1
|
Liu M, Ji Q, Zhang S, Qian J, Li B, Li J, Xiao P, Hu S. Hematopoietic stem cell transplantation for purine nucleoside phosphorylase deficiency with two novel mutations: a case report and review of literature. Hematology 2025; 30:2445404. [PMID: 39772979 DOI: 10.1080/16078454.2024.2445404] [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: 05/17/2024] [Accepted: 12/14/2024] [Indexed: 01/14/2025] Open
Abstract
PURPOSE We report the case of a 6-year-old boy who presented with muscular hypertonia, impaired growth, and recurrent infections, who was diagnosed with purine nucleoside phosphorylase (PNP) deficiency with two novel mutations in the PNP gene. He underwent a hematopoietic stem cell transplantation (HSCT) from an unrelated donor, and we observed the clinical outcome. METHODS We retrospectively analyzed the clinical manifestations and outcomes of this patient who underwent HSCT. We analyzed the results of whole exome sequencing (WES) on the patient. RESULTS The patient experienced repeated serious respiratory and gastrointestinal infections since birth and presented with neurological symptoms. He was found to have two novel pathogenic mutations in the PNP gene through WES. One hemizygous variant was c.385dup (p.Ile129Asnfs*6) in exon 4. The other was a heterozygous deletion in exon 2-6. He underwent HSCT with clinical improvement. CONCLUSIONS We presented a patient with two novel mutations in the PNP gene and clinical improvement following an allo-HSCT.
Collapse
Affiliation(s)
- Minyuan Liu
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Qi Ji
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Senlin Zhang
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jing Qian
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Bohan Li
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jie Li
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Peifang Xiao
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Shaoyan Hu
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
- Jiangsu Pediatric Hematology and Oncology Center, Suzhou, People's Republic of China
| |
Collapse
|
2
|
Song H, Kim D, Jang SJ, Hwang HS, Song JS. Clinicopathologic features of histologic transformation in lung adenocarcinoma after treatment with epidermal growth factor receptor-tyrosine kinase inhibitors. Ann Diagn Pathol 2025; 77:152478. [PMID: 40215564 DOI: 10.1016/j.anndiagpath.2025.152478] [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: 03/12/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) may lead to drug resistance, and the underlying mechanism may involve histologic transformations to small cell carcinoma (SCC), squamous cell carcinoma (SqCC), and sarcomatoid carcinoma (SC). Although there are reports regarding these transformations, comprehensive analyses are limited. METHODS A total of 233 patients with primary lung adenocarcinoma treated with EGFR-TKIs were reviewed. Among them, 26 patients (11.1 %) showed histologic transformation. RESULTS Eleven patients (42.3 %) showed SCC and SqCC transformations respectively, and four patients (15.4 %) showed SC transformation. The median time from TKI initiation to transformation was 19.8 months (6.8-51.4) for SCC, 45.3 months (2.4-101.5) for SqCC, and 11.8 months (6.8-15.7) for SC. The median overall survival (OS) was 41.8 months (12.5-78.9), 72.6 months (18.8-112.7), and 23.7 months (17.4-34.4), respectively. The survival from transformation was 12.3 months (2.1-28.3), 16.9 months (0.7-43.2), and 11.4 months (1.6-23.5), respectively. The most common mutations were TP53, PTEN, and RB1 in SCC; TP53 and RB1 in SqCC; and TP53 and KMT2D in SC. SC transformation had the worst OS, followed by SCC and SqCC (p < 0.001). This prognosis difference was also reflected in the time to transformation after EGFR-TKI treatment (p = 0.005). However, survival after transformation was not associated with tumor subtypes (p = 0.536). CONCLUSIONS The analysis of mutation profiles and survival outcomes revealed that the transformation subtype affects prognosis. Additionally, the time taken to undergo transformation is critical for patient outcomes.
Collapse
Affiliation(s)
- Halim Song
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea
| | - Deokhoon Kim
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea
| | - Se Jin Jang
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea
| | - Hee Sang Hwang
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea
| | - Joon Seon Song
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea; Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, South Korea.
| |
Collapse
|
3
|
Schwerd‐Kleine P, Würth R, Cheytan T, Michel L, Thewes V, Gutjahr E, Seker‐Cin H, Kazdal D, Neuberth S, Thiel V, Schwickert J, Vorberg T, Wischhusen J, Stenzinger A, Zapatka M, Lichter P, Schneeweiss A, Trumpp A, Sprick MR. Biopsy-derived organoids in personalised early breast cancer care: Challenges of tumour purity and normal cell overgrowth cap their practical utility. Int J Cancer 2025; 156:2200-2209. [PMID: 40022208 PMCID: PMC11970545 DOI: 10.1002/ijc.35386] [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: 07/05/2024] [Revised: 12/05/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025]
Abstract
The ability to establish organoids composed exclusively of tumour rather than healthy cells is essential for their implementation into clinical practice. Organoids have recently emerged as a powerful tool to expand patient material in culture and generate modifiable 3D models derived from humans or animal models. For translational research, they enable the creation of model systems for an ever-increasing number of cell types and diseases. And in personalised medicine, they potentially allow for functional drug testing with high predictive power in certain settings. We found that using biopsy material from untreated, early-stage primary breast cancer patients poses significant challenges for consistently culturing tumour cells as organoids. Specifically, we observed frequent outgrowth of genetically normal, non-cancerous epithelial cells. We analysed >100 biopsy samples from early-stage breast cancer and present our large collection of >70 organoid lines. We also show methods of assessing successful tumour cell culture in a time, and cost-efficient manner, proving a high rate (>85%) of normal cell overgrowth in early-stage breast cancer organoids. Finally, we show a number of successful attempts to culture cancer organoids from mastectomy-derived tissue of advanced, metastatic breast cancer. We conclude that the usefulness of organoids from early breast cancer for translational research and personalised medicine, especially guidance of adjuvant or post-surgical maintenance therapy, is strongly limited by the low success rate of culturing cancerous cells under organoid conditions.
Collapse
Affiliation(s)
- Paul Schwerd‐Kleine
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Roberto Würth
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
| | - Tasneem Cheytan
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Laura Michel
- Division of Gynecologic OncologyNational Center for Tumor Diseases (NCT)HeidelbergGermany
| | - Verena Thewes
- Division of Molecular GeneticsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Ewgenija Gutjahr
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Institute of Pathology, Heidelberg University HospitalHeidelbergGermany
| | - Huriye Seker‐Cin
- Institute of Pathology, Heidelberg University HospitalHeidelbergGermany
| | - Daniel Kazdal
- Institute of Pathology, Heidelberg University HospitalHeidelbergGermany
| | - Sarah‐Jane Neuberth
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Vera Thiel
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Jonas Schwickert
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Tim Vorberg
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Jennifer Wischhusen
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
| | | | - Marc Zapatka
- Division of Molecular GeneticsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Peter Lichter
- Division of Molecular GeneticsGerman Cancer Research Center (DKFZ)HeidelbergGermany
- National Center for Tumor Diseases (NCT)HeidelbergGermany
| | - Andreas Schneeweiss
- Division of Gynecologic OncologyNational Center for Tumor Diseases (NCT)HeidelbergGermany
| | - Andreas Trumpp
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
| | - Martin R. Sprick
- Division of Stem Cells and CancerGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI‐STEM gGmbH)HeidelbergGermany
| |
Collapse
|
4
|
Ranaweera DM, de Silva DC, Samarasinghe D, Perera S, Kugalingam N, Samarasinghe SR, Madushani WY, Jayaweera HHE, Gunewardene S, Muneeswaran K, Gnanam VS, Chandrasekharan NV. Development of a TaqMan-based dosage analysis PCR assay for the molecular diagnosis of 22q11.2 deletion syndrome. Genes Genet Syst 2025; 100:n/a. [PMID: 39842802 DOI: 10.1266/ggs.24-00142] [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] [Indexed: 01/24/2025] Open
Abstract
A hemizygous 1.5-3.0-Mb microdeletion of human chromosome 22q11.2 with the loss of multiple genes including histone cell cycle regulator (HIRA) causes 22q11.2 deletion syndrome (22q11.2 DS), a common disorder with variable manifestations including congenital malformations affecting the heart, palate and kidneys in association with neurodevelopmental, psychiatric, endocrine and autoimmune abnormalities. The aim of this study was to develop a TaqMan-based dosage analysis PCR (TaqMan qPCR) for use as a rapid, cost-effective test for clinically suspected patients fulfilling previously described criteria for molecular diagnosis of 22q11.2 DS in a lower middle-income country where the cost of testing limits its use in routine clinical practice. Nineteen patients were recruited with informed consent following ethical approval from the Ethics Review Committee, Lady Ridgeway Hospital for Children, Colombo. Dosage analysis of extracted DNA was performed using a TaqMan qPCR assay by amplifying regions within the target (HIRA) and control (testin LIM domain protein (TES)) genes of suspected patient (P) and unaffected person (N) samples. For detection of a deletion, the normalized value (HIRA/TES dosage) of a P sample was compared with that of an N sample. A ratio of P:N of 0.5 confirmed the presence of a deletion while a ratio of 1.0 refuted this. Seven of the 19 patients were found to have a HIRA deletion, confirming the diagnosis of 22q11.2 DS, with these results being in complete agreement with those of fluorescence in situ hybridization (FISH) (performed in nine of the 19 cases) and whole-exome sequencing (all 19 samples tested). This TaqMan qPCR assay was able to reliably distinguish HIRA-deleted cases from non-deleted ones. The assay was both cheaper and faster compared to commercially available alternatives in our setting, including FISH and multiple ligation-dependent probe amplification.
Collapse
|
5
|
Berghaus N, Hielscher T, Savran D, Schrimpf D, Maas SLN, Preusser M, Weller M, Acker T, Herold-Mende C, Wick W, von Deimling A, Sahm F. Meningiomas: Sex-specific differences and prognostic implications of a chromosome X loss. Neuro Oncol 2025; 27:1019-1028. [PMID: 39535060 PMCID: PMC12083225 DOI: 10.1093/neuonc/noae239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Meningiomas are the most common primary intracranial tumors in adults. Several studies proposed new stratification systems with a more accurate risk prediction than the WHO grading, eg, based on methylation and copy-number variations (CNVs). Yet, common shortcomings in these analyses are either a lack of stratification by sex of patients or excluding the gonosomes from CNV assessment. METHODS Within this study, DNA methylation array data from 7424 meningioma samples as well as targeted sequencing, clinical annotations, and morphology subtyping of 796 samples were examined for differences between females and males regarding mutations, methylation classes, CNVs, and histology. RESULTS Meningiomas from females accounted for about 53% of the malignant tumors and present a loss of one X chromosome in 57% of these malignant cases. In the group of benign tumors, females comprised about 75% of the patients. Therein, a loss of one X chromosome was detected in only about 10% of the cases but was associated with a significantly worse progression-free survival. CONCLUSIONS Although genomic instability is a common feature of malignant meningiomas, particularly loss of the X chromosome in tumors of female patients in otherwise histologically and molecularly low-risk tumors confers higher risk. Hence, the gonosomal copy-number status can be leveraged for increased diagnostic accuracy.
Collapse
Affiliation(s)
- Natalie Berghaus
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dilan Savran
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Schrimpf
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sybren L N Maas
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Austria
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Till Acker
- Institute of Neuropathology, Justus-Liebig University Gießen, Germany
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Wolfgang Wick
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas von Deimling
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Felix Sahm
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
6
|
Akarca FG, Grenert JP, Kakar S. Role of genomic analysis in the classification of well differentiated hepatocellular lesions. Hum Pathol 2025; 158:105794. [PMID: 40374146 DOI: 10.1016/j.humpath.2025.105794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND The distinction of focal nodular hyperplasia (FNH) and hepatocellular adenoma (HCA) from well-differentiated hepatocellular carcinoma (WD-HCC) in noncirrhotic liver can be challenging. High-grade dysplastic nodule (HGDN) in cirrhosis can have overlapping features with WD-HCC. In some cases, HCA diagnosis is evident but glutamine synthetase (GS) staining is indeterminate for β-catenin activation, which does not allow reliable risk assessment. This study examines the role of genomic analysis in better categorization of WD hepatocellular lesions (WDHL). DESIGN Genomic analysis using capture-based NGS assay was done in 23 WDHLs that could not be definitely classified based on morphology, reticulin stain and IHC, and were designated as 'atypical hepatocellular neoplasms' (AHNs). GS staining was classified as diffuse homogeneous (moderate to strong staining in >90 % of tumor cells), diffuse heterogeneous (50-90 %), not diffuse (<50 %) and borderline (not clear if more or less than 50 %). RESULTS The genomic profile provided additional information for the diagnosis and/or risk assessment enabling a benign diagnosis in 15/23 cases (66 %) and HCC in 4/23 cases (17 %), while the diagnosis remained as atypical in the remaining 4 cases. Of the 4 cases with final HCC diagnosis, findings were suspicious but not diagnostic based on morphology/IHC; additional changes like TERT promoter mutation (n = 2), AXIN mutation (n = 1), CDKN2A loss (n = 2) and copy number alterations (n = 3) helped to support HCC. Of the 15 cases with a final benign diagnosis, the status of β-catenin activation was unclear based on GS stain in 8 cases, 2 of which showed CTNNB1 exon 7 mutation, 1 showed CTNNB1 exon 8 mutation, while genomic changes in 5 cases did not show any evidence of Wnt activation. FNH-like features were seen in 2 cases, but the genomic changes excluded FNH (CTNNB1 and ARID1A mutation). The final diagnosis was unchanged from the initial diagnosis of AHN in 4/23 cases (17 %) as the molecular findings did not favor HCC. CONCLUSION Genomic changes were helpful in characterization of WDHLs, supporting HCC in 17 % of cases and clarifying β-catenin activation status in all 7 cases with borderline GS staining. Genomic changes are not specific but can provide diagnostic clues in selected challenging cases that cannot be classified on morphology and IHC. Given the significant treatment implications of distinguishing between HCC and benign/premalignant entities, routine use of genomic analysis in diagnostically challenging settings should be considered.
Collapse
|
7
|
Van Swearingen AED, Lee MR, Rogers LW, Sibley AB, Shi P, Qin X, Goodin M, Seale K, Owzar K, Anders CK. Genomic and immune profiling of breast cancer brain metastases. Acta Neuropathol Commun 2025; 13:99. [PMID: 40355907 PMCID: PMC12070617 DOI: 10.1186/s40478-025-02001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 04/06/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Brain metastases (BrM) arising from breast cancer (BC) are an increasing consequence of advanced disease, with up to half of patients with metastatic HER2 + or triple negative BC experiencing central nervous system (CNS) recurrence. The genomic alterations driving CNS recurrence, along with contributions of the immune microenvironment, particularly by intrinsic subtype, remain unclear. METHODS We characterized the genomic and immune landscape of BCBrM from a cohort of 42 patients by sequencing whole-exome DNA (WES) and total RNA libraries from frozen and FFPE BrM and FFPE extracranial tumors (ECT). Analyses included PAM50 intrinsic subtypes, somatic mutations, copy number variations (CNV), pathway alterations, immune cell type deconvolution, and associations with clinical outcomes RESULTS: Intrinsic subtype calls were concordant for the majority of BrM-ECT pairs (60%). Across all BrM and ECT samples, the most common somatic gene mutation was TP53 (64%, 30/47). For patients with matched FFPE BrM-FFPE ECT, alterations tended to be conserved across tissue type, although differential somatic mutations and CNV in specific genes were observed. Several genomic pathways were differentially expressed between patient-matched BrM-ECT; MYC targets, DNA damage repair, cholesterol homeostasis, and oxidative phosphorylation were higher in BrM, while immune-related pathways were lower in BrM. Deconvolution of immune populations between BrM-ECT demonstrated activated dendritic cell populations were higher in BrM compared to ECT. Increased expression of several oncogenic preselected pathways in BrM were associated with inferior survival, including DNA damage repair, inflammatory response, and oxidative phosphorylation CONCLUSIONS: Collectively, this study illustrates that while some genomic alterations are shared between BrM and ECT, there are also unique aspects of BrM including somatic mutations, CNV, pathway alterations, and immune landscape. A deeper understanding of differences inherent to BrM will contribute to the development of BrM-tailored therapeutic strategies. Additional analyses are warranted in larger cohorts, particularly with additional matched BrM-ECT.
Collapse
Affiliation(s)
| | - Marissa R Lee
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Layne W Rogers
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Alexander B Sibley
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Pixu Shi
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Xiaodi Qin
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Michael Goodin
- Duke Center for Brain and Spine Metastasis, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Katelyn Seale
- Duke Cancer Institute, Duke University Hospital, Durham, NC, USA
| | - Kouros Owzar
- Department of Biostatistics and Bioinformatics, Duke Center for Brain and Spine Metastasis, Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Carey K Anders
- Department of Medical Oncology, Duke Center for Brain and Spine Metastasis, Duke Cancer Institute, Duke University, 10 Searle Center Drive, Campus Box 3881, Durham, NC, 27710, USA.
| |
Collapse
|
8
|
Evans E, Dholakia J, Abraham J, Hinton A, Zhang J, Xiu J, Maney T, Oberley M, Thacker P, Herzog TJ, Spetzler D, Arend RC. Whole-exome sequencing provides assessment of homologous recombination deficiency for identification of PARPi-responsive ovarian tumors. Gynecol Oncol 2025; 197:139-145. [PMID: 40344964 DOI: 10.1016/j.ygyno.2025.04.586] [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: 02/18/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND/OBJECTIVES Homologous recombination repair deficiency (HRD) is frequently detected in gynecological cancers and is associated with sensitivity to poly-ADP ribose polymerase inhibition (PARPi). BRCA1/2 mutations have been approved as biomarkers for PARPi therapy, along with genomic patterns such as genomic loss of heterozygosity (gLOH) and large-scale transitions (LSTs). Clinical applications of various HRD assays are still under investigation. Here we validate the performance of a novel HRD assay based on whole-exome sequencing (WES). METHODS WES was used to evaluate gLOH, LST, and BRCA mutations and this data was compared to standard assays. An optimized genomic scar score (GSS) was based on combined gLOH and LST. A comprehensive HRD score was then developed, combining GSS and BRCA status. Survival data from 1661 PARPi-treated ovarian cancer patients was queried to optimize GSS and HRD scores associated with PARPi response. RESULTS A comparison of WES results to the OncoScan CNV assay and Myriad MyChoice assay showed high concordance for LOH values and GSS values, respectively. Median overall survival in PARPi-treated patients was 50.8 months for GSS-high, BRCA1/2-mut, 42.7 months for GSS-high, BRCA1/2-WT, and 36.6 months for GSS-low, BRCA1/2-WT patients with significant differences between each group. Combining the BRCA1/2-mut and WT GSS-high groups resulted in a median OS value of 47.8 months, significantly higher than the GSS-low BRCA1/2-WT group. CONCLUSIONS The use of a WES assay to assess BRCA results, along with a GSS method incorporating gLOH and LST, produced a HRD test that is predictive for PARPi therapy.
Collapse
Affiliation(s)
- Elizabeth Evans
- Mercy Hospital Coletta Cancer Center, Oklahoma City, OK, USA
| | | | | | | | | | | | | | | | - Premal Thacker
- Washington University School of Medicine, St. Louis, MO, USA
| | - Thomas J Herzog
- Washington University School of Medicine, St. Louis, MO, USA
| | | | | |
Collapse
|
9
|
Gridina M, Lagunov T, Belokopytova P, Torgunakov N, Nuriddinov M, Nurislamov A, Nazarenko LP, Kashevarova AA, Lopatkina ME, Vasilyev S, Zuev A, Belyaeva EO, Salyukova OA, Cheremnykh AD, Sukhanova NN, Minzhenkova ME, Markova ZG, Demina NA, Stepanchuk Y, Khabarova A, Yan A, Valeev E, Koksharova G, Grigor'eva EV, Kokh N, Lukjanova T, Maximova Y, Musatova E, Shabanova E, Kechin A, Khrapov E, Boyarskih U, Ryzhkova O, Suntsova M, Matrosova A, Karoli M, Manakhov A, Filipenko M, Rogaev E, Shilova NV, Lebedev IN, Fishman V. Combining chromosome conformation capture and exome sequencing for simultaneous detection of structural and single-nucleotide variants. Genome Med 2025; 17:47. [PMID: 40336115 PMCID: PMC12060427 DOI: 10.1186/s13073-025-01471-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 04/10/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Effective molecular diagnosis of congenital diseases hinges on comprehensive genomic analysis, traditionally reliant on various methodologies specific to each variant type-whole exome or genome sequencing for single nucleotide variants (SNVs), array CGH for copy-number variants (CNVs), and microscopy for structural variants (SVs). METHODS We introduce a novel, integrative approach combining exome sequencing with chromosome conformation capture, termed Exo-C. This method enables the concurrent identification of SNVs in clinically relevant genes and SVs across the genome and allows analysis of heterozygous and mosaic carriers. Enhanced with targeted long-read sequencing, Exo-C evolves into a cost-efficient solution capable of resolving complex SVs at base-pair accuracy. RESULTS Applied to 66 human samples Exo-C achieved 100% recall and 73% precision in detecting chromosomal translocations and SNVs. We further benchmarked its performance for inversions and CNVs and demonstrated its utility in detecting mosaic SVs and resolving diagnostically challenging cases. CONCLUSIONS Through several case studies, we demonstrate how Exo-C's multifaceted application can effectively uncover diverse causative variants and elucidate disease mechanisms in patients with rare disorders.
Collapse
Affiliation(s)
- Maria Gridina
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia.
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia.
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia.
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia.
| | - Timofey Lagunov
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
| | - Polina Belokopytova
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Nikita Torgunakov
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
| | - Miroslav Nuriddinov
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
| | - Artem Nurislamov
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia
| | - Lyudmila P Nazarenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Anna A Kashevarova
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Maria E Lopatkina
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Stanislav Vasilyev
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Andrey Zuev
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Elena O Belyaeva
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Olga A Salyukova
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Aleksandr D Cheremnykh
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Natalia N Sukhanova
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | | | | | - Nina A Demina
- Research Centre for Medical Genetics, Moscow, 115522, Russia
| | - Yana Stepanchuk
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
| | - Anna Khabarova
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
| | - Alexandra Yan
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
| | - Emil Valeev
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
| | - Galina Koksharova
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia
| | - Elena V Grigor'eva
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
| | - Natalia Kokh
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
| | - Tatiana Lukjanova
- Center for Family Care and Reproduction, 1 Kiyevskaya Str, Novosibirsk, 6300136, Russia
| | - Yulia Maximova
- Center for Family Care and Reproduction, 1 Kiyevskaya Str, Novosibirsk, 6300136, Russia
- Novosibirsk State Medical University, Novosibirsk, 630091, Russia
| | - Elizaveta Musatova
- Genetics and Reproductive Medicine Center, "GENETICO" PJSC, Moscow, 119333, Russia
| | - Elena Shabanova
- North-Western State Medical University named after I.I. Mechnikov, Saint-Petersburg, 191015, Russia
| | - Andrey Kechin
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
| | - Evgeniy Khrapov
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
| | - Uliana Boyarskih
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
| | - Oxana Ryzhkova
- Research Centre for Medical Genetics, Moscow, 115522, Russia
| | - Maria Suntsova
- Sechenov First Moscow State Medical University, Moscow, 119435, Russia
- Endocrinology Research Center, Moscow, 117292, Russia
| | - Alina Matrosova
- Sechenov First Moscow State Medical University, Moscow, 119435, Russia
- Endocrinology Research Center, Moscow, 117292, Russia
| | - Mikhail Karoli
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia
| | - Andrey Manakhov
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia
| | - Maxim Filipenko
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
| | - Evgeny Rogaev
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia
- UMass Chan Medical School, Worcester, 01655, USA
| | | | - Igor N Lebedev
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Veniamin Fishman
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia.
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia.
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia.
- Artificial Intelligence Research Institute, Moscow, Russia, 121170.
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia.
| |
Collapse
|
10
|
Zhu H, Zhang J, Rao S, Durbin MD, Li Y, Lang R, Liu J, Xiao B, Shan H, Meng Z, Wang J, Tang X, Shi Z, Cox LL, Zhao S, Ware SM, Tan TY, de Silva M, Gallacher L, Liu T, Mi J, Zeng C, Zheng HF, Zhang Q, Antonarakis SE, Cox TC, Zhang YB. Common cis-regulatory variation modifies the penetrance of pathogenic SHROOM3 variants in craniofacial microsomia. Genome Res 2025; 35:1065-1079. [PMID: 40234029 PMCID: PMC12047249 DOI: 10.1101/gr.280047.124] [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: 10/07/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025]
Abstract
Pathogenic coding variants have been identified in thousands of genes, yet the mechanisms underlying the incomplete penetrance in individuals carrying these variants are poorly understood. In this study, in a cohort of 2009 craniofacial microsomia (CFM) patients of Chinese ancestry and 2625 Han Chinese controls, we identified multiple predicted pathogenic coding variants in SHROOM3 in both CFM patients and healthy individuals. We found that the penetrance of CFM correlates with specific haplotype combinations containing likely pathogenic-coding SHROOM3 variants and CFM-associated expression quantitative trait loci (eQTLs) of SHROOM3 expression. Further investigations implicate specific eQTL combinations, such as rs1001322 or rs344131, in combination with other significant CFM-associated eQTLs, which we term combined eQTL phenotype modifiers (CePMods). We additionally show that rs344131, located within a regulatory enhancer region of SHROOM3, demonstrates allele-specific effects on enhancer activity and thus impacts expression levels of the associated SHROOM3 allele harboring any rare coding variant. Our findings also suggest that CePMods may serve as pathogenic determinants, even in the absence of rare deleterious coding variants in SHROOM3 This highlights the critical role of allelic expression in determining the penetrance and severity of craniofacial abnormalities, including microtia and facial asymmetry. Additionally, using quantitative phenotyping, we demonstrate that both microtia and facial asymmetry are present in two separate Shroom3 mouse models, the severity of which is dependent on gene dosage. Our study establishes SHROOM3 as a likely pathogenic gene for CFM and demonstrates eQTLs as determinants of modified penetrance in the manifestation of the disease in individuals carrying likely pathogenic rare coding variants.
Collapse
Affiliation(s)
- Hao Zhu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Jiao Zhang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Beijing 100144, China
| | - Soumya Rao
- Department of Oral & Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA
| | - Matthew D Durbin
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Ying Li
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100051, China
| | - Ruirui Lang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Jiqiang Liu
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Baichuan Xiao
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Hailin Shan
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Ziqiu Meng
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Jinmo Wang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Xiaokai Tang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Zhenni Shi
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Liza L Cox
- Department of Oral & Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA
| | - Shouqin Zhao
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100051, China
| | - Stephanie M Ware
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Tiong Y Tan
- Victorian Clinical Genetics Service, Royal Children's Hospital and Department of Pediatrics, University of Melbourne, Victoria 3052, Australia
| | - Michelle de Silva
- Victorian Clinical Genetics Service, Royal Children's Hospital and Department of Pediatrics, University of Melbourne, Victoria 3052, Australia
| | - Lyndon Gallacher
- Victorian Clinical Genetics Service, Royal Children's Hospital and Department of Pediatrics, University of Melbourne, Victoria 3052, Australia
| | - Ting Liu
- Department of Ophthalmology, Daping Hospital, Army Medical University, Chongqing 400000, China
| | - Jie Mi
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Changqing Zeng
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Hou-Feng Zheng
- Center for Health and Data Science (CHDS), the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
- Diseases & Population (DaP) Geninfo Laboratory, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Qingguo Zhang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Beijing 100144, China
| | - Stylianos E Antonarakis
- Department of Genetic Medicine and Development, University of Geneva Medical Faculty, Geneva 1211, Switzerland;
- Medigenome, Swiss Institute of Genomic Medicine, 1207 Geneva, Switzerland
- iGE3 Institute of Genetics and Genomes in Geneva, Geneva 1211, Switzerland
| | - Timothy C Cox
- Department of Oral & Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA;
- Department of Pediatrics, University of Missouri-Kansas City, Kansas City, Missouri 64108, USA
| | - Yong-Biao Zhang
- School of Engineering Medicine, Beihang University, Beijing 100191, China;
- Key Laboratory of Big Data-Based Precision Medicine and Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing 100191, China
- National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), Beihang University, Beijing 100083, China
| |
Collapse
|
11
|
Mendoza RP, Tjota MY, Choi DN, Chapel DB, Kolin DL, Euscher ED, Barroeta JE, Numan TA, Xing D, Afkhami M, Bakkar R, Lastra RR. Clinicopathologic and Molecular Characterization of Gynecologic Carcinosarcomas With a Mesonephric-Like Carcinomatous Component. Am J Surg Pathol 2025; 49:439-447. [PMID: 39931796 DOI: 10.1097/pas.0000000000002368] [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] [Indexed: 04/17/2025]
Abstract
Carcinosarcoma with a mesonephric-like carcinomatous component (MLCS) is a rare subtype of gynecologic malignancy recently described in the literature. This study aims to expand the genomic characterization of MLCS by performing independent molecular analysis of the carcinomatous and sarcomatous components in a series of MLCS. Eight cases of gynecologic MLCS (endometrial, lower uterine segment, and ovarian) were identified and underwent clinicopathologic evaluation. Genomic DNA extraction and next-generation sequencing (NGS) were performed separately from the carcinomatous and sarcomatous components of 4 tumors, while 2 tumors underwent NGS of combined carcinomatous and sarcomatous components. The average age at diagnosis was 65.6 years (range 50 to 83 years). MLCS patients were diagnosed at FIGO stage I (n=3), stage II (n=2), stage III (n=2), and stage IV (n=1). The carcinomatous and sarcomatous components were observed to harbor the same single nucleotide variations. All cases had less than 10 mutations/Mb and were microsatellites stable. All cases (6/6, 100%) harbored KRAS point mutations in codon 12, including the following variants: p.G12D (n=2), p.G12A (n=2), and p.G12V (n=2). Five cases showed additional alterations in ARID1A (case 1), PTEN (case 2), PIK3CA (case 4), SPOP (case 6), TET1 (case 6), BUB1 (case 7), LYN (case 7) and PTPRD (case 7). The presence of both KRAS and PTEN / PIK3CA alterations suggests a combined endometrioid and mesonephric differentiation in MLCS.
Collapse
Affiliation(s)
- Rachelle P Mendoza
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Melisa Y Tjota
- Department of Pathology, University of Chicago Medical Center, Chicago, IL
| | - Donghyuk N Choi
- Department of Pathology, University of Rochester Medical Center, Rochester, NY
| | - David B Chapel
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | - David L Kolin
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
| | | | - Julieta E Barroeta
- Department of Pathology, University of Chicago Medical Center, Chicago, IL
| | - Tricia A Numan
- Department of Pathology, Johns Hopkins Medical Institute, Baltimore, MD
| | - Deyin Xing
- Department of Pathology, Johns Hopkins Medical Institute, Baltimore, MD
| | - Michelle Afkhami
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Rania Bakkar
- Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Ricardo R Lastra
- Department of Pathology, University of Chicago Medical Center, Chicago, IL
| |
Collapse
|
12
|
Bravo-Perez C, Gurnari C, Huuhtanen J, Kawashima N, Guarnera L, Mandala A, Williams ND, Haddad C, Witt M, Unlu S, Brady Z, Ogbue O, Orland M, Ahmed A, Kubota Y, Pagliuca S, Durmaz A, Mustjoki S, Visconte V, Maciejewski JP. Inborn errors of immunity underlie clonal T cell expansions in large granular lymphocyte leukemia. J Clin Invest 2025; 135:e184431. [PMID: 40309770 PMCID: PMC12043085 DOI: 10.1172/jci184431] [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: 07/01/2024] [Accepted: 02/27/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUNDT cell large granular lymphocyte leukemia (T-LGLL) is a lymphoproliferative disorder of cytotoxic T lymphocytes (CTLs), often with gain-of-function STAT3 mutations. T-LGLL represents a unique model for the study of persistent CTL expansions. Albeit autoimmunity is implied, various paradoxical observations led us to investigate whether immunodeficiency traits underpin T-LGLL.METHODSThis is a comprehensive immunogenomic study of 92 consecutive patients from a large T-LGLL cohort with full laboratory-clinical characterization (n = 271). Whole-exome profiling of variants associated with inborn errors of immunity (IEI) and somatic mutations in T cell lymphoid drivers was analyzed. Single-cell RNA-Seq and TCR-Seq in T-LGLL samples and RNA-Seq in T cell cancer cell lines were utilized to establish biological correlations.RESULTSLymphocytopenia and/or hypogammaglobulinemia were identified in 186 of 241 (77%) T-LGLL patients. Genetic screening for IEI revealed 43 rare heterozygous variants in 38 different immune genes in 34 of 92 (36%) patients (vs. 167/63,026 [0.26%] in controls). High-confidence deleterious variants associated with dominant, adult-onset IEIs were detected in 15 of 92 (16%) patients. Carriers showed atypical features otherwise tied to the cryptic IEI, such as earlier onset, lower lymphocyte counts, lower STAT3 mutational rate, and higher proportions of hypogammaglobulinemia and immune cytopenia/bone marrow failure than noncarriers. Somatic mutational landscape, RNA-Seq, and TCR-Seq analyses supported immune imbalance caused by the IEI variants and interactions with somatic mutations in T cell lymphoid drivers.CONCLUSIONSOur findings in T-LGLL reveal that maladaptive CTL expansions may stem from cryptic immunodeficiency traits and open the horizon of IEIs to clonal hematopoiesis and bone marrow failure.FUNDINGNIH; Aplastic Anemia and MDS International Foundation; VeloSano; Edward P. Evans Foundation; Instituto de Salud Carlos III; European Research Council; European Research Area Network on Personalised Medicine; Academy Finland; Cancer Foundation Finland.
Collapse
Affiliation(s)
- Carlos Bravo-Perez
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, University of Murcia, IMIB-Pascual Parrilla, CIBERER–Instituto de Salud Carlos III, Murcia, Spain
| | - Carmelo Gurnari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Jani Huuhtanen
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Department of Computer Science, Aalto University, Espoo, Finland
| | - Naomi Kawashima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Luca Guarnera
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Aashray Mandala
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Nakisha D. Williams
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Christopher Haddad
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Michaela Witt
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Serhan Unlu
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Zachary Brady
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Olisaemeka Ogbue
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mark Orland
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Arooj Ahmed
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Yasuo Kubota
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Simona Pagliuca
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Hematology Department, Nancy University Hospital and UMR 7365 CNRS University of Lorraine, France
| | - Arda Durmaz
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Valeria Visconte
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
13
|
Zhu G, Sun Z, Liu Y, Liu J, Guo L, Pei G, Jiang Y, Miao B, Li Z, Zhang P, Tang D, Zhang W, Wang C. Rational Design and Organoid-Based Evaluation of a Cocktail CAR-γδ T Cell Therapy for Heterogeneous Glioblastoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2501772. [PMID: 40112194 PMCID: PMC12097020 DOI: 10.1002/advs.202501772] [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] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Indexed: 03/22/2025]
Abstract
Various challenges, including tumor heterogeneity and inadequate T cell infiltration, impede the progress of chimeric antigen receptor T cell (CAR-T) therapy for glioblastoma (GBM). To address these obstacles, a multiple step strategy is designed. Initially, literature review and bioinformatics analysis to screen a set of antigens that are heterogeneously expressed in GBM, which are designated as the target-bank, are leveraged. Then, according to the multiplex immunohistochemistry results of each patient's tumor sample, a personalized panel of antigens based on the principle that most cancer cells in tumor tissues can be covered from the target-bank is selected. To target these antigens, Vδ1 T cells are chosen as CAR vehicles because of its high tissue infiltration and off-the-shelf properties, and an optimized protocol for engineering CAR-Vδ1 T cells with high purity and cytotoxicity, low exhaustion, and cytokine release is developed. Next, the specific panel of cocktail CAR-Vδ1 T cells in the GBM organoids that are directly derived from the same patient's tumor is tested. The term "prof" cocktail therapy is coined to describe the approach using precise and rational combination of tumor antigens, organoid-based evaluation, and fitness of Vδ1 T cells. It may accelerate development of effective CAR-T drugs for heterogeneous solid tumors.
Collapse
Affiliation(s)
- Guidong Zhu
- Department of Neurosurgery, The Second Hospital, Cheeloo College of MedicineShandong UniversityJinan250031P.R. China
| | - Zhongzheng Sun
- Department of Neurosurgery, The Second Hospital, Cheeloo College of MedicineShandong UniversityJinan250031P.R. China
| | - Yingchao Liu
- Department of NeurosurgeryProvincial Hospital Affiliated to Shandong First Medical UniversityJinan250031P.R. China
| | - Jiang Liu
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of MedicineShandong UniversityJinan250031P.R. China
| | - Linpei Guo
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of MedicineShandong UniversityJinan250031P.R. China
| | - Guojing Pei
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of MedicineShandong UniversityJinan250031P.R. China
| | - Ying Jiang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of MedicineShandong UniversityJinan250031P.R. China
| | - Baowang Miao
- Department of Neurosurgery, The Second Hospital, Cheeloo College of MedicineShandong UniversityJinan250031P.R. China
| | - Zhen Li
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of MedicineShandong UniversityJinan250031P.R. China
| | - Ping Zhang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of MedicineShandong UniversityJinan250031P.R. China
| | - Dongqi Tang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of MedicineShandong UniversityJinan250031P.R. China
- Multidisciplinary Innovation Center for Nephrology of the Second Hospital of Shandong UniversityJinan250031China
| | - Wen Zhang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of MedicineShandong UniversityJinan250031P.R. China
| | - Chengwei Wang
- Department of Neurosurgery, The Second Hospital, Cheeloo College of MedicineShandong UniversityJinan250031P.R. China
| |
Collapse
|
14
|
Boulos H, Lo C, Zhu W, Driessen TM, Yamada-Hanff J, Harding T, Lozac'hmeur A, Pereira T, Sonnenschein A, Och J, Jin A, Patel N, Blidner R, Tell R, Freaney J, Beaubier N, Mahon B. Analytical Validation of Next-Generation Sequencing-Based Comprehensive Liquid Biopsy Assay for Therapy Selection. J Mol Diagn 2025; 27:383-394. [PMID: 40287222 DOI: 10.1016/j.jmoldx.2025.02.005] [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: 04/03/2024] [Revised: 10/29/2024] [Accepted: 02/06/2025] [Indexed: 04/29/2025] Open
Abstract
Liquid biopsies are an increasingly important tool for the real-time monitoring of biomarkers, cancer recurrence, and disease burden in oncology practice. Tempus xF+ is a liquid biopsy assay that detects cell-free DNA in blood samples of patients with advanced solid tumors. The xF+ panel covers 523 genes spanning approximately 1.8 Mb of the human genome and can detect single-nucleotide variants and insertions-deletions in 522 genes. It also detects copy number gains in 7 genes and translocations (gene rearrangements) in 10 genes. Furthermore, the larger panel size allows for the calculation of blood tumor mutational burden. This work highlights the analytical validation performed for the xF+ assay, comparing it with a smaller panel liquid biopsy assay, calculating blood tumor mutational burden, and exploring its potential clinical utility.
Collapse
Affiliation(s)
| | | | - Wei Zhu
- Tempus AI, Inc., Chicago, Illinois
| | | | | | | | | | | | | | - Josh Och
- Tempus AI, Inc., Chicago, Illinois
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Shi T, Gao Z, Zhang Y, Rausher MD, Chen J. A Strategy of Assessing Gene Copy Number Differentiation Between Populations Using Ultra-Fast De Novo Assembly of Next-Generation Sequencing Data. Mol Ecol Resour 2025; 25:e14080. [PMID: 39925235 DOI: 10.1111/1755-0998.14080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 01/10/2025] [Accepted: 01/27/2025] [Indexed: 02/11/2025]
Abstract
Gene duplication and loss play pivotal roles in the evolutionary dynamics of genomes, contributing to species phenotypic diversity and adaptation. However, detecting copy number variations (CNVs) in homoploid populations and newly-diverged species using short reads from next-generation sequencing (NGS) with traditional methods can often be challenging due to uneven read coverage caused by variations in GC content and the presence of repetitive sequences. To address these challenges, we developed a novel pipeline, ST4gCNV, which leverages ultra-fast de novo assemblies of NGS data to detect gene-specific CNVs between populations. The pipeline effectively reduces the variance of read coverage due to technical factors such as GC bias, providing a reliable CNV detection with a minimum sequencing depth of 10. We successfully apply ST4gCNV to the resequencing analysis of homoploid species Nelumbo nucifera and Nelumbo lutea (lotus). We reveal significant CNV-driven differentiation between these species, particularly in genes related to petal colour diversity such as those involved in the anthocyanin pathway. By highlighting the extensive gene duplication and loss events in Nelumbo, our study demonstrates the utility of ST4gCNV in population genomics and underscores its potential of integrating genomic CNV analysis with traditional SNP-based resequencing analysis.
Collapse
Affiliation(s)
- Tao Shi
- State Key Laboratory of Plant Diversity and Specialty Crops, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan, China
- Hubei Key Laboratory of Wetland Evolution & Ecological Restoration, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan, China
| | - Zhiyan Gao
- State Key Laboratory of Plant Diversity and Specialty Crops, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan, China
- Hubei Key Laboratory of Wetland Evolution & Ecological Restoration, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan, China
| | - Yue Zhang
- State Key Laboratory of Plant Diversity and Specialty Crops, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan, China
- Hubei Key Laboratory of Wetland Evolution & Ecological Restoration, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan, China
- Aquatic Plant Research Center, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan, China
| | - Mark D Rausher
- Department of Biology, Duke University, Durham, North Carolina, USA
| | - Jinming Chen
- State Key Laboratory of Plant Diversity and Specialty Crops, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan, China
- Hubei Key Laboratory of Wetland Evolution & Ecological Restoration, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan, China
- Aquatic Plant Research Center, Wuhan Botanical Garden, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
16
|
Yost KE, Zhao Y, Hung KL, Zhu K, Xu D, Corces MR, Shams S, Louie BH, Sarmashghi S, Sundaram L, Luebeck J, Clarke S, Doane AS, Granja JM, Choudhry H, Imieliński M, Cherniack AD, Khurana E, Bafna V, Felau I, Zenklusen JC, Laird PW, Curtis C, Greenleaf WJ, Chang HY. Three-dimensional genome landscape of primary human cancers. Nat Genet 2025; 57:1189-1200. [PMID: 40355593 DOI: 10.1038/s41588-025-02188-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/02/2025] [Indexed: 05/14/2025]
Abstract
Genome conformation underlies transcriptional regulation by distal enhancers, and genomic rearrangements in cancer can alter critical regulatory interactions. Here we profiled the three-dimensional genome architecture and enhancer connectome of 69 tumor samples spanning 15 primary human cancer types from The Cancer Genome Atlas. We discovered the following three archetypes of enhancer usage for over 100 oncogenes across human cancers: static, selective gain or dynamic rewiring. Integrative analyses revealed the enhancer landscape of noncancer cells in the tumor microenvironment for genes related to immune escape. Deep whole-genome sequencing and enhancer connectome mapping provided accurate detection and validation of diverse structural variants across cancer genomes and revealed distinct enhancer rewiring consequences from noncoding point mutations, genomic inversions, translocations and focal amplifications. Extrachromosomal DNA promoted more extensive enhancer rewiring among several types of focal amplification mechanisms. These results suggest a systematic approach to understanding genome topology in cancer etiology and therapy.
Collapse
Affiliation(s)
- Kathryn E Yost
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Yanding Zhao
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - King L Hung
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kaiyuan Zhu
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Duo Xu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York City, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York City, NY, USA
- Pathos AI, Chicago, IL, USA
| | - M Ryan Corces
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Shadi Shams
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Bryan H Louie
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Laksshman Sundaram
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Illumina AI laboratory, Illumina Inc, Foster City, CA, USA
- NVIDIA Bio Research, NVIDIA, Santa Clara, CA, USA
| | - Jens Luebeck
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Stanley Clarke
- Vilcek Institute of Graduate Biomedical Sciences, NYU Grossman School of Medicine, New York City, NY, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York City, NY, USA
- Department of Pathology, New York University Langone Health, New York City, NY, USA
- New York Genome Center, New York City, NY, USA
| | - Ashley S Doane
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Jeffrey M Granja
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, Cancer and Mutagenesis Unit, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Marcin Imieliński
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York City, NY, USA
- Department of Pathology, New York University Langone Health, New York City, NY, USA
- New York Genome Center, New York City, NY, USA
| | - Andrew D Cherniack
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ekta Khurana
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York City, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York City, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York City, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Vineet Bafna
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Ina Felau
- National Cancer Institute, NIH, Bethesda, MD, USA
| | | | - Peter W Laird
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Christina Curtis
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Applied Physics, Stanford University, Stanford, CA, USA.
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
17
|
Tao T, Liu S, He M, Zhao M, Chen C, Peng J, Wang Y, Cai J, Xiong J, Lai C, Gu W, Ying M, Mao J, Li L, Jia X, Wu X, Peng W, Zhang X, Li Y, Li T, Wang J, Shu Q. Synchronous bilateral Wilms tumors are prone to develop independently and respond differently to preoperative chemotherapy. Int J Cancer 2025; 156:1746-1755. [PMID: 39723643 DOI: 10.1002/ijc.35297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/20/2024] [Accepted: 11/28/2024] [Indexed: 12/28/2024]
Abstract
Wilms tumor (WT) is the most common kidney cancer in infants and young children. The determination of the clonality of bilateral WTs is critical to the treatment, because lineage-independent and metastatic tumors may require different treatment strategies. Here we found synchronous bilateral WT (n = 24 tumors from 12 patients) responded differently to preoperative chemotherapy. Transcriptome, whole-exome and whole-genome analysis (n = 12 tumors from 6 patients) demonstrated that each side of bilateral WT was clonally independent in terms of somatic driver mutations, copy number variations and transcriptomic profile. Molecular timing analysis revealed distinct timing and patterns of chromosomal evolution and mutational processes between the two sides of WT. Mutations in WT1, CTNNB1 and copy-neutral loss of heterozygosity of 11p15.5 provide possible genetic predisposition for the early initiation of bilateral WT. Our results provide comprehensive evidence and new insights regarding the separate initiation and early embryonic development of bilateral WT, which may benefit clinical practices in treating metastatic or refractory bilateral WT.
Collapse
Affiliation(s)
- Ting Tao
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Shuangai Liu
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- The First Clinical Institute, Zunyi Medical University, Zunyi, China
| | - Min He
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Manli Zhao
- Department of Pathology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chen Chen
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jinkai Peng
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yilong Wang
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Neurology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jiabin Cai
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jieni Xiong
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Can Lai
- Department of Radiology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Weizhong Gu
- Department of Pathology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Meidan Ying
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
- Nanhu Brain-computer Interface Institute, Hangzhou, China
| | - Junqing Mao
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Linjie Li
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xuan Jia
- Department of Radiology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xuan Wu
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Wanxin Peng
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Xiang Zhang
- The Affiliated Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yong Li
- Hunan Children's Hospital, Changsha, China
| | - Tao Li
- Department of Oncology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jinhu Wang
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Qiang Shu
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Children's Hospital Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Key Laboratory of Diagnosis and Treatment of Neonatal Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
18
|
Ikeda J, Shiba N, Kato S, Kunimoto H, Saito Y, Sagisaka M, Ito M, Goto H, Okuno Y, Nakamura W, Yoshitomi M, Takeuchi M, Ito S, Nakajima H, Kato M, Tsujimoto SI. Establishment of a high-risk pediatric AML-derived cell line YCU-AML2 with genetic and metabolic vulnerabilities. Int J Hematol 2025; 121:694-705. [PMID: 39891826 DOI: 10.1007/s12185-025-03929-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/03/2025]
Abstract
The prognosis of acute myeloid leukemia (AML) with KMT2A::MLLT3 rearrangement and MECOM overexpression and/or KRAS mutation is dismal, and the optimal treatment strategy remains unclear. However, to the best of our knowledge, a suitable model (such as a cell line or its xenograft model) for research on this subtype has not been established. We established a novel AML cell line, YCU-AML2, and its xenograft model harboring KMT2A::MLLT3 rearrangement, MECOM overexpression, and KRAS G12A mutation. YCU-AML2 xenograft mice models developed AML and mimicked the clinical phenotype of the original patient. YCU-AML2 expressed high sensitivity to MEK inhibitors, such as trametinib and selumetinib. Moreover, YCU-AML2 also exhibited high sensitivity to L-asparaginase with glutaminase activity, perhaps because of its reliance on oxidative phosphorylation via glutaminolysis as its main energy source. We believe that the YCU-AML2 cell line and its xenograft model can serve as models to explore the molecular pathogenesis of high-risk AML with KMT2A::MLLT3 rearrangement, MECOM overexpression, and/or KRAS mutation and develop new treatment strategies.
Collapse
Affiliation(s)
- Junji Ikeda
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
- Department of Stem Cell and Immune Regulation, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Norio Shiba
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Shota Kato
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyoshi Kunimoto
- Department of Stem Cell and Immune Regulation, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Yusuke Saito
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan
| | - Maiko Sagisaka
- Clinical Research Institute, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Mieko Ito
- Clinical Research Institute, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Hiroaki Goto
- Division of Hematology/Oncology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yusuke Okuno
- Department of Virology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Wataru Nakamura
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Masahiro Yoshitomi
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Masanobu Takeuchi
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Shuichi Ito
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Hideaki Nakajima
- Department of Stem Cell and Immune Regulation, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Motohiro Kato
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shin-Ichi Tsujimoto
- Department of Pediatrics, Graduate School of Medicine, Yokohama City University, 3-9, Fukuura, Kanazawa-Ku, Yokohama, Kanagawa, 236-0004, Japan.
| |
Collapse
|
19
|
Liu B, Tao W, Zhou X, Xu LD, Luo Y, Yang X, Min Q, Huang M, Zhu Y, Cui X, Wang Y, Gong T, Zhang E, Huang YS, Chen W, Yan S, Wu N. Multi‑omics analysis identifies different molecular subtypes with unique outcomes in early-stage poorly differentiated lung adenocarcinoma. Mol Cancer 2025; 24:129. [PMID: 40312720 PMCID: PMC12044723 DOI: 10.1186/s12943-025-02333-7] [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: 02/22/2025] [Accepted: 04/12/2025] [Indexed: 05/03/2025] Open
Abstract
INTRODUCTION Early-stage poorly differentiated lung adenocarcinoma (LUAD) is plagued by a high risk of postoperative recurrence, and its prognostic heterogeneity complicates treatment and surveillance planning. We conducted this integrative multi-omics study to identify those patients with a truly high risk of adverse outcomes. METHODS Whole-exome, RNA and whole methylome sequencing were carried out on 101 treatment-naïve early-stage poorly differentiated LUADs. Integrated analyses were conducted to disclose molecular characteristics and explore molecular subtyping. Functional validation of key molecules was carried out through in vitro and in vivo experiments. RESULTS Recurrent tumors exhibited significantly higher ploidy (p = 0.024), the fraction of the genome altered (FGA, p = 0.042), and aneuploidy (p < 0.05) compared to non-recurrent tumors, as well as a higher frequency of CNVs. Additionally, recurrent tumors showed hypomethylation at both the global level and in CpG island regions. Integrative transcriptomic and methylation analyses identified three molecular subtypes (C1, C2, and C3), with the C1 subtype presenting the worst prognosis (p = 0.024). Although frequently mutated genes showed similar mutation frequencies across the three subtypes, the C1 subtype exhibited the highest tumor mutation burden (TMB), mutant-allele tumor heterogeneity (MATH), aneuploidy, and HLA loss of heterozygosity (HLA-LOH), along with relatively lower immune cell infiltration. Furthermore, GINS1 and CPT1C were found to promote LUAD progression, and their high expression correlated with a poor prognosis. CONCLUSIONS This multi-omics study identified three integrative subtypes with distinct prognostic implications, paving the way for more precise management and postoperative monitoring of early-stage poorly differentiated LUAD.
Collapse
Affiliation(s)
- Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Wei Tao
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Xuantong Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Li-Di Xu
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Yanrui Luo
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Xin Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qingjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Miao Huang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yuge Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xinrun Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yaqi Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Tongyang Gong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Enli Zhang
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Yu S Huang
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Weizhi Chen
- Genecast Biotechnology Co, Ltd, Wuxi, Jiangsu, China
| | - Shi Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Nan Wu
- State Key Laboratory of Molecular Oncology, Frontiers Science Center for Cancer Integrative Omics, Department of Thoracic Surgery II, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan, China.
| |
Collapse
|
20
|
Papke DJ, Chrisinger JSA, French CA, Crymes A, Krivak TC, Estape RE, Seetharam M, Patel RA, O'Connor WN, Chi AW, Gutman P, Singer S, Kim C, Bryant DA, Oberley MJ, Adeyelu T, Bridge JA, Evans MG. MAD::NUT Fusion Sarcoma: A Sarcoma Class With NUTM1, NUTM2A, and NUTM2G Fusions and Possibly Distinctive Subtypes. Mod Pathol 2025; 38:100729. [PMID: 39921028 DOI: 10.1016/j.modpat.2025.100729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/31/2024] [Accepted: 01/27/2025] [Indexed: 02/10/2025]
Abstract
NUT fusion-associated cancers are heterogeneous and include NUT carcinoma and an emerging group with non-BRD4/BRD3/NSD3 fusion partners. In this study, we characterized 11 tumors harboring MAD::NUT fusions (10/11 in female patients; median age: 48 years; range: 1-67 years), all histologically different from NUT carcinoma. Eight cases were identified via sequencing database review and 3 were diagnosed prospectively. Eight (73%) patients presented with multifocal disease, including 6 with disseminated peritoneal tumors; 3 (27%) presented with solitary colonic, pulmonary, or orbital masses. Nine (82%) tumors harbored NUTM1 fusions, with MXI1 (5/9; 56%), MXD4 (2/9; 22%), and MGA (2/9; 22%). One tumor each harbored MXD4::NUTM2G and MXI1::NUTM2A fusions. The 9 MXD4/MXI1-rearranged sarcomas were high-grade, with epithelioid-to-spindle cell cytomorphology, amphophilic cytoplasm, vesicular nuclei, and prominent nucleoli. Histologic features included infiltrative growth (7/7 assessable tumors), rhabdoid morphology (7/9; 78%), prominent collagen (3/9; 33%), multinucleated tumor cells (2/9; 22%), and myxoid stroma (1/9; 11%). MXD4/MXI1-rearranged sarcomas expressed desmin (3/7; 43%) and keratin(s) (3/7; 43%), and not p63 (6 tumors), CD34 (5 tumors), or S-100 (5 tumors). The adult MGA::NUTM1 fusion sarcoma exhibited some cytologic overlap with MXD4/MXI1-rearranged sarcomas but showed lower grade myxoid spindle cell regions, microcystic spaces, and S-100 expression. The pediatric MGA::NUTM1 fusion sarcoma was low-grade with CD34/S-100 coexpression. Immunohistochemistry demonstrated NUTM1 expression in NUTM1-rearranged sarcomas (5/5), and weak and no expression in NUTM2A- and NUTM2G-rearranged sarcomas, respectively. Gene expression profiling demonstrated sarcomas with MXD4/MXI1::NUTM1/NUTM2A/NUTM2G fusions clustered separately from NUT carcinoma. Follow-up was available for 9 patients (82%; median length: 1.8 years; range: 2 months to 8.2 years). Four of 7 patients with MXD4/MXI1-rearranged sarcomas died of disease (median survival: 1.3 years; range: 5 months to 4.8 years), 1 entered hospice at 2 months, 1 was alive with pericardial masses at 2.8 years, and 1 was alive with no evidence of disease at 8.2 years. The adult with the MGA::NUTM1 fusion sarcoma died of other causes at 4.5 years; the child was alive without disease at 11 months. We conclude that MAD::NUT fusions define a sarcoma class distinct from NUT carcinoma. Among this group, MGA::NUTM1 fusion sarcomas might represent a distinctive subset. NUTM1 immunohistochemistry does not reliably detect NUTM2A/NUTM2G-rearranged sarcomas.
Collapse
Affiliation(s)
- David J Papke
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| | - John S A Chrisinger
- Division of Anatomic and Molecular Pathology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Christopher A French
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Anthony Crymes
- Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Thomas C Krivak
- Division of Gynecologic Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, Pennsylvania
| | - Ricardo E Estape
- Hospital Corporation of America (HCA) Florida Institute for Gynecologic Oncology, HCA Florida Healthcare, Miami, Florida
| | - Mahesh Seetharam
- Division of Hematology and Oncology, Mayo Clinic Arizona, Phoenix, Arizona
| | - Reema A Patel
- Division of Medical Oncology, Department of Internal Medicine, University of Kentucky College of Medicine, Lexington, Kentucky
| | - William N O'Connor
- Department of Pathology and Laboratory Medicine, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Anthony W Chi
- Rockville Regional Lab, Kaiser Permanente Mid-Atlantic Medical Group, Rockville, Maryland
| | - Pablo Gutman
- Department of Pathology, Holy Cross Hospital, Silver Spring, Maryland
| | - Stephan Singer
- Institute of Pathology, University Hospital Tübingen, Tübingen, Germany
| | - Chul Kim
- Department of Medicine, Division of Hematology and Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | | | | | | | - Julia A Bridge
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, Nebraska; ProPath, Dallas, Texas
| | | |
Collapse
|
21
|
Zhang Y, Liu T, Yang Q, Hu X, Li W, Wei A. Unusual Autosomal Dominant Inheritance of Oculocutaneous Albinism Type 4 (OCA-4): Clinical and Functional Features From A Chinese Family. Pigment Cell Melanoma Res 2025; 38:e70013. [PMID: 40195871 DOI: 10.1111/pcmr.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/08/2025] [Accepted: 03/22/2025] [Indexed: 04/09/2025]
Abstract
Oculocutaneous albinism (OCA) is a complex genetic disorder characterized by reduced or absent pigmentation in the skin, hair, and eyes. Among the eight known subtypes, OCA-4 is caused by a mutation in SLC45A2, which plays a crucial role in melanin biosynthesis. While autosomal recessive inheritance is the most common pattern for all OCA subtypes, autosomal dominant cases are extremely rare. We report three patients from a Chinese family exhibiting autosomal dominant OCA-4. Clinical assessments evaluated pigmentation and ocular features in affected family members. Next-generation sequencing was performed to identify pathogenic variants, and functional studies in MNT-1 cells were performed to explore the variant's biological effects. Patients exhibited mild hypopigmentation and foveal hypoplasia, consistent with the OCA-4 phenotype. Genetic analysis identified a heterozygous c.208T>C (p.Tyr70His) variant in SLC45A2, the same variant that has been previously reported in association with autosomal dominant OCA-4. Functional studies demonstrated that this variant caused protein retention in the endoplasmic reticulum, resulting in reduced melanin production. This family represents the first documented cases of autosomal dominant OCA-4 in the Chinese population and only the second reported worldwide. Our findings confirm that the p.Tyr70His variant causes autosomal dominant OCA-4. This study deepens our understanding of OCA-4's genetic mechanisms and increases the complexity of its inheritance patterns in genetic counseling.
Collapse
Affiliation(s)
- Yingzi Zhang
- Department of Dermatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Teng Liu
- Department of Dermatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Qingsong Yang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Genetics and Birth Defects Control Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Xuyun Hu
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Genetics and Birth Defects Control Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Wei Li
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Genetics and Birth Defects Control Center, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Aihua Wei
- Department of Dermatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Bradshaw MS, Raychaudhuri J, Murphy L, Barnard R, Firman T, Gaskell AA, Layer RM. Rapid, Reliable, and Interpretable Copy Number Variant Curation Visualizations for Diagnostic Settings with SeeNV. J Mol Diagn 2025; 27:336-345. [PMID: 40044036 DOI: 10.1016/j.jmoldx.2025.01.008] [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: 05/09/2024] [Revised: 12/05/2024] [Accepted: 01/15/2025] [Indexed: 03/10/2025] Open
Abstract
Copy number variants (CNVs), structural alterations in the genome involving duplication or deletion of DNA segments, are implicated in various health conditions. Despite their clinical significance, accurate identification and interpretation of CNVs remain challenging, especially in the context of whole-exome sequencing (WES), which is commonly used in clinical diagnostic laboratories. Although WES offers economic advantages over whole-genome sequencing, it struggles with CNV detection because of technical noise introduced by laboratory and analytic processes. Manual curation of CNV calls generated by these tools is labor intensive and error prone. To address this, SeeNV, a command-line tool, is introduced to aid manual curation of CNVs at scale. SeeNV is one solution to these issues, developed in collaboration with and used by the Precision Diagnostics Laboratory at Children's Hospital Colorado. SeeNV generates static infographics for each CNV, incorporating sample and cohort sequencing coverage statistics, CNV population frequency, and, more, facilitating rapid and precise assessment. Using CNV calls identified in publicly available WES and whole-genome sequencing samples, users can rapidly and reliably curate CNV calls, needing only 4.3 seconds to curate a call, achieving 0.95 recall (analytical sensitivity) and 0.74 precision (positive predictive value). SeeNV is freely available for download on GitHub.
Collapse
Affiliation(s)
- Michael S Bradshaw
- Department of Computer Science, University of Colorado Boulder, Boulder, Colorado
| | - Jishnu Raychaudhuri
- Department of Computer Science, University of Colorado Boulder, Boulder, Colorado
| | - Lachlan Murphy
- Department of Computer Science, University of Colorado Boulder, Boulder, Colorado
| | - Rebecca Barnard
- Precision Medicine Institute, Children's Hospital Colorado, Aurora, Colorado
| | - Taylor Firman
- Precision Medicine Institute, Children's Hospital Colorado, Aurora, Colorado
| | - Alisa A Gaskell
- Precision Medicine Institute, Children's Hospital Colorado, Aurora, Colorado.
| | - Ryan M Layer
- Department of Computer Science, University of Colorado Boulder, Boulder, Colorado.
| |
Collapse
|
23
|
Xie B, Li Q, Ma Q, Huang Y, Wu Z, Hu Z, Feng J, Tong K. Clinicopathological and molecular features of so-called low-grade oncocytic fumarate hydratase-deficient renal cell carcinoma: a study of 5 cases. Virchows Arch 2025:10.1007/s00428-025-04111-8. [PMID: 40304778 DOI: 10.1007/s00428-025-04111-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/12/2025] [Accepted: 04/16/2025] [Indexed: 05/02/2025]
Abstract
Low-grade oncocytic fumarate hydratase-deficient renal cell carcinoma (FHdRCC), resembling succinate dehydrogenase-deficient RCC morphologically, is a recently described uncommon variant of FHdRCC. We report five additional cases to enhance the understanding of this rare tumor type. These tumors had variably thick fibromuscular capsules with frequent intracapsular invasions and had prominently compact nests and/or tubules. The tumor cells were characterized by abundant dense eosinophilic cytoplasm imparting a ground glass-like appearance and by variable bubbly cytoplasm. Despite a predominantly low histology grade, focal nuclear pleomorphism was observed in two cases. Two cases showed FH negativity and 2SC positivity, while the remaining cases exhibited both FH and 2SC positivity. All cases were positive for PAX8, Vimentin, P504s, and SDHB, but negative for CD117, CA9, CK20, ALK, and cathepsin-K. CK7 and TFE3 positivity were seen in 2 and 4 of 5 cases, respectively. The whole exome sequencing and multiplex PCR-based next generation sequencing identified pathogenic or likely pathogenic FH mutations in three cases and failed in one case showing the FH-negative and 2SC-positive immunophenotype. The remaining case exhibiting both FH and 2SC positivity had a germline FH mutation of uncertain significance and single copy loss of chromosomes 1, 14, and 18. None of three cases who underwent FISH detections had TFE3 translocations. Follow-up time for the 5 cases ranged from 7 to 94 months (median 46 months; mean 52 months). All cases were alive without disease, except for case 4 being alive with a stable contralateral renal mass. While low-grade oncocytic FHdRCC has a relatively better prognosis than conventional FHdRCCs, further research is required to confirm this in the future.
Collapse
Affiliation(s)
- Bin Xie
- Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410078, Hunan, China
| | - Qi Li
- Department of Pathology, Bishan Hospital of Chongqing, Bishan Hospital of Chongqing Medical University, Chongqing, 402760, China
| | - Qiang Ma
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ying Huang
- Clinical Molecular Medical Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ze Wu
- Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410078, Hunan, China
| | - Zhongliang Hu
- Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410078, Hunan, China
| | - Junming Feng
- Department of Pathology, Bishan Hospital of Chongqing, Bishan Hospital of Chongqing Medical University, Chongqing, 402760, China
| | - Kuo Tong
- Department of Pathology, Bishan Hospital of Chongqing, Bishan Hospital of Chongqing Medical University, Chongqing, 402760, China.
| |
Collapse
|
24
|
Martín-Arana J, Gimeno-Valiente F, Henriksen TV, García-Micó B, Martínez-Castedo B, Gambardella V, Martínez-Ciarpaglini C, Palomar B, Huerta M, Camblor DG, García Bartolomé M, Carbonell-Asins JA, Frydendahl A, Gotchalck KA, Fleitas T, Tébar-Martínez R, Moro D, Pla V, Pérez-Santiago L, Martín-Arévalo J, Casado D, García-Botello S, Espí A, Roselló S, Roda D, Andersen CL, Cervantes A, Tarazona N. Whole-exome tumor-agnostic ctDNA analysis enhances minimal residual disease detection and reveals relapse mechanisms in localized colon cancer. NATURE CANCER 2025:10.1038/s43018-025-00960-z. [PMID: 40301653 DOI: 10.1038/s43018-025-00960-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 03/25/2025] [Indexed: 05/01/2025]
Abstract
In stage 2-3 colon cancer (CC), postsurgery circulating tumor DNA (ctDNA) assessment is crucial for guiding adjuvant chemotherapy (ACT) decisions. While existing assays detect ctDNA and help identify high-risk persons with CC for recurrence, their limited sensitivity after surgery poses challenges in deciding on ACT. Additionally, a substantial portion of persons with CC fail to clear ctDNA after ACT, leading to recurrence. In this study, we performed whole-exome sequencing (WES) of ctDNA at different time points in participants with relapsed CC in two independent cohorts, alongside transcriptomic and proteomic analyses of metastases, to enhance comprehension of progression mechanisms. A plasma WES-based tumor-agnostic assay demonstrated higher sensitivity in detecting minimal residual disease (MRD) compared to current assays. Immune evasion appears to be the primary driver of progression in the localized CC setting, indicating the potential efficacy of immunotherapy for microsatellite stability in persons with CC. Organoid modeling further supports the promising potential of targeted therapy in eradicating MRD, surpassing conventional treatments.
Collapse
Affiliation(s)
- Jorge Martín-Arana
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - Francisco Gimeno-Valiente
- Cancer Evolution and Genome Instability Laboratory, University College London Cancer Institute, London, UK
| | - Tenna Vesterman Henriksen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Blanca García-Micó
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - Belén Martínez-Castedo
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - Valentina Gambardella
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - Carolina Martínez-Ciarpaglini
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
- Department of Pathology, INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Brenda Palomar
- Department of Pathology, INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Marisol Huerta
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - Daniel G Camblor
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - Miguel García Bartolomé
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | | | - Amanda Frydendahl
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Tania Fleitas
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - Roberto Tébar-Martínez
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | - David Moro
- Colorectal Surgery Unit, Department of General and Digestive Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario, Valencia, Spain
- Department of Surgery, University of Valencia, Valencia, Spain
| | - Vicente Pla
- Colorectal Surgery Unit, Department of General and Digestive Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario, Valencia, Spain
- Department of Surgery, University of Valencia, Valencia, Spain
| | - Leticia Pérez-Santiago
- Colorectal Surgery Unit, Department of General and Digestive Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario, Valencia, Spain
- Department of Surgery, University of Valencia, Valencia, Spain
| | - José Martín-Arévalo
- Colorectal Surgery Unit, Department of General and Digestive Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario, Valencia, Spain
- Department of Surgery, University of Valencia, Valencia, Spain
| | - David Casado
- Colorectal Surgery Unit, Department of General and Digestive Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario, Valencia, Spain
- Department of Surgery, University of Valencia, Valencia, Spain
| | - Stephanie García-Botello
- Colorectal Surgery Unit, Department of General and Digestive Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario, Valencia, Spain
- Department of Surgery, University of Valencia, Valencia, Spain
| | - Alejandro Espí
- Colorectal Surgery Unit, Department of General and Digestive Surgery, INCLIVA Biomedical Research Institute, Hospital Clínico Universitario, Valencia, Spain
- Department of Surgery, University of Valencia, Valencia, Spain
| | - Susana Roselló
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - Desamparados Roda
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain
| | - Claus Lindbjerg Andersen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Andrés Cervantes
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain.
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain.
| | - Noelia Tarazona
- Department of Medical Oncology, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain.
- Instituto de Salud Carlos III, CIBERONC, Madrid, Spain.
| |
Collapse
|
25
|
Phan-Canh T, Nguyen-Le DM, Luu PL, Khunweeraphong N, Kuchler K. Rapid in vitro evolution of flucytosine resistance in Candida auris. mSphere 2025; 10:e0097724. [PMID: 40099908 PMCID: PMC12039228 DOI: 10.1128/msphere.00977-24] [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: 12/27/2024] [Accepted: 02/14/2025] [Indexed: 03/20/2025] Open
Abstract
The pan-antifungal-resistant pathogen Candida auris has been causing high-mortality infection outbreaks in hospitals and healthcare settings. The prodrug 5-fluorocytosine (5FC) is one of four chemical entities, but its clinical use as an antifungal drug has been limited owing to pronounced resistance. However, antifungal combination therapy with 5FC appears as a promising strategy for treating C. auris infections. Here, we show that a C. auris clinical isolate can rapidly acquire genetic mutations to mount 5FC resistance after only one to two passages under drug selection. We exploit a new bioinformatics workflow to identify genetic polymorphisms from RNA-seq data. Strikingly, we identify several mutations in the FUR1 gene encoding the 5-fluorouracil convertase that normally generates the active drug. A single nonsense mutation truncates the enzyme at residue Q30*, leading to 5FC resistance due to inactive Fur1. Whole-genome sequencing analysis revealed that an indel mutation in FCY2 also contributes to 5FC resistance. Furthermore, at least one out of seven adapted strains acquired enhanced 5FC tolerance without mutations in the 5FC conversion pathway. Thus, we demonstrate that FUR1 mutations are critical drivers of 5FC resistance in C. auris.IMPORTANCECandida auris is a high-priority human fungal pathogen, causing infection outbreaks of high mortality in healthcare settings. Antifungal combination therapy with 5-fluorocytosine (5FC) is one of the emerging approaches in treatment. However, acquired 5FC resistance traits have been a matter of concern. 5FC is taken up by fungal cells via a cytosine permease and further metabolized by a cytosine deaminase to 5-fluorouracil (5FU). 5FU is then converted by the Fur1 uracil phosphoribosyltransferase into a toxic antimetabolite that disrupts fungal RNA and DNA syntheses. Mutations in these proteins are commonly associated with 5FC resistance in fungal species. Here, we show that C. auris can rapidly develop resistance under 5FC selective stress owing to mutational inactivation of Fur1 function. Moreover, other mechanisms that bypass mutations in the 5FC conversion pathway may also contribute to 5FC resistance traits. Finally, we have developed a tailored bioinformatics workflow that facilitates the identification of polymorphisms associated with 5FC resistance in clinical isolates.
Collapse
Affiliation(s)
- Trinh Phan-Canh
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Vienna, Austria
| | - Duc-Minh Nguyen-Le
- Institute for Applied Research in Health Sciences and Aging (ARiHA)-Thong Nhat Hospital, Ho Chi Minh City, Vietnam
| | - Phuc-Loi Luu
- Institute for Applied Research in Health Sciences and Aging (ARiHA)-Thong Nhat Hospital, Ho Chi Minh City, Vietnam
- Mathematics Department, Faculty of Fundamental Sciences, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Narakorn Khunweeraphong
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Vienna, Austria
| | - Karl Kuchler
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Medical University of Vienna, Center for Medical Biochemistry, Vienna, Austria
| |
Collapse
|
26
|
Li Y, Xu X, She Y, Su Z, Liu X, Chen Y, Ye C, Zhang Y, Yu H, Chen C, Chen S, Zhou L. Genetic Etiology of Epilepsy: A Retrospective Study From a Single-Center Cohort. Clin Genet 2025. [PMID: 40296287 DOI: 10.1111/cge.14757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025]
Abstract
Next generation sequencing (NGS) technology has made significant progress in the genetic diagnosis and treatment of epilepsy. However, genetic studies on epilepsy with different etiologies remain relatively limited. In this study, whole-genome or whole-exome sequencing was performed on 158 unrelated patients with epilepsy of various etiologies, and the identified variants were analyzed for their association with 1356 seizure-related genes in the database. Additionally, the pathogenicity or likely pathogenicity of those variants associated with known epilepsy genes was evaluated. The results showed that pathogenic or likely pathogenic variants were detected in 31.65% (50/158) of the patients in our cohort study. Further analysis revealed significant differences in the diagnostic rates among different epilepsy categories: 29.60% (37/125) for idiopathic epilepsy and 39.39% (13/33) for symptomatic epilepsy. Moreover, the genes PRRT2, KMT2C, PRKRA, NOTCH3, NAGLU, and SCN1A were identified as potentially important for epilepsy, suggesting they could become key targets for clinical diagnosis and treatment. In conclusion, NGS technology demonstrates high diagnostic efficiency for epilepsy of different etiologies and highlights significant differences among various types. This provides novel genetic insights for the diagnosis and treatment of epilepsy.
Collapse
Affiliation(s)
- Yinchao Li
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Xiaowei Xu
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yingfang She
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhengwei Su
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Xianyue Liu
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Ying Chen
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Chenghui Ye
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yuanchao Zhang
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Hang Yu
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Chun Chen
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Shuda Chen
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Liemin Zhou
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
27
|
Wu S, Chen X, Chen Y, Li C, Yang R, Zhang T, Ma J. Genetic characteristics associated with isolated Microtia revealed through whole exome sequencing of 201 pedigrees. Hum Mol Genet 2025:ddaf063. [PMID: 40275486 DOI: 10.1093/hmg/ddaf063] [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: 03/24/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Microtia is one of the most common congenital craniofacial malformations, characterized by the maldevelopment of the external and middle ear. While numerous genes have been implicated in syndromic forms of microtia, the genetic underpinnings of isolated microtia remain poorly understood. In this study, we conducted whole exome sequencing (WES) on 201 pedigrees with isolated microtia to investigate its genetic basis. Bioinformatics analysis identified 1362 deleterious variants corresponding to 332 candidate genes, including 40 previously associated with microtia-related phenotypes. Among these, variants in FOXI3, the most frequently identified pathogenic gene for isolated microtia so far, were detected. Remarkably, the remaining 39 genes, which have been recognized as pathogenic in syndromes with microtia, are also suggested to play a role in isolated microtia. However, the precise molecular mechanisms by which these genes contribute to microtia remain to be elucidated. Furthermore, through protein-protein interaction network analysis, functional annotation, and zebrafish expression profiling, we identified two novel genes, MCM2 and BDNF, as the most promising contributors to the pathogenesis of isolated microtia. Our findings, based on the largest WES study of isolated microtia pedigrees to date, provide new insights into the genetic architecture of isolated microtia and suggest promising avenues for future research.
Collapse
Affiliation(s)
- Siyi Wu
- ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, No. 83 Fenyang Road, Xuhui District, Shanghai 200031, China
| | - Xin Chen
- ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, No. 83 Fenyang Road, Xuhui District, Shanghai 200031, China
| | - Ying Chen
- ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, No. 83 Fenyang Road, Xuhui District, Shanghai 200031, China
| | - Chenlong Li
- ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, No. 83 Fenyang Road, Xuhui District, Shanghai 200031, China
| | - Run Yang
- ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, No. 83 Fenyang Road, Xuhui District, Shanghai 200031, China
| | - Tianyu Zhang
- ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, No. 83 Fenyang Road, Xuhui District, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine (Fudan University), No. 83 Fenyang Road, Xuhui District, Shanghai 200031, China
| | - Jing Ma
- ENT Institute, Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, No. 83 Fenyang Road, Xuhui District, Shanghai 200031, China
- Institute of Medical Genetics & Genomics, Fudan University, No. 131 Dong'an Road, Xuhui District, Shanghai 200032, China
| |
Collapse
|
28
|
Kang X, Li X, Zhou J, Zhang Y, Qiu L, Tian C, Deng Z, Liang X, Zhang Z, Du S, Hu S, Wang N, Yue Z, Xu Y, Gao Y, Dai J, Wang Z, Yu C, Chen J, Wu Y, Chen L, Yao Y, Yao S, Yang X, Yan L, Wen Q, Depies OM, Chan K, Liang X, Li G, Zi Z, Liu X, Gan H. Extrachromosomal DNA replication and maintenance couple with DNA damage pathway in tumors. Cell 2025:S0092-8674(25)00414-3. [PMID: 40300601 DOI: 10.1016/j.cell.2025.04.012] [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: 10/18/2024] [Revised: 02/19/2025] [Accepted: 04/06/2025] [Indexed: 05/01/2025]
Abstract
Extrachromosomal DNA (ecDNA) drives the evolution of cancer cells. However, the functional significance of ecDNA and the molecular components involved in its replication and maintenance remain largely unknown. Here, using CRISPR-C technology, we generated ecDNA-carrying (ecDNA+) cell models. By leveraging these models alongside other well-established systems, we demonstrated that ecDNA can replicate and be maintained in ecDNA+ cells. The replication of ecDNA activates the ataxia telangiectasia mutated (ATM)-mediated DNA damage response (DDR) pathway. Topoisomerases, such as TOP1 and TOP2B, play a role in ecDNA replication-induced DNA double-strand breaks (DSBs). A subset of these elevated DSBs persists into the mitotic phase and is primarily repaired by the alternative non-homologous end joining (alt-NHEJ) pathway, which involves POLθ and LIG3. Correspondingly, ecDNA maintenance requires DDR, and inhibiting DDR impairs the circularization of ecDNA. In summary, we demonstrate reciprocal interactions between ecDNA maintenance and DDR, providing new insights into the detection and treatment of ecDNA+ tumors.
Collapse
Affiliation(s)
- Xing Kang
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xinran Li
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaqi Zhou
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yang Zhang
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lingyu Qiu
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Congcong Tian
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhiwen Deng
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, China
| | - Xiaoyan Liang
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ziwei Zhang
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Songlin Du
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang 712100, Shaanxi, China
| | - Suili Hu
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Nan Wang
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen Yue
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yajing Xu
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuan Gao
- Department of Pharmacology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Junbiao Dai
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics and Shenzhen Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Zhiquan Wang
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Chuanhe Yu
- Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Jinyi Chen
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuchun Wu
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; School of Basic Medicine, Qingdao University, Qingdao, China
| | - Liangming Chen
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuan Yao
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Sitong Yao
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xinran Yang
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lixia Yan
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Wen
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Olivia M Depies
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kuiming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Xiaohuan Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Gang Li
- Centre of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Zhike Zi
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiangyu Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, China
| | - Haiyun Gan
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
29
|
Bhandari M, He F, Rogojina A, Li F, Zou Y, Jiang J, Lai Z, Houghton P, Kurmasheva RT, Chen Y, Wang X, Zheng S. Benchmarking mouse contamination removing protocols in patient-derived xenografts genomic profiling. NPJ Precis Oncol 2025; 9:113. [PMID: 40247091 PMCID: PMC12006369 DOI: 10.1038/s41698-025-00902-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/05/2025] [Indexed: 04/19/2025] Open
Abstract
Patient-derived xenograft (PDX) models are widely used in cancer research. Genomic and transcriptomic profiling of PDXs are inevitably contaminated by sequencing reads originated from mouse cells. Here, we examine the impact of mouse read contamination on RNA sequencing (RNAseq), Whole Exome Sequencing (WES), and Whole Genome Sequencing (WGS) data of 21 PDXs. We also systematically benchmark the performance of 12 computational protocols for removing mouse reads from PDXs. We find that mouse read contamination increases expression of immune and stromal related genes, and inflates the number of somatic mutations. However, detection of gene fusions and copy number alterations is minimally affected by mouse read contamination. Using gold standard datasets, we find that pseudo-alignment protocols often demonstrate better prediction performance and computing efficiency. The best performing tool is a relatively new tool Xengsort. Our results emphasize the importance of removing mouse reads from PDXs and the need to adopt new tools in PDX genomic studies.
Collapse
Affiliation(s)
- Mukund Bhandari
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
| | - Funan He
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Population Health Sciences, San Antonio, TX, USA
| | - Anna Rogojina
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
| | - Fuyang Li
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Molecular Medicine, San Antonio, TX, USA
| | - Yi Zou
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
| | - Jing Jiang
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Population Health Sciences, San Antonio, TX, USA
| | - Zhao Lai
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Molecular Medicine, San Antonio, TX, USA
- Mays Cancer Center, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Peter Houghton
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Molecular Medicine, San Antonio, TX, USA
- Mays Cancer Center, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Raushan T Kurmasheva
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Molecular Medicine, San Antonio, TX, USA
- Mays Cancer Center, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Yidong Chen
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA
- Department of Population Health Sciences, San Antonio, TX, USA
- Mays Cancer Center, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Xiaojing Wang
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA.
- Department of Population Health Sciences, San Antonio, TX, USA.
- Mays Cancer Center, University of Texas Health at San Antonio, San Antonio, TX, USA.
| | - Siyuan Zheng
- Greehey Children's Cancer Research Institute, San Antonio, TX, USA.
- Department of Population Health Sciences, San Antonio, TX, USA.
- Mays Cancer Center, University of Texas Health at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
30
|
Ortiz-Martínez D. DeBasher: a flow-based programming bash extension for the implementation of complex and interactive workflows with stateful processes. BMC Bioinformatics 2025; 26:106. [PMID: 40240939 PMCID: PMC12004750 DOI: 10.1186/s12859-025-06108-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/07/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Bioinformatics data analysis faces significant challenges. As data analysis often takes the form of pipelines or workflows, workflow managers (WfMs) have become essential. Data flow programming constitutes the preferred approach in WfMs, enabling parallel processes activated reactively based on input availability. While this paradigm typically follows a linear, acyclic progression, cyclic workflows are sometimes necessary in bioinformatics analyses. These cyclic workflows also present an opportunity to explore workflow interactivity, a feature not widely implemented in existing WfMs. RESULTS We propose DeBasher, a tool that adopts the flow-based programming (FBP) paradigm, in which the workflow components are in control of their life cycle and can store state information, allowing the execution of complex workflows that include cycles. DeBasher also incorporates a powerful model of interactivity, where the user can alter the behavior of a running workflow. Additionally, DeBasher allows the user to define triggers so as to initiate the execution of a complete workflow or a part of it. The ability to execute processes with state and in control of their life cycle also has applications in dynamic scheduling tasks. Furthermore, DeBasher presents a series of extra features, including the combination of multiple workflows at runtime through a feature we have called runtime piping, switching to static scheduling to increase scalability, or implementing processes in multiple languages. DeBasher has been successfully used to process 131.7 TB of genomic data by means of a variant calling pipeline. CONCLUSIONS DeBasher is an FBP Bash extension that can be useful in a wide range of situations and in particular when implementing complex workflows, workflows with interactivity or triggers, or when a high scalability is required.
Collapse
Affiliation(s)
- Daniel Ortiz-Martínez
- Department of Mathematics and Computer Science, Universitat de Barcelona, Gran Via de les Corts Catalanes, 585, 08007, Barcelona, Spain.
| |
Collapse
|
31
|
Chen Y, Yang R, Chen X, Zhang T, Li C, Ma J. Identification of novel TCOF1 mutations in Treacher Collins syndrome and their functional characterization. Orphanet J Rare Dis 2025; 20:184. [PMID: 40240907 PMCID: PMC12001626 DOI: 10.1186/s13023-025-03667-7] [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: 08/27/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Treacher Collins syndrome (TCS) is a congenital disorder primarily caused by the mutation in the Treacle Ribosome Biogenesis Factor 1 (TCOF1) gene. However, the significance of many TCOF1 mutations remains uncertain. RESULTS We report two novel mutations identified in two TCS families and assess their pathogenicity alongside two previously reported mutations. Both novel mutations, c.2115dupG (p.T706DfsTer52) and c.2142+23_2142+52 del (p.A715VfsTer31), result in truncated proteins lacking nuclear location signals (NLSs), which impedes their entry into the nucleus and reduces mRNA expression level. Notably, the mutation c.2142+23_2142+52 del, leading to the retention of a 62 bp intron and disrupting RNA splicing, represents the first documented case of intron retention in TCS patients. Additionally, the previously reported mutation c.136 C> G (p.L46V) hinders protein nuclear location, while mutation c.1719del (p.N574TfsTer22) significantly decreases mRNA levels. CONCLUSIONS Our research expands the spectrum of TCOF1 mutations and provides evidence clarifying their pathogenic nature. These findings are crucial for genetic counseling and prenatal diagnosis for TCS patients.
Collapse
Affiliation(s)
- Ying Chen
- Department of Facial Plastic and Reconstructive Surgery, ENT Institute, Eye and ENT Hospital of Fudan University, Shanghai, 200031, People's Republic of China
| | - Run Yang
- Department of Facial Plastic and Reconstructive Surgery, ENT Institute, Eye and ENT Hospital of Fudan University, Shanghai, 200031, People's Republic of China
| | - Xin Chen
- Department of Facial Plastic and Reconstructive Surgery, ENT Institute, Eye and ENT Hospital of Fudan University, Shanghai, 200031, People's Republic of China
| | - Tianyu Zhang
- Department of Facial Plastic and Reconstructive Surgery, ENT Institute, Eye and ENT Hospital of Fudan University, Shanghai, 200031, People's Republic of China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai, 200031, People's Republic of China
| | - Chenlong Li
- Department of Facial Plastic and Reconstructive Surgery, ENT Institute, Eye and ENT Hospital of Fudan University, Shanghai, 200031, People's Republic of China.
| | - Jing Ma
- Department of Facial Plastic and Reconstructive Surgery, ENT Institute, Eye and ENT Hospital of Fudan University, Shanghai, 200031, People's Republic of China.
- Institute of Medical Genetics and Genomics, Fudan University, Shanghai, 200032, People's Republic of China.
- Surgery Laboratory, Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, People's Republic of China.
| |
Collapse
|
32
|
Chia S, Wen Seow JJ, Peres da Silva R, Suphavilai C, Shirgaonkar N, Murata-Hori M, Zhang X, Yong EY, Pan J, Thangavelu MT, Periyasamy G, Yap A, Anand P, Muliaditan D, Chan YS, Siyu W, Yong CW, Hong N, Ran G, Sim NL, Guo YA, Yi Teh AX, Wei Ling CC, Wei Tan EK, Pei Cherylin FW, Chang M, Han S, Seow-En I, Chen Hui LR, Hsia Gan AH, Yap CK, Ng HH, Skanderup AJ, Chinswangwatanakul V, Riansuwan W, Trakarnsanga A, Pithukpakorn M, Tanjak P, Chaiboonchoe A, Park D, Kim DK, Iyer NG, Tsantoulis P, Tejpar S, Kim JE, Kim TI, Sampattavanich S, Tan IB, Nagarajan N, DasGupta R. CAN-Scan: A multi-omic phenotype-driven precision oncology platform identifies prognostic biomarkers of therapy response for colorectal cancer. Cell Rep Med 2025; 6:102053. [PMID: 40187357 PMCID: PMC12047494 DOI: 10.1016/j.xcrm.2025.102053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 12/10/2024] [Accepted: 03/10/2025] [Indexed: 04/07/2025]
Abstract
Application of machine learning (ML) on cancer-specific pharmacogenomic datasets shows immense promise for identifying predictive response biomarkers to enable personalized treatment. We introduce CAN-Scan, a precision oncology platform, which applies ML on next-generation pharmacogenomic datasets generated from a freeze-viable biobank of patient-derived primary cell lines (PDCs). These PDCs are screened against 84 Food and Drug Administration (FDA)-approved drugs at clinically relevant doses (Cmax), focusing on colorectal cancer (CRC) as a model system. CAN-Scan uncovers prognostic biomarkers and alternative treatment strategies, particularly for patients unresponsive to first-line chemotherapy. Specifically, it identifies gene expression signatures linked to resistance against 5-fluorouracil (5-FU)-based drugs and a focal copy-number gain on chromosome 7q, harboring critical resistance-associated genes. CAN-Scan-derived response signatures accurately predict clinical outcomes across four independent, ethnically diverse CRC cohorts. Notably, drug-specific ML models reveal regorafenib and vemurafenib as alternative treatments for BRAF-expressing, 5-FU-insensitive CRC. Altogether, this approach demonstrates significant potential in improving biomarker discovery and guiding personalized treatments.
Collapse
Affiliation(s)
- Shumei Chia
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.
| | - Justine Jia Wen Seow
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Rafael Peres da Silva
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Chayaporn Suphavilai
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Niranjan Shirgaonkar
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Maki Murata-Hori
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Xiaoqian Zhang
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Elena Yaqing Yong
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Jiajia Pan
- National Cancer Centre, Singapore, Singapore
| | - Matan Thangavelu Thangavelu
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Experimental Drug Development Centre (EDDC), A∗STAR, Singapore, Singapore
| | - Giridharan Periyasamy
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Experimental Drug Development Centre (EDDC), A∗STAR, Singapore, Singapore
| | - Aixin Yap
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Padmaja Anand
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Daniel Muliaditan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Yun Shen Chan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Wang Siyu
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Chua Wei Yong
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Nguyen Hong
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Gao Ran
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Ngak Leng Sim
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Yu Amanda Guo
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | | | | | - Emile Kwong Wei Tan
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Singapore
| | - Fu Wan Pei Cherylin
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Singapore
| | - Meihuan Chang
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Singapore
| | - Shuting Han
- National Cancer Centre, Singapore, Singapore
| | - Isaac Seow-En
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Singapore
| | | | - Anna Hwee Hsia Gan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Choon Kong Yap
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Huck Hui Ng
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anders Jacobsen Skanderup
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Vitoon Chinswangwatanakul
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Woramin Riansuwan
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Atthaphorn Trakarnsanga
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Manop Pithukpakorn
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol, Bangkok, Thailand
| | - Pariyada Tanjak
- Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Cancer Center, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Amphun Chaiboonchoe
- Siriraj Center of Research Excellence for Precision Medicine and Systems Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Daye Park
- Division of Gastroenterology, Department of Internal Medicine, Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dong Keon Kim
- Division of Gastroenterology, Department of Internal Medicine, Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | | | - Petros Tsantoulis
- Hôpitaux Universitaires de Genève, University of Geneva, Geneva, Switzerland
| | - Sabine Tejpar
- Department of Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jung Eun Kim
- R&D center PODO Therapeutics Co. 338 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13493, Republic of Korea
| | - Tae Il Kim
- R&D center PODO Therapeutics Co. 338 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13493, Republic of Korea; Division of Gastroenterology, Department of Internal Medicine, Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Somponnat Sampattavanich
- Siriraj Center of Research Excellence for Precision Medicine and Systems Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Iain Beehuat Tan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; National Cancer Centre, Singapore, Singapore; Duke-National University of Singapore Medical School, Singapore, Singapore.
| | - Niranjan Nagarajan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Ramanuj DasGupta
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore; CRUK Scotland Institute, School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK.
| |
Collapse
|
33
|
Fiore D, Cappelli LV, Zhaoqi L, Kotlov N, Sorokina M, Phillip J, Zumbo P, Yoffe L, Ghione P, Wang A, Han X, Taylor A, Chiu W, Fragliasso V, Tabbo F, Zamponi N, Di Siervi N, Kayembe C, Medico G, Patel RP, Gaudiano M, Machiorlatti R, Astone G, Cacciapuoti MT, Zanetti G, Pignataro C, Eric RA, Patel S, Zammarchi F, Zanettini C, Queiroz L, Nikitina A, Kudryashova O, Karelin A, Nikitin D, Tychinin D, Postovalova E, Bagaev A, Svekolkin V, Belova E, Tikhonova K, Degryse S, Xu C, Novero D, Ponzoni M, Tiacci E, Falini B, Song J, Khodos I, De Stanchina E, Macari G, Cafforio L, Gardini S, Piva R, Medico E, Ng SY, Moskowitz A, Epstein Z, Intlekofer A, Ahmed D, Chan WC, Martin P, Ruan J, Bertoni F, Foà R, Brody JD, Weinstock DM, Osan J, Santambrogio L, Elemento O, Betel D, Tam W, Ruella M, Cerchietti L, Rabadan R, Horwitz S, Inghirami G. A patient-derived T cell lymphoma biorepository uncovers pathogenetic mechanisms and host-related therapeutic vulnerabilities. Cell Rep Med 2025; 6:102029. [PMID: 40147445 PMCID: PMC12047492 DOI: 10.1016/j.xcrm.2025.102029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 04/24/2024] [Accepted: 02/21/2025] [Indexed: 03/29/2025]
Abstract
Peripheral T cell lymphomas (PTCLs) comprise heterogeneous malignancies with limited therapeutic options. To uncover targetable vulnerabilities, we generate a collection of PTCL patient-derived tumor xenografts (PDXs) retaining histomorphology and molecular donor-tumor features over serial xenografting. PDX demonstrates remarkable heterogeneity, complex intratumor architecture, and stepwise trajectories mimicking primary evolutions. Combining functional transcriptional stratification and multiparametric imaging, we identify four distinct PTCL microenvironment subtypes with prognostic value. Mechanistically, we discover a subset of PTCLs expressing Epstein-Barr virus-specific T cell receptors and uncover the capacity of cancer-associated fibroblasts of counteracting treatments. PDXs' pre-clinical testing captures individual vulnerabilities, mirrors donor patients' clinical responses, and defines effective patient-tailored treatments. Ultimately, we assess the efficacy of CD5KO- and CD30- Chimeric Antigen Receptor T Cells (CD5KO-CART and CD30_CART, respectively), demonstrating their therapeutic potential and the synergistic role of immune checkpoint inhibitors for PTCL treatment. This repository represents a resource for discovering and validating intrinsic and extrinsic factors and improving the selection of drugs/combinations and immune-based therapies.
Collapse
Affiliation(s)
- Danilo Fiore
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; Institute for Experimental Endocrinology and Oncology, "G.Salvatore" IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Luca Vincenzo Cappelli
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Liu Zhaoqi
- Program for Mathematical Genomics, Department of Systems Biology, Department of Biomedical Informatics, Columbia University, New York, NY 10027 USA; China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | | | | | - Jude Phillip
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US; Chemical and Biomolecular Engineering, Oncology, Sidney Kimmel Comprehensive Cancer Center, Core Member, Institute for Nanobiotechnology (INBT), Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Paul Zumbo
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, US
| | - Liron Yoffe
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Paola Ghione
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anqi Wang
- Program for Mathematical Genomics, Department of Systems Biology, Department of Biomedical Informatics, Columbia University, New York, NY 10027 USA
| | - Xueshuai Han
- Program for Mathematical Genomics, Department of Systems Biology, Department of Biomedical Informatics, Columbia University, New York, NY 10027 USA; China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Abigail Taylor
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - William Chiu
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Valentina Fragliasso
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Laboratory of translational research, Azienda USL - IRCCS di Reggio Emilia, 42122 Reggio Emila, Italy
| | - Fabrizio Tabbo
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; SC Oncologia ASL CN2 Alba Bra Ospedale Michele e Pietro Ferrero, 12060 Verduno, (CN), Italy
| | - Nahuel Zamponi
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US
| | - Nicolás Di Siervi
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US
| | - Clarisse Kayembe
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Giovanni Medico
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ruchi P Patel
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Perelman Center for Advanced Medicine, SPE 8-112, Philadelphia, PA 19104, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marcello Gaudiano
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Rodolfo Machiorlatti
- Department of Pathology, Center for Experimental Research and Medical Studies, University of Torino, 10126 Torino, Italy
| | - Giuseppina Astone
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Maria Teresa Cacciapuoti
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Giorgia Zanetti
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Claudia Pignataro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Ruiz Arvin Eric
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sanjay Patel
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Claudio Zanettini
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lucio Queiroz
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Chengqi Xu
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Domenico Novero
- Division of Pathological Anatomy, Quality and Safety of Diagnosis and Treatment, Città della Salute e della Scienza, 10126 Turin, Italy
| | - Maurilio Ponzoni
- Pathology Unit, San Raffaele Scientific Institute, Milan, Italy; Unit of Lymphoid Malignancies, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Enrico Tiacci
- Institute of Hematology, University of Perugia, Ospedale S. Maria della Misericordia, S. Andrea delle Fratte, 06156 Perugia Italy
| | - Brunangelo Falini
- Institute of Hematology, University of Perugia, Ospedale S. Maria della Misericordia, S. Andrea delle Fratte, 06156 Perugia Italy
| | - Joo Song
- Department of Pathology, City of Hope Medical Center, Duarte, CA 91010, US
| | - Inna Khodos
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, US
| | - Elisa De Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, US
| | | | | | | | - Roberto Piva
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; Medical Genetics Unit, Città della Salute e della Scienza University Hospital, 10126 Turin, Italy
| | - Enzo Medico
- Department of Oncology, University of Torino, Candiolo, TO, Italy; Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo, TO, Italy
| | - Samuel Y Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; National Cancer Institute, Bethesda, MD 20892, USA
| | - Allison Moskowitz
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zachary Epstein
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrew Intlekofer
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dogan Ahmed
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Wing C Chan
- Department of Pathology, City of Hope Medical Center, Duarte, CA 91010, US
| | - Peter Martin
- Lymphoma Service, Weill Cornell Medical Center, New York, NY 10065, USA
| | - Jia Ruan
- Lymphoma Service, Weill Cornell Medical Center, New York, NY 10065, USA
| | - Francesco Bertoni
- Lymphoma Genomics, Institute of Oncology Research, Faculty of Biomedical Sciences, USI, 6500 Bellinzona, Switzerland; Oncology Institute of Southern Switzerland, EOC,6500 Bellinzona, Switzerland
| | - Robin Foà
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Joshua D Brody
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, US; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Jaspreet Osan
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Oliver Elemento
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Doron Betel
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US; Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, US
| | - Wayne Tam
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA; Division of Hematopathology, Northwell Health, New York, NY 11740, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Perelman Center for Advanced Medicine, SPE 8-112, Philadelphia, PA 19104, USA; Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA; Lymphoma Program, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Leandro Cerchietti
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY 10065 US
| | - Raul Rabadan
- Program for Mathematical Genomics, Department of Systems Biology, Department of Biomedical Informatics, Columbia University, New York, NY 10027 USA
| | - Steven Horwitz
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Giorgio Inghirami
- Pathology and Laboratory Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
34
|
Aronson SL, Thijssen B, Lopez-Yurda M, Koole SN, van der Leest P, León-Castillo A, Harkes R, Seignette IM, Sanders J, Alkemade M, Kemper I, Holtkamp MJ, Mandjes IAM, Broeks A, Lahaye MJ, Rijlaarsdam MA, van den Broek D, Wessels LFA, Horlings HM, van Driel WJ, Sonke GS. Neo-adjuvant pembrolizumab in stage IV high-grade serous ovarian cancer: the phase II Neo-Pembro trial. Nat Commun 2025; 16:3520. [PMID: 40229272 PMCID: PMC11997049 DOI: 10.1038/s41467-025-58440-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/24/2025] [Indexed: 04/16/2025] Open
Abstract
While immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, their efficacy in high-grade serous ovarian cancer (HGSOC) remains limited. Some patients, however, achieve lasting responses, emphasizing the need to understand how tumor microenvironment and molecular characteristics influence ICI response. The phase 2 Neo-Pembro study (NCT03126812) included 33 untreated stage IV HGSOC patients, who were scheduled for 6 cycles of carboplatin-paclitaxel and interval cytoreductive surgery. Pembrolizumab (pembro) was added from cycle two and continued for one year. The primary objective was to assess intratumoral immune activation using multiplexed immunofluorescence and immune-related gene expression. Our findings show immune activation, evidenced by an increase in CD3 + , CD8 + , CD8 + /FOXP3+ ratio, TNF-α and interferon-γ signaling. Treatment was well-tolerated. We observed major pathologic responses in 9/33 patients (27%, 95%CI 14-46), with pathologic response strongly associated with immune activation and OS. At a median follow-up of 52.8 months, 8/9 major responders were alive, with 6 patients recurrence-free. In contrast, 4/24 minor responders survived, including one recurrence-free. ctDNA clearance was observed in all major responders and was associated with prolonged PFS and OS. PD-L1 expression and homologous recombination deficiency were predictive of major response and may serve as biomarkers, warranting further exploration. These results suggest major responders may benefit from neo-adjuvant pembro.
Collapse
Affiliation(s)
- S L Aronson
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Center for Gynecologic Oncology Amsterdam, Department of Gynecologic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - B Thijssen
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - M Lopez-Yurda
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - S N Koole
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Center for Gynecologic Oncology Amsterdam, Department of Gynecologic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - P van der Leest
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A León-Castillo
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R Harkes
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - I M Seignette
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M Alkemade
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - I Kemper
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - M J Holtkamp
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - I A M Mandjes
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - A Broeks
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - M J Lahaye
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - M A Rijlaarsdam
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - D van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - L F A Wessels
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - H M Horlings
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - W J van Driel
- Center for Gynecologic Oncology Amsterdam, Department of Gynecologic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - G S Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
35
|
Nørgaard M, Rusan M, Kondrup K, Sørensen EMG, Weiss S, Bjerre MT, Fredsøe J, Vang S, Jensen JB, De Laere B, Grönberg H, Borre M, Lindberg J, Sørensen KD. Deep targeted sequencing of circulating tumor DNA to inform treatment in patients with metastatic castration-resistant prostate cancer. J Exp Clin Cancer Res 2025; 44:120. [PMID: 40229848 PMCID: PMC11998381 DOI: 10.1186/s13046-025-03356-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/04/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Intrinsic and acquired resistance to second-generation anti-androgens pose a significant clinical challenge in the treatment of metastatic castration-resistant prostate cancer (mCRPC). Novel biomarkers to predict treatment response and inform alternative treatment options are urgently needed. METHODS Deep targeted sequencing, with a prostate cancer-specific gene panel, was performed on circulating tumor DNA (ctDNA) and germline DNA from blood of mCRPC patients recruited in Denmark (n = 53), prior to starting first-line treatment with enzalutamide or abiraterone acetate, and for a subset of patients also at progression (n = 18). Likely clonal hematopoietic variants were filtered out. Genomic findings were correlated to clinical outcomes (PSA progression-free survival (PFS), overall survival (OS)). Intrinsic resistance candidate biomarkers were considered by enrichment analysis of nonresponders vs. responders. Genomic alterations at progression were considered as possible drivers of acquired resistance. Clinical actionability was assessed based on OncoKB and ESCAT. RESULTS Somatic alterations in PTEN, cell cycle regulators (CCND1, CDKN1B, CDKN2A, and RB1) and chromatin modulators (CHD1, ARID1A) were associated with significantly shorter PFS and OS, also after adjusting for ctDNA% in multivariate Cox regression analysis. The associations with poorer outcomes for alterations in PTEN and chromatin modulators were validated in an external dataset. Patients with primary resistance to enzalutamide/abiraterone had enrichment for BRAF amplification and CHD1 loss, while responders had enrichment for TMPRSS2 fusions. AR resistance mutations emerged in 22% of patients at progression. These were mutually exclusive with other alterations that may confer resistance (i.e., activating CTNNB1 mutations, combined TP53/RB1 loss). Clinically actionable alterations, primarily in homologous recombination repair genes, were found in 54.7% and 49.0% of patients (OncoKB and ESCAT, respectively), with few additional alterations detected at progression. Level I alterations were identified in 41.5% of patients employing OncoKB, however only in 13.2% based on ESCAT. CONCLUSIONS Our study identifies known and novel prognostic and predictive biomarker candidates in patients with mCRPC undergoing first-line treatment with enzalutamide or abiraterone acetate. It further provides real-world evidence of the significant potential of genomic profiling of ctDNA to inform treatment in this setting. Clinical trials are warranted to advance the implementation of ctDNA-based biomarkers into clinical practice.
Collapse
Affiliation(s)
- Maibritt Nørgaard
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Maria Rusan
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus, Denmark
| | - Karoline Kondrup
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ea Marie Givskov Sørensen
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Simone Weiss
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Marianne Trier Bjerre
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
- Department of Urology, Gødstrup Hospital, Gødstrup, Denmark
| | - Jacob Fredsøe
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Søren Vang
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jørgen Bjerggaard Jensen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Urology, Gødstrup Hospital, Gødstrup, Denmark
| | - Bram De Laere
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Gent (CRIG), Ghent University, Ghent, Belgium
| | - Henrik Grönberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Michael Borre
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Urology, Aarhus University Hospital, Aarhus, Denmark
| | - Johan Lindberg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Karina Dalsgaard Sørensen
- Department of Molecular Medicine, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus, 8200, Denmark.
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
36
|
Mayr L, Neyazi S, Schwark K, Trissal M, Beck A, Labelle J, Eder SK, Weiler-Wichtl L, Marques JG, de Biagi-Junior CAO, Lo Cascio C, Chapman O, Sridhar S, Kenkre R, Dutta A, Wang S, Wang J, Hack O, Nascimento A, Nguyen CM, Castellani S, Rozowsky JS, Groves A, Panditharatna E, Cruzeiro GAV, Haase RD, Tabatabai K, Madlener S, Wadden J, Adam T, Kong S, Miclea M, Patel T, Bruckner K, Senfter D, Lämmerer A, Supko J, Guntner AS, Palova H, Neradil J, Stepien N, Lötsch-Gojo D, Berger W, Leiss U, Rosenmayr V, Dorfer C, Dieckmann K, Peyrl A, Azizi AA, Baumgartner A, Slaby O, Pokorna P, Clark LM, Cameron A, Nguyen QD, Wakimoto H, Dubois F, Greenwald NF, Bandopadhayay P, Beroukhim R, Ligon K, Kramm C, Bronsema A, Bailey S, Stucklin AG, Mueller S, Skrypek M, Martinez N, Bowers DC, Jones DTW, Jones C, Jäger N, Sterba J, Müllauer L, Haberler C, Kumar-Sinha C, Chinnaiyan A, Mody R, Chavez L, Furtner J, Koschmann C, Gojo J, Filbin MG. Effective targeting of PDGFRA-altered high-grade glioma with avapritinib. Cancer Cell 2025; 43:740-756.e8. [PMID: 40086436 DOI: 10.1016/j.ccell.2025.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/27/2024] [Accepted: 02/12/2025] [Indexed: 03/16/2025]
Abstract
PDGFRA is crucial to tumorigenesis and frequently genomically altered in high-grade glioma (HGG). In a comprehensive dataset of pediatric HGG (n = 261), we detect PDGFRA mutations and/or amplifications in 15% of cases, suggesting PDGFRA as a therapeutic target. We reveal that the PDGFRA/KIT inhibitor avapritinib shows (1) selectivity for PDGFRA inhibition, (2) distinct patterns of subcellular effects, (3) in vitro and in vivo activity in patient-derived HGG models, and (4) effective blood-brain barrier penetration in mice and humans. Furthermore, we report preliminary clinical real-world experience using avapritinib in pediatric and young adult patients with predominantly recurrent/refractory PDGFRA-altered HGG (n = 8). Our early data demonstrate that avapritinib is well tolerated and results in radiographic response in 3/7 cases, suggesting a potential role for avapritinib in the treatment of HGG with specific PDGFRA alterations. Overall, these translational results underscore the therapeutic potential of PDGFRA inhibition with avapritinib in HGG.
Collapse
Affiliation(s)
- Lisa Mayr
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA; Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Sina Neyazi
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Kallen Schwark
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria Trissal
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Alexander Beck
- Center for Neuropathology and Prion Research, Ludwig Maximilians University Munich, Faculty of Medicine, Muenchen, 80539 Bayern, Germany
| | - Jenna Labelle
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Sebastian K Eder
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA; St. Anna Children's Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna and St. Anna Children's Cancer Research Institute (CCRI), 1090 Vienna, Austria
| | - Liesa Weiler-Wichtl
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Joana G Marques
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Carlos A O de Biagi-Junior
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Costanza Lo Cascio
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Owen Chapman
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Sunita Sridhar
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Rishaan Kenkre
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Aditi Dutta
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Shanqing Wang
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Jessica Wang
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Olivia Hack
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Andrezza Nascimento
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Cuong M Nguyen
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Sophia Castellani
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Jacob S Rozowsky
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Andrew Groves
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Eshini Panditharatna
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Gustavo Alencastro Veiga Cruzeiro
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Rebecca D Haase
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Kuscha Tabatabai
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Sibylle Madlener
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Jack Wadden
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tiffany Adam
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Seongbae Kong
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Madeline Miclea
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Tirth Patel
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Katharina Bruckner
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; Department of Neurosurgery, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Daniel Senfter
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Anna Lämmerer
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, 1090 Vienna, Austria
| | - Jeffrey Supko
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Armin S Guntner
- Institute for Analytical and General Chemistry, Johannes Kepler University, 4040 Linz, Austria
| | - Hana Palova
- Central European Institute of Technology, Masaryk University, 60177 Brno, Czech Republic
| | - Jakub Neradil
- Department of Experimental Biology, Faculty of Science, Masaryk University, 61137 Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Natalia Stepien
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Daniela Lötsch-Gojo
- Department of Neurosurgery, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Walter Berger
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria; Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, 1090 Vienna, Austria
| | - Ulrike Leiss
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Verena Rosenmayr
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Christian Dorfer
- Department of Neurosurgery, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Karin Dieckmann
- Department of Radiotherapy, Medical University of Vienna, 1090 Vienna, Austria
| | - Andreas Peyrl
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Amedeo A Azizi
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Alicia Baumgartner
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA; Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, 60177 Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Petra Pokorna
- Central European Institute of Technology, Masaryk University, 60177 Brno, Czech Republic
| | - Louise M Clark
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, MA 02210, USA
| | - Amy Cameron
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, MA 02210, USA
| | - Quang-De Nguyen
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, MA 02210, USA
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Frank Dubois
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Noah F Greenwald
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Cancer Biology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02115, USA
| | - Pratiti Bandopadhayay
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Rameen Beroukhim
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Cancer Biology and Medical Oncology, Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA 02115, USA
| | - Keith Ligon
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA
| | - Christof Kramm
- Division of Pediatric Hematology and Oncology, University Medical Center Göttingen, 37099 Göttingen, Germany
| | - Annika Bronsema
- Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Simon Bailey
- Great North Childrens Hospital and Newcastle University, Newcastle upon Tyne, UK; Newcastle Hospitals NHS Foundation Trust, NE1 4LP Newcastle, UK
| | - Ana Guerreiro Stucklin
- Department of Oncology and Children's Research Center, University Children's Hospital Zurich, 8008 Zurich, Switzerland
| | - Sabine Mueller
- Departments of Pediatrics, Neurology, and Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Mary Skrypek
- Department of Pediatric Hematology-Oncology, Children's Minnesota, Minneapolis, MN 55404, USA
| | - Nina Martinez
- Department of Neurology & Neurological Surgery, Jefferson University, Philadelphia, PA 19107, USA
| | - Daniel C Bowers
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany; Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Chris Jones
- Division of Molecular Pathology, Institute of Cancer Research, SM2 5NG London, UK
| | - Natalie Jäger
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jaroslav Sterba
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 662630 Brno, Czech Republic
| | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria
| | - Christine Haberler
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Chandan Kumar-Sinha
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Arul Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Rajen Mody
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Lukas Chavez
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Julia Furtner
- Division of Neuroradiology and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria; Research Center of Medical Image Analysis and Artificial Intelligence, Danube Private University, 3500 Krems an der Donau, Austria
| | - Carl Koschmann
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Johannes Gojo
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria.
| | - Mariella G Filbin
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorder Center and Harvard Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
37
|
Lee M, Han SH, Kim D, Yun S, Yeom J, Kyeong M, Park SY, Lee DY. Systematic identification of genomic hotspots for high-yield protein production in CHO cells. N Biotechnol 2025; 88:61-72. [PMID: 40228657 DOI: 10.1016/j.nbt.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/24/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
The efficient and stable production of therapeutic proteins in Chinese hamster ovary (CHO) cells hinges on robust cell line development (CLD). Traditional methods relying on random transgene integration often result in clonal variability, requiring extensive and resource-intensive screening. To address this limitation, we established a systematic, multiomics-driven framework that integrates 202 RNA-sequencing datasets and whole-genome sequencing data to identify genomic "hotspot" loci for precise and high-yield transgene integration. From an initial pool of 20 candidate loci, 5 top-performing hotspots were validated using site-specific integration in CHO-DG44 cells via the CRISPR/Cas9 system with Recombinase-mediated cassette exchange (RMCE). These genomic hotspots achieved 2.2- to 15.0-fold higher relative specific productivity compared to previously known controls (Fer1L4 and Locus1 sites), across multiple therapeutic proteins, including a lysosomal storage disorder-related enzyme and an Immunoglobulin G (IgG)-related monoclonal antibody (mAb) expression. This study offers a transformative approach to CLD, achieving significant improvements in productivity, genomic stability, and efficiency, as well as paving the way for enhanced biopharmaceutical manufacturing.
Collapse
Affiliation(s)
- Minouk Lee
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Sung-Hyuk Han
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea; Cell-based Process Engineering, R&D, GC Biopharma, 93, Ihyun-ro, 30beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 16924, Republic of Korea
| | - Dongseok Kim
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Seongtae Yun
- Cell-based Process Engineering, R&D, GC Biopharma, 93, Ihyun-ro, 30beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 16924, Republic of Korea
| | - Jinho Yeom
- Cell-based Process Engineering, R&D, GC Biopharma, 93, Ihyun-ro, 30beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 16924, Republic of Korea
| | - Minji Kyeong
- Cell-based Process Engineering, R&D, GC Biopharma, 93, Ihyun-ro, 30beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 16924, Republic of Korea
| | - Seo-Young Park
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Dong-Yup Lee
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| |
Collapse
|
38
|
Merrick BA, Brooks AM, Foley JF, Martin NP, Fannin RD, Gladwell W, Gerrish KE. hTERT and SV40LgT Renal Cell Lines Adjust Their Transcriptional Responses After Copy Number Changes from the Parent Proximal Tubule Cells. Int J Mol Sci 2025; 26:3607. [PMID: 40332109 PMCID: PMC12027150 DOI: 10.3390/ijms26083607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Primary mouse renal proximal tubule epithelial cells (moRPTECs) were immortalized by lentivirus transduction to create hTERT or SV40LgT (LgT) cell lines. Prior work showed a more pronounced injury and repair response in LgT versus hTERT cells after chemical challenge. We hypothesized that unique genomic changes occurred after immortalization, altering critical genes and pathways. RNA-seq profiling and whole-genome sequencing (WGS) of parent, hTERT, and LgT cells showed that 92.5% of the annotated transcripts were shared, suggesting a conserved proximal tubule expression pattern. However, the cell lines exhibited unique transcriptomic and genomic profiles different from the parent cells. Three transcript classes were quite relevant for chemical challenge response-Cyps, ion channels, and metabolic transporters-each important for renal function. A pathway analysis of the hTERT cells suggested alterations in intermediary and energy metabolism. LgT cells exhibited pathway activation in cell cycle and DNA repair that was consistent with replication stress. Genomic karyotyping by combining WGS and RNA-seq data showed increased gene copy numbers in chromosome 5 for LgT cells, while hTERT cells displayed gene copy losses in chromosomes 4 and 9. These data suggest that the exaggerated transcriptional responses of LgT cells versus hTERT cells result from differences in gene copy numbers, replication stress, and the unique selection processes underlying LgT or hTERT immortalization.
Collapse
Affiliation(s)
- Bruce Alex Merrick
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA;
| | - Ashley M. Brooks
- Biostatistics and Computational Biology Branch, Integrative Bioinformatics Support Group, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA;
| | - Julie F. Foley
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA;
| | - Negin P. Martin
- Viral Vector Core, Neurobiology Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA;
| | - Rick D. Fannin
- Molecular Genomics Core Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA; (R.D.F.); (W.G.); (K.E.G.)
| | - Wesley Gladwell
- Molecular Genomics Core Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA; (R.D.F.); (W.G.); (K.E.G.)
| | - Kevin E. Gerrish
- Molecular Genomics Core Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA; (R.D.F.); (W.G.); (K.E.G.)
| |
Collapse
|
39
|
Moia R, Talotta D, Terzi Di Bergamo L, Almasri M, Dondolin R, Salehi M, Cosentino C, Soscia R, Della Starza I, Bruscaggin A, Andorno A, Mercalli F, Cresta S, Bomben R, Bittolo T, Vit F, Bulian P, Zucchetto A, Bruna R, Rivolta GM, Schipani M, Secomandi E, Kogila S, Bellia M, Mouhssine S, Nabki J, Al Deeban B, Ghanej J, Cividini L, Maher N, Melle F, Motta G, Leutner M, Lorenzi A, Mahmoud AM, Al Essa W, Deambrogi C, Rasi S, Petrucci L, Boldorini RL, Di Rocco A, Del Giudice I, Spina M, Palazzolo S, Canal F, Canzonieri V, Martelli M, Pileri S, Gattei V, Foà R, Rossi D, Gaidano G. Molecular clustering on ctDNA improves the prognostic stratification of patients with DLBCL compared with ctDNA levels. Blood Adv 2025; 9:1692-1701. [PMID: 39825831 PMCID: PMC11999195 DOI: 10.1182/bloodadvances.2024014136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/25/2024] [Accepted: 12/21/2024] [Indexed: 01/20/2025] Open
Abstract
ABSTRACT Circulating tumor DNA (ctDNA) levels can help predict outcomes in diffuse large B-cell lymphoma (DLBCL), but its integration with DLBCL molecular clusters remains unexplored. Using the LymphGen tool in 77 DLBCL cases with both ctDNA and tissue biopsy, a 95.8% concordance rate in molecular cluster assignment was observed, showing the reproducibility of molecular clustering on ctDNA. A multicenter, prospective cohort of 166 patients with newly diagnosed DLBCL was analyzed for ctDNA levels and molecular clusters using cancer personalized profiling by deep sequencing. Patients with ctDNA levels of <2.5 log10 haploid genome equivalents (hGE)/mL had a 4-year progression-free survival (PFS) and overall survival (OS) of 71.7% and 85.7%, respectively, compared with 50.3% and 61.0% for those with higher ctDNA levels (P = .0018 and P = .0017). Recursive partitioning showed that patients with ctDNA levels of ≥2.5 log10 hGE/mL were further stratified by clusters ST2/BN2. In this group, ST2/BN2 patients associated with a favorable outcome with a 4-year PFS and OS of 87.5% and 100%, respectively, compared to 38.0% and 47.1% for other clusters (P = .003 and P = .001). Combining ctDNA levels and ST2/BN2 clusters improved outcome prediction. Low-risk patients (n = 51), characterized by ctDNA levels of <2.5 log10 hGE/mL and/or BN2/ST2 cluster, had a 4-year PFS and OS of 75.3% and 87.8%, respectively. High-risk patients (n = 115), with ctDNA levels of ≥2.5 log10 hGE/mL and no BN2/ST2 cluster, had a 4-year PFS and OS of 38.0% and 47.1%, respectively. Adding cluster assignment to ctDNA levels improved the model's C statistics (0.63 vs 0.59 for PFS; 0.68 vs 0.63 for OS). Liquid biopsy thus provides a multilayered approach for outcome prediction in DLBCL.
Collapse
MESH Headings
- Humans
- Circulating Tumor DNA/genetics
- Circulating Tumor DNA/blood
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/blood
- Prognosis
- Female
- Male
- Middle Aged
- Aged
- Adult
- Biomarkers, Tumor
- High-Throughput Nucleotide Sequencing
- Aged, 80 and over
- Prospective Studies
Collapse
Affiliation(s)
- Riccardo Moia
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Donatella Talotta
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | | | - Mohammad Almasri
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Riccardo Dondolin
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Matin Salehi
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Chiara Cosentino
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Roberta Soscia
- Institute of Hematology, Hematology Unit, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Irene Della Starza
- Institute of Hematology, Hematology Unit, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Alessio Bruscaggin
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Annalisa Andorno
- Division of Pathology, Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Francesca Mercalli
- Division of Pathology, Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Stefania Cresta
- Division of Pathology, Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Riccardo Bomben
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Tamara Bittolo
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Filippo Vit
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Pietro Bulian
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Antonella Zucchetto
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Riccardo Bruna
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Giulia Maria Rivolta
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Mattia Schipani
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Eleonora Secomandi
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Sreekar Kogila
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Matteo Bellia
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Samir Mouhssine
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Jana Nabki
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Bashar Al Deeban
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Joseph Ghanej
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Luca Cividini
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Nawar Maher
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Federica Melle
- Hematopathology Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Giovanna Motta
- Hematopathology Unit, Istituto di Ricovero e Cura a Carattere Scientifico Azienda Ospedaliero-Universitaria of Bologna, Bologna, Italy
| | - Monica Leutner
- Division of Pathology, Azienda Sanitaria Locale del VCO, Verbania, Italy
| | - Angela Lorenzi
- Division of Hematology, Azienda Sanitaria Locale del VCO, Verbania, Italy
| | - Abdurraouf Mokhtar Mahmoud
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Wael Al Essa
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Clara Deambrogi
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Silvia Rasi
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Luigi Petrucci
- Institute of Hematology, Hematology Unit, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Renzo Luciano Boldorini
- Division of Pathology, Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Alice Di Rocco
- Institute of Hematology, Hematology Unit, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Ilaria Del Giudice
- Institute of Hematology, Hematology Unit, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Michele Spina
- Division of Medical Oncology and Immune-related Tumors, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Stefano Palazzolo
- Pathology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Fabio Canal
- Pathology and Histology Unit, Azienda Sanitaria Friuli Occidentale, Pordenone, Italy
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Maurizio Martelli
- Institute of Hematology, Hematology Unit, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Stefano Pileri
- Hematopathology Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Valter Gattei
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Robin Foà
- Institute of Hematology, Hematology Unit, Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Davide Rossi
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
- Clinic of Hematology, Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
- Faculty of Biomedicine, Universita' della Svizzera italiana, Lugano, Switzerland
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
40
|
Tanaka M, Letchworth R, Barnes AA, Lum L, Hughes S, Schlauderaff G, Chaudhary P, Ng KM, Superville D, Luna CM, Gonzalez M, Grossmann A, Reeves MQ. CIT tumor lines: A novel series of immunogenic squamous cell skin carcinoma cell lines derived from chemical carcinogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.647071. [PMID: 40291731 PMCID: PMC12026586 DOI: 10.1101/2025.04.03.647071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Immunotherapy is now widely used to treat cancer, but its efficacy in many cancer types remains modest. To overcome current barriers, preclinical mouse models that faithfully recapitulate the diversity of cancer types, tumor genetics, mutation burdens, and neoantigen patterns of human tumors are essential. Currently, there are relatively few transplantable murine models of squamous cell carcinomas (SCC). Here we describe a novel series of 11 skin SCC tumor lines, the Carcinogen-Induced Tumor (CIT) lines, syngeneic to the FVB strain. The CIT lines were established from skin carcinomas induced by DMBA and TPA treatment, and harbor genetic drivers and overall tumor mutational burdens that recapitulate those found across multiple human SCCs. Each CIT line gives rise to tumors with a consistent immune infiltration pattern, ranging from T cell-rich "hot" tumors to T cell-poor "cold" tumors. Hot CIT lines exhibit partial responses to treatment with immune checkpoint inhibitors, and we have identified two neoantigens present in an immunotherapy-responsive CIT line. The CIT lines thus provide a valuable new series of preclinical models for studying anti-tumor immune responses and developing strategies to improve immunotherapy efficacy in SCCs.
Collapse
|
41
|
Buianova AA, Lashkova YS, Kulichenko TV, Kuznetsov IS, Ivanov AA, Parshina OP, Suchalko ON, Vakhlyarskaya SS, Korostin DO. Heterozygous deletion of 10q24.31-q24.33- a new syndrome associated with multiple congenital anomalies: case report and literature review. Neurol Res Pract 2025; 7:22. [PMID: 40189573 PMCID: PMC11974182 DOI: 10.1186/s42466-025-00378-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/19/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Congenital anomalies and neurodevelopmental disorders are complex conditions often requiring comprehensive diagnostic approaches. Next-generation sequencing (NGS), particularly whole-exome sequencing (WES), has greatly improved the detection of pathogenic variants, including copy number variations (CNVs), which account for up to 35% of genetic causes in neurological patients. Combining CNV and single nucleotide variant (SNV) analysis through WES enhances diagnostic accuracy, especially in cases with unclassified congenital anomalies. CASE PRESENTATION AND LITERATURE REVIEW This study reports a 14-year-old male patient with multiple congenital anomalies, including hypospadias, complete cleft palate, and recurrent pneumonia. His clinical presentation includes significant physical and intellectual developmental delays, autism-like symptoms, and spastic diplegia. Whole-exome sequencing (WES) was performed due to these complex symptoms, revealing a novel heterozygous deletion on chromosome 10q24.31-q24.33. Laboratory findings indicated agammaglobulinemia, leading to prophylactic antibiotic therapy and immunoglobulin replacement. Additional imaging studies showed cystic malformation of the middle lobe of the right lung, sliding hiatal hernia with prolapse of the gastric mucosa, and brain anomalies consistent with Joubert syndrome. CONCLUSIONS This case underscores the importance of genetic analysis in understanding the etiology of congenital anomalies and neurodevelopmental disorders, providing critical insights into the molecular mechanisms driving complex phenotypes. The identified chromosomal deletion contributes to the existing literature on genomic imbalances associated with similar phenotypes.
Collapse
Affiliation(s)
- Anastasiia A Buianova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 1, bld. 1, Ostrovityanova St., Moscow, 117513, Russia.
| | - Yulia S Lashkova
- Russian Children's Clinical Hospital, 117 Leninsky Prospect, Moscow, 119571, Russia.
| | - Tatiana V Kulichenko
- Russian Children's Clinical Hospital, 117 Leninsky Prospect, Moscow, 119571, Russia
| | - Ivan S Kuznetsov
- Russian Children's Clinical Hospital, 117 Leninsky Prospect, Moscow, 119571, Russia
| | - Artem A Ivanov
- Russian Children's Clinical Hospital, 117 Leninsky Prospect, Moscow, 119571, Russia
| | - Olga P Parshina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 1, bld. 1, Ostrovityanova St., Moscow, 117513, Russia
| | - Oleg N Suchalko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 1, bld. 1, Ostrovityanova St., Moscow, 117513, Russia
| | | | - Dmitriy O Korostin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 1, bld. 1, Ostrovityanova St., Moscow, 117513, Russia
| |
Collapse
|
42
|
Liu X, Liang C, Ding L, Zhang Q, Liu Y, Wang W. Analysis of the clinical application value of cfDNA methylation and fragmentation in early diagnosis of esophageal cancer. Genomics 2025; 117:111034. [PMID: 40188889 DOI: 10.1016/j.ygeno.2025.111034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/02/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND This study explores the clinical value of cfDNA methylation and fragmentation for the early diagnosis of esophageal cancer using liquid biopsy. METHODS Whole genome bisulfite sequencing and low-pass whole genome sequencing were utilized to detect cfDNA biomarkers, comparing 30 esophageal cancer patients with 10 healthy controls. RESULTS Significant differences in cfDNA methylation and fragmentation were observed between cancerous and non-cancerous samples (p < 0.05). A volcano plot identified 822 differentially methylated markers (817 upregulated, 5 downregulated), with SOX17, SOX1, ZNF382, ZNF667-AS1, and TFPI2 highly associated with esophageal cancer. Fragmentation markers (EDM, FSD, FSR, TFBS, CNV) showed 95 % specificity and sensitivity, with EDM demonstrating the best performance. CONCLUSION Our study highlights the clinical potential of cfDNA methylation and fragmentation biomarkers for the early diagnosis of esophageal cancer.
Collapse
Affiliation(s)
- Xin Liu
- Department of Cardiothoracic Surgery, Xiangyang Central Hospital, Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, Hubei, China; Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, China
| | - Chen Liang
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, China
| | - Lingwen Ding
- Department of Vaccination clinic, Xiangyang Center for Disease Control and Prevention, Xiangyang, Hubei, China
| | - Qian Zhang
- Department of Cardiothoracic Surgery, Xiangyang Central Hospital, Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Yi Liu
- Department of Cardiothoracic Surgery, Xiangyang Central Hospital, Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Wei Wang
- Department of Cardiothoracic Surgery, Xiangyang Central Hospital, Hospital Affiliated to Hubei University of Arts and Science, Xiangyang, Hubei, China.
| |
Collapse
|
43
|
Peng T, Ma X, Hua W, Wang C, Chu Y, Sun M, Fermi V, Hamelmann S, Lindner K, Shao C, Zaman J, Tian W, Zhuo Y, Harim Y, Stöffler N, Hammann L, Xiao Q, Jin X, Warta R, Lotsch C, Zhuang X, Feng Y, Fu M, Zhang X, Zhang J, Xu H, Qiu F, Xie L, Zhang Y, Zhu W, Du Z, Salgueiro L, Schneider M, Eichhorn F, Lefevre A, Pusch S, Grinevich V, Ratliff M, Loges S, Bunse L, Sahm F, Xiang Y, Unterberg A, von Deimling A, Platten M, Herold-Mende C, Wu Y, Liu HK, Mao Y. Individualized patient tumor organoids faithfully preserve human brain tumor ecosystems and predict patient response to therapy. Cell Stem Cell 2025; 32:652-669.e11. [PMID: 39938519 DOI: 10.1016/j.stem.2025.01.002] [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: 12/22/2023] [Revised: 09/27/2024] [Accepted: 01/03/2025] [Indexed: 02/14/2025]
Abstract
Tumor organoids are important tools for cancer research, but current models have drawbacks that limit their applications for predicting response to therapy. Here, we developed a fast, efficient, and complex culture system (IPTO, individualized patient tumor organoid) that accurately recapitulates the cellular and molecular pathology of human brain tumors. Patient-derived tumor explants were cultured in induced pluripotent stem cell (iPSC)-derived cerebral organoids, thus enabling culture of a wide range of human tumors in the central nervous system (CNS), including adult, pediatric, and metastatic brain cancers. Histopathological, genomic, epigenomic, and single-cell RNA sequencing (scRNA-seq) analyses demonstrated that the IPTO model recapitulates cellular heterogeneity and molecular features of original tumors. Crucially, we showed that the IPTO model predicts patient-specific drug responses, including resistance mechanisms, in a prospective patient cohort. Collectively, the IPTO model represents a major breakthrough in preclinical modeling of human cancers, which provides a path toward personalized cancer therapy.
Collapse
Affiliation(s)
- Tianping Peng
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiujian Ma
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Changwen Wang
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Youjun Chu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Meng Sun
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Valentina Fermi
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Stefan Hamelmann
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Katharina Lindner
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Tanslational Neuroscience (MCTN), Heidelberg University, Heidelberg 69120, Germany; Immune Monitoring Unit, National Center for Tumor Diseases (NCT), Heidelberg 69120, Germany
| | - Chunxuan Shao
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Julia Zaman
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Weili Tian
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Yue Zhuo
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Yassin Harim
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Nadja Stöffler
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Linda Hammann
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Qungen Xiao
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Xiaoliang Jin
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Rolf Warta
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Catharina Lotsch
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Xuran Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuan Feng
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Minjie Fu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Xin Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Jinsen Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Hao Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Fufang Qiu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Liqian Xie
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Yi Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Zunguo Du
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lorena Salgueiro
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim 68167, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Mark Schneider
- Translational Research Unit, Thoraxklinik at Heidelberg University, Heidelberg 69120, Germany; Translational Lung Research Center Heidelberg (TRLC), German Center for Lung Research (DZL), Heidelberg 69120, Germany
| | - Florian Eichhorn
- Department of Thoracic Surgery, Thoraxklinik, University Hospital Heidelberg, Roentgenstrasse 1, Heidelberg 69126, Germany; Translational Lung Research Center Heidelberg (TRLC), German Center for Lung Research (DZL), Heidelberg 69120, Germany
| | - Arthur Lefevre
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Stefan Pusch
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Miriam Ratliff
- DKTK Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Research Center (DKFZ), Department of Neurosurgery, University Hospital Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Sonja Loges
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim 68167, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany; Translational Lung Research Center Heidelberg (TRLC), German Center for Lung Research (DZL), Heidelberg 69120, Germany
| | - Lukas Bunse
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Tanslational Neuroscience (MCTN), Heidelberg University, Heidelberg 69120, Germany; Immune Monitoring Unit, National Center for Tumor Diseases (NCT), Heidelberg 69120, Germany
| | - Felix Sahm
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Yangfei Xiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Andreas Unterberg
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Andreas von Deimling
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Michael Platten
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Helmholtz Institute of Translational Oncology Mainz (HI-TRON Mainz) - a Helmholtz Institute of the DKFZ, Mainz 55131, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Tanslational Neuroscience (MCTN), Heidelberg University, Heidelberg 69120, Germany; Immune Monitoring Unit, National Center for Tumor Diseases (NCT), Heidelberg 69120, Germany; German Cancer Consortium (DKTK), DKFZ, Core Center, Heidelberg 69120, Germany
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Yonghe Wu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China.
| | - Hai-Kun Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China; Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China.
| |
Collapse
|
44
|
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] [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
|
45
|
Song IH, Ahn B, Park YS, Kim DH, Hong SM. Presence of RB1 or Absence of LRP1B Mutation Predicts Poor Overall Survival in Patients with Gastric Neuroendocrine Carcinoma and Mixed Adenoneuroendocrine Carcinoma. Cancer Res Treat 2025; 57:492-506. [PMID: 39327909 PMCID: PMC12016830 DOI: 10.4143/crt.2024.667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/25/2024] [Indexed: 09/28/2024] Open
Abstract
PURPOSE Neuroendocrine carcinomas (NECs) of the stomach are extremely rare, but fatal. However, our understanding of the genetic alterations in gastric NECs is limited. We aimed to evaluate genomic and clinicopathological characteristics of gastric NECs and mixed adenoneuroendocrine carcinomas (MANECs). MATERIALS AND METHODS Fourteen gastric NECs, three gastric MANECs, and 1,381 gastric adenocarcinomas were retrieved from the departmental next-generation sequencing database between 2017 and 2022. Clinicopathological parameters and next-generation sequencing test results were retrospectively collected and reviewed. RESULTS Gastric NECs and MANECs frequently harbored alterations of TP53, RB1, SMARCA4, RICTOR, APC, TOP1, SLX4, EGFR, BRCA2, and TERT. In contrast, gastric adenocarcinomas exhibited alterations of TP53, CDH1, LRP1B, ARID1A, ERBB2, GNAS, CCNE1, NOTCH, and MYC. Mutations of AKT3, RB1, and SLX4; amplification of BRCA2 and RICTOR; and deletion of ADAMTS18, DDX11, KLRC3, KRAS, MAX, NFKBIA, NUDT7, and RB1 were significantly more frequent in gastric NECs and MANECs than in gastric adenocarcinomas. The presence of LRP1B mutation was significantly associated with longer overall survival (OS), whereas RB1 mutation and advanced TNM stage were associated with shorter OS. CONCLUSION We identified frequently mutated genes and potential predictors of survival in patients with gastric NECs and MANECs.
Collapse
Affiliation(s)
- In Hye Song
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Bokyung Ahn
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Young Soo Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Deok Hoon Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
46
|
Fiorini E, Malinova A, Schreyer D, Pasini D, Bevere M, Alessio G, Rosa D, D'Agosto S, Azzolin L, Milite S, Andreani S, Lupo F, Veghini L, Grimaldi S, Pedron S, Castellucci M, Nourse C, Salvia R, Malleo G, Ruzzenente A, Guglielmi A, Milella M, Lawlor RT, Luchini C, Agostini A, Carbone C, Pilarsky C, Sottoriva A, Scarpa A, Tuveson DA, Bailey P, Corbo V. MYC ecDNA promotes intratumour heterogeneity and plasticity in PDAC. Nature 2025; 640:811-820. [PMID: 40074906 PMCID: PMC12003172 DOI: 10.1038/s41586-025-08721-9] [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: 10/06/2023] [Accepted: 01/30/2025] [Indexed: 03/14/2025]
Abstract
Intratumour heterogeneity and phenotypic plasticity drive tumour progression and therapy resistance1,2. Oncogene dosage variation contributes to cell-state transitions and phenotypic heterogeneity3, thereby providing a substrate for somatic evolution. Nonetheless, the genetic mechanisms underlying phenotypic heterogeneity are still poorly understood. Here we show that extrachromosomal DNA (ecDNA) is a major source of high-level focal amplification in key oncogenes and a major contributor of MYC heterogeneity in pancreatic ductal adenocarcinoma (PDAC). We demonstrate that ecDNAs drive varying levels of MYC dosage, depending on their regulatory landscape, enabling cancer cells to rapidly and reversibly adapt to microenvironmental changes. In the absence of selective pressure, a high ecDNA copy number imposes a substantial fitness cost on PDAC cells. We also show that MYC dosage affects cell morphology and dependence of cancer cells on stromal niche factors. Our work provides a detailed analysis of ecDNAs in PDAC and describes a new genetic mechanism driving MYC heterogeneity in PDAC.
Collapse
Affiliation(s)
- Elena Fiorini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Antonia Malinova
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Daniel Schreyer
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Davide Pasini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- Department of Medicine, University of Verona, Verona, Italy
| | - Michele Bevere
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Giorgia Alessio
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- Department of Medicine, University of Verona, Verona, Italy
| | - Diego Rosa
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- Department of Medicine, University of Verona, Verona, Italy
| | - Sabrina D'Agosto
- ARC-Net Research Centre, University of Verona, Verona, Italy
- Human Technopole, Milan, Italy
| | - Luca Azzolin
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | - Salvatore Milite
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | - Silvia Andreani
- ARC-Net Research Centre, University of Verona, Verona, Italy
- Department of Biochemistry and Molecular Biology, University of Würzburg, Würzburg, Germany
| | - Francesca Lupo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Lisa Veghini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Sonia Grimaldi
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Serena Pedron
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Craig Nourse
- Cancer Research UK Beatson Institute, Glasgow, UK
- Botton-Champalimaud Pancreatic Cancer Centre, Lisbon, Portugal
| | - Roberto Salvia
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona, Verona, Italy
| | - Giuseppe Malleo
- Department of General and Pancreatic Surgery, The Pancreas Institute, University of Verona, Verona, Italy
| | - Andrea Ruzzenente
- Department of Surgical Sciences, Division of General and Hepatobiliary Surgery, University of Verona, Verona, Italy
| | - Alfredo Guglielmi
- Department of Surgical Sciences, Division of General and Hepatobiliary Surgery, University of Verona, Verona, Italy
| | - Michele Milella
- Section of Medical Oncology, Department of Medicine, University of Verona, Verona, Italy
| | - Rita T Lawlor
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
- ARC-Net Research Centre, University of Verona, Verona, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Antonio Agostini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Bioinformatics Research Core Facility, Gemelli Science and Technology Park (GSTeP), Rome, Italy
| | - Carmine Carbone
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | - Andrea Sottoriva
- Computational Biology Research Centre, Human Technopole, Milan, Italy
| | - Aldo Scarpa
- ARC-Net Research Centre, University of Verona, Verona, Italy
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Peter Bailey
- School of Cancer Sciences, University of Glasgow, Glasgow, UK.
- Botton-Champalimaud Pancreatic Cancer Centre, Lisbon, Portugal.
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy.
- ARC-Net Research Centre, University of Verona, Verona, Italy.
| |
Collapse
|
47
|
Martínez-Jiménez F, Chowell D. Genetic immune escape in cancer: timing and implications for treatment. Trends Cancer 2025; 11:286-294. [PMID: 39632211 PMCID: PMC11981860 DOI: 10.1016/j.trecan.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Genetic immune escape (GIE) alterations pose a significant challenge in cancer by enabling tumors to evade immune detection. These alterations, which can vary significantly across cancer types, may often arise early in clonal evolution and contribute to malignant transformation. As tumors evolve, GIE alterations are positively selected, allowing immune-resistant clones to proliferate. In addition to genetic changes, the tumor microenvironment (TME) and non-genetic factors such as inflammation, smoking, and environmental exposures play crucial roles in promoting immune evasion. Understanding the timing and mechanisms of GIE, alongside microenvironmental influences, is crucial for improving early detection and developing more effective therapeutic interventions. This review highlights the implications of GIE in cancer development and immunotherapy resistance, and emphasizes the need for integrative approaches.
Collapse
Affiliation(s)
- Francisco Martínez-Jiménez
- Systems Oncology Program, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain; Hartwig Medical Foundation, Amsterdam, The Netherlands.
| | - Diego Chowell
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
48
|
Chen P, Zhou JB, Chu XP, Feng YY, Zeng QB, Lei JH, Wong KP, Chan TI, Lam CW, Zhu WL, Chu WK, Hu F, Luo GH, Chan KI, Deng CX. Establishing a cryopreserved biobank of living tumor tissues for drug sensitivity testing. Bioact Mater 2025; 46:582-596. [PMID: 40061435 PMCID: PMC11889390 DOI: 10.1016/j.bioactmat.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 07/05/2024] [Accepted: 09/04/2024] [Indexed: 03/17/2025] Open
Abstract
The cryopreservation of cancer tissues to generate frozen libraries is a common practice used worldwide for storing patient samples for later applications. However, frozen samples stored by existing methods cannot be used for initiating living cell cultures, such as patient-derived tumor organoids (PDOs), which offer great potential for personalized treatment. To overcome this challenge, we developed a novel procedure for culturing PDOs using frozen live tumor tissues. We show that tumor specimens stored using this technique maintain their viability and can be successfully used to generate organoids even after long-term freezing, with an impressive success rate of 95.2 %. Importantly, we found that the structural features, tumor marker expression, and drug responses of organoids derived from frozen tissues are similar to those derived from fresh tissues. Moreover, organoids derived from frozen tissues can be routinely passaged and frozen, making them ideal for high-throughput drug screening at any time. Notably, cryopreserved tumor tissues can also be utilized in air-liquid interface (ALI) culture. This method allows for preserving the original tumor microenvironment, making it an invaluable resource for conducting tests on antitumor drug responses, including immune checkpoint inhibitors (ICIs). This innovation has the potential to enable the identification of potentially effective drugs for patients and facilitate the development of novel therapeutic drugs. Thus, we have established protocols for the long-term cryopreservation of cancer tissues to maintain their viability and microenvironment, which are useful for personalized therapy.
Collapse
Affiliation(s)
- Ping Chen
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jing-Bo Zhou
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Xiang-Peng Chu
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Yang-Yang Feng
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Qi-Bing Zeng
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Josh-Haipeng Lei
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Ka-Pou Wong
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | | | | | - Wen-Li Zhu
- Kiang Wu Hospital, Macau SAR 999078, China
| | | | - Feng Hu
- Kiang Wu Hospital, Macau SAR 999078, China
| | | | | | - Chu-Xia Deng
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| |
Collapse
|
49
|
Jiao D, Dong X, Fan S, Liu X, Yu Y, Wei C. Gastric cancer genomics study using reference human pangenomes. Life Sci Alliance 2025; 8:e202402977. [PMID: 39870503 PMCID: PMC11772497 DOI: 10.26508/lsa.202402977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/29/2025] Open
Abstract
A pangenome is the sum of the genetic information of all individuals in a species or a population. Genomics research has been gradually shifted to a paradigm using a pangenome as the reference. However, in disease genomics study, pangenome-based analysis is still in its infancy. In this study, we introduced a graph-based pangenome GGCPan from 185 patients with gastric cancer. We then systematically compared the cancer genomics study results using GGCPan, a linear pangenome GCPan, and the human reference genome as the reference. For small variant detection and microsatellite instability status identification, there is little difference in using three different genomes. Using GGCPan as the reference had a significant advantage in structural variant identification. A total of 24 candidate gastric cancer driver genes were detected using three different reference genomes, of which eight were common and five were detected only based on pangenomes. Our results showed that disease-specific pangenome as a reference is promising and a whole set of tools are still to be developed or improved for disease genomics study in the pangenome era.
Collapse
Affiliation(s)
- Du Jiao
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaorui Dong
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyu Fan
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyi Liu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yingyan Yu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaochun Wei
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
50
|
Zhang Q, Wang X, Shao Z, Zhang Y, Zhang L, Chen M, Zhou X, Zhu H, Zhou Y, Lu X, Li P, Chi W, Li L, Shao Z, Huang S, Xue J, Chi Y, Wu J, Xiu B. LINC01235 Promotes Clonal Evolution through DNA Replication Licensing-Induced Chromosomal Instability in Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413527. [PMID: 39950924 PMCID: PMC11984920 DOI: 10.1002/advs.202413527] [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: 10/23/2024] [Revised: 01/19/2025] [Indexed: 04/12/2025]
Abstract
Despite the development of HER2-targeting drugs such as trastuzumab and T-DXd, treatment resistance is a substantial challenge, often leading to relapse and distant metastasis. Tumor heterogeneity in HER2-positive breast cancer drives the evolution of resistant clones following therapeutic stress. However, the targetable drivers of anti-HER2 treatment resistance are not thoroughly identified. This study aims to use neoadjuvant-targeted therapy cohorts and a patient-derived organoid in vitro treatment model to uncover the potential targetable drivers of anti-HER2 treatment resistance. it is found that LINC01235 significantly enhances DNA replication licensing and chromosomal instability, fostering clonal expansion and evolution, and ultimately increasing resistance to therapeutic interventions. LINC01235 regulates global H3K27ac, H3K9ac, and H3K36me3 modifications, promotes H2A.Z expression in regulatory regions, and increases the accessibility of DNA licensing factors to their promoter regions. XRCC5 is identified as a key component for maintaining genomic stability, crucial for LINC01235's role in replication licensing. Furthermore, therapeutic strategies targeting LINC01235, including the use of antisense oligonucleotides or ATR inhibitors, which showed promise in overcoming treatment resistance are explored. These findings underscore the pivotal role of LINC01235 in driving resistance mechanisms and highlight novel avenues for targeted therapies to improve the outcomes of patients with HER2-positive breast cancer.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Xuliren Wang
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Zhibo Shao
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Yi Zhang
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Liyi Zhang
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Ming Chen
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Xujie Zhou
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Han Zhu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
| | - Yue Zhou
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Xinya Lu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Pei Li
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Weiru Chi
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Lun Li
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
- Department of General SurgeryThe Second Xiangya HospitalCentral South UniversityHunan410011China
| | - Zhi‐Ming Shao
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Shenglin Huang
- Fudan University Shanghai Cancer CenterKey Laboratory of Medical Epigenetics and MetabolismInstitutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Jingyan Xue
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| | - Yayun Chi
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
- Pathology CenterShanghai General HospitalShanghai Jiaotong University School of MedicineShanghai200080China
| | - Jiong Wu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
- Collaborative Innovation Center for Cancer MedicineShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Bingqiu Xiu
- Department of Breast SurgeryKey Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghai200032China
- Department of OncologyFudan University Shanghai Medical CollegeShanghai200032China
| |
Collapse
|