University of Hong KongHong Kong999077China
| | - Linda Huang
- Invitrocue Hong Kong LtdHong KongSAR999077China
| | | | - Ka‐Leung Wong
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityHong Kong999077China
| | - Ho‐Fai Chau
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityHong Kong999077China
| | - Ivan Ching‐Ho Ko
- S.H. Ho Urology CentreDepartment of SurgeryThe Chinese University of Hong KongHong Kong999077China
| | - Chris Ho‐Ming Wong
- S.H. Ho Urology CentreDepartment of SurgeryThe Chinese University of Hong KongHong Kong999077China
| | - David Ka‐Wai Leung
- S.H. Ho Urology CentreDepartment of SurgeryThe Chinese University of Hong KongHong Kong999077China
| | - Steffi Kar‐Kei Yuen
- S.H. Ho Urology CentreDepartment of SurgeryThe Chinese University of Hong KongHong Kong999077China
| | - Dinglan Wu
- S.H. Ho Urology CentreDepartment of SurgeryThe Chinese University of Hong KongHong Kong999077China
| | - Xiaofan Ding
- Department of Biomedical SciencesFaculty of Health SciencesUniversity of MacauTaipaMacaoSAR999078China
| | - Chi Fai Ng
- S.H. Ho Urology CentreDepartment of SurgeryThe Chinese University of Hong KongHong Kong999077China
| | - Jeremy Yuen‐Chun Teoh
- S.H. Ho Urology CentreDepartment of SurgeryThe Chinese University of Hong KongHong Kong999077China
- Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong999077China
- Department of UrologyMedical University of ViennaVienna1090Austria
| |
Collapse
|
21
|
Blomain ES, Soudi S, Wang Z, Somani A, Subramanian A, Nouth SCL, Oladipo E, New C, Kenney DE, Nemat-Gorgani N, Kindler T, Avedian RS, Steffner RJ, Mohler DG, Hiniker SM, Chin AL, Kalbasi A, Binkley MS, Fried M, Gaida MM, van de Rijn M, Moding EJ. Evolutionary Pressures Shape Undifferentiated Pleomorphic Sarcoma Development and Radiotherapy Response. Cancer Res 2025; 85:1162-1174. [PMID: 39808162 DOI: 10.1158/0008-5472.can-24-3281] [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: 09/10/2024] [Revised: 10/19/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
Radiotherapy is an integral component in the treatment of many types of cancer, with approximately half of patients with cancer receiving radiotherapy. Systemic therapy applies pressure that can select for resistant tumor subpopulations, underscoring the importance of understanding how radiation impacts tumor evolution to improve treatment outcomes. We integrated temporal genomic profiling of 120 spatially distinct tumor regions from 20 patients with undifferentiated pleomorphic sarcomas (UPS), longitudinal circulating tumor DNA analysis, and evolutionary biology computational pipelines to study UPS evolution during tumorigenesis and in response to radiotherapy. Most unirradiated UPSs displayed initial linear evolution, followed by subsequent branching evolution with distinct mutational processes during early and late development. Metrics of genetic divergence between regions provided evidence of strong selection pressures during UPS development that further increased during radiotherapy. Subclone abundance changed after radiotherapy with subclone contraction tied to alterations in calcium signaling, and inhibiting calcium transporters radiosensitized sarcoma cells. Finally, circulating tumor DNA analysis accurately measured subclone abundance and enabled noninvasive monitoring of subclonal changes. These results demonstrate that radiation exerts selective pressures on UPSs and suggest that targeting radioresistant subclonal populations could improve outcomes after radiotherapy. Significance: Radiotherapy mediates tumor evolution by leading to the expansion of resistant subclonal cancer cell populations, indicating that developing approaches to target resistant subclones will be crucial to improve radiotherapy response.
Collapse
Affiliation(s)
- Erik S Blomain
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Shaghayegh Soudi
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Ziwei Wang
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Anish Somani
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Ajay Subramanian
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Serey C L Nouth
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Eniola Oladipo
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Christin New
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Deborah E Kenney
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Neda Nemat-Gorgani
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Thomas Kindler
- Department of Internal Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- TRON, Translational Oncology at the University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Raffi S Avedian
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Robert J Steffner
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - David G Mohler
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Susan M Hiniker
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Alexander L Chin
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Anusha Kalbasi
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Michael S Binkley
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Marius Fried
- Department of Internal Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias M Gaida
- TRON, Translational Oncology at the University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Pathology, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matt van de Rijn
- Department of Pathology, Stanford University, Stanford, California
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, California
- Stanford Cancer Institute, Stanford University, Stanford, California
| |
Collapse
|
22
|
Sethna Z, Guasp P, Reiche C, Milighetti M, Ceglia N, Patterson E, Lihm J, Payne G, Lyudovyk O, Rojas LA, Pang N, Ohmoto A, Amisaki M, Zebboudj A, Odgerel Z, Bruno EM, Zhang SL, Cheng C, Elhanati Y, Derhovanessian E, Manning L, Müller F, Rhee I, Yadav M, Merghoub T, Wolchok JD, Basturk O, Gönen M, Epstein AS, Momtaz P, Park W, Sugarman R, Varghese AM, Won E, Desai A, Wei AC, D'Angelica MI, Kingham TP, Soares KC, Jarnagin WR, Drebin J, O'Reilly EM, Mellman I, Sahin U, Türeci Ö, Greenbaum BD, Balachandran VP. RNA neoantigen vaccines prime long-lived CD8 + T cells in pancreatic cancer. Nature 2025; 639:1042-1051. [PMID: 39972124 PMCID: PMC11946889 DOI: 10.1038/s41586-024-08508-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 12/10/2024] [Indexed: 02/21/2025]
Abstract
A fundamental challenge for cancer vaccines is to generate long-lived functional T cells that are specific for tumour antigens. Here we find that mRNA-lipoplex vaccines against somatic mutation-derived neoantigens may solve this challenge in pancreatic ductal adenocarcinoma (PDAC), a lethal cancer with few mutations. At an extended 3.2-year median follow-up from a phase 1 trial of surgery, atezolizumab (PD-L1 inhibitory antibody), autogene cevumeran1 (individualized neoantigen vaccine with backbone-optimized uridine mRNA-lipoplex nanoparticles) and modified (m) FOLFIRINOX (chemotherapy) in patients with PDAC, we find that responders with vaccine-induced T cells (n = 8) have prolonged recurrence-free survival (RFS; median not reached) compared with non-responders without vaccine-induced T cells (n = 8; median RFS 13.4 months; P = 0.007). In responders, autogene cevumeran induces CD8+ T cell clones with an average estimated lifespan of 7.7 years (range 1.5 to roughly 100 years), with approximately 20% of clones having latent multi-decade lifespans that may outlive hosts. Eighty-six percent of clones per patient persist at substantial frequencies approximately 3 years post-vaccination, including clones with high avidity to PDAC neoepitopes. Using PhenoTrack, a novel computational strategy to trace single T cell phenotypes, we uncover that vaccine-induced clones are undetectable in pre-vaccination tissues, and assume a cytotoxic, tissue-resident memory-like T cell state up to three years post-vaccination with preserved neoantigen-specific effector function. Two responders recurred and evidenced fewer vaccine-induced T cells. Furthermore, recurrent PDACs were pruned of vaccine-targeted cancer clones. Thus, in PDAC, autogene cevumeran induces de novo CD8+ T cells with multiyear longevity, substantial magnitude and durable effector functions that may delay PDAC recurrence. Adjuvant mRNA-lipoplex neoantigen vaccines may thus solve a pivotal obstacle for cancer vaccination.
Collapse
MESH Headings
- Humans
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/cytology
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/therapy
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/surgery
- Cancer Vaccines/immunology
- Cancer Vaccines/genetics
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/therapeutic use
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/therapy
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/surgery
- Female
- Fluorouracil/therapeutic use
- Fluorouracil/administration & dosage
- Antibodies, Monoclonal, Humanized
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Male
- Oxaliplatin
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- Middle Aged
- Follow-Up Studies
- Disease-Free Survival
- Immunologic Memory
- Aged
- Irinotecan
- Leucovorin
Collapse
Affiliation(s)
- Zachary Sethna
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- The Olayan Center for Cancer Vaccines, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pablo Guasp
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charlotte Reiche
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martina Milighetti
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicholas Ceglia
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Erin Patterson
- The Olayan Center for Cancer Vaccines, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jayon Lihm
- The Olayan Center for Cancer Vaccines, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - George Payne
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Olga Lyudovyk
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luis A Rojas
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nan Pang
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Akihiro Ohmoto
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Masataka Amisaki
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Abderezak Zebboudj
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zagaa Odgerel
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emmanuel M Bruno
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Siqi Linsey Zhang
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charlotte Cheng
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yuval Elhanati
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | - Ina Rhee
- Genentech, San Francisco, CA, USA
| | | | - Taha Merghoub
- Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Jedd D Wolchok
- Meyer Cancer Center, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Olca Basturk
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew S Epstein
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Parisa Momtaz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wungki Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryan Sugarman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anna M Varghese
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elizabeth Won
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Avni Desai
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alice C Wei
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael I D'Angelica
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - T Peter Kingham
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin C Soares
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William R Jarnagin
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeffrey Drebin
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Ugur Sahin
- BioNTech, Mainz, Germany
- HI-TRON, Helmholtz Institute for Translational Oncology, Mainz, Germany
| | - Özlem Türeci
- BioNTech, Mainz, Germany
- HI-TRON, Helmholtz Institute for Translational Oncology, Mainz, Germany
| | - Benjamin D Greenbaum
- The Olayan Center for Cancer Vaccines, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Vinod P Balachandran
- Immuno-Oncology Service, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Hepatopancreatobiliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- The Olayan Center for Cancer Vaccines, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
23
|
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: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
24
|
Ma J, Shah R, Bell AC, McDermott N, Pei X, Selenica P, Haseltine J, Delsite R, Khan AJ, Lok BH, Ellis MJ, Aft RF, Setton J, Reis-Filho JS, Riaz N, Powell SN. Increased Synthetic Cytotoxicity of Combinatorial Chemoradiation Therapy in Homologous Recombination Deficient Tumors. Int J Radiat Oncol Biol Phys 2025; 121:768-779. [PMID: 38997095 PMCID: PMC11717987 DOI: 10.1016/j.ijrobp.2024.06.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 06/10/2024] [Accepted: 06/29/2024] [Indexed: 07/14/2024]
Abstract
PURPOSE Homologous recombination deficient (HRD) tumors are exquisitely sensitive to platinum-based chemotherapy and when combined with radiation therapy (RT), leads to improved overall survival in multiple cancer types. Whether a subset of tumors with distinct molecular characteristics demonstrate increased benefit from cisplatin and RT (c-RT) is unclear. We hypothesized that HRD tumors, whether associated with BRCA mutations or genomic scars of HRD, exhibit exquisite sensitivity to c-RT, and that HRD may be a significant driver of c-RT benefit. METHODS AND MATERIALS Sensitivity to c-RT was examined using isogenic and sporadic breast cancer cell lines. HRD was assessed using 4 assays: RT-induced Rad51 foci, a DR-GFP reporter assay, a genomic scar score (large-scale state transitions [LST]), and clonogenic survival assays. Whole-genome sequencing of 4 breast tumors from a phase 2 clinical trial of neoadjuvant c-RT in triple-negative breast cancer was performed and HRD was defined using HRDetect. RESULTS BRCA1/2 deficient cell lines displayed functional HRD based on the Rad51 functional assay, with c-RT to RT or cisplatin interaction ratios (IR) of 1.11 and 26.84 for the BRCA1 isogenic pair at 2 μM cisplatin and 6 Gy, respectively. The highest LST lines demonstrated HRD and synthetic cytotoxicity to c-RT with IR at 2 Gy and cisplatin 20 μM of 7.50, and the lowest LST line with IR of 0.65. Of 4 evaluable patients in the phase 2 trial, one achieved a pathologic complete response with corresponding HRD based on multiple genomic scar scores including HRDetect and LST scores, compared with patients without a pathologic complete response. CONCLUSIONS HRD breast cancers, whether identified by BRCA1/2 mutation status, functional tests, or mutational signatures, appear to be significantly more sensitive to c-RT compared with isogenic controls or tumors without HRD mutational signatures. HRD tumors may be exquisitely sensitive to c-RT which warrants further clinical investigation to guide a precision oncology approach.
Collapse
Affiliation(s)
- Jennifer Ma
- Departments of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rachna Shah
- Departments of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew C Bell
- Departments of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Niamh McDermott
- Departments of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Xin Pei
- Departments of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pier Selenica
- Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Justin Haseltine
- Departments of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert Delsite
- Departments of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Atif J Khan
- Departments of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Benjamin H Lok
- Departments of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Departments of Radiation Oncology; Medical Biophysics; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Matthew J Ellis
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Rebecca F Aft
- Department of General Surgery, Washington University, St Louis, Missouri
| | - Jeremy Setton
- Departments of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Nadeem Riaz
- Departments of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Simon N Powell
- Departments of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
25
|
Kim SR, Oldfield L, Kim RH, Espin‐Garcia O, Han K, Vicus D, Eiriksson L, Tone A, Pollett A, Cesari M, Clarke B, Bernardini MQ, Pugh TJ, Ferguson SE. Molecular Classification of Endometrial Cancers Using an Integrative DNA Sequencing Panel. J Surg Oncol 2025; 131:734-741. [PMID: 39501493 PMCID: PMC12065444 DOI: 10.1002/jso.27973] [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: 06/06/2024] [Revised: 08/08/2024] [Accepted: 10/12/2024] [Indexed: 05/11/2025]
Abstract
BACKGROUND AND OBJECTIVES Adoption of molecular classification in endometrial cancer (EC) into clinical practice remains challenging due to complexity in coordination of multiple assays. We aimed to develop a simple molecular technique to classify ECs into four subgroups using our custom-designed targeted sequencing panel. METHODS Patients with newly diagnosed ECs were prospectively recruited from three cancer centres in Ontario, Canada. Using our panel, 181 ECs were sequenced. Variants were analysed for pathogenicity and clinicopathologic information was collected through medical records retrospectively. RESULTS Of 181, 86 (48%) were mismatch repair deficient (MMRd), of which 62 (72%) harboured MLH1 promoter methylation and 24 (28%) had pathogenic variants in MMR genes. Of single classifiers, three (1.8%) had pathogenic POLE (POLEmut), 15 (9%) had TP53 mutations (p53abn) and 61 (37%) had no specific molecular profile subtype (NSMP). Sixteen (9%) had more than one molecular classifying feature, with eight (4%) MMRd-p53abn, six (3%) POLEmut-MMRd, one (0.5%) POLEmut-MMRd-p53abn and one (0.5%) POLEmut-p53abn. When MMRd group was further subclassified according to mechanism of MMR loss, MLH1 promoter methylated group had worse outcomes than those with somatic MMR pathogenic variants. CONCLUSIONS Our panel can classify ECs into four subgroups through a simplified process and can be implemented reflexively in clinical practice.
Collapse
Affiliation(s)
- Soyoun R. Kim
- Division of Gynecologic OncologyPrincess Margaret Cancer Centre, Sinai Health Systems, University Health NetworkTorontoOntarioCanada
- Department of Obstetrics and GynaecologyUniversity of TorontoTorontoOntarioCanada
| | - Leslie Oldfield
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioCanada
| | - Raymond H. Kim
- Fred A Litwin Family Centre for Genetic MedicineUniversity Health NetworkTorontoOntarioCanada
- Zane Cohen Centre for Digestive DiseasesFamilial Gastrointestinal Cancer Registry, Mount Sinai HospitalTorontoOntarioCanada
- Division of Medical Oncology and HematologyPrincess Margaret Cancer Centre, Sinai Health Systems, University Health NetworkTorontoOntarioCanada
- Ontario Institute for Cancer ResearchUniversity Health NetworkTorontoOntarioCanada
| | - Osvaldo Espin‐Garcia
- Department of BiostatisticsPrincess Margaret Cancer Centre, University Health Network, University of TorontoTorontoOntarioCanada
| | - Kathy Han
- Department of Radiation OncologyUniversity of TorontoTorontoOntarioCanada
| | - Danielle Vicus
- Department of Obstetrics and GynaecologyUniversity of TorontoTorontoOntarioCanada
- Division of Gynecologic OncologyOdette Cancer CentreTorontoOntarioCanada
| | - Lua Eiriksson
- Division of Gynecologic OncologyJuravinski Cancer CentreHamiltonOntarioCanada
| | - Alicia Tone
- Division of Gynecologic OncologyPrincess Margaret Cancer Centre, Sinai Health Systems, University Health NetworkTorontoOntarioCanada
| | - Aaron Pollett
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Matthew Cesari
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Blaise Clarke
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Marcus Q. Bernardini
- Division of Gynecologic OncologyPrincess Margaret Cancer Centre, Sinai Health Systems, University Health NetworkTorontoOntarioCanada
- Department of Obstetrics and GynaecologyUniversity of TorontoTorontoOntarioCanada
| | - Trevor J. Pugh
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioCanada
- Ontario Institute for Cancer ResearchUniversity Health NetworkTorontoOntarioCanada
| | - Sarah E. Ferguson
- Division of Gynecologic OncologyPrincess Margaret Cancer Centre, Sinai Health Systems, University Health NetworkTorontoOntarioCanada
- Department of Obstetrics and GynaecologyUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
26
|
Gormley M, Adhikari A, Dudding T, Pring M, Hurley K, Macfarlane GJ, Lagiou P, Lagiou A, Polesel J, Agudo A, Alemany L, Ahrens W, Healy CM, Conway DI, Canova C, Holcatova I, Richiardi L, Znaor A, Olshan AF, Hung RJ, Liu G, Bratman S, Zhao X, Holt J, Cortez R, Gaborieau V, McKay JD, Waterboer T, Brennan P, Hayes N, Diergaarde B, Virani S. VOYAGER: an international consortium investigating the role of human papilloma virus and genetics in oral and oropharyngeal cancer risk and survival. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.17.25322399. [PMID: 40034767 PMCID: PMC11875266 DOI: 10.1101/2025.02.17.25322399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Head and neck cancer (HNC) is the sixth most common cancer globally. Incidence and survival rates vary significantly across geographic regions and tumor subsites. This is partly due to differences in risk factor exposure, which includes tobacco smoking, alcohol consumption and human papillomavirus (HPV) infection, alongside detection and treatment strategies. The VOYAGER (human papillomaVirus, Oral and oropharYngeal cAncer GEnomic Research) consortium is a collaboration between five large North American and European studies which generated data on 10,530 participants (7,233 cases and 3,297 controls). The primary goal of the collaboration was to improve understing of the role of HPV and genetic factors in oral cavity and oropharyngeal cancer risk and outcome. Demographic and clinical data collected by the five studies were harmonized, and HPV status was determined for the majority of cases. In addition, 999 tumors were sequenced to define somatic mutations. These activities generated a comprehensive biomedical resource that can be utilized to answer critical outsting research questions to help improve HNC prevention, early detection, treatment, and surveillance.
Collapse
Affiliation(s)
- M Gormley
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Bristol Dental School, University of Bristol, Bristol, UK
- University Hospitals Bristol NHS Foundation Trust Bristol Dental Hospital, Bristol, UK
| | - A Adhikari
- University Hospitals Bristol NHS Foundation Trust Bristol Dental Hospital, Bristol, UK
| | - T Dudding
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Bristol Dental School, University of Bristol, Bristol, UK
| | - M Pring
- Bristol Dental School, University of Bristol, Bristol, UK
- University Hospitals Bristol NHS Foundation Trust Bristol Dental Hospital, Bristol, UK
| | - K Hurley
- University Hospitals Bristol NHS Foundation Trust Bristol Dental Hospital, Bristol, UK
| | - G J Macfarlane
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, UK
| | - P Lagiou
- School of Medicine, National and Kapodistrian University of Athens, Greece
| | - A Lagiou
- School of Public Health, University of West Attica, Greece
| | - J Polesel
- Unit of Cancer Epidemiology, Centro di Riferimento Oncologico di Aviano (CRO) National Cancer Institute, IRCCS, Italy
| | - A Agudo
- Nutrition and Cancer Unit, Cancer Epidemiology Research Program, Catalan Institute of Oncology/IDIBELL, Barcelona, Spain
| | - L Alemany
- Infections and Cancer Unit, Cancer Epidemiology Research Program, Catalan Institute of Oncology/IDIBELL, Barcelona, Spain
- Centro de Investigación Biomédica en Red: Epidemiología y Salud Pública (CIBERESP CB06/02/0073), Madrid, Spain
| | - W Ahrens
- Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology - BIPS, Germany
| | - C M Healy
- School of Dental Science, Dublin Dental University Hospital, Trinity College Dublin, Irel
| | - D I Conway
- School of Medicine, Dentistry, and Nursing, University of Glasgow, UK
| | - C Canova
- Department of Cardiac, Thoracic and Vascular Sciences University of Padova, Italy
| | - I Holcatova
- Institute of Hygiene and Epidemiology, Charles University Prague, Czech Republic
| | - L Richiardi
- Reference Centre for Epidemiology and Cancer Prevention in Piemonte, Italy
| | - A Znaor
- Cancer Surveillance, International Agency for Research on Cancer, France
| | - A F Olshan
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, US
| | - R J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - G Liu
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Computational Biology and Medicine Program, Princess Margaret Cancer Centre, Toronto Canada
| | - S Bratman
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - X Zhao
- Department of Medicine, University of Tennessee, USA
| | - J Holt
- Department of Medicine, University of Tennessee, USA
| | - R Cortez
- Genomic Epidemiology Group, World Health Organization, International Agency for Research on Cancer, Lyon, France
| | - V Gaborieau
- Genomic Epidemiology Group, World Health Organization, International Agency for Research on Cancer, Lyon, France
| | - J D McKay
- Genomic Epidemiology Group, World Health Organization, International Agency for Research on Cancer, Lyon, France
| | - T Waterboer
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - P Brennan
- Genomic Epidemiology Group, World Health Organization, International Agency for Research on Cancer, Lyon, France
| | - N Hayes
- Department of Medicine, University of Tennessee, USA
| | - B Diergaarde
- Department of Human Genetics, School of Public Health, University of Pittsburgh, and UPMC Hillman Cancer Center, Pittsburgh, US
| | - S Virani
- Genomic Epidemiology Group, World Health Organization, International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
27
|
Shen X, Zuo L, Ye Z, Yuan Z, Huang K, Li Z, Yu Q, Zou X, Wei X, Xu P, Deng Y, Jin X, Xu X, Wu L, Zhu H, Qin P. Inferring cell trajectories of spatial transcriptomics via optimal transport analysis. Cell Syst 2025; 16:101194. [PMID: 39904341 DOI: 10.1016/j.cels.2025.101194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 09/30/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025]
Abstract
The integration of cell transcriptomics and spatial position to organize differentiation trajectories remains a challenge. Here, we introduce SpaTrack, which leverages optimal transport to reconcile both gene expression and spatial position from spatial transcriptomics into the transition costs, thereby reconstructing cell differentiation. SpaTrack can construct detailed spatial trajectories that reflect the differentiation topology and trace cell dynamics across multiple samples over temporal intervals. To capture the dynamic drivers of differentiation, SpaTrack models cell fate as a function of expression profiles influenced by transcription factors over time. By applying SpaTrack, we successfully disentangle spatiotemporal trajectories of axolotl telencephalon regeneration and mouse midbrain development. Diverse malignant lineages expanding within a primary tumor are uncovered. One lineage, characterized by upregulated epithelial mesenchymal transition, implants at the metastatic site and subsequently colonizes to form a secondary tumor. Overall, SpaTrack efficiently advances trajectory inference from spatial transcriptomics, providing valuable insights into differentiation processes.
Collapse
Affiliation(s)
- Xunan Shen
- BGI Research, Chongqing 401329, China; BGI Research, Beijing 102601, China
| | | | | | - Zhongyang Yuan
- BGI Research, Chongqing 401329, China; State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ke Huang
- BGI Research, Chongqing 401329, China
| | - Zeyu Li
- BGI Research, Chongqing 401329, China
| | - Qichao Yu
- BGI Research, Chongqing 401329, China
| | - Xuanxuan Zou
- BGI Research, Chongqing 401329, China; Department of Neurology, Hubei Provincial Clinical Research Center for Parkinson's Disease, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | | | - Ping Xu
- BGI Research, Chongqing 401329, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Yaqi Deng
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China
| | - Xin Jin
- BGI Research, Shenzhen 518083, China
| | - Xun Xu
- BGI Research, Shenzhen 518083, China.
| | - Liang Wu
- BGI Research, Chongqing 401329, China; BGI Research, Shenzhen 518083, China.
| | | | - Pengfei Qin
- BGI Research, Chongqing 401329, China; BGI Research, Shenzhen 518083, China.
| |
Collapse
|
28
|
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: 2] [Impact Index Per Article: 2.0] [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
|
29
|
Sun C, Yang X, Chen L, Bie Z, Kang R, Ai B, Ma J, Zheng Z, Liu H, Liu J, Zhong J, Yu J. A Multi-Center Real-World Study of Clinicopathologic Characteristics and Efficacy of the Malignant Mesothelioma in Chinese Population. Thorac Cancer 2025; 16:e15533. [PMID: 39940064 PMCID: PMC11821455 DOI: 10.1111/1759-7714.15533] [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: 12/01/2024] [Revised: 12/30/2024] [Accepted: 01/05/2025] [Indexed: 02/14/2025] Open
Abstract
OBJECTIVE Malignant mesothelioma (MM) is a rare malignant tumor. To explore the clinicopathological characteristics and efficacy of Chinese population with MM in the real-world. METHODS Two hundred and forty-eight patients diagnosed with MM between September 2007 and August 2024 from three large medical centers (Beijing Hospital, Peking University Cancer Hospital, and Chinese Academy of Medical Sciences Cancer Hospital) were retrospectively analyzed. Kaplan-Meier and Cox regression were performed. Breast cancer gene 1-associated protein 1 (BAP1) status was evaluated. RESULTS Chinese population with MM had a lower diagnostic age, higher proportion of youth and female, more advanced stage and lower expression of characteristic markers. The median progression-free survival (mPFS) and median overall survival (mOS) were 8.90 and 25.60 months for the first-line treatment, and 3.28 and 19.50 months for the second-line. The first-line immunotherapy provided a relatively higher objective response rate (33.3% vs. 20.5%, p = 0.402) and a trend to prolong mPFS (12.10 vs. 9.20 months, p = 0.345) and mOS (NA vs. 23.90, p = 0.185) compared with chemotherapy. Bevacizumab combined with chemotherapy relatively prolonged mPFS (10.47 vs. 7.93 months, p = 0.074) and mOS (31.30 vs. 23.20 months, p = 0.673) than chemotherapy alone. Carboplatin relatively improved mPFS than cisplatin (10.87 vs. 8.87 months, p = 0.185). Age and histologic type were predictors for PFS, and gender, histologic subtype, and CK5/6 were prognosis factors for OS. Briefly, 17.78% patients existed BAP1 deletions and correlated with OS benefit. CONCLUSION Chinese population with MM present unique clinicopathologic characteristics and could benefit from the first-line immunotherapy and bevacizumab combined with chemotherapy. Gender, histologic subtype, and CK5/6 are prognosis factors for OS. BAP1 deletions correlate with OS benefit.
Collapse
Affiliation(s)
- Chenrui Sun
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| | - Xue Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Lan Chen
- Department of Pathology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| | - Zhixin Bie
- Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| | - Runting Kang
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| | - Bin Ai
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical ScienceBeijingChina
| | - Junling Ma
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical ScienceBeijingChina
| | - Zitong Zheng
- Department of OncologyBinzhou Medical University HospitalBinzhouChina
| | - Haolan Liu
- Department of OncologyBinzhou Medical University HospitalBinzhouChina
| | - Juanjuan Liu
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| | - Jia Zhong
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences Peking Union Medical CollegeBeijingChina
| | - Jiangyong Yu
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
30
|
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 PMCID: PMC12121501 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] [Download PDF] [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
|
31
|
Walker R, Joo JE, Mahmood K, Clendenning M, Como J, Preston SG, Joseland S, Pope BJ, Medeiros ABD, Murillo BV, Pachter N, Sweet K, Spigelman AD, Groves A, Gleeson M, Bernatowicz K, Poplawski N, Andrews L, Healey E, Gallinger S, Grant RC, Win AK, Hopper JL, Jenkins MA, Torrezan GT, Rosty C, Macrae FA, Winship IM, Buchanan DD, Georgeson P. Adenomas from individuals with pathogenic biallelic variants in the MUTYH and NTHL1 genes demonstrate base excision repair tumour mutational signature profiles similar to colorectal cancers, expanding potential diagnostic and variant classification applications. Transl Oncol 2025; 52:102266. [PMID: 39793275 PMCID: PMC11774829 DOI: 10.1016/j.tranon.2024.102266] [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: 08/31/2024] [Revised: 11/11/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Colorectal cancers (CRCs) from people with biallelic germline likely pathogenic/pathogenic variants in MUTYH or NTHL1 exhibit specific single base substitution (SBS) mutational signatures, namely combined SBS18 and SBS36 (SBS18+SBS36), and SBS30, respectively. The aim was to determine if adenomas from biallelic cases demonstrated these mutational signatures at diagnostic levels. METHODS Whole-exome sequencing of FFPE tissue and matched blood-derived DNA was performed on 9 adenomas and 15 CRCs from 13 biallelic MUTYH cases, on 7 adenomas and 2 CRCs from 5 biallelic NTHL1 cases and on 27 adenomas and 26 CRCs from 46 non-hereditary (sporadic) participants. All samples were assessed for COSMIC v3.2 SBS mutational signatures. RESULTS In biallelic MUTYH cases, SBS18+SBS36 signature proportions in adenomas (mean±standard deviation, 65.6 %±29.6 %) were not significantly different to those observed in CRCs (76.2 % ± 20.5 %, p-value=0.37), but were significantly higher compared with non-hereditary adenomas (7.6 % ± 7.0 %, p-value=3.4 × 10-4). Similarly, in biallelic NTHL1 cases, SBS30 signature proportions in adenomas (74.5 %±9.4 %) were similar to those in CRCs (78.8 % ± 2.4 %) but significantly higher compared with non-hereditary adenomas (2.8 % ± 3.6 %, p-value=5.1 × 10-7). Additionally, a compound heterozygote with the c.1187G>A p.(Gly396Asp) pathogenic variant and the c.533G>C p.(Gly178Ala) variant of unknown significance (VUS) in MUTYH demonstrated high levels of SBS18+SBS36 in four adenomas and one CRC, providing evidence for reclassification of the VUS to pathogenic. CONCLUSIONS SBS18+SBS36 and SBS30 were enriched in adenomas at comparable proportions to those observed in CRCs from biallelic MUTYH and biallelic NTHL1 cases, respectively. Therefore, testing adenomas may improve the identification of biallelic cases and facilitate variant classification, ultimately enabling opportunities for CRC prevention.
Collapse
Affiliation(s)
- Romy Walker
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3010, Australia.
| | - Jihoon E Joo
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3010, Australia
| | - Khalid Mahmood
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3010, Australia; Melbourne Bioinformatics, The University of Melbourne, Melbourne, VIC, 3053, Australia
| | - Mark Clendenning
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3010, Australia
| | - Julia Como
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3010, Australia
| | - Susan G Preston
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3010, Australia
| | - Sharelle Joseland
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3010, Australia
| | - Bernard J Pope
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia; Melbourne Bioinformatics, The University of Melbourne, Melbourne, VIC, 3053, Australia
| | - Ana B D Medeiros
- Clinical and Functional Genomics Group, International Research Centre/CIPE, A.C. Camargo Cancer Centre, Sao Paulo, 01508-010, Brazil
| | - Brenely V Murillo
- Genetic Services of Western Australia, King Edward Memorial Hospital, Perth, WA, 6008, Australia
| | - Nicholas Pachter
- Genetic Services of Western Australia, King Edward Memorial Hospital, Perth, WA, 6008, Australia; Medical School, University of Western Australia, Perth, WA, 6009, Australia; School of Medicine, Curtin University, Perth, WA, 6845, Australia
| | - Kevin Sweet
- Division of Human Genetics, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Allan D Spigelman
- Hunter Family Cancer Service, Newcastle, NSW, 2298, Australia; St Vincent's Cancer Genetics Unit, Sydney, NSW, 2290, Australia; Surgical Professorial Unit, UNSW Clinical School of Clinical Medicine, Sydney, NSW, 2052, Australia
| | | | | | | | - Nicola Poplawski
- Adult Genetics Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Lesley Andrews
- Hereditary Cancer Centre, Prince of Wales Hospital, Randwick, New South Wales, Australia; School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
| | - Emma Healey
- Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Randwick, New South Wales 2031 Australia; Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, New South Wales 2500 Australia
| | - Steven Gallinger
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Robert C Grant
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Aung K Win
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3010, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, VIC, 3053, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, VIC, 3053, Australia
| | - Mark A Jenkins
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3010, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, VIC, 3053, Australia
| | - Giovana T Torrezan
- Clinical and Functional Genomics Group, International Research Centre/CIPE, A.C. Camargo Cancer Centre, Sao Paulo, 01508-010, Brazil; National Institute of Science and Technology in Oncogenomics and Therapeutic Innovation, Sao Paulo 01508-010, Brazil
| | - Christophe Rosty
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3010, Australia; Envoi Specialist Pathologists, Brisbane, QLD, 4059, Australia; University of Queensland, Brisbane, QLD, 4072, Australia
| | - Finlay A Macrae
- Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, VIC, 3000, Australia; Department of Medicine, The University of Melbourne, Parkville, VIC, 3000, Australia; Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Parkville, VIC, 3000, Australia
| | - Ingrid M Winship
- Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, VIC, 3000, Australia; Department of Medicine, The University of Melbourne, Parkville, VIC, 3000, Australia
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3010, Australia; Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, VIC, 3000, Australia. https://twitter.com/dan_buchanan
| | - Peter Georgeson
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC, 3010, Australia. https://twitter.com/petergeorgeson
| |
Collapse
|
32
|
Li Y, Simonds WF, Chen H. A Comparative Genomic Analysis of Parathyroid Adenomas and Carcinomas Harboring Heterozygous Germline CDC73 Mutations. J Clin Endocrinol Metab 2025; 110:429-440. [PMID: 39044678 PMCID: PMC11747674 DOI: 10.1210/clinem/dgae506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/06/2024] [Accepted: 07/22/2024] [Indexed: 07/25/2024]
Abstract
CONTEXT Parathyroid cancer has been linked to germline mutations of the Cell Division Cycle 73 (CDC73) gene. However, carriers harboring cancer-associated germline CDC73 mutations may develop only parathyroid adenoma or no parathyroid disease. This incomplete penetrance indicates that additional genomic events are required for parathyroid tumorigenesis. OBJECTIVE (1) Determine the status of the second CDC73 allele in parathyroid tumors harboring germline CDC73 mutations and (2) compare the genomic landscapes between parathyroid carcinomas and adenomas. DESIGN Whole-exome and RNA sequencing of 12 parathyroid tumors harboring germline CDC73 mutations (6 adenomas and 6 carcinomas) and their matched normal tissues. RESULTS All 12 parathyroid tumors had gained 1 somatic event predicted to cause a complete inactivation of the second CDC73 allele. Several distinctive genomic features were identified in parathyroid carcinomas compared to adenomas, including more single nucleotide variants bearing the C > G transversion and APOBEC deamination signatures, frequent mutations of the genes involved in the PI-3K/mTOR signaling, a greater number of copy number variations, and substantially more genes with altered expression. Parathyroid carcinomas also share some genomic features with adenomas. For instance, both have recurrent somatic mutations and copy number loss that impact the genes involved in T-cell receptor signaling and tumor antigen presentation, suggesting a shared strategy to evade immune surveillance. CONCLUSION Biallelic inactivation of CDC73 is essential for parathyroid tumorigenesis in carriers harboring germline mutations of this gene. Despite sharing some genomic features with adenomas, parathyroid carcinomas have more distinctive alterations in the genome, some of which may be critical for cancer formation.
Collapse
Affiliation(s)
- Yulong Li
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - William F Simonds
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haobin Chen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
33
|
Landry AP, Wang JZ, Liu J, Patil V, Gui C, Patel Z, Ajisebutu A, Ellenbogen Y, Wei Q, Singh O, Sosa J, Mansouri S, Wilson C, Cohen-Gadol AA, Zaazoue MA, Tabatabai G, Tatagiba M, Behling F, Barnholtz-Sloan JS, Sloan AE, Chotai S, Chambless LB, Rebchuk AD, Makarenko S, Yip S, Mansouri A, Tsang DS, Aldape K, Gao A, Nassiri F, Zadeh G. Development and validation of a molecular classifier of meningiomas. Neuro Oncol 2025:noae242. [PMID: 39775867 DOI: 10.1093/neuonc/noae242] [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: 08/06/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Meningiomas exhibit considerable clinical and biological heterogeneity. We previously identified four distinct molecular groups (immunogenic, NF2-wildtype, hypermetabolic, proliferative) that address much of this heterogeneity. Despite the utility of these groups, the stochasticity of clustering methods and the use of multi-omics data for discovery limits the potential for classifying prospective cases. We sought to address this with a dedicated classifier. METHODS Using an international cohort of 1698 meningiomas, we constructed and rigorously validated a machine learning-based molecular classifier using only DNA methylation data as input. Original and newly-predicted molecular groups were compared using DNA methylation, RNA sequencing, copy number profiles, whole exome sequencing, and clinical outcomes. RESULTS We show that group-specific outcomes in the validation cohort are nearly identical to those originally described, with median PFS of 7.4 (4.9-Inf) years in hypermetabolic tumors and 2.5 (2.3-5.3) years in proliferative tumors (not reached in the other groups). Tumors classified as NF2-wildtype had no NF2 mutations, and 51.4% had canonical mutations previously described in this group. RNA pathway analysis revealed upregulation of immune-related pathways in the immunogenic group, metabolic pathways in the hypermetabolic group and cell-cycle programs in the proliferative group. Bulk deconvolution similarly revealed enrichment of macrophages in immunogenic tumours and neoplastic cells in hypermetabolic and proliferative tumours with similar proportions to those originally described. CONCLUSIONS Our DNA methylation-based classifier, which is publicly available for immediate clinical use, recapitulates the biology and outcomes of the original molecular groups as assessed using multiple metrics/platforms that were not used in its training.
Collapse
Affiliation(s)
- Alexander P Landry
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Justin Z Wang
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jeff Liu
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Vikas Patil
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Chloe Gui
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Zeel Patel
- Temerty School of Medicine, University of Toronto, Toronto, ON, Canada
| | - Andrew Ajisebutu
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Yosef Ellenbogen
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Qingxia Wei
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Olivia Singh
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Julio Sosa
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Sheila Mansouri
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Christopher Wilson
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Tulsa, OK, United States
| | | | - Mohamed A Zaazoue
- Department of Neurological Surgery, Indiana University, Indianapolis, IN, United States
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Ghazaleh Tabatabai
- German Cancer Consortium (DKTK), DKFZ Partner Site Tübingen, Tübingen, Germany
- Cluster of Excellence (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Neurology and Interdisciplinary Neuro-Oncology, Center for Neuro-Oncology, Comprehensive Cancer Center, Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Marcos Tatagiba
- Department of Neurosurgery, Center for Neuro-Oncology, Comprehensive Cancer Center, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Felix Behling
- Department of Neurosurgery, Center for Neuro-Oncology, Comprehensive Cancer Center, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Jill S Barnholtz-Sloan
- Central Brain Tumor Registry of the United States, Hinsdale, IL, United States
- Trans Divisional Research Program (TDRP), Division of Cancer Epidemiology and Genetics (DCEG), National Cancer Institute, Bethesda, MD, United States
- Center for Biomedical Informatics & Information Technology (CBIIT), National Cancer Institute, Bethesda, MD, United States
| | - Andrew E Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH United States
- Department of Neurological Surgery & Piedmont Brain Tumor Center, Atlanta, GA
| | - Silky Chotai
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Lola B Chambless
- Department of Neurosurgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Alexander D Rebchuk
- Division of Neurosurgery, University of British Columbia, Vancouver, BC, Canada
| | - Serge Makarenko
- Division of Neurosurgery, University of British Columbia, Vancouver, BC, Canada
| | - Stephen Yip
- Department of Pathology & Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Derek S Tsang
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Kenneth Aldape
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Andrew Gao
- Division of Laboratory Medicine and Pathobiology, University Health Network, Toronto, ON, Canada
| | - Farshad Nassiri
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Gelareh Zadeh
- MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
34
|
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
|
35
|
Jamieson A, McConechy MK, Lum A, Senz J, Dowhy T, Huntsman DG, McAlpine JN. Selective utilization of circulating tumor DNA testing enables disease monitoring in endometrial and ovarian carcinomas. J Gynecol Oncol 2025; 36:e5. [PMID: 38909641 PMCID: PMC11791001 DOI: 10.3802/jgo.2025.36.e5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/06/2024] [Accepted: 05/13/2024] [Indexed: 06/25/2024] Open
Abstract
OBJECTIVE Biomarkers reflecting real-time response to therapy and recurrence are lacking. We assessed the clinical value of detecting cell-free circulating tumor DNA (ctDNA) mutations in endometrial cancer (EC) and ovarian cancer (OC) patients. METHODS EC/OC patients undergoing primary surgery were consented for tissue banking and 2-year serial blood draws. Tumor tissue DNA and plasma ctDNA underwent next generation sequencing using a targeted gene panel for somatic mutations. RESULTS Of 44 patients (24 EC, 17 OC, 2 synchronous endometrial and ovarian carcinomas [SEOC] and 1 endocervical adenocarcinoma [EA]) at least one somatic mutation was identified in tumor tissue in 40 (91%, 20/24 EC, all OC/SEOC/EA), and in preoperative plasma ctDNA in 12 (27%) patients (6/24 [25%] EC and 6/17 [35%] OC). Detection of preoperative ctDNA mutations was associated with advanced stage, higher preoperative CA125, and subsequent disease recurrence. In 5/12 (42%) patients with preoperative ctDNA mutations, examination/imaging suggested clinical stage I however final pathology revealed stage II/III. In 11 patients where serial timepoints were assessed during treatment for ctDNA and CA125, ctDNA clearance preceded normalization of CA125. Thirteen patients developed recurrent disease (4 EC, 8 OC, 1 EA); 8 in whom ctDNA mutations were detected postoperatively, and 4 followed through time of recurrence with ctDNA mutations identified 2-5 months prior to clinical/radiologic/biomarker progression in 3. CONCLUSION ctDNA can reflect larger tumor volume/metastases, treatment response and subsequent disease recurrence in EC and OC. Careful patient selection is critical to direct resources to patients most likely to benefit, considering disease burden and risk group.
Collapse
Affiliation(s)
- Amy Jamieson
- Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, University of British Columbia, Vancouver, Canada
| | | | - Amy Lum
- Department of Molecular Oncology, University of British Columbia, Vancouver, Canada
| | - Janine Senz
- Department of Molecular Oncology, University of British Columbia, Vancouver, Canada
| | | | - David G Huntsman
- Imagia Canexia Health, Inc., Vancouver, Canada
- Department of Molecular Oncology, University of British Columbia, Vancouver, Canada
| | - Jessica N McAlpine
- Division of Gynecologic Oncology, Department of Gynecology and Obstetrics, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
36
|
Cao X, Luo Y, Shen S, Ren X. Primary mucinous cystadenocarcinoma of the breast: A case report and literature review. Oncol Lett 2025; 29:60. [PMID: 39611063 PMCID: PMC11602826 DOI: 10.3892/ol.2024.14806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 10/30/2024] [Indexed: 11/30/2024] Open
Abstract
Mucinous cystadenocarcinoma (MCA) is a rare breast cancer. The present study reports a case of primary MCA of the breast with a comprehensive evaluation of this rare tumour. A 51-year-old woman sought medical attention for a mass in the left breast. A core needle biopsy revealed an infiltrating adenocarcinoma with mucus secretion and papillary formation. The macroscopic appearance was of a greyish-white, tough and well-circumscribed solid mass, without a notable cyst. Microscopically, the tumour consisted of ducts and cysts of varying sizes. Varying degrees of branching papillary structures were observed in the lumen and cyst cavities. The tumour cells were highly columnar in shape, with high-grade nuclei arranged in a single-layer. Immunohistochemistry revealed that the tumour was a basal-like triple-negative breast cancer with a high proliferation index and tumour protein p53 diffuse strong expression. Mutations in breast cancer 1-associated RING domain 1 (BARD1), kinase domain containing receptor (KDR), mucin-6 (MUC6), tumour protein 53 (TP53) and breast cancer 1-interacting protein C-terminal helicase 1 (BRIP1) were identified using DNA analysis. The patient was followed up for 26 months and showed no signs of recurrence or metastasis. In conclusion, the current study presents a case of MCA of breast accompanied by mutations in the BARD1, KDR, MUC6, TP53 and BRIP1 genes, with no recurrence after a 26-month follow-up. Combining this case with a review of the literature helps us to better understand the clinicopathological and genetic characteristics of MCA, and guide treatment.
Collapse
Affiliation(s)
- Xi Cao
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Yongchao Luo
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Songjie Shen
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Xinyu Ren
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| |
Collapse
|
37
|
Prip F, Lamy P, Lindskrog SV, Strandgaard T, Nordentoft I, Birkenkamp-Demtröder K, Birkbak NJ, Kristjánsdóttir N, Kjær A, Andreasen TG, Ahrenfeldt J, Pedersen JS, Rasmussen AM, Hermann GG, Mogensen K, Petersen AC, Hartmann A, Grimm MO, Horstmann M, Nawroth R, Segersten U, Sikic D, van Kessel KEM, Zwarthoff EC, Maurer T, Simic T, Malmström PU, Malats N, Jensen JB, UROMOL Consortium, Real FX, Dyrskjøt L. Comprehensive genomic characterization of early-stage bladder cancer. Nat Genet 2025; 57:115-125. [PMID: 39753772 PMCID: PMC11735393 DOI: 10.1038/s41588-024-02030-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 10/31/2024] [Indexed: 01/18/2025]
Abstract
Understanding the molecular landscape of nonmuscle-invasive bladder cancer (NMIBC) is essential to improve risk assessment and treatment regimens. We performed a comprehensive genomic analysis of patients with NMIBC using whole-exome sequencing (n = 438), shallow whole-genome sequencing (n = 362) and total RNA sequencing (n = 414). A large genomic variation within NMIBC was observed and correlated with different molecular subtypes. Frequent loss of heterozygosity in FGFR3 and 17p (affecting TP53) was found in tumors with mutations in FGFR3 and TP53, respectively. Whole-genome doubling (WGD) was observed in 15% of the tumors and was associated with worse outcomes. Tumors with WGD were genomically unstable, with alterations in cell-cycle-related genes and an altered immune composition. Finally, integrative clustering of multi-omics data highlighted the important role of genomic instability and immune cell exhaustion in disease aggressiveness. These findings advance our understanding of genomic differences associated with disease aggressiveness in NMIBC and may ultimately improve patient stratification.
Collapse
Affiliation(s)
- Frederik Prip
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Philippe Lamy
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Sia Viborg Lindskrog
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Trine Strandgaard
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Iver Nordentoft
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Karin Birkenkamp-Demtröder
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Nicolai Juul Birkbak
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Nanna Kristjánsdóttir
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Asbjørn Kjær
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Tine G Andreasen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Johanne Ahrenfeldt
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jakob Skou Pedersen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Asta Mannstaedt Rasmussen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Gregers G Hermann
- Department of Urology, Herlev Hospital, Copenhagen University, Copenhagen, Denmark
| | - Karin Mogensen
- Department of Urology, Herlev Hospital, Copenhagen University, Copenhagen, Denmark
| | - Astrid C Petersen
- Department of Pathology, Aalborg University Hospital, Aalborg, Denmark
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Comprehensive Cancer Center EMN, Erlangen, Germany
| | | | - Marcus Horstmann
- Department of Urology, University Hospital Essen, Essen, Germany
| | - Roman Nawroth
- Department of Urology, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Ulrika Segersten
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Danijel Sikic
- Department of Urology and Pediatric Urology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Kim E M van Kessel
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Urology, Amphia Ziekenhuis, Breda, the Netherlands
| | - Ellen C Zwarthoff
- Department of Pathology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Tobias Maurer
- Department of Urology and Martini-Klinik, University of Hamburg-Eppendorf, Hamburg, Germany
| | - Tatjana Simic
- Institute of Medical and Clinical Biochemistry, Center for Redox Medicine, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Per-Uno Malmström
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Center (CNIO) and CIBERONC, Madrid, Spain
| | - Jørgen Bjerggaard Jensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Francisco X Real
- Epithelial Carcinogenesis Group, Spanish National Cancer Research Center (CNIO) and CIBERONC, Madrid, Spain
- Medicine and Life Sciences Department, Universitat Pompeu Fabra, Barcelona, Spain
| | - Lars Dyrskjøt
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
Collaborators
Kim E M van Kessel,
Collapse
|
38
|
Li R, Villa NY, Yu X, Johnson JO, Borjas G, Dhillon J, Moran-Segura CM, Kim Y, Francis N, Dorman D, Powers JJ, Sexton WJ, Spiess PE, Poch MA, Zemp L, Gilbert SM, Zhang J, Pow-Sang JM, Anderson ARA, Li T, Wang X, Grass GD, Burke JM, Dinney CPN, Rodriguez PC, Jain RK, Mulé JJ, Conejo-Garcia JR. Oncolytic immunotherapy with nivolumab in muscle-invasive bladder cancer: a phase 1b trial. Nat Med 2025; 31:176-188. [PMID: 39521884 DOI: 10.1038/s41591-024-03324-9] [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: 01/14/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024]
Abstract
There is a critical unmet need for safe and efficacious neoadjuvant treatment for cisplatin-ineligible patients with muscle-invasive bladder cancer. Here we launched a phase 1b study using the combination of intravesical cretostimogene grenadenorepvec (oncolytic serotype 5 adenovirus encoding granulocyte-macrophage colony-stimulating factor) with systemic nivolumab in cisplatin-ineligible patients with cT2-4aN0-1M0 muscle-invasive bladder cancer. The primary objective was to measure safety, and the secondary objective was to assess the anti-tumor efficacy as measured by pathologic complete response along with 1-year recurrence-free survival. No dose-limiting toxicity was encountered in 21 patients enrolled and treated. Combination treatment achieved a pathologic complete response rate of 42.1% and a 1-year recurrence-free survival rate of 70.4%. Pathologic response was associated with baseline free E2F activity and tumor mutational burden but not PD-L1 status. Although T cell infiltration was broadly induced after intravesical oncolytic immunotherapy, the formation, enlargement and maturation of tertiary lymphoid structures was specifically associated with complete response, supporting the importance of coordinated humoral and cellular immune responses. Together, these results highlight the potential of this combination regimen to enhance therapeutic efficacy in cisplatin-ineligible patients with muscle-invasive bladder cancer, warranting additional study as a neoadjuvant therapeutic option. ClinicalTrials.gov identifier: NCT04610671 .
Collapse
Affiliation(s)
- Roger Li
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA.
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA.
| | - Nancy Y Villa
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Joseph O Johnson
- Analytic Microscopy Core, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Gustavo Borjas
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Jasreman Dhillon
- Department of Pathology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Carlos M Moran-Segura
- Advanced Analytical and Digital Laboratory, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Youngchul Kim
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | - Denise Dorman
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - John J Powers
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Wade J Sexton
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Philippe E Spiess
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Michael A Poch
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Logan Zemp
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Scott M Gilbert
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Jingsong Zhang
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Julio M Pow-Sang
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Alexander R A Anderson
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Tingyi Li
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - G Daniel Grass
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | - Colin P N Dinney
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Rohit K Jain
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - James J Mulé
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
- Department of Integrative Immunobiology, Duke School of Medicine, Durham, NC, USA
| |
Collapse
|
39
|
Ganguli P, Basanta CC, Acha-Sagredo A, Misetic H, Armero M, Mendez A, Zahra A, Devonshire G, Kelly G, Freeman A, Green M, Nye E, Bichisecchi A, Bonfanti P, Rodriguez-Justo M, Spencer J, Fitzgerald RC, Ciccarelli FD. Context-dependent effects of CDKN2A and other 9p21 gene losses during the evolution of esophageal cancer. NATURE CANCER 2025; 6:158-174. [PMID: 39753721 PMCID: PMC11779637 DOI: 10.1038/s43018-024-00876-0] [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: 01/05/2024] [Accepted: 11/07/2024] [Indexed: 01/31/2025]
Abstract
CDKN2A is a tumor suppressor located in chromosome 9p21 and frequently lost in Barrett's esophagus (BE) and esophageal adenocarcinoma (EAC). How CDKN2A and other 9p21 gene co-deletions affect EAC evolution remains understudied. We explored the effects of 9p21 loss in EACs and cancer progressor and non-progressor BEs with matched genomic, transcriptomic and clinical data. Despite its cancer driver role, CDKN2A loss in BE prevents EAC initiation by counterselecting subsequent TP53 alterations. 9p21 gene co-deletions predict poor patient survival in EAC but not BE through context-dependent effects on cell cycle, oxidative phosphorylation and interferon response. Immune quantifications using bulk transcriptome, RNAscope and high-dimensional tissue imaging showed that IFNE loss reduces immune infiltration in BE, but not EAC. Mechanistically, CDKN2A loss suppresses the maintenance of squamous epithelium, contributing to a more aggressive phenotype. Our study demonstrates context-dependent roles of cancer genes during disease evolution, with consequences for cancer detection and patient management.
Collapse
Affiliation(s)
- Piyali Ganguli
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, UK
- Barts Cancer Institute - Centre for Cancer Evolution, Queen Mary University of London, London, UK
| | - Celia C Basanta
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, UK
- Barts Cancer Institute - Centre for Cancer Evolution, Queen Mary University of London, London, UK
| | - Amelia Acha-Sagredo
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, UK
- Barts Cancer Institute - Centre for Cancer Evolution, Queen Mary University of London, London, UK
| | - Hrvoje Misetic
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, UK
- Barts Cancer Institute - Centre for Cancer Evolution, Queen Mary University of London, London, UK
| | - Maria Armero
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, UK
- Barts Cancer Institute - Centre for Cancer Evolution, Queen Mary University of London, London, UK
| | - Akram Mendez
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, UK
- Barts Cancer Institute - Centre for Cancer Evolution, Queen Mary University of London, London, UK
| | - Aeman Zahra
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, UK
- Barts Cancer Institute - Centre for Cancer Evolution, Queen Mary University of London, London, UK
| | - Ginny Devonshire
- Early Cancer Institute, Hutchison Research Centre, University of Cambridge, Cambridge, UK
| | - Gavin Kelly
- Bioinformatics & Biostatistics STP, The Francis Crick Institute, London, UK
| | - Adam Freeman
- Early Cancer Institute, Hutchison Research Centre, University of Cambridge, Cambridge, UK
| | - Mary Green
- Experimental Histopathology STP, The Francis Crick Institute, London, UK
| | - Emma Nye
- Experimental Histopathology STP, The Francis Crick Institute, London, UK
| | - Anita Bichisecchi
- Epithelial Stem Cell Biology & Regenerative Medicine Laboratory, The Francis Crick Institute, London, UK
- Institute of Immunity & Transplantation, Division of Infection & Immunity, UCL, London, UK
| | - Paola Bonfanti
- Epithelial Stem Cell Biology & Regenerative Medicine Laboratory, The Francis Crick Institute, London, UK
- Institute of Immunity & Transplantation, Division of Infection & Immunity, UCL, London, UK
| | | | - Jo Spencer
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Rebecca C Fitzgerald
- Early Cancer Institute, Hutchison Research Centre, University of Cambridge, Cambridge, UK
| | - Francesca D Ciccarelli
- Cancer Systems Biology Laboratory, The Francis Crick Institute, London, UK.
- Barts Cancer Institute - Centre for Cancer Evolution, Queen Mary University of London, London, UK.
| |
Collapse
|
40
|
Noguchi R, Ono T, Osaki J, Adachi Y, Iwata S, Shiota Y, Yanagihara K, Nishino S, Funada T, Ogura K, Yoshida A, Kawai A, Kondo T. Establishment and characterization of a novel patient-derived cell line from conventional central grade 3 chondrosarcoma, NCC-CS1-C1. Hum Cell 2024; 38:28. [PMID: 39645627 DOI: 10.1007/s13577-024-01152-0] [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: 09/12/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024]
Abstract
Chondrosarcoma (CS) is a malignant tumor that produces cartilaginous matrix and is the second most common primary bone sarcoma. CS encompasses a range of histological subtypes, with high-grade conventional central CS being particularly rare, occurring at a rate of 1.81 cases per 1 million person-years. Complete surgical resection is the standard curative treatment for this subtype, as radiation therapy and chemotherapy have proven ineffective. High-grade conventional central CS is highly metastatic and prone to recurrence, resulting in a poor prognosis. Therefore, effective multidisciplinary treatment strategies are urgently needed. Patient-derived cell lines offer promising tools for exploring new therapeutic approaches. However, only two cell lines of high-grade CSs are currently available in public cell banks. In this study, we aimed to establish a novel cell line for high-grade conventional central CS. We successfully developed the NCC-CS1-C1 cell line using surgically resected tumor tissues from a patient with conventional central grade 3 CS. This cell line harbored an IDH1 mutation (p.R132S), commonly found in 50% of CS cases, and exhibited complex copy number variants. A high-throughput screening of 221 anti-cancer drugs identified five candidates-bortezomib, carfilzomib, doxorubicin, panobinostat, and romidepsin-that demonstrated low IC50 values, indicating potential efficacy in treating CS. These findings suggest that NCC-CS1-C1 is a valuable tool for both preclinical and basic research on high-grade conventional central CS.
Collapse
Affiliation(s)
- Rei Noguchi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Takuya Ono
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Julia Osaki
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yuki Adachi
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Shuhei Iwata
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Yomogi Shiota
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Kazuyoshi Yanagihara
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Shogo Nishino
- Department of Diagnostic Pathology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Takaya Funada
- Department of Musculoskeletal Oncology and Rehabilitation, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Koichi Ogura
- Department of Musculoskeletal Oncology and Rehabilitation, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Akihiko Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology and Rehabilitation, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
41
|
Zhu B, Tapinos A, Koka H, Yi Lee PM, Zhang T, Zhu W, Wang X, Klein A, Lee D, Tse GM, Tsang KH, Wu C, Hua M, Highfill CA, Lenz P, Zhou W, Wang D, Luo W, Jones K, Hutchinson A, Hicks B, Garcia-Closas M, Chanock S, Tse LA, Wedge DC, Yang XR. Genomes and epigenomes of matched normal and tumor breast tissue reveal diverse evolutionary trajectories and tumor-host interactions. Am J Hum Genet 2024; 111:2773-2788. [PMID: 39492056 PMCID: PMC11639081 DOI: 10.1016/j.ajhg.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/07/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024] Open
Abstract
Normal tissues adjacent to the tumor (NATs) may harbor early breast carcinogenesis events driven by field cancerization. Although previous studies have characterized copy-number (CN) and transcriptomic alterations, the evolutionary history of NATs in breast cancer (BC) remains poorly characterized. Utilizing whole-genome sequencing (WGS), methylation profiling, and RNA sequencing (RNA-seq), we analyzed paired germline, NATs, and tumor samples from 43 individuals with BC in Hong Kong (HK). We found that single-nucleotide variants (SNVs) were common in NATs, with one-third of NAT samples exhibiting SNVs in driver genes, many of which were present in paired tumor samples. The most frequently mutated genes in both tumor and NAT samples were PIK3CA, TP53, GATA3, and AKT1. In contrast, large-scale aberrations such as somatic CN alterations (SCNAs) and structural variants (SVs) were rarely detected in NAT samples. We generated phylogenetic trees to investigate the evolutionary history of paired NAT and tumor samples. They could be categorized into tumor only, shared, and multiple-tree groups, the last of which is concordant with non-genetic field cancerization. These groups exhibited distinct genomic and epigenomic characteristics in both NAT and tumor samples. Specifically, NAT samples in the shared-tree group showed higher number of mutations, while NAT samples belonging to the multiple-tree group showed a less inflammatory tumor microenvironment (TME), characterized by a higher proportion of regulatory T cells (Tregs) and lower presence of CD14 cell populations. In summary, our findings highlight the diverse evolutionary history in BC NAT/tumor pairs and the impact of field cancerization and TME in shaping the genomic evolutionary history of tumors.
Collapse
Affiliation(s)
- Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Avraam Tapinos
- Manchester Cancer Research Centre, The University of Manchester, Manchester, UK
| | - Hela Koka
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Priscilla Ming Yi Lee
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Wei Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Xiaoyu Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Alyssa Klein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - DongHyuk Lee
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Department of Statistics, Pusan National University, Busan, Korea
| | - Gary M Tse
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Koon-Ho Tsang
- Department of Pathology, Yan Chai Hospital, Hong Kong, China
| | - Cherry Wu
- Department of Pathology, North District Hospital, Hong Kong, China
| | - Min Hua
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Chad A Highfill
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Petra Lenz
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Weiyin Zhou
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Difei Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Wen Luo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Kristine Jones
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Amy Hutchinson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Belynda Hicks
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Montserrat Garcia-Closas
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Lap Ah Tse
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - David C Wedge
- Manchester Cancer Research Centre, The University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester, UK.
| | - Xiaohong R Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
42
|
Xu G, Yu J, Lyu J, Zhan M, Xu J, Huang M, Zhao R, Li Y, Zhu J, Feng J, Tan S, Ran P, Su Z, Liu X, Zhao J, Zhang H, Xu C, Chang J, Hou Y, Ding C. Proteogenomic Landscape of Breast Ductal Carcinoma Reveals Tumor Progression Characteristics and Therapeutic Targets. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401041. [PMID: 39418072 PMCID: PMC11633542 DOI: 10.1002/advs.202401041] [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: 01/28/2024] [Revised: 09/04/2024] [Indexed: 10/19/2024]
Abstract
Multi-omics studies of breast ductal carcinoma (BRDC) have advanced the understanding of the disease's biology and accelerated targeted therapies. However, the temporal order of a series of biological events in the progression of BRDC is still poorly understood. A comprehensive proteogenomic analysis of 224 samples from 168 patients with malignant and benign breast diseases is carried out. Proteogenomic analysis reveals the characteristics of linear multi-step progression of BRDC, such as tumor protein P53 (TP53) mutation-associated estrogen receptor 1 (ESR1) overexpression is involved in the transition from ductal hyperplasia (DH) to ductal carcinoma in situ (DCIS). 6q21 amplification-associated nuclear receptor subfamily 3 group C member 1 (NR3C1) overexpression helps DCIS_Pure (pure DCIS, no histologic evidence of invasion) cells avoid immune destruction. The T-cell lymphoma invasion and metastasis 1, androgen receptor, and aldo-keto reductase family 1 member C1 (TIAM1-AR-AKR1C1) axis promotes cell invasion and migration in DCIS_adjIDC (DCIS regions of invasive cancers). In addition, AKR1C1 is identified as a potential therapeutic target and demonstrated the inhibitory effect of aspirin and dydrogesterone as its inhibitors on tumor cells. The integrative multi-omics analysis helps to understand the progression of BRDC and provides an opportunity to treat BRDC in different stages.
Collapse
Affiliation(s)
- Ganfei Xu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Juan Yu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jiacheng Lyu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Mengna Zhan
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jie Xu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Minjing Huang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Rui Zhao
- Institute for Developmental and Regenerative Cardiovascular MedicineMOE‐Shanghai Key Laboratory of Children's Environmental HealthXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
| | - Yan Li
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jiajun Zhu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jinwen Feng
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Subei Tan
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Peng Ran
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Zhenghua Su
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Xinhua Liu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jianyuan Zhao
- Institute for Developmental and Regenerative Cardiovascular MedicineMOE‐Shanghai Key Laboratory of Children's Environmental HealthXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
| | - Hongwei Zhang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Chen Xu
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Jun Chang
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Yingyong Hou
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
| | - Chen Ding
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesHuman Phenome InstituteDepartment of PathologyZhongshan Hospital, Fudan UniversityShanghai200433China
- Departments of Cancer Research InstituteAffiliated Cancer Hospital of Xinjiang Medical UniversityXinjiang Key Laboratory of Translational Biomedical EngineeringUrumqi830000P. R. China
| |
Collapse
|
43
|
Pokorna P, Palova H, Adamcova S, Jugas R, Al Tukmachi D, Kyr M, Knoflickova D, Kozelkova K, Bystry V, Mejstrikova S, Merta T, Trachtova K, Podlipna E, Mudry P, Pavelka Z, Bajciova V, Tinka P, Jarosova M, Catela Ivkovic T, Madlener S, Pal K, Stepien N, Mayr L, Tichy B, Drabova K, Jezova M, Kozakova S, Vanackova J, Radova L, Steininger K, Haberler C, Gojo J, Sterba J, Slaby O. Real-World Performance of Integrative Clinical Genomics in Pediatric Precision Oncology. J Transl Med 2024; 104:102161. [PMID: 39442669 DOI: 10.1016/j.labinv.2024.102161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/16/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Despite significant improvement in the survival of pediatric patients with cancer, treatment outcomes for high-risk, relapsed, and refractory cancers remain unsatisfactory. Moreover, prolonged survival is frequently associated with long-term adverse effects due to intensive multimodal treatments. Accelerating the progress of pediatric oncology requires both therapeutic advances and strategies to mitigate the long-term cytotoxic side effects, potentially through targeting specific molecular drivers of pediatric malignancies. In this report, we present the results of integrative genomic and transcriptomic profiling of 230 patients with malignant solid tumors (the "primary cohort") and 18 patients with recurrent or otherwise difficult-to-treat nonmalignant conditions (the "secondary cohort"). The integrative workflow for the primary cohort enabled the identification of clinically significant single nucleotide variants, small insertions/deletions, and fusion genes, which were found in 55% and 28% of patients, respectively. For 38% of patients, molecularly informed treatment recommendations were made. In the secondary cohort, known or potentially driving alteration was detected in 89% of cases, including a suspected novel causal gene for patients with inclusion body infantile digital fibromatosis. Furthermore, 47% of findings also brought therapeutic implications for subsequent management. Across both cohorts, changes or refinements to the original histopathological diagnoses were achieved in 4% of cases. Our study demonstrates the efficacy of integrating advanced genomic and transcriptomic analyses to identify therapeutic targets, refine diagnoses, and optimize treatment strategies for challenging pediatric and young adult malignancies and underscores the need for broad implementation of precision oncology in clinical settings.
Collapse
Affiliation(s)
- Petra Pokorna
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic; Center for Precision Medicine, University Hospital Brno, Brno, Czech Republic
| | - Hana Palova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sona Adamcova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Robin Jugas
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Dagmar Al Tukmachi
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Michal Kyr
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Dana Knoflickova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Katerina Kozelkova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Vojtech Bystry
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Sona Mejstrikova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Tomas Merta
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Karolina Trachtova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Eliska Podlipna
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Peter Mudry
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zdenek Pavelka
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Viera Bajciova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavel Tinka
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marie Jarosova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Tina Catela Ivkovic
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Sibylle Madlener
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Karol Pal
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Natalia Stepien
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Lisa Mayr
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Boris Tichy
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Klara Drabova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marta Jezova
- Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sarka Kozakova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Department of Pharmacy, University Hospital Brno, Brno, Czech Republic
| | - Jitka Vanackova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Lenka Radova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Karin Steininger
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christine Haberler
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Johannes Gojo
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Jaroslav Sterba
- Center for Precision Medicine, University Hospital Brno, Brno, Czech Republic; Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Center for Precision Medicine, University Hospital Brno, Brno, Czech Republic; Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
44
|
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
|
45
|
Zhao Y, Zhang D, Meng B, Zhang Y, Ma S, Zeng J, Wang X, Peng T, Gong X, Zhai R, Dong L, Jiang Y, Dai X, Fang X, Jia W. Integrated proteomic and glycoproteomic analysis reveals heterogeneity and molecular signatures of brain metastases from lung adenocarcinomas. Cancer Lett 2024; 605:217262. [PMID: 39341452 DOI: 10.1016/j.canlet.2024.217262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/26/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024]
Abstract
Brain metastasis is a major cause of poor prognosis and death in lung adenocarcinoma (LUAD); however, the understanding of therapeutic strategies and mechanisms for brain metastases from LUAD (BM-LUAD) remains notably limited, especially at the proteomics levels. To address this issue, we conducted integrated proteomic and glycoproteomic analyses on 49 BM-LUAD tumors, revealing two distinct subtypes of the disease: BM-S1 and BM-S2. Whole exome sequencing analysis revealed that somatic mutations in STK11 and KEAP1, as well as copy number deletions on chr19p13.3, such as STK11, UQCR11, and SLC25A23, were more frequently detected in BM-S2. In BM-S1 tumors, we observed significant infiltration of GFAP + astrocytes, as evidenced by elevated levels of GFAP, GABRA2, GABRG1 and GAP43 proteins and an enrichment of astrocytic signatures in both our proteomic data and external spatial transcriptomic data. Conversely, BM-S2 tumors demonstrated higher levels of PD-1 immune cell infiltration, supported by the upregulation of PD-1 and LAG-3 genes. These findings suggest distinct microenvironmental adaptations required by the different BM-LUAD subtypes. Additionally, we observed unique glycosylation patterns between the subtypes, with increased fucosylation in BM-S1 and enhanced sialylation in BM-S2, primarily affected by glycosylation enzymes such as FUT9, B4GALT1, and ST6GAL1. Specifically, in BM-S2, these sialylation modifications are predominantly localized to the lysosomes, underscoring the critical role of N-glycosylation in the tumor progression of BM-LUAD. Overall, our study not only provides a comprehensive multi-omic data resource but also offers valuable biological insights into BM-LUAD, highlighting potential mechanisms and therapeutic targets for further investigation.
Collapse
Affiliation(s)
- Yang Zhao
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Dainan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Bo Meng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Yong Zhang
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shunchang Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Jiaming Zeng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Xi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Tao Peng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Xiaoyun Gong
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Rui Zhai
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Lianhua Dong
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - You Jiang
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China
| | - Xinhua Dai
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China.
| | - Xiang Fang
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, 100029, China.
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
46
|
Camp SY, He MX, Cuoco MS, Saad E, Pimenta E, Meli K, Bakouny Z, Labaki C, Titchen BM, Kang YJ, Horst J, Trowbridge R, Shannon E, Helvie K, Thorner AR, Vigneau S, Mayorga A, Kodali J, Lachmayr H, Bemus M, Park J, Choueiri T, Bi K, Van Allen EM. Single-cell epigenetic profiling reveals an interferon response-high program associated with BAP1 deficiency in kidney cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623837. [PMID: 39605676 PMCID: PMC11601305 DOI: 10.1101/2024.11.15.623837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Renal cell carcinoma (RCC) is characterized by recurrent somatic mutations in epigenetic regulators, which stratify patients into clinically significant subgroups with distinct prognoses and treatment responses. However, the cell type-specific epigenetic landscape of RCC-broadly and in the context of these mutations-is incompletely understood. To investigate these open questions, we integrated single nucleus ATAC sequencing data from RCC tumors across four independent cohorts. In clear cell RCC tumors, we identified four shared malignant epigenetic programs related to angiogenesis, proximal tubule-like features, interferon (IFN) signaling, and one that lacked distinct genomic regions with increased accessibility. Among the mutated epigenetic regulators, BAP1 mutation exhibited the most significant impact on chromatin accessibility in tumor cells, and the associated epigenetic changes were linked to IFN response. We identify multiple potential sources of elevated IFN signaling in these lesions, such as increased immune infiltration and increased accessibility and expression of an IFN-associated ERV, ERV3-16A3_LTR. We find that the expression of ERV3-16A3_LTR may itself be a negative prognostic biomarker in ccRCC. Our findings highlight the convergence of malignant epigenetic programs across ccRCC tumors and suggest that BAP1 loss, potentially through ERV3-16A3_LTR dysregulation, is associated with an IFN response-high epigenetic program.
Collapse
Affiliation(s)
- Sabrina Y. Camp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Meng Xiao He
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Graduate Program in Biophysics, Boston, MA 02115, USA
| | - Michael S. Cuoco
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, CA 92093, USA
| | - Eddy Saad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Erica Pimenta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Kevin Meli
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Division of Medical Sciences PhD Program in Biological and Biomedical Sciences, Boston, MA 02115, USA
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Chris Labaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Breanna M. Titchen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Harvard Division of Medical Sciences PhD Program in Biological and Biomedical Sciences, Boston, MA 02115, USA
| | - Yun Jee Kang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Jack Horst
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Rachel Trowbridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Erin Shannon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Karla Helvie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Aaron R. Thorner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Sébastien Vigneau
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Angie Mayorga
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jahnavi Kodali
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Hannah Lachmayr
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Meredith Bemus
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jihye Park
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Toni Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Kevin Bi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Eliezer M. Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, 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
| |
Collapse
|
47
|
Zhou ZJ, Ye YH, Hu ZQ, Hou YR, Liu KX, Sun RQ, Wang PC, Luo CB, Li J, Zou JX, Zhou J, Fan J, Song CL, Zhou SL. Whole-exome sequencing reveals genomic landscape of intrahepatic cholangiocarcinoma and identifies SAV1 as a potential driver. Nat Commun 2024; 15:9960. [PMID: 39551842 PMCID: PMC11570600 DOI: 10.1038/s41467-024-54387-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 11/07/2024] [Indexed: 11/19/2024] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is the second most common primary hepatic malignancy after hepatocellular carcinoma, with poor prognosis and limited treatment options. The genomic features of ICC in Chinese patients remain largely unknown. In this study, we perform deep whole-exome sequencing of 204 Chinese primary ICCs and characterize genomic alterations and clonal evolution, and reveal their associations with patient outcomes. We identify six mutational signatures, including Signatures A and F, which are highly similar to previously described signatures linked to aristolochic acid and aflatoxin exposures, respectively. We also identify 13 significantly mutated genes in the ICC samples, including SAV1. We find that SAV1 was mutated in 2.9% (20/672) of 672 ICC samples. SAV1 mutation is associated with lower SAV1 protein levels, higher rates of tumor recurrence, and shorter overall patient survival. Biofunctional investigations reveal a tumor-suppressor role of SAV1: its inactivation suppresses Hippo signaling, leading to YAP activation, thereby promoting tumor growth and metastasis. Collectively, our results delineate the genomic landscape of Chinese ICCs and identify SAV1 as a potential driver of ICC.
Collapse
Affiliation(s)
- Zheng-Jun Zhou
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu-Hang Ye
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhi-Qiang Hu
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue-Ru Hou
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China
| | - Kai-Xuan Liu
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rong-Qi Sun
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng-Cheng Wang
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chu-Bin Luo
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia Li
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ji-Xue Zou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng-Li Song
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning, China.
| | - Shao-Lai Zhou
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China.
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
48
|
Lee H, Jeong SH, Baek YW, Lee H, Sa JK, Lee JY, Lee YS, Nam YJ, Kim J, Kim J, Choi JY, Park SA, Kim JH, Park YH, Lim J, Kim YH, Park EK, Kim C, Lee JH. Deciphering the toxicity of polyhexamethylene guanidine phosphate in lung carcinogenesis: Mutational profiles and molecular mechanisms. CHEMOSPHERE 2024; 368:143785. [PMID: 39577803 DOI: 10.1016/j.chemosphere.2024.143785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/24/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Polyhexamethylene guanidine (PHMG) is widely utilized in personal hygiene products due to its bactericidal, non-volatile, and hydrophilic properties. However, the long-term toxic effects and underlying mechanisms associated with respiratory exposure to the commonly used form, PHMG phosphate (PHMG-p), are still insufficiently understood. This study aims to elucidate the types of pulmonary lesions and the incidence of lung cancer associated with varying concentrations of PHMG-p and observation periods, along with the molecular mechanisms underlying this relationship. To assess these effects, CT scans and pathological analyses were conducted for up to 54 weeks following initial exposure to PHMG-p. Furthermore, to investigate the underlying causes of pulmonary toxicity, TGF-beta-activated kinase 1 was identified as a PHMG-p-binding protein, and its associated signaling pathways, including necroptosis, apoptosis, and MKK7, were explored. Somatic mutational signature, and gene ontology (GO) analyses were performed to investigate the genetic characteristics of PHMG-p-induced lung carcinogenesis. PHMG-p exposure led to somatic mutations in lung cancer-related genes, including TP53, SOS1, KMT2D, MDM2, ERBB2, SETD2, MET, ARID1A, RBM10, and CDKN2A as well as in genes such as RAB31, WASHC1, DDX11, ECD, STAB2, MUC2, and MUC5AC. The mutated genes were primarily associated with impaired DNA repair mechanisms. GO analysis highlighted the activation of pathways related to cell cycle checkpoints, necroptosis, MAPK, and idiopathic pulmonary fibrosis, while also revealing the suppression of signaling pathways associated with natural killer cells, GADD45, LXR/RXR activation, and IL-15 production. Gain-of-function experiments confirmed the oncogenic roles of PLAU and HMGA2, as well as the tumor-suppressive functions of TBX4 and GPX3. These findings suggest that PHMG-p activates necroptosis and MAPK signaling, increases the frequency of somatic mutations, and inhibits apoptosis, thus fostering an environment conducive to carcinogenesis. This underscores the importance of understanding the potential health risks associated with PHMG-p exposure and provides insights for future research and regulatory considerations regarding the safety of personal hygiene products.
Collapse
Affiliation(s)
- Hong Lee
- Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi, 15355, Republic of Korea.
| | - Sang Hoon Jeong
- Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi, 15355, Republic of Korea.
| | - Yong-Wook Baek
- Humidifier Disinfectant Health Center, National Institute of Environmental Research, Incheon, 22689, Republic of Korea.
| | - Hyejin Lee
- Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi, 15355, Republic of Korea.
| | - Jason K Sa
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
| | - Ji Yoon Lee
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
| | - Yu-Seon Lee
- Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi, 15355, Republic of Korea.
| | - Yoon Jeong Nam
- Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi, 15355, Republic of Korea.
| | - Jaeyoung Kim
- Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi, 15355, Republic of Korea.
| | - Jonghoon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06531, Republic of Korea.
| | - Jin Young Choi
- Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi, 15355, Republic of Korea.
| | - Su A Park
- Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi, 15355, Republic of Korea.
| | - Je Hyeong Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi, 15355, Republic of Korea.
| | - Yoon Hee Park
- Medical Science Research Center, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi, 15355, Republic of Korea.
| | - Jungyun Lim
- Humidifier Disinfectant Health Center, National Institute of Environmental Research, Incheon, 22689, Republic of Korea.
| | - Young-Hee Kim
- Humidifier Disinfectant Health Center, National Institute of Environmental Research, Incheon, 22689, Republic of Korea.
| | - Eun-Kee Park
- Department of Medical Humanities and Social Medicine, College of Medicine, Kosin University, Busan, 49267, Republic of Korea.
| | - Cherry Kim
- Department of Radiology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi, 15355, Republic of Korea.
| | - Ju-Han Lee
- Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Gyeonggi, 15355, Republic of Korea.
| |
Collapse
|
49
|
Quezada-Maldonado EM, Cerrato-Izaguirre D, Morales-Bárcenas R, Bautista-Ocampo Y, Santibáñez-Andrade M, Quintana-Belmares R, Chirino YI, Basurto-Lozada P, Robles-Espinoza CD, Sánchez-Pérez Y, García-Cuellar CM. Mutational landscape induced by chronic exposure to environmental PM 10 and PM 2.5 in A549 lung epithelial cell. CHEMOSPHERE 2024; 368:143766. [PMID: 39551196 DOI: 10.1016/j.chemosphere.2024.143766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/05/2024] [Accepted: 11/15/2024] [Indexed: 11/19/2024]
Abstract
Exposure to particulate matter (PM) has been linked to an increased risk of multiple diseases, primarily lung cancer, through various molecular mechanisms. However, the mutagenic potential of PM remains unclear. This study aimed to provide a comprehensive description of genetic mutations and mutagenic signatures resulting from chronic exposure to PM10 or PM2.5. Using whole exome sequencing, we identified driver mutations and mutational signatures in A549 cells, a lung epithelial cell model subjected to weekly exposure to either PM10 or PM2.5, for a period of 28 weeks. The number of single nucleotide variations, insertions, and deletions increased depending on the duration of exposure. PM10 generated the highest number of genomic alterations. Amplifications in SYK (oncogene) and mutations in NCOR1 (tumor suppressor gene) were prevalent in cells exposed to either PM10 or PM2.5; however, other mutations were exclusive, such as TP53 and ANK3 for PM10, and ERCC1 and ERCC2 for PM2.5. Different p53-related signaling pathways were most enriched by driver mutations upon exposure to both PM10 and PM2.5, particularly the glucose deprivation pathway. Exposure to either PM10 or PM2.5 resulted in high frequencies of C > A substitutions and one-base insertions/deletions in microhomology sites. The single-base substitution (SBS) signature SBS05, related to the nucleotide excision DNA repair pathway, contributed the most to both PM10-and PM2.5-exposed cells. The contribution of signature SBS18, related to oxidative stress, was observed in cells exposed to either PM10 or PM2.5, but a greater contribution was observed in PM2.5-exposed cells. In addition, SBS03 and SBS36, which are related to different DNA damage repair mechanisms, were observed more frequently in PM10-exposed cells. We assessed the mutagenic potential of PM10 and PM2.5, as a complete mixture, identifying mutated driver genes and mutational signatures generated by chronic PM exposure, which could contribute to the development of cancer, cardiovascular, and digestive diseases.
Collapse
Affiliation(s)
- Ericka Marel Quezada-Maldonado
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, San Fernando No. 22. Tlalpan. México CP 14080. CDMX, Mexico
| | - Dennis Cerrato-Izaguirre
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, San Fernando No. 22. Tlalpan. México CP 14080. CDMX, Mexico
| | - Rocío Morales-Bárcenas
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, San Fernando No. 22. Tlalpan. México CP 14080. CDMX, Mexico
| | - Yanueh Bautista-Ocampo
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, San Fernando No. 22. Tlalpan. México CP 14080. CDMX, Mexico
| | - Miguel Santibáñez-Andrade
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, San Fernando No. 22. Tlalpan. México CP 14080. CDMX, Mexico
| | - Raúl Quintana-Belmares
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, San Fernando No. 22. Tlalpan. México CP 14080. CDMX, Mexico
| | - Yolanda I Chirino
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Los Reyes Iztacala, Tlalnepantla de Baz, Estado de México 54090, CP, Mexico
| | - Patricia Basurto-Lozada
- Laboratorio Internacional de Investigación Sobre el Genoma Humano, Universidad Nacional Autónoma de México, Santiago de Querétaro, 76010, CP, Mexico
| | - Carla Daniela Robles-Espinoza
- Laboratorio Internacional de Investigación Sobre el Genoma Humano, Universidad Nacional Autónoma de México, Santiago de Querétaro, 76010, CP, Mexico; Wellcome Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Yesennia Sánchez-Pérez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, San Fernando No. 22. Tlalpan. México CP 14080. CDMX, Mexico.
| | - Claudia M García-Cuellar
- Dirección de Investigación, Instituto Nacional de Cancerología (INCan), San Fernando No. 22, Tlalpan, Ciudad de México, 14080, CP, Mexico.
| |
Collapse
|
50
|
Vijayraghavan S, Blouin T, McCollum J, Porcher L, Virard F, Zavadil J, Feghali-Bostwick C, Saini N. Widespread mutagenesis and chromosomal instability shape somatic genomes in systemic sclerosis. Nat Commun 2024; 15:8889. [PMID: 39406724 PMCID: PMC11480385 DOI: 10.1038/s41467-024-53332-z] [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: 04/06/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Systemic sclerosis is a connective tissue disorder characterized by excessive fibrosis that primarily affects women, and can present as a multisystem pathology. Roughly 4-22% of patients with systemic sclerosis develop cancer, which drastically worsens prognosis. However, the mechanisms underlying systemic sclerosis initiation, propagation, and cancer development are poorly understood. We hypothesize that the inflammation and immune response associated with systemic sclerosis can trigger DNA damage, leading to elevated somatic mutagenesis, a hallmark of pre-cancerous tissues. To test our hypothesis, we culture clonal lineages of fibroblasts from the lung tissues of controls and systemic sclerosis patients and compare their mutation burdens and spectra. We find an overall increase in all major mutation types in systemic sclerosis samples compared to control lung samples, from small-scale events such as single base substitutions and insertions/deletions, to chromosome-level changes, including copy-number changes and structural variants. In the genomes of patients with systemic sclerosis, we find evidence of somatic hypermutation or kategis (typically only seen in cancer genomes), we identify mutation signatures closely resembling the error-prone translesion polymerase Polη activity, and observe an activation-induced deaminase-like mutation signature, which overlaps with genomic regions displaying kataegis.
Collapse
Affiliation(s)
- Sriram Vijayraghavan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Thomas Blouin
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - James McCollum
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Latarsha Porcher
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - François Virard
- University Claude Bernard Lyon 1, INSERM U1052-CNRS UMR5286, Cancer Research Center, Centre Léon Bérard, Lyon, France
| | - Jiri Zavadil
- International Agency for Research on Cancer WHO, Epigenomics and Mechanisms Branch, Lyon, France
| | - Carol Feghali-Bostwick
- Department of Medicine, Division of Rheumatology, Medical University of South Carolina, Charleston, SC, USA
| | - Natalie Saini
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|