53 results on '"Sigouros M"'
Search Results
2. Clinico-genomic profiling and outcome prediction of neuroendocrine prostate cancer (NEPC)
- Author
-
Conteduca, V., primary, Oromendia, C., additional, Sigouros, M., additional, Sborner, A., additional, Nanus, D.M., additional, Tagawa, S.T., additional, Ballman, K., additional, and Beltran, H., additional
- Published
- 2018
- Full Text
- View/download PDF
3. 839P - Clinico-genomic profiling and outcome prediction of neuroendocrine prostate cancer (NEPC)
- Author
-
Conteduca, V., Oromendia, C., Sigouros, M., Sborner, A., Nanus, D.M., Tagawa, S.T., Ballman, K., and Beltran, H.
- Published
- 2018
- Full Text
- View/download PDF
4. mrLab: Leveraging Mixed Reality in a Precision Medicine Laboratory to Increase Safety and Productivity of Healthcare Workers during the COVID-19 Pandemic
- Author
-
Sboner, A., Alexandros Sigaras, Davis, J., Roshal, S., Wilkes, D., Hebding, C., Bockelman, D., Kane, T., Ackermann, S., Sigouros, M., Catalano, J., Martin, M., Song, W., Mosquera, J., Sternberg, C., Loda, M., Weinstein, H., and Elemento, O.
5. Emerging molecular phenotypes and potential therapeutic targets in esophageal and gastric adenocarcinoma unearthed by whole genome and transcriptome analyses.
- Author
-
Windon A, Al Assaad M, Hadi K, Mendelson N, Hissong E, Deshpande A, Tranquille M, Mclee J, Levine MF, Patel M, Medina-Martínez JS, Chiu K, Manohar J, Sigouros M, Ocean AJ, Sboner A, Jessurun J, Elemento O, Shah M, and Mosquera JM
- Subjects
- Humans, Male, Female, Transcriptome, Middle Aged, Aged, Phenotype, Whole Genome Sequencing, Mutation, Adult, Adenocarcinoma genetics, Adenocarcinoma pathology, Stomach Neoplasms genetics, Stomach Neoplasms pathology, Esophageal Neoplasms genetics, Esophageal Neoplasms pathology, Gene Expression Profiling methods
- Abstract
Background: Adenocarcinoma of the esophagus and stomach demands a deeper molecular understanding to advance treatment strategies and improve patient outcomes. Here, we profiled the genome and transcriptome landscape of these cancers, explored molecular characteristics that are undetectable by other sequencing platforms, and analyzed their potential clinical ramifications., Methods: Our study employed state-of-the-art integrative analyses of whole genome and transcriptome sequencing on 51 matched tumor and germline samples from 46 patients. Mutations and rearrangements in clinically relevant cancer genes were investigated and correlated with OncoKB, a knowledge-based precision oncology database, to identify treatment implications. Genome-wide signatures and manually curated molecular profiles were also determined., Results: The analyses revealed 90 targetable oncogenic mutations and fusions in 63 % of the patients, including novel NTRK, NRG1, ALK, and MET fusions, and structural variants in cancer genes like RAD51B. Also, molecular signatures associated with mismatch repair and homologous recombination deficiency were elucidated. Notably, we identified CDK12-type genomic instability associated with CDK12 fusions., Conclusions: Our findings support the potential of whole genome and transcriptome sequencing analyses as a comprehensive approach to identify treatment targets in adenocarcinoma of the stomach and the esophagus, and their application in precision oncology., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests. Kevin Hadi reports a relationship with Isabl that includes: employment. Aditya Deshpande reports a relationship with Isabl that includes: employment. Max F. Levine reports a relationship with Isabl that includes: employment. Minal Patel reports a relationship with Isabl that includes: employment. Juan Medina Martinez reports a relationship with Isabl that includes: employment. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier GmbH. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
6. Whole genome profiling of primary and metastatic adrenocortical carcinoma unravels significant molecular events.
- Author
-
Kalomeris T, Assaad MA, la Mora JD, Gundem G, Levine MF, Boyraz B, Manohar J, Sigouros M, Medina-Martínez JS, Sboner A, Elemento O, Scognamiglio T, and Mosquera JM
- Subjects
- Humans, Female, Male, Middle Aged, Adult, Mutation, Whole Genome Sequencing, Loss of Heterozygosity genetics, Aged, Adrenocortical Carcinoma genetics, Adrenocortical Carcinoma pathology, Adrenal Cortex Neoplasms genetics, Adrenal Cortex Neoplasms pathology
- Abstract
Adrenocortical carcinoma (ACC) is a rare, aggressive malignancy with limited treatment options and poor prognosis, with a 5-year survival rate of about 15 %. This study used whole genome sequencing to characterize the genomic landscape of five patients, one of them with both primary and metastatic samples. Key driver mutations were detected, including APC, JAK1, RFWD3 as well as other genes. Notably, a primary tumor harbored a RAD51 biallelic deleterious translocation, associated with homologous recombination deficiency signature. Large-scale copy neutral loss of heterozygosity (LOH) was identified in four tumors, three had TP53 mutations, with structural variants impacting genes as RB1, CDKN2A, and NF1. A genomic signature specific to mismatch repair was observed in a sample with MHS6 mutation. Two tumors presented novel fusions at TERT locus, including TERT::ZNF521. Comparative analysis between conventional and oncocytic ACC subtypes revealed no significant differences in mutation load, microsatellite instability, or specific gene enrichment. This comprehensive WGS analysis broadens the spectrum of genomic alterations in ACC, highlighting potential molecular targets and differences across subtypes that may inform future therapeutic strategies., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Max F. Levine, Gunes Gundem and Juan S. Medina-Martinez reports a relationship with Isabl, Inc. that includes: employment. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier GmbH. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
7. Enhancing the detection of clinically relevant biomarkers in advanced uterine and tubo-ovarian carcinomas through genome-wide analysis.
- Author
-
Al Assaad M, Hadi K, Tu J, Levine MF, Patel M, Deshpande A, Manohar J, Sigouros M, Sboner A, Chapman-Davis E, Elemento O, Holcomb K, Boyraz B, and Mosquera JM
- Subjects
- Humans, Female, Middle Aged, Aged, Adult, Genome-Wide Association Study, Whole Genome Sequencing, Aged, 80 and over, Biomarkers, Tumor genetics, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Uterine Neoplasms genetics, Uterine Neoplasms pathology
- Abstract
Background: Advanced-stage tube-ovarian cancers (TOC) and uterine cancers (UC) significantly contribute to cancer mortality. While surgery achieves clinical remission in most cases, recurrence often necessitates systemic therapy. Recent molecular phenotype studies have advanced targeted therapies. We employed whole genome sequencing (WGS) to investigate biomarkers in gynecologic malignancies., Design: Ninety-one tumor samples (45 TOC, 46 UC) were analyzed for germline mutations, somatic driver mutations, rearrangements, genome-wide signatures, and molecular phenotypes. A WGS-based high-confidence classifier for homologous recombination deficiency (HRD) was applied. Genomic profiles were correlated with clinical outcomes., Results: The HRD phenotype was identified in serous carcinoma components, with 50 % of HRD cases showing BRCA1/2 wildtype (33 %) or variants of unknown significance (17 %). HRD correlated with better overall survival in tubo-ovarian serous carcinoma and was linked to responses to platinum therapy and PARP inhibitors. Endometrioid carcinomas showed no HRD phenotype despite BRCA1/2 variants. CDK12-type genomic instability (10 %) occurred in cases with CDK12 rearrangements, and CCNE1 gain (8 %) was observed in CCNE1-amplified cases, independent of copy number. In endometrioid carcinoma, mismatch repair (MMR) deficiency single base substitution signatures, particularly SBS44, contributed to high tumor mutation burden (TMB). Ultra-high TMB was found in two cases with pathogenic POLE and POLQ mutations, exhibiting multiple SBS signatures, including MMR deficiency., Conclusion: Comprehensive genomic profiling of advanced-stage TOC and UC via WGS reveals key biomarkers and therapeutic targets, enhancing diagnostic accuracy and advancing personalized medicine in gynecological cancers., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Kevin Hadi reports a relationship with Isabl that includes: employment. Max F. Levine reports a relationship with Isabl that includes: employment. Minal Patel reports a relationship with Isabl that includes: employment. Aditya Deshpande reports a relationship with Isabl that includes: employment. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier GmbH. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
8. Novel structural variants that impact cell cycle genes are elucidated in metastatic gastrointestinal stromal tumors.
- Author
-
Delgado-de la Mora J, Al Assaad M, Quitian S, Levine MF, Deshpande A, Sigouros M, Manohar J, Medina-Martínez JS, Sboner A, Elemento O, Jessurun J, Hissong E, and Mosquera JM
- Subjects
- Humans, Middle Aged, Male, Female, Aged, Cyclin-Dependent Kinases genetics, Cyclin-Dependent Kinase Inhibitor p15 genetics, Cyclin-Dependent Kinase Inhibitor p16 genetics, Whole Genome Sequencing, Protein Kinase Inhibitors therapeutic use, Genes, cdc genetics, Gastrointestinal Stromal Tumors genetics, Gastrointestinal Stromal Tumors pathology, Gastrointestinal Stromal Tumors drug therapy, Gastrointestinal Neoplasms genetics, Gastrointestinal Neoplasms pathology
- Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal neoplasm of the digestive tract. Despite multiple therapeutic advances, patients with advanced disease frequently develop resistance to tyrosine kinase inhibitors (TKIs), and therefore represent a therapeutic challenge. We employed whole genome sequencing (WGS) on three metastatic GISTs refractory to various TKIs and explored a publicly available cohort of 499 GISTs. This study sheds light on the clinical importance of alterations in cell cycle genes such as cyclin-dependent kinase 2 A (CDKN2A), and cyclin-dependent kinase 2B (CDKN2B), their frequent alteration in metastatic GISTs and their potential role in tumor progression of this neoplasm. Likewise, new structural variations were identified in cyclin-dependent kinase 12 (CDK12). Whole genome profiling of metastatic GIST provides new insights to advance precision care of the disease, focusing on new therapeutic possibilities, especially for emerging targets such as CDK12., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Max F. Levine reports a relationship with Isabl Inc. that includes: employment. Aditya Deshpande reports a relationship with Isabel Inc. that includes: employment. Juan S. Medina-Martinez reports a relationship with Isabl Inc that includes: employment. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier GmbH. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
9. Whole genome and transcriptome analysis of pancreatic acinar cell carcinoma elucidates mechanisms of homologous recombination deficiency and unravels novel relevant fusion events.
- Author
-
Delgado-de la Mora J, Al Assaad M, Karaaslan S, Hadi K, Halima A, Deshpande A, Manohar J, Sigouros M, Medina-Martínez JS, Lieberman MD, Sboner A, Popa EC, Jessurun J, Elemento O, Ocean AJ, Hissong E, and Mosquera JM
- Subjects
- Humans, Male, Female, Middle Aged, Homologous Recombination genetics, Transcriptome genetics, Whole Genome Sequencing, Aged, Gene Fusion, Adult, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Carcinoma, Acinar Cell genetics, Carcinoma, Acinar Cell pathology, Gene Expression Profiling methods
- Abstract
Pancreatic acinar cell carcinoma (PACC) is a rare pancreatic tumor with a heterogeneous clinical course and, except for radical surgery, limited treatment options. We present a comprehensive study encompassing whole-genome and RNA sequencing of 7 tumor samples from 3 metastatic PACC patients to further delineate its genomic landscape and potential therapeutic implications. Our findings reveal distinct signatures of homologous recombination deficiency (HRD) in patients harboring pathogenic germline BRCA1/2 and FANCL mutations, demonstrating favorable responses to poly (ADP-ribose) polymerase 1 (PARP) inhibitors with prolonged disease-free intervals. Additionally, we first describe structural variants in PACC, including BRCA1::TRIM47 fusion and another variant impacting FANCC, both events related to HRD, and we also identify alterations in the mitogen-activated protein kinase (MAPK) pathway, including RAF1 duplication as well as novel BRAF::SORBS2 and MAP7D2::SND1 gene fusions, offering potential targets for therapy. Our study underscores the importance of genome and transcriptome-wide profiling of PACC, to help guide personalized treatment strategies to improve patient outcomes., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Kevin Hadi reports a relationship with Isabl Inc that includes: employment. Juan S. Medina-Martinez reports a relationship with Isabl Inc that includes: employment. Aditya Deshpande reports a relationship with Isabl Inc that includes: employment. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. Competing interest Kevin Hadi, Aditya Deshpande and Juan S. Medina-Martinez are employees at Isabl, Inc., (Copyright © 2024 Elsevier GmbH. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
10. Corrigendum to "Whole genome and transcriptome analysis of pancreatic acinar cell carcinoma elucidates mechanisms of homologous recombination deficiency and unravels novel relevant fusion events" [Pathol. - Res. Pract. 266 (2025) 155798].
- Author
-
Delgado-de la Mora J, Al Assaad M, Karaaslan S, Hadi K, Halima A, Deshpande A, Manohar J, Sigouros M, Medina-Martínez JS, Lieberman MD, Sboner A, Popa EC, Jessurun J, Elemento O, Ocean AJ, Hissong E, and Mosquera JM
- Published
- 2025
- Full Text
- View/download PDF
11. Kinome-Focused CRISPR-Cas9 Screens in African Ancestry Patient-Derived Breast Cancer Organoids Identify Essential Kinases and Synergy of EGFR and FGFR1 Inhibition.
- Author
-
Madorsky Rowdo FP, Martini R, Ackermann SE, Tang CP, Tranquille M, Irizarry A, Us I, Alawa O, Moyer JE, Sigouros M, Nguyen J, Al Assaad M, Cheng E, Ginter PS, Manohar J, Stonaker B, Boateng R, Oppong JK, Adjei EK, Awuah B, Kyei I, Aitpillah FS, Adinku MO, Ankomah K, Osei-Bonsu EB, Gyan KK, Hoda S, Newman L, Mosquera JM, Sboner A, Elemento O, Dow LE, Davis MB, and Martin ML
- Subjects
- Humans, Female, Protein Kinase Inhibitors pharmacology, Black People genetics, Organoids pathology, Organoids metabolism, Breast Neoplasms genetics, Breast Neoplasms pathology, Breast Neoplasms drug therapy, CRISPR-Cas Systems, Receptor, Fibroblast Growth Factor, Type 1 genetics, Receptor, Fibroblast Growth Factor, Type 1 antagonists & inhibitors, Receptor, Fibroblast Growth Factor, Type 1 metabolism, ErbB Receptors genetics, ErbB Receptors antagonists & inhibitors, ErbB Receptors metabolism
- Abstract
Precision medicine approaches to cancer treatment aim to exploit genomic alterations that are specific to individual patients to tailor therapeutic strategies. Yet, some targetable genes and pathways are essential for tumor cell viability even in the absence of direct genomic alterations. In underrepresented populations, the mutational landscape and determinants of response to existing therapies are poorly characterized because of limited inclusion in clinical trials and studies. One way to reveal tumor essential genes is with genetic screens. Most screens are conducted on cell lines that bear little resemblance to patient tumors, after years of culture under nonphysiologic conditions. To address this problem, we aimed to develop a CRISPR screening pipeline in three-dimensionally grown patient-derived tumor organoid (PDTO) models. A breast cancer PDTO biobank that focused on underrepresented populations, including West African patients, was established and used to conduct a negative-selection kinome-focused CRISPR screen to identify kinases essential for organoid growth and potential targets for combination therapy with EGFR or MEK inhibitors. The screen identified several previously unidentified kinase targets, and the combination of FGFR1 and EGFR inhibitors synergized to block organoid proliferation. Together, these data demonstrate the feasibility of CRISPR-based genetic screens in patient-derived tumor models, including PDTOs from underrepresented patients with cancer, and identify targets for cancer therapy. Significance: Generation of a breast cancer patient-derived tumor organoid biobank focused on underrepresented populations enabled kinome-focused CRISPR screening that identified essential kinases and potential targets for combination therapy with EGFR or MEK inhibitors. See related commentary by Trembath and Spanheimer, p. 407., (©2025 American Association for Cancer Research.)
- Published
- 2025
- Full Text
- View/download PDF
12. Mesonephric Adenocarcinoma in a Young Male Patient.
- Author
-
Guevara D, Shin N, Boiko A, Valiev I, Elsaeed AG, Mosquera JM, Al Assaad M, Manohar J, Sigouros M, Zaichikova A, Fomchenkova V, Yunusova L, Smirnova S, Elemento O, Nauseef J, and Sternberg CN
- Subjects
- Humans, Male, Adult, Tuberous Sclerosis Complex 2 Protein genetics, Class I Phosphatidylinositol 3-Kinases genetics, Mutation, Adenocarcinoma pathology, Adenocarcinoma genetics, Adenocarcinoma diagnosis
- Abstract
Background/aim: Mesonephric adenocarcinoma in males is an exceptionally rare malignancy arising from mesonephric remnants. Accurate diagnosis requires thorough histopathological and molecular analysis., Case Report: A 33-year-old male presented with right lower quadrant pain, and imaging revealed a pelvic mass compressing adjacent structures. Biopsy confirmed mesonephric adenocarcinoma. Genetic profiling revealed mutations in tuberous sclerosis complex 2 (TSC2) and phosphatidylinositol-4,5-biphosphate 3-kinase catalytic subunit alpha (PIK3CA), offering potential for targeted therapy., Conclusion: This case highlights the diagnostic and therapeutic challenges of mesonephric adenocarcinoma in males. Molecular profiling provided insights into tumor biology and potential targeted treatments. Multidisciplinary collaboration and surveillance remain essential for managing rare malignancies effectively., (Copyright © 2025 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
13. Unusual Presentation of Advanced Urothelial Cancer in a Young Patient.
- Author
-
Guevara D, Shin N, Boiko A, Valiev I, Elsaeed AG, Mosquera JM, Al Assaad M, Manohar J, Sigouros M, Zaichikova A, Fomchenkova V, Yunusova L, Smirnova S, Elemento O, Nanus D, and Sternberg CN
- Subjects
- Humans, Male, Adult, Tumor Suppressor Protein p53 genetics, Carcinoma, Transitional Cell drug therapy, Carcinoma, Transitional Cell pathology, Carcinoma, Transitional Cell genetics, Mutation, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Retinoblastoma Binding Proteins genetics, Immunotherapy methods, Urethral Neoplasms pathology, Urethral Neoplasms genetics, Urethral Neoplasms drug therapy, Urethral Neoplasms therapy, Antibodies, Monoclonal, Ubiquitin-Protein Ligases, Antibodies, Monoclonal, Humanized therapeutic use
- Abstract
Background/aim: Urothelial carcinoma, common in older adults, is rare in younger populations and even less common in the prostatic urethra. Advanced disease is typically managed with platinum-based chemotherapy, immune checkpoint inhibitors, and targeted therapies. However, rare presentations in young patients with aggressive disease highlight the need for innovative and personalized treatment strategies., Case Report: This case report presents a rare instance of metastatic urothelial carcinoma originating in the prostatic urethra of a 37-year-old male. Initial symptoms led to diagnosis through imaging, biopsy, and genetic profiling, revealing mutations in TP53 and RB1. The patient underwent multiple treatments, including dose-dense chemotherapy, pembrolizumab immunotherapy, and targeted antibody-drug conjugates (Enfortumab Vedotin and Sacituzumab Govitecan). Despite aggressive therapies, disease management remained challenging, leading to experimental treatments, including a personalized vaccine., Conclusion: This case underscores the importance of precision medicine and the need for innovative treatment options for rare and aggressive cancers., (Copyright © 2025 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
14. The interplay of mutagenesis and ecDNA shapes urothelial cancer evolution.
- Author
-
Nguyen DD, Hooper WF, Liu W, Chu TR, Geiger H, Shelton JM, Shah M, Goldstein ZR, Winterkorn L, Helland A, Sigouros M, Manohar J, Moyer J, Al Assaad M, Semaan A, Cohen S, Madorsky Rowdo F, Wilkes D, Osman M, Singh RR, Sboner A, Valentine HL, Abbosh P, Tagawa ST, Nanus DM, Nauseef JT, Sternberg CN, Molina AM, Scherr D, Inghirami G, Mosquera JM, Elemento O, Robine N, and Faltas BM
- Subjects
- Female, Humans, Male, APOBEC Deaminases genetics, APOBEC Deaminases metabolism, Cyclin D1 genetics, Cyclin D1 metabolism, DNA Copy Number Variations genetics, Drug Resistance, Neoplasm genetics, Urothelium metabolism, Urothelium pathology, Whole Genome Sequencing, Middle Aged, DNA, Circular genetics, Evolution, Molecular, Mutagenesis genetics, Mutation, Urinary Bladder Neoplasms drug therapy, Urinary Bladder Neoplasms genetics, Urinary Bladder Neoplasms pathology
- Abstract
Advanced urothelial cancer is a frequently lethal disease characterized by marked genetic heterogeneity
1 . In this study, we investigated the evolution of genomic signatures caused by endogenous and external mutagenic processes and their interplay with complex structural variants (SVs). We superimposed mutational signatures and phylogenetic analyses of matched serial tumours from patients with urothelial cancer to define the evolutionary dynamics of these processes. We show that APOBEC3-induced mutations are clonal and early, whereas chemotherapy induces mutational bursts of hundreds of late subclonal mutations. Using a genome graph computational tool2 , we observed frequent high copy-number circular amplicons characteristic of extrachromosomal DNA (ecDNA)-forming SVs. We characterized the distinct temporal patterns of APOBEC3-induced and chemotherapy-induced mutations within ecDNA-forming SVs, gaining new insights into the timing of these mutagenic processes relative to ecDNA biogenesis. We discovered that most CCND1 amplifications in urothelial cancer arise within circular ecDNA-forming SVs. ecDNA-forming SVs persisted and increased in complexity, incorporating additional DNA segments and contributing to the evolution of treatment resistance. Oxford Nanopore Technologies long-read whole-genome sequencing followed by de novo assembly mapped out CCND1 ecDNA structure. Experimental modelling of CCND1 ecDNA confirmed its role as a driver of treatment resistance. Our findings define fundamental mechanisms that drive urothelial cancer evolution and have important therapeutic implications., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
15. Whole genome sequencing elucidates etiological differences in MCPyV-negative Merkel cell carcinoma.
- Author
-
Stephan C, Al Assaad M, Levine MF, Deshpande A, Sigouros M, Manohar J, Sboner A, Elemento O, Pavlick AC, and Mosquera JM
- Subjects
- Humans, Male, Aged, Female, Mutation, Aged, 80 and over, Middle Aged, Carcinoma, Merkel Cell genetics, Carcinoma, Merkel Cell virology, Carcinoma, Merkel Cell pathology, Skin Neoplasms genetics, Skin Neoplasms virology, Skin Neoplasms pathology, Merkel cell polyomavirus genetics, Merkel cell polyomavirus isolation & purification, Whole Genome Sequencing
- Abstract
Merkel cell carcinoma (MCC) is an aggressive neuroendocrine neoplasm of the skin. Immunosuppression, ultraviolet radiation and the integration of Merkel cell polyomavirus (MCPyV) have all been shown to be involved in the pathogenesis of this malignancy. We performed whole genome sequencing on two MCPyV-negative cases of MCC that demonstrated very different clinical presentations and outcomes, and mutational profiles. The first case exhibited a highly aggressive clinical course, absence of UV-signature mutations and a low tumor mutational burden. A rearrangement in the tumor suppressor gene SUFU was identified, a likely driver and potential target of the Hedgehog signaling pathway. Meanwhile, the second case exhibited a less aggressive behavior, harbored UV-signature mutations, and a high mutational burden including mutations in TP53 and RB1., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Max F. Levine and Aditya Deshpande are employees at Isabl, Inc. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier GmbH. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
16. Integrative Transcriptomic and Single-Cell Protein Characterization of Colorectal Carcinoma Delineates Distinct Tumor Immune Microenvironments Associated with Overall Survival.
- Author
-
Hissong E, Bhinder B, Kim J, Ohara K, Ravichandran H, Assaad MA, Elsoukkary S, Shusterman M, Khan U, Eng KW, Bareja R, Manohar J, Sigouros M, Rendeiro AF, Jessurun J, Ocean AJ, Sboner A, Elemento O, Mosquera JM, and Shah MA
- Abstract
Colorectal carcinoma (CRC) is a heterogeneous group of tumors with varying therapeutic response and prognosis, and evidence suggests the tumor immune microenvironment (TIME) plays a pivotal role. Using advanced molecular and spatial biology technologies, we aimed to evaluate the TIME in patients with CRC to determine whether specific alterations in the immune composition correlated with prognosis. We identified primary and metastatic tumor samples from 31 consented patients, which were profiled with whole-exome sequencing and bulk RNA-seq. Immune cell deconvolution followed by gene set enrichment analysis and unsupervised clustering was performed. A subset of tumors underwent in situ analysis of the TIME spatial composition at single-cell resolution through Imaging Mass Mass Cytometry. Gene set enrichment analysis revealed two distinct groups of advanced CRC, one with an immune activated phenotype and the other with a suppressed immune microenvironment. The activated TIME phenotype contained increased Th1 cells, activated dendritic cells, tertiary lymphoid structures, and higher counts of CD8+ T cells whereas the inactive or suppressed TIME contained increased macrophages and a higher M2/M1 ratio. Our findings were further supported by RNA-seq data analysis from the TCGA CRC database, in which unsupervised clustering also identified two separate groups. The immunosuppressed CRC TIME had a lower overall survival probability (HR 1.66, p=0.007). This study supports the pertinent role of the CRC immune microenvironment in tumor progression and patient prognosis. We characterized the immune cell composition to better understand the complexity and vital role that immune activity states of the TIME play in determining patient outcome., Competing Interests: Conflicts of Interest: O.E. holds equity in OneThree Biotech, Volastra Therapeutics, Owkin, Champions Oncology, Pionyr Immunotherapeutics, Harmonic Discovery and Freenome. The remaining authors declare no competing interests. No patents have been filed or are related to this manuscript. Competing Interests O.E. holds equity in OneThree Biotech, Volastra Therapeutics, Owkin, Champions Oncology, Pionyr Immunotherapeutics, Harmonic Discovery and Freenome. The remaining authors declare no competing interests. No patents have been filed or are related to this manuscript.
- Published
- 2024
- Full Text
- View/download PDF
17. Whole-Genome Sequencing Analysis of Male Breast Cancer Unveils Novel Structural Events and Potential Therapeutic Targets.
- Author
-
Al Assaad M, Michaud O, Semaan A, Sigouros M, Tranquille M, Phan A, Levine MF, Gundem G, Medina-Martínez JS, Papaemmanuil E, Manohar J, Wilkes D, Sboner A, Hoda SAF, Elemento O, and Mosquera JM
- Subjects
- Humans, Male, Female, In Situ Hybridization, Fluorescence, Mutation, Oncogenes, Germ-Line Mutation, DNA Helicases genetics, Nuclear Proteins genetics, Transcription Factors genetics, Breast Neoplasms, Male genetics, Breast Neoplasms, Male therapy, Breast Neoplasms pathology
- Abstract
The molecular characterization of male breast cancer (MaBC) has received limited attention in research, mostly because of its low incidence rate, accounting for only 0.5% to 1% of all reported cases of breast cancer each year. Managing MaBC presents significant challenges, with most treatment protocols being adapted from those developed for female breast cancer. Utilizing whole-genome sequencing (WGS) and state-of-the-art analyses, the genomic features of 10 MaBC cases (n = 10) were delineated and correlated with clinical and histopathologic characteristics. Using fluorescence in situ hybridization, an additional cohort of 18 patients was interrogated to supplement WGS findings. The genomic landscape of MaBC uncovered significant genetic alterations that could influence diagnosis and treatment. We found common somatic mutations in key driver genes, such as FAT1, GATA3, SMARCA4, and ARID2. Our study also mapped out structural variants that impact cancer-associated genes, such as ARID1A, ESR1, GATA3, NTRK1, and NF1. Using a WGS-based classifier, homologous recombination deficiency (HRD) was identified in 2 cases, both presenting with deleterious variants in BRCA2. Noteworthy was the observation of FGFR1 amplification in 21% of cases. Altogether, we identified at least 1 potential therapeutic target in 8 of the 10 cases, including high tumor mutational burden, FGFR1 amplification, and HRD. Our study is the first WGS characterization of MaBC, which uncovered potentially relevant variants, including structural events in cancer genes, HRD signatures, and germline pathogenic mutations. Our results demonstrate unique genetic markers and potential treatment targets in MaBC, thereby underlining the necessity of tailoring treatment strategies for this understudied patient population. These WGS-based findings add to the growing knowledge of MaBC genomics and highlight the need to expand research on this type of cancer., (Copyright © 2024 United States & Canadian Academy of Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Noninvasive Detection of Neuroendocrine Prostate Cancer through Targeted Cell-free DNA Methylation.
- Author
-
Franceschini GM, Quaini O, Mizuno K, Orlando F, Ciani Y, Ku SY, Sigouros M, Rothmann E, Alonso A, Benelli M, Nardella C, Auh J, Freeman D, Hanratty B, Adil M, Elemento O, Tagawa ST, Feng FY, Caffo O, Buttigliero C, Basso U, Nelson PS, Corey E, Haffner MC, Attard G, Aparicio A, Demichelis F, and Beltran H
- Subjects
- Male, Humans, DNA Methylation, Prospective Studies, Biopsy, Prostatic Neoplasms, Castration-Resistant diagnosis, Prostatic Neoplasms, Castration-Resistant genetics, Cell-Free Nucleic Acids genetics
- Abstract
Castration-resistant prostate cancer (CRPC) is a heterogeneous disease associated with phenotypic subtypes that drive therapy response and outcome differences. Histologic transformation to castration-resistant neuroendocrine prostate cancer (CRPC-NE) is associated with distinct epigenetic alterations, including changes in DNA methylation. The current diagnosis of CRPC-NE is challenging and relies on metastatic biopsy. We developed a targeted DNA methylation assay to detect CRPC-NE using plasma cell-free DNA (cfDNA). The assay quantifies tumor content and provides a phenotype evidence score that captures diverse CRPC phenotypes, leveraging regions to inform transcriptional state. We tested the design in independent clinical cohorts (n = 222 plasma samples) and qualified it achieving an AUC > 0.93 for detecting pathology-confirmed CRPC-NE (n = 136). Methylation-defined cfDNA tumor content was associated with clinical outcomes in two prospective phase II clinical trials geared towards aggressive variant CRPC and CRPC-NE. These data support the application of targeted DNA methylation for CRPC-NE detection and patient stratification., Significance: Neuroendocrine prostate cancer is an aggressive subtype of treatment-resistant prostate cancer. Early detection is important, but the diagnosis currently relies on metastatic biopsy. We describe the development and validation of a plasma cell-free DNA targeted methylation panel that can quantify tumor fraction and identify patients with neuroendocrine prostate cancer noninvasively. This article is featured in Selected Articles from This Issue, p. 384., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
19. NIPBL::NACC1 Fusion Hepatic Carcinoma.
- Author
-
Hissong E, Al Assaad M, Bal M, Reed KA, Fornelli A, Levine MF, Gundem G, Semaan A, Orr CE, Sakhadeo U, Manohar J, Sigouros M, Wilkes D, Sboner A, Montgomery EA, Graham RP, Medina-Martínez JS, Robine N, Fang JM, Choi EK, Westerhoff M, Delgado-de la Mora J, Caudell P, Yantiss RK, Papaemmanuil E, Elemento O, Sigel C, Jessurun J, and Mosquera JM
- Subjects
- Adult, Humans, Female, In Situ Hybridization, Fluorescence, Bile Ducts, Intrahepatic pathology, Inhibins, Biomarkers, Tumor genetics, Biomarkers, Tumor analysis, Cell Cycle Proteins genetics, Neoplasm Proteins genetics, Repressor Proteins genetics, Carcinoma, Hepatocellular, Cholangiocarcinoma genetics, Cholangiocarcinoma pathology, Liver Neoplasms pathology, Bile Duct Neoplasms pathology
- Abstract
Several reports describing a rare primary liver tumor with histologic features reminiscent of follicular thyroid neoplasms have been published under a variety of descriptive terms including thyroid-like, solid tubulocystic, and cholangioblastic cholangiocarcinoma. Although these tumors are considered to represent histologic variants, they lack classic features of cholangiocarcinoma and have unique characteristics, namely immunoreactivity for inhibin and NIPBL::NACC1 fusions. The purpose of this study is to present clinicopathologic and molecular data for a large series of these tumors to better understand their pathogenesis. We identified 11 hepatic tumors with these features. Immunohistochemical and NACC1 and NIPBL fluorescence in situ hybridization assays were performed on all cases. Four cases had available material for whole-genome sequencing (WGS) analysis. Most patients were adult women (mean age: 42 y) who presented with abdominal pain and large hepatic masses (mean size: 14 cm). Ten patients had no known liver disease. Of the patients with follow-up information, 3/9 (33%) pursued aggressive behavior. All tumors were composed of bland cuboidal cells with follicular and solid/trabecular growth patterns in various combinations, were immunoreactive for inhibin, showed albumin mRNA by in situ hybridization, and harbored the NIPBL::NACC1 fusion by fluorescence in situ hybridization. WGS corroborated the presence of the fusion in all 4 tested cases, high tumor mutational burden in 2 cases, and over 30 structural variants per case in 3 sequenced tumors. The cases lacked mutations typical of conventional intrahepatic cholangiocarcinoma. In this report, we describe the largest series of primary inhibin-positive hepatic neoplasms harboring a NIPBL::NACC1 fusion and the first WGS analysis of these tumors. We propose to name this neoplasm NIPBL:NACC1 fusion hepatic carcinoma., Competing Interests: Conflicts of Interest and Source of Funding: This work was supported by the Englander Institute for Precision Medicine and by the Department of Pathology and Laboratory Medicine at Weill Cornell Medicine. The work at Memorial Sloan Kettering Cancer Center was funded in part by the Marie-Josée and Henry R. Kravis Center for Molecular Oncology, the Cancer Center Support Grant of the National Institutes of Health/National Cancer Institute under award number P30CA008748. M.F.L., J.S.M.M., and E.P. are employees at Isabl, Inc. G.G. is a consultant for Isabl. For the remaining authors none were declared., (Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
20. Deciphering the origin and therapeutic targets of cancer of unknown primary: a case report that illustrates the power of integrative whole-exome and transcriptome sequencing analysis.
- Author
-
Al Assaad M, Shin N, Sigouros M, Manohar J, Antysheva Z, Kotlov N, Kiriy D, Nikitina A, Kleimenov M, Tsareva A, Makarova A, Fomchenkova V, Dubinina J, Boyko A, Almog N, Wilkes D, Escalon JG, Saxena A, Elemento O, Sternberg CN, Nanus DM, and Mosquera JM
- Abstract
Cancer of unknown primary (CUP) represents a significant diagnostic and therapeutic challenge, being the third to fourth leading cause of cancer death, despite advances in diagnostic tools. This article presents a successful approach using a novel genomic analysis in the evaluation and treatment of a CUP patient, leveraging whole-exome sequencing (WES) and RNA sequencing (RNA-seq). The patient, with a history of multiple primary tumors including urothelial cancer, exhibited a history of rapid progression on empirical chemotherapy. The application of our approach identified a molecular target, characterized the tumor expression profile and the tumor microenvironment, and analyzed the origin of the tumor, leading to a tailored treatment. This resulted in a substantial radiological response across all metastatic sites and the predicted primary site of the tumor. We argue that a comprehensive genomic and molecular profiling approach, like the BostonGene
© Tumor Portrait, can provide a more definitive, personalized treatment strategy, overcoming the limitations of current predictive assays. This approach offers a potential solution to an unmet clinical need for a standardized approach in identifying the tumor origin for the effective management of CUP., Competing Interests: NS, ZA, NK, DK, AN, MK, AT, AM, VF, JD, AB, and NA are employees of Bostongene. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Al Assaad, Shin, Sigouros, Manohar, Antysheva, Kotlov, Kiriy, Nikitina, Kleimenov, Tsareva, Makarova, Fomchenkova, Dubinina, Boyko, Almog, Wilkes, Escalon, Saxena, Elemento, Sternberg, Nanus and Mosquera.)- Published
- 2024
- Full Text
- View/download PDF
21. Molecular features of prostate cancer after neoadjuvant therapy in the phase 3 CALGB 90203 trial.
- Author
-
Sumiyoshi T, Wang X, Warner EW, Sboner A, Annala M, Sigouros M, Beja K, Mizuno K, Ku S, Fazli L, Eastham J, Taplin ME, Simko J, Halabi S, Morris MJ, Gleave ME, Wyatt AW, and Beltran H
- Subjects
- Male, Humans, Neoadjuvant Therapy, Docetaxel, Androgen Antagonists therapeutic use, Androgens therapeutic use, Treatment Outcome, Neoplasm Recurrence, Local surgery, Prostate-Specific Antigen, Prostatectomy, Nuclear Proteins, Repressor Proteins, Prostatic Neoplasms drug therapy, Prostatic Neoplasms genetics, Prostatic Neoplasms surgery
- Abstract
Background: The phase 3 CALGB 90203 (Alliance) trial evaluated neoadjuvant chemohormonal therapy for high-risk localized prostate cancer before radical prostatectomy. We dissected the molecular features of post-treated tumors with long-term clinical outcomes to explore mechanisms of response and resistance to chemohormonal therapy., Methods: We evaluated 471 radical prostatectomy tumors, including 294 samples from 166 patients treated with 6 cycles of docetaxel plus androgen deprivation therapy before radical prostatectomy and 177 samples from 97 patients in the control arm (radical prostatectomy alone). Targeted DNA sequencing and RNA expression of tumor foci and adjacent noncancer regions were analyzed in conjunction with pathologic changes and clinical outcomes., Results: Tumor fraction estimated from DNA sequencing was significantly lower in post-treated tumor tissues after chemohormonal therapy compared with controls. Higher tumor fraction after chemohormonal therapy was associated with aggressive pathologic features and poor outcomes, including prostate-specific antigen-progression-free survival. SPOP alterations were infrequently detected after chemohormonal therapy, while TP53 alterations were enriched and associated with shorter overall survival. Residual tumor fraction after chemohormonal therapy was linked to higher expression of androgen receptor-regulated genes, cell cycle genes, and neuroendocrine genes, suggesting persistent populations of active prostate cancer cells. Supervised clustering of post-treated high-tumor-fraction tissues identified a group of patients with elevated cell cycle-related gene expression and poor clinical outcomes., Conclusions: Distinct recurrent prostate cancer genomic and transcriptomic features are observed after exposure to docetaxel and androgen deprivation therapy. Tumor fraction assessed by DNA sequencing quantifies pathologic response and could be a useful trial endpoint or prognostic biomarker. TP53 alterations and high cell cycle transcriptomic activity are linked to aggressive residual disease, despite potent chemohormonal therapy., (© The Author(s) 2023. Published by Oxford University Press. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
22. Author Correction: Evolution of structural rearrangements in prostate cancer intracranial metastases.
- Author
-
Khani F, Hooper WF, Wang X, Chu TR, Shah M, Winterkorn L, Sigouros M, Conteduca V, Pisapia D, Wobker S, Walker S, Graff JN, Robinson B, Mosquera JM, Sboner A, Elemento O, Robine N, and Beltran H
- Published
- 2023
- Full Text
- View/download PDF
23. Whole-genome Analysis Elucidates Complex Genomic Events in GLI1 -rearranged Enteric Tumor.
- Author
-
Mendelson NL, Al Assaad M, Hadi K, Manohar J, Sigouros M, Sboner A, Medina-Martínez JS, Elemento O, Jessurun J, and Mosquera JM
- Subjects
- Humans, Zinc Finger Protein GLI1 genetics, Genomics, Cell Line, Tumor, Neoplasms
- Published
- 2023
- Full Text
- View/download PDF
24. Evolution of structural rearrangements in prostate cancer intracranial metastases.
- Author
-
Khani F, Hooper WF, Wang X, Chu TR, Shah M, Winterkorn L, Sigouros M, Conteduca V, Pisapia D, Wobker S, Walker S, Graff JN, Robinson B, Mosquera JM, Sboner A, Elemento O, Robine N, and Beltran H
- Abstract
Intracranial metastases in prostate cancer are uncommon but clinically aggressive. A detailed molecular characterization of prostate cancer intracranial metastases would improve our understanding of their pathogenesis and the search for new treatment strategies. We evaluated the clinical and molecular characteristics of 36 patients with metastatic prostate cancer to either the dura or brain parenchyma. We performed whole genome sequencing (WGS) of 10 intracranial prostate cancer metastases, as well as WGS of primary prostate tumors from men who later developed metastatic disease (n = 6) and nonbrain prostate cancer metastases (n = 36). This first whole genome sequencing study of prostate intracranial metastases led to several new insights. First, there was a higher diversity of complex structural alterations in prostate cancer intracranial metastases compared to primary tumor tissues. Chromothripsis and chromoplexy events seemed to dominate, yet there were few enrichments of specific categories of structural variants compared with non-brain metastases. Second, aberrations involving the AR gene, including AR enhancer gain were observed in 7/10 (70%) of intracranial metastases, as well as recurrent loss of function aberrations involving TP53 in 8/10 (80%), RB1 in 2/10 (20%), BRCA2 in 2/10 (20%), and activation of the PI3K/AKT/PTEN pathway in 8/10 (80%). These alterations were frequently present in tumor tissues from other sites of disease obtained concurrently or sequentially from the same individuals. Third, clonality analysis points to genomic factors and evolutionary bottlenecks that contribute to metastatic spread in patients with prostate cancer. These results describe the aggressive molecular features underlying intracranial metastasis that may inform future diagnostic and treatment approaches., (© 2023. Nature Publishing Group UK.)
- Published
- 2023
- Full Text
- View/download PDF
25. Long-molecule scars of backup DNA repair in BRCA1- and BRCA2-deficient cancers.
- Author
-
Setton J, Hadi K, Choo ZN, Kuchin KS, Tian H, Da Cruz Paula A, Rosiene J, Selenica P, Behr J, Yao X, Deshpande A, Sigouros M, Manohar J, Nauseef JT, Mosquera JM, Elemento O, Weigelt B, Riaz N, Reis-Filho JS, Powell SN, and Imieliński M
- Subjects
- Humans, Chromosome Inversion, Translocation, Genetic genetics, Homologous Recombination, Cytogenetic Analysis, BRCA1 Protein deficiency, BRCA1 Protein genetics, BRCA2 Protein deficiency, BRCA2 Protein genetics, DNA Repair genetics, Neoplasms genetics, Chromosome Aberrations classification
- Abstract
Homologous recombination (HR) deficiency is associated with DNA rearrangements and cytogenetic aberrations
1 . Paradoxically, the types of DNA rearrangements that are specifically associated with HR-deficient cancers only minimally affect chromosomal structure2 . Here, to address this apparent contradiction, we combined genome-graph analysis of short-read whole-genome sequencing (WGS) profiles across thousands of tumours with deep linked-read WGS of 46 BRCA1- or BRCA2-mutant breast cancers. These data revealed a distinct class of HR-deficiency-enriched rearrangements called reciprocal pairs. Linked-read WGS showed that reciprocal pairs with identical rearrangement orientations gave rise to one of two distinct chromosomal outcomes, distinguishable only with long-molecule data. Whereas one (cis) outcome corresponded to the copying and pasting of a small segment to a distant site, a second (trans) outcome was a quasi-balanced translocation or multi-megabase inversion with substantial (10 kb) duplications at each junction. We propose an HR-independent replication-restart repair mechanism to explain the full spectrum of reciprocal pair outcomes. Linked-read WGS also identified single-strand annealing as a repair pathway that is specific to BRCA2 deficiency in human cancers. Integrating these features in a classifier improved discrimination between BRCA1- and BRCA2-deficient genomes. In conclusion, our data reveal classes of rearrangements that are specific to BRCA1 or BRCA2 deficiency as a source of cytogenetic aberrations in HR-deficient cells., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
26. Immunogenomic Landscape of Neuroendocrine Prostate Cancer.
- Author
-
Bhinder B, Ferguson A, Sigouros M, Uppal M, Elsaeed AG, Bareja R, Alnajar H, Eng KW, Conteduca V, Sboner A, Mosquera JM, Elemento O, and Beltran H
- Subjects
- Male, Humans, Retrospective Studies, Tumor Microenvironment genetics, Prostatic Neoplasms genetics, Prostatic Neoplasms therapy, Prostatic Neoplasms pathology, Neuroendocrine Tumors genetics, Neuroendocrine Tumors therapy, Neuroendocrine Tumors metabolism, Carcinoma, Neuroendocrine pathology
- Abstract
Purpose: Patients with neuroendocrine prostate cancer (NEPC) are often managed with immunotherapy regimens extrapolated from small-cell lung cancer (SCLC). We sought to evaluate the tumor immune landscape of NEPC compared with other prostate cancer types and SCLC., Experimental Design: In this retrospective study, a cohort of 170 patients with 230 RNA-sequencing and 104 matched whole-exome sequencing data were analyzed. Differences in immune and stromal constituents, frequency of genomic alterations, and associations with outcomes were evaluated., Results: In our cohort, 36% of the prostate tumors were identified as CD8+ T-cell inflamed, whereas the remaining 64% were T-cell depleted. T-cell-inflamed tumors were enriched in anti-inflammatory M2 macrophages and exhausted T cells and associated with shorter overall survival relative to T-cell-depleted tumors (HR, 2.62; P < 0.05). Among all prostate cancer types in the cohort, NEPC was identified to be the most immune depleted, wherein only 9 out of the 36 total NEPC tumors were classified as T-cell inflamed. These inflamed NEPC cases were enriched in IFN gamma signaling and PD-1 signaling compared with other NEPC tumors. Comparison of NEPC with SCLC revealed that NEPC had poor immune content and less mutations compared with SCLC, but expression of checkpoint genes PD-L1 and CTLA-4 was comparable between NEPC and SCLC., Conclusions: NEPC is characterized by a relatively immune-depleted tumor immune microenvironment compared with other primary and metastatic prostate adenocarcinoma except in a minority of cases. These findings may inform development of immunotherapy strategies for patients with advanced prostate cancer., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
27. Multicenter phase 2 study of oral azacitidine (CC-486) plus CHOP as initial treatment for PTCL.
- Author
-
Ruan J, Moskowitz A, Mehta-Shah N, Sokol L, Chen Z, Kotlov N, Nos G, Sorokina M, Maksimov V, Sboner A, Sigouros M, van Besien K, Horwitz S, Rutherford SC, Mulvey E, Revuelta MV, Xiang J, Alonso A, Melnick A, Elemento O, Inghirami G, Leonard JP, Cerchietti L, and Martin P
- Subjects
- Humans, Azacitidine adverse effects, Doxorubicin, Prednisone adverse effects, Vincristine, Cyclophosphamide adverse effects, Immunologic Factors therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Tumor Microenvironment, Lymphoma, T-Cell, Peripheral pathology
- Abstract
Peripheral T-cell lymphomas (PTCL) with T-follicular helper phenotype (PTCL-TFH) has recurrent mutations affecting epigenetic regulators, which may contribute to aberrant DNA methylation and chemoresistance. This phase 2 study evaluated oral azacitidine (CC-486) plus cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) as initial treatment for PTCL. CC-486 at 300 mg daily was administered for 7 days before C1 of CHOP, and for 14 days before CHOP C2-6. The primary end point was end-of-treatment complete response (CR). Secondary end points included safety and survival. Correlative studies assessed mutations, gene expression, and methylation in tumor samples. Grade 3 to 4 hematologic toxicities were mostly neutropenia (71%), with febrile neutropenia uncommon (14%). Nonhematologic toxicities included fatigue (14%) and gastrointestinal symptoms (5%). In 20 evaluable patients, CR was 75%, including 88.2% for PTCL-TFH (n = 17). The 2-year progression-free survival (PFS) was 65.8% for all and 69.2% for PTCL-TFH, whereas 2-year overall survival (OS) was 68.4% for all and 76.1% for PTCL-TFH. The frequencies of the TET2, RHOA, DNMT3A, and IDH2 mutations were 76.5%, 41.1%, 23.5%, and 23.5%, respectively, with TET2 mutations significantly associated with CR (P = .007), favorable PFS (P = .004) and OS (P = .015), and DNMT3A mutations associated with adverse PFS (P = .016). CC-486 priming contributed to the reprograming of the tumor microenvironment by upregulation of genes related to apoptosis (P < .01) and inflammation (P < .01). DNA methylation did not show significant shift. This safe and active regimen is being further evaluated in the ALLIANCE randomized study A051902 in CD30-negative PTCL. This trial was registered at www.clinicaltrials.gov as #NCT03542266., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
28. Case report: Metastatic urothelial cancer with an exceptional response to immunotherapy and comprehensive understanding of the tumor and the tumor microenvironment.
- Author
-
Sternberg CN, Shin N, Chernyshov K, Calabro F, Cerbone L, Procopio G, Miheecheva N, Sagaradze G, Zaichikova A, Samarina N, Boyko A, Brown JH, Yunusova L, Guevara D, Manohar J, Sigouros M, Al Assaad M, Elemento O, and Mosquera JM
- Abstract
Although immune checkpoint inhibitors (ICIs) are increasingly used as second-line treatments for urothelial cancer (UC), only a small proportion of patients respond. Therefore, understanding the mechanisms of response to ICIs is critical to improve clinical outcomes for UC patients. The tumor microenvironment (TME) is recognized as a key player in tumor progression and the response to certain anti-cancer treatments. This study aims to investigate the mechanism of response using integrated genomic and transcriptomic profiling of a UC patient who was part of the KEYNOTE-045 trial and showed an exceptional response to pembrolizumab. Diagnosed in 2014 and receiving first-line chemotherapy without success, the patient took part in the KEYNOTE-045 trial for 2 years. She showed dramatic improvement and has now been free of disease for over 6 years. Recently described by Bagaev et al., the Molecular Functional (MF) Portrait was utilized to dissect genomic and transcriptomic features of the patient's tumor and TME. The patient's tumor was characterized as Immune Desert, which is suggestive of a non-inflamed microenvironment. Integrated whole-exome sequencing (WES) and RNA sequencing (RNA-seq) analysis identified an ATM mutation and high TMB level (33.9 mut/mb), which are both positive biomarkers for ICI response. Analysis further revealed the presence of the APOBEC complex, indicating the potential for use of APOBEC signatures as predictive biomarkers for immunotherapy response. Overall, comprehensive characterization of the patient's tumor and TME with the MF Portrait revealed important insights that could potentially be hypothesis generating to identify clinically useful biomarkers and improve treatment for UC patients., Competing Interests: Authors NaiS, KC, NM, GS, AZ, NarS, AB, JB and LY were employed by BostonGene, Corp. CNS has received honoraria from MERCK and participated as an author in the Keynote-045 trial. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The authors declare that this study received funding from BostonGene, Merck and Sharp and Dohme, LLC. Corp. BostonGene had the following involvement with the study: analysis, interpretation of data, and the writing of this article. Merck, Sharp and Dohme, LLC had the following involvement with the study: study design of Keynote 045, sample collection of tissue from San Camillo Hospital, and the decision to submit this article for publication., (Copyright © 2022 Sternberg, Shin, Chernyshov, Calabro, Cerbone, Procopio, Miheecheva, Sagaradze, Zaichikova, Samarina, Boyko, Brown, Yunusova, Guevara, Manohar, Sigouros, Al Assaad, Elemento and Mosquera.)
- Published
- 2022
- Full Text
- View/download PDF
29. Weakly-supervised tumor purity prediction from frozen H&E stained slides.
- Author
-
Brendel M, Getseva V, Assaad MA, Sigouros M, Sigaras A, Kane T, Khosravi P, Mosquera JM, Elemento O, and Hajirasouliha I
- Subjects
- Cohort Studies, Genome, High-Throughput Nucleotide Sequencing, Humans, Male, Prostatic Neoplasms
- Abstract
Background: Estimating tumor purity is especially important in the age of precision medicine. Purity estimates have been shown to be critical for correction of tumor sequencing results, and higher purity samples allow for more accurate interpretations from next-generation sequencing results. Molecular-based purity estimates using computational approaches require sequencing of tumors, which is both time-consuming and expensive., Methods: Here we propose an approach, weakly-supervised purity (wsPurity), which can accurately quantify tumor purity within a digitally captured hematoxylin and eosin (H&E) stained histological slide, using several types of cancer from The Cancer Genome Atlas (TCGA) as a proof-of-concept., Findings: Our model predicts cancer type with high accuracy on unseen cancer slides from TCGA and shows promising generalizability to unseen data from an external cohort (F1-score of 0.83 for prostate adenocarcinoma). In addition we compare performance of our model on tumor purity prediction with a comparable fully-supervised approach on our TCGA held-out cohort and show our model has improved performance, as well as generalizability to unseen frozen slides (0.1543 MAE on an independent test cohort). In addition to tumor purity prediction, our approach identified high resolution tumor regions within a slide, and can also be used to stratify tumors into high and low tumor purity, using different cancer-dependent thresholds., Interpretation: Overall, we demonstrate our deep learning model's different capabilities to analyze tumor H&E sections. We show our model is generalizable to unseen H&E stained slides from data from TCGA as well as data processed at Weill Cornell Medicine., Funding: Starr Cancer Consortium Grant (SCC I15-0027) to Iman Hajirasouliha., (Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
30. Allele-informed copy number evaluation of plasma DNA samples from metastatic prostate cancer patients: the PCF_SELECT consortium assay.
- Author
-
Orlando F, Romanel A, Trujillo B, Sigouros M, Wetterskog D, Quaini O, Leone G, Xiang JZ, Wingate A, Tagawa S, Jayaram A, Linch M, Jamal-Hanjani M, Swanton C, Rubin MA, Wyatt AW, Beltran H, Attard G, and Demichelis F
- Abstract
Sequencing of cell-free DNA (cfDNA) in cancer patients' plasma offers a minimally-invasive solution to detect tumor cell genomic alterations to aid real-time clinical decision-making. The reliability of copy number detection decreases at lower cfDNA tumor fractions, limiting utility at earlier stages of the disease. To test a novel strategy for detection of allelic imbalance, we developed a prostate cancer bespoke assay, PCF_SELECT, that includes an innovative sequencing panel covering ∼25 000 high minor allele frequency SNPs and tailored analytical solutions to enable allele-informed evaluation. First, we assessed it on plasma samples from 50 advanced prostate cancer patients. We then confirmed improved detection of genomic alterations in samples with <10% tumor fractions when compared against an independent assay. Finally, we applied PCF_SELECT to serial plasma samples intensively collected from three patients previously characterized as harboring alterations involving DNA repair genes and consequently offered PARP inhibition. We identified more extensive pan-genome allelic imbalance than previously recognized in prostate cancer. We confirmed high sensitivity detection of BRCA2 allelic imbalance with decreasing tumor fractions resultant from treatment and identified complex ATM genomic states that may be incongruent with protein losses. Overall, we present a framework for sensitive detection of allele-specific copy number changes in cfDNA., (© The Author(s) 2022. Published by Oxford University Press on behalf of NAR Cancer.)
- Published
- 2022
- Full Text
- View/download PDF
31. Tumor-immune microenvironment revealed by Imaging Mass Cytometry in a metastatic sarcomatoid urothelial carcinoma with a prolonged response to pembrolizumab.
- Author
-
Alnajar H, Ravichandran H, Figueiredo Rendeiro A, Ohara K, Al Zoughbi W, Manohar J, Greco N, Sigouros M, Fox J, Muth E, Angiuoli S, Faltas B, Shusterman M, Sternberg CN, Elemento O, and Mosquera JM
- Subjects
- Antibodies, Monoclonal, Humanized, B7-H1 Antigen, Female, Humans, Image Cytometry, Male, Tumor Microenvironment, Antineoplastic Agents, Immunological therapeutic use, Carcinoma, Transitional Cell drug therapy, Carcinoma, Transitional Cell pathology, Sarcoma drug therapy, Soft Tissue Neoplasms, Urinary Bladder Neoplasms drug therapy, Urinary Bladder Neoplasms pathology
- Abstract
Sarcomatoid urothelial carcinoma (SUC) is a rare subtype of urothelial carcinoma (UC) that typically presents at an advanced stage compared to more common variants of UC. Locally advanced and metastatic UC have a poor long-term survival following progression on first-line platinum-based chemotherapy. Antibodies directed against the programmed cell death 1 protein (PD-1) or its ligand (PD-L1) are now approved to be used in these scenarios. The need for reliable biomarkers for treatment stratification is still under research. Here, we present a novel case report of the first Imaging Mass Cytometry (IMC) analysis done in SUC to investigate the immune cell repertoire and PD-L1 expression in a patient who presented with metastatic SUC and experienced a prolonged response to the anti-PD1 immune checkpoint inhibitor pembrolizumab after progression on first-line chemotherapy. This case report provides an important platform for translating these findings to a larger cohort of UC and UC variants., (© 2022 Alnajar et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2022
- Full Text
- View/download PDF
32. Detecting Neuroendocrine Prostate Cancer Through Tissue-Informed Cell-Free DNA Methylation Analysis.
- Author
-
Berchuck JE, Baca SC, McClure HM, Korthauer K, Tsai HK, Nuzzo PV, Kelleher KM, He M, Steinharter JA, Zacharia S, Spisak S, Seo JH, Conteduca V, Elemento O, Auh J, Sigouros M, Corey E, Hirsch MS, Taplin ME, Choueiri TK, Pomerantz MM, Beltran H, and Freedman ML
- Subjects
- DNA Methylation, Humans, Male, Prostate pathology, Carcinoma, Neuroendocrine genetics, Cell-Free Nucleic Acids genetics, Neuroendocrine Tumors pathology, Prostatic Neoplasms diagnosis, Prostatic Neoplasms genetics, Prostatic Neoplasms metabolism
- Abstract
Purpose: Neuroendocrine prostate cancer (NEPC) is a resistance phenotype that emerges in men with metastatic castration-resistant prostate adenocarcinoma (CR-PRAD) and has important clinical implications, but is challenging to detect in practice. Herein, we report a novel tissue-informed epigenetic approach to noninvasively detect NEPC., Experimental Design: We first performed methylated immunoprecipitation and high-throughput sequencing (MeDIP-seq) on a training set of tumors, identified differentially methylated regions between NEPC and CR-PRAD, and built a model to predict the presence of NEPC (termed NEPC Risk Score). We then performed MeDIP-seq on cell-free DNA (cfDNA) from two independent cohorts of men with NEPC or CR-PRAD and assessed the accuracy of the model to predict the presence NEPC., Results: The test cohort comprised cfDNA samples from 48 men, 9 with NEPC and 39 with CR-PRAD. NEPC Risk Scores were significantly higher in men with NEPC than CR-PRAD (P = 4.3 × 10-7) and discriminated between NEPC and CR-PRAD with high accuracy (AUROC 0.96). The optimal NEPC Risk Score cutoff demonstrated 100% sensitivity and 90% specificity for detecting NEPC. The independent, multi-institutional validation cohort included cfDNA from 53 men, including 12 with NEPC and 41 with CR-PRAD. NEPC Risk Scores were significantly higher in men with NEPC than CR-PRAD (P = 7.5×10-12) and perfectly discriminated NEPC from CR-PRAD (AUROC 1.0). Applying the predefined NEPC Risk Score cutoff to the validation cohort resulted in 100% sensitivity and 95% specificity for detecting NEPC., Conclusions: Tissue-informed cfDNA methylation analysis is a promising approach for noninvasive detection of NEPC in men with advanced prostate cancer., (©2021 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
33. Extracellular Matrix in Synthetic Hydrogel-Based Prostate Cancer Organoids Regulate Therapeutic Response to EZH2 and DRD2 Inhibitors.
- Author
-
Mosquera MJ, Kim S, Bareja R, Fang Z, Cai S, Pan H, Asad M, Martin ML, Sigouros M, Rowdo FM, Ackermann S, Capuano J, Bernheim J, Cheung C, Doane A, Brady N, Singh R, Rickman DS, Prabhu V, Allen JE, Puca L, Coskun AF, Rubin MA, Beltran H, Mosquera JM, Elemento O, and Singh A
- Subjects
- Androgen Receptor Antagonists pharmacology, Androgen Receptor Antagonists therapeutic use, Animals, Cell Line, Tumor, Enhancer of Zeste Homolog 2 Protein, Extracellular Matrix metabolism, Humans, Hydrogels pharmacology, Hydrogels therapeutic use, Male, Mice, Receptors, Dopamine D2 genetics, Receptors, Dopamine D2 therapeutic use, Organoids metabolism, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant metabolism, Prostatic Neoplasms, Castration-Resistant pathology
- Abstract
Following treatment with androgen receptor (AR) pathway inhibitors, ≈20% of prostate cancer patients progress by shedding their AR-dependence. These tumors undergo epigenetic reprogramming turning castration-resistant prostate cancer adenocarcinoma (CRPC-Adeno) into neuroendocrine prostate cancer (CRPC-NEPC). No targeted therapies are available for CRPC-NEPCs, and there are minimal organoid models to discover new therapeutic targets against these aggressive tumors. Here, using a combination of patient tumor proteomics, RNA sequencing, spatial-omics, and a synthetic hydrogel-based organoid, putative extracellular matrix (ECM) cues that regulate the phenotypic, transcriptomic, and epigenetic underpinnings of CRPC-NEPCs are defined. Short-term culture in tumor-expressed ECM differentially regulated DNA methylation and mobilized genes in CRPC-NEPCs. The ECM type distinctly regulates the response to small-molecule inhibitors of epigenetic targets and Dopamine Receptor D2 (DRD2), the latter being an understudied target in neuroendocrine tumors. In vivo patient-derived xenograft in immunocompromised mice showed strong anti-tumor response when treated with a DRD2 inhibitor. Finally, we demonstrate that therapeutic response in CRPC-NEPCs under drug-resistant ECM conditions can be overcome by first cellular reprogramming with epigenetic inhibitors, followed by DRD2 treatment. The synthetic organoids suggest the regulatory role of ECM in therapeutic response to targeted therapies in CRPC-NEPCs and enable the discovery of therapies to overcome resistance., (© 2021 Wiley-VCH GmbH.)
- Published
- 2022
- Full Text
- View/download PDF
34. Validation of a Circulating Tumor DNA-Based Next-Generation Sequencing Assay in a Cohort of Patients with Solid tumors: A Proposed Solution for Decentralized Plasma Testing.
- Author
-
Al Zoughbi W, Fox J, Beg S, Papp E, Hissong E, Ohara K, Keefer L, Sigouros M, Kane T, Bockelman D, Nichol D, Patchell E, Bareja R, Karandikar A, Alnajar H, Cerqueira G, Guthrie VB, Verner E, Manohar J, Greco N, Wilkes D, Tagawa S, Malbari MS, Holcomb K, Eng KW, Shah M, Altorki NK, Sboner A, Nanus D, Faltas B, Sternberg CN, Simmons J, Houvras Y, Molina AM, Angiuoli S, Elemento O, and Mosquera JM
- Subjects
- High-Throughput Nucleotide Sequencing, Humans, Microsatellite Instability, Retrospective Studies, Circulating Tumor DNA genetics, Neoplasms genetics
- Abstract
Background: Characterization of circulating tumor DNA (ctDNA) has been integrated into clinical practice. Although labs have standardized validation procedures to develop single locus tests, the efficacy of on-site plasma-based next-generation sequencing (NGS) assays still needs to be proved., Materials and Methods: In this retrospective study, we profiled DNA from matched tissue and plasma samples from 75 patients with cancer. We applied an NGS test that detects clinically relevant alterations in 33 genes and microsatellite instability (MSI) to analyze plasma cell-free DNA (cfDNA)., Results: The concordance between alterations detected in both tissue and plasma samples was higher in patients with metastatic disease. The NGS test detected 77% of sequence alterations, amplifications, and fusions that were found in metastatic samples compared with 45% of those alterations found in the primary tumor samples (p = .00005). There was 87% agreement on MSI status between the NGS test and tumor tissue results. In three patients, MSI-high ctDNA correlated with response to immunotherapy. In addition, the NGS test revealed an FGFR2 amplification that was not detected in tumor tissue from a patient with metastatic gastric cancer, emphasizing the importance of profiling plasma samples in patients with advanced cancer., Conclusion: Our validation experience of a plasma-based NGS assay advances current knowledge about translating cfDNA testing into clinical practice and supports the application of plasma assays in the management of oncology patients with metastatic disease. With an in-house method that minimizes the need for invasive procedures, on-site cfDNA testing supplements tissue biopsy to guide precision therapy and is entitled to become a routine practice., Implications for Practice: This study proposes a solution for decentralized liquid biopsy testing based on validation of a next-generation sequencing (NGS) test that detects four classes of genomic alterations in blood: sequence mutations (single nucleotide substitutions or insertions and deletions), fusions, amplifications, and microsatellite instability (MSI). Although there are reference labs that perform single-site comprehensive liquid biopsy testing, the targeted assay this study validated can be established locally in any lab with capacity to offer clinical molecular pathology assays. To the authors' knowledge, this is the first report that validates evaluating an on-site plasma-based NGS test that detects the MSI status along with common sequence alterations encountered in solid tumors., (© 2021 AlphaMed Press.)
- Published
- 2021
- Full Text
- View/download PDF
35. Author Correction: Circulating tumor cell heterogeneity in neuroendocrine prostate cancer by single cell copy number analysis.
- Author
-
Conteduca V, Ku SY, Fernandez L, Dago-Rodriquez A, Lee J, Jendrisak A, Slade M, Gilbertson C, Manohar J, Sigouros M, Wang Y, Dittamore R, Wenstrup R, Mosquera JM, Schonhoft JD, and Beltran H
- Published
- 2021
- Full Text
- View/download PDF
36. Circulating tumor cell heterogeneity in neuroendocrine prostate cancer by single cell copy number analysis.
- Author
-
Conteduca V, Ku SY, Fernandez L, Dago-Rodriquez A, Lee J, Jendrisak A, Slade M, Gilbertson C, Manohar J, Sigouros M, Wang Y, Dittamore R, Wenstrup R, Mosquera JM, Schonhoft JD, and Beltran H
- Abstract
Neuroendocrine prostate cancer is an aggressive variant of prostate cancer that may arise de novo or develop from pre-existing prostate adenocarcinoma as a mechanism of treatment resistance. The combined loss of tumor suppressors RB1, TP53, and PTEN are frequent in NEPC but also present in a subset of prostate adenocarcinomas. Most clinical and preclinical studies support a trans-differentiation process, whereby NEPC arises clonally from a prostate adenocarcinoma precursor during the course of treatment resistance. Here we highlight a case of NEPC with significant intra-patient heterogeneity observed across metastases. We further demonstrate how single-cell genomic analysis of circulating tumor cells combined with a phenotypic evaluation of cellular diversity can be considered as a window into tumor heterogeneity in patients with advanced prostate cancer., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
37. Functional comparison of exome capture-based methods for transcriptomic profiling of formalin-fixed paraffin-embedded tumors.
- Author
-
Shohdy KS, Bareja R, Sigouros M, Wilkes DC, Dorsaint P, Manohar J, Bockelman D, Xiang JZ, Kim R, Ohara K, Eng K, Mosquera JM, Elemento O, Sboner A, Alonso A, and Faltas BM
- Abstract
The availability of fresh frozen (FF) tissue is a barrier for implementing RNA sequencing (RNA-seq) in the clinic. The majority of clinical samples are stored as formalin-fixed, paraffin-embedded (FFPE) tissues. Exome capture platforms have been developed for RNA-seq from FFPE samples. However, these methods have not been systematically compared. We performed transcriptomic analysis of 32 FFPE tumor samples from 11 patients using three exome capture-based methods: Agilent SureSelect V6, TWIST NGS Exome, and IDT XGen Exome Research Panel. We compared these methods to the TruSeq RNA-seq of fresh frozen (FF-TruSeq) tumor samples from the same patients. We assessed the recovery of clinically relevant biological features. The Spearman's correlation coefficients between the global expression profiles of the three capture-based methods from FFPE and matched FF-TruSeq were high (rho = 0.72-0.9, p < 0.05). A significant correlation between the expression of key immune genes between individual capture-based methods and FF-TruSeq (rho = 0.76-0.88, p < 0.05) was observed. All exome capture-based methods reliably detected outlier expression of actionable gene transcripts, including ERBB2, MET, NTRK1, and PPARG. In urothelial cancer samples, the Agilent assay was associated with the highest molecular subtype concordance with FF-TruSeq (Cohen's k = 0.7, p < 0.01). The Agilent and IDT assays detected all the clinically relevant fusions that were initially identified in FF-TruSeq. All FFPE exome capture-based methods had comparable performance and concordance with FF-TruSeq. Our findings will enable the implementation of RNA-seq in the clinic to guide precision oncology approaches., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
38. Androgen receptor variant shows heterogeneous expression in prostate cancer according to differentiation stage.
- Author
-
Gjyrezi A, Galletti G, Zhang J, Worroll D, Sigouros M, Kim S, Cooley V, Ballman KV, Ocean AJ, Shah MA, Scandura JM, Sboner A, Nanus DM, Beltran H, Tagawa S, and Giannakakou P
- Subjects
- Cell Line, Tumor, Humans, Male, Neoplasm Staging, Prostatic Neoplasms chemistry, RNA-Seq, Receptors, Androgen analysis, Receptors, Androgen physiology, Neoplastic Cells, Circulating chemistry, Prostatic Neoplasms pathology, Receptors, Androgen genetics
- Abstract
Quantitation of androgen receptor variant (AR-V) expression in circulating tumor cells (CTCs) from patients with metastatic castration-resistant prostate cancer (mCRPC) has great potential for treatment customization. However, the absence of a uniform CTC isolation platform and consensus on an analytical assay has prevented the incorporation of these measurements in routine clinical practice. Here, we present a single-CTC sensitive digital droplet PCR (ddPCR) assay for the quantitation of the two most common AR-Vs, AR-V7, and AR-v567es, using antigen agnostic CTC enrichment. In a cohort of 29 mCRPC patients, we identify AR-V7 in 66% and AR-v567es in 52% of patients. These results are corroborated using another gene expression platform (NanoString
TM ) and by analysis of RNA-Seq data from patients with mCRPC (SU2C- PCF Dream Team). We next quantify AR-V expression in matching EpCAM-positive vs EpCAM-negative CTCs, as EpCAM-based CTC enrichment is commonly used. We identify lower AR-V prevalence in the EpCAM-positive fraction, suggesting that EpCAM-based CTC enrichment likely underestimates AR-V prevalence. Lastly, using single CTC analysis we identify enrichment for AR-v567es in patients with neuroendocrine prostate cancer (NEPC) indicating that AR-v567es may be involved in lineage plasticity, which warrants further mechanistic interrogation.- Published
- 2021
- Full Text
- View/download PDF
39. Temporal evolution of cellular heterogeneity during the progression to advanced AR-negative prostate cancer.
- Author
-
Brady NJ, Bagadion AM, Singh R, Conteduca V, Van Emmenis L, Arceci E, Pakula H, Carelli R, Khani F, Bakht M, Sigouros M, Bareja R, Sboner A, Elemento O, Tagawa S, Nanus DM, Loda M, Beltran H, Robinson B, and Rickman DS
- Subjects
- Adenocarcinoma metabolism, Animals, Carcinoma, Neuroendocrine metabolism, Cell Line, Tumor, Disease Progression, Humans, Male, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, N-Myc Proto-Oncogene Protein genetics, N-Myc Proto-Oncogene Protein metabolism, Organ Culture Techniques methods, Prognosis, Prostate pathology, Prostatic Neoplasms metabolism, Receptors, Androgen metabolism, Retinoblastoma Protein genetics, Retinoblastoma Protein metabolism, Mice, Adenocarcinoma genetics, Carcinoma, Neuroendocrine genetics, Gene Expression Regulation, Neoplastic, Prostate metabolism, Prostatic Neoplasms genetics, Receptors, Androgen genetics
- Abstract
Despite advances in the development of highly effective androgen receptor (AR)-directed therapies for the treatment of men with advanced prostate cancer, acquired resistance to such therapies frequently ensues. A significant subset of patients with resistant disease develop AR-negative tumors that lose their luminal identity and display neuroendocrine features (neuroendocrine prostate cancer (NEPC)). The cellular heterogeneity and the molecular evolution during the progression from AR-positive adenocarcinoma to AR-negative NEPC has yet to be characterized. Utilizing a new genetically engineered mouse model, we have characterized the synergy between Rb1 loss and MYCN (encodes N-Myc) overexpression which results in the formation of AR-negative, poorly differentiated tumors with high metastatic potential. Single-cell-based approaches revealed striking temporal changes to the transcriptome and chromatin accessibility which have identified the emergence of distinct cell populations, marked by differential expression of Ascl1 and Pou2f3, during the transition to NEPC. Moreover, global DNA methylation and the N-Myc cistrome are redirected following Rb1 loss. Altogether, our data provide insight into the progression of prostate adenocarcinoma to NEPC.
- Published
- 2021
- Full Text
- View/download PDF
40. Integration of whole-exome and anchored PCR-based next generation sequencing significantly increases detection of actionable alterations in precision oncology.
- Author
-
Beg S, Bareja R, Ohara K, Eng KW, Wilkes DC, Pisapia DJ, Zoughbi WA, Kudman S, Zhang W, Rao R, Manohar J, Kane T, Sigouros M, Xiang JZ, Khani F, Robinson BD, Faltas BM, Sternberg CN, Sboner A, Beltran H, Elemento O, and Mosquera JM
- Abstract
Background: Frequency of clinically relevant mutations in solid tumors by targeted and whole-exome sequencing is ∼30%. Transcriptome analysis complements detection of actionable gene fusions in advanced cancer patients. Goal of this study was to determine the added value of anchored multiplex PCR (AMP)-based next-generation sequencing (NGS) assay to identify further potential drug targets, when coupled with whole-exome sequencing (WES)., Methods: Selected series of fifty-six samples from 55 patients enrolled in our precision medicine study were interrogated by WES and AMP-based NGS. RNA-seq was performed in 19 cases. Clinically relevant and actionable alterations detected by three methods were integrated and analyzed., Results: AMP-based NGS detected 48 fusions in 31 samples (55.4%); 31.25% (15/48) were classified as targetable based on published literature. WES revealed 29 samples (51.8%) harbored targetable alterations. TMB-high and MSI-high status were observed in 12.7% and 1.8% of cases. RNA-seq from 19 samples identified 8 targetable fusions (42.1%), also captured by AMP-based NGS. When number of actionable fusions detected by AMP-based NGS were added to WES targetable alterations, 66.1% of samples had potential drug targets. When both WES and RNA-seq were analyzed, 57.8% of samples had targetable alterations., Conclusions: This study highlights importance of an integrative genomic approach for precision oncology, including use of different NGS platforms with complementary features. Integrating RNA data (whole transcriptome or AMP-based NGS) significantly enhances detection of potential targets in cancer patients. In absence of fresh frozen tissue, AMP-based NGS is a robust method to detect actionable fusions using low-input RNA from archival tissue., Competing Interests: Declaration of Competing Interest The authors declare no conflict of interest., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
41. Distinct Classes of Complex Structural Variation Uncovered across Thousands of Cancer Genome Graphs.
- Author
-
Hadi K, Yao X, Behr JM, Deshpande A, Xanthopoulakis C, Tian H, Kudman S, Rosiene J, Darmofal M, DeRose J, Mortensen R, Adney EM, Shaiber A, Gajic Z, Sigouros M, Eng K, Wala JA, Wrzeszczyński KO, Arora K, Shah M, Emde AK, Felice V, Frank MO, Darnell RB, Ghandi M, Huang F, Dewhurst S, Maciejowski J, de Lange T, Setton J, Riaz N, Reis-Filho JS, Powell S, Knowles DA, Reznik E, Mishra B, Beroukhim R, Zody MC, Robine N, Oman KM, Sanchez CA, Kuhner MK, Smith LP, Galipeau PC, Paulson TG, Reid BJ, Li X, Wilkes D, Sboner A, Mosquera JM, Elemento O, and Imielinski M
- Subjects
- Chromosome Inversion genetics, Chromothripsis, DNA Copy Number Variations genetics, Gene Rearrangement genetics, Genome, Human genetics, Humans, Mutation genetics, Whole Genome Sequencing methods, Genomic Structural Variation genetics, Genomics methods, Neoplasms genetics
- Abstract
Cancer genomes often harbor hundreds of somatic DNA rearrangement junctions, many of which cannot be easily classified into simple (e.g., deletion) or complex (e.g., chromothripsis) structural variant classes. Applying a novel genome graph computational paradigm to analyze the topology of junction copy number (JCN) across 2,778 tumor whole-genome sequences, we uncovered three novel complex rearrangement phenomena: pyrgo, rigma, and tyfonas. Pyrgo are "towers" of low-JCN duplications associated with early-replicating regions, superenhancers, and breast or ovarian cancers. Rigma comprise "chasms" of low-JCN deletions enriched in late-replicating fragile sites and gastrointestinal carcinomas. Tyfonas are "typhoons" of high-JCN junctions and fold-back inversions associated with expressed protein-coding fusions, breakend hypermutation, and acral, but not cutaneous, melanomas. Clustering of tumors according to genome graph-derived features identified subgroups associated with DNA repair defects and poor prognosis., Competing Interests: Declaration of Interests J.S.R.-F. reports receiving personal/consultancy fees from VolitionRx, Paige.AI, Goldman Sachs, REPARE Therapeutics, GRAIL, Ventana Medical Systems, Roche, Genentech, and InviCRO outside of the scope of the submitted work., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
42. SLFN11 Expression in Advanced Prostate Cancer and Response to Platinum-based Chemotherapy.
- Author
-
Conteduca V, Ku SY, Puca L, Slade M, Fernandez L, Hess J, Bareja R, Vlachostergios PJ, Sigouros M, Mosquera JM, Sboner A, Nanus DM, Elemento O, Dittamore R, Tagawa ST, and Beltran H
- Subjects
- Aged, Aged, 80 and over, Antineoplastic Agents, Biomarkers, Tumor genetics, Bone Neoplasms drug therapy, Bone Neoplasms metabolism, Bone Neoplasms secondary, Follow-Up Studies, Humans, Male, Middle Aged, Nuclear Proteins genetics, Prognosis, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant metabolism, Retrospective Studies, Survival Rate, Tumor Cells, Cultured, Biomarkers, Tumor metabolism, Cisplatin pharmacology, Gene Expression Regulation, Neoplastic drug effects, Nuclear Proteins metabolism, Prostatic Neoplasms, Castration-Resistant pathology
- Abstract
Expression of the DNA/RNA helicase schlafen family member 11 (SLFN11) has been identified as a sensitizer of tumor cells to DNA-damaging agents including platinum chemotherapy. We assessed the impact of SLFN11 expression on response to platinum chemotherapy and outcomes in patients with metastatic castration-resistant prostate cancer (CRPC). Tumor expression of SLFN11 was assessed in 41 patients with CRPC treated with platinum chemotherapy by RNA sequencing (RNA-seq) of metastatic biopsy tissue ( n = 27) and/or immunofluorescence in circulating tumor cells (CTC; n = 20). Cox regression and Kaplan-Meier methods were used to evaluate the association of SLFN11 expression with radiographic progression-free survival (rPFS) and overall survival (OS). Multivariate analysis included tumor histology (i.e., adenocarcinoma or neuroendocrine) and the presence or absence of DNA repair aberrations. Patient-derived organoids with SLFN11 expression and after knockout by CRISPR-Cas9 were treated with platinum and assessed for changes in dose response. Patients were treated with platinum combination ( N = 38) or platinum monotherapy ( N = 3). Median lines of prior therapy for CRPC was two. Median OS was 8.7 months. Overexpression of SLFN11 in metastatic tumors by RNA-seq was associated with longer rPFS compared with those without overexpression (6.9 vs. 2.8 months, HR = 3.72; 95% confidence interval (CI), 1.56-8.87; P < 0.001); similar results were observed for patients with SLFN11-positive versus SLFN11-negative CTCs (rPFS 6.0 vs. 2.2 months, HR = 4.02; 95% CI, 0.77-20.86; P = 0.002). A prostate-specific antigen (PSA) decline of ≥50% was observed in all patients with SLFN11 overexpression. No association was observed between SLFN11 expression and OS. On multivariable analysis, SLFN11 was an independent factor associated with rPFS on platinum therapy. Platinum response of organoids expressing SLFN11 was reduced after SLFN11 knockout. Our data suggest that SLFN11 expression might identify patients with CRPC with a better response to platinum chemotherapy independent of histology or other genomic alterations. Additional studies, also in the context of PARP inhibitors, are warranted., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
43. Circulating tumor DNA profile recognizes transformation to castration-resistant neuroendocrine prostate cancer.
- Author
-
Beltran H, Romanel A, Conteduca V, Casiraghi N, Sigouros M, Franceschini GM, Orlando F, Fedrizzi T, Ku SY, Dann E, Alonso A, Mosquera JM, Sboner A, Xiang J, Elemento O, Nanus DM, Tagawa ST, Benelli M, and Demichelis F
- Subjects
- Gene Expression Profiling, Humans, Male, Prospective Studies, Adenocarcinoma blood, Adenocarcinoma genetics, Carcinoma, Neuroendocrine blood, Carcinoma, Neuroendocrine genetics, Circulating Tumor DNA blood, Circulating Tumor DNA genetics, DNA Methylation, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Neoplasm Proteins blood, Neoplasm Proteins genetics, Prostatic Neoplasms, Castration-Resistant blood, Prostatic Neoplasms, Castration-Resistant genetics
- Abstract
Loss of androgen receptor (AR) signaling dependence occurs in approximately 15%-20% of advanced treatment-resistant prostate cancers, and this may manifest clinically as transformation from a prostate adenocarcinoma histology to a castration-resistant neuroendocrine prostate cancer (CRPC-NE). The diagnosis of CRPC-NE currently relies on a metastatic tumor biopsy, which is invasive for patients and sometimes challenging to diagnose due to morphologic heterogeneity. By studying whole-exome sequencing and whole-genome bisulfite sequencing of cell free DNA (cfDNA) and of matched metastatic tumor biopsies from patients with metastatic prostate adenocarcinoma and CRPC-NE, we identified CRPC-NE features detectable in the circulation. Overall, there was markedly higher concordance between cfDNA and biopsy tissue genomic alterations in patients with CRPC-NE compared with castration-resistant adenocarcinoma, supporting greater intraindividual genomic consistency across metastases. Allele-specific copy number and serial sampling analyses allowed for the detection and tracking of clonal and subclonal tumor cell populations. cfDNA methylation was indicative of circulating tumor content fraction, reflective of methylation patterns observed in biopsy tissues, and was capable of detecting CRPC-NE-associated epigenetic changes (e.g., hypermethylation of ASXL3 and SPDEF; hypomethylation of INSM1 and CDH2). A targeted set combining genomic (TP53, RB1, CYLD, AR) and epigenomic (hypo- and hypermethylation of 20 differential sites) alterations applied to ctDNA was capable of identifying patients with CRPC-NE.
- Published
- 2020
- Full Text
- View/download PDF
44. Androgen deprivation upregulates SPINK1 expression and potentiates cellular plasticity in prostate cancer.
- Author
-
Tiwari R, Manzar N, Bhatia V, Yadav A, Nengroo MA, Datta D, Carskadon S, Gupta N, Sigouros M, Khani F, Poutanen M, Zoubeidi A, Beltran H, Palanisamy N, and Ateeq B
- Subjects
- Androgen Receptor Antagonists therapeutic use, Animals, Casein Kinase I antagonists & inhibitors, Casein Kinase I metabolism, Cell Line, Tumor, Co-Repressor Proteins metabolism, HEK293 Cells, Humans, Male, Mice, Nerve Tissue Proteins metabolism, Neuroendocrine Tumors drug therapy, Neuroendocrine Tumors pathology, Prostate drug effects, Prostate pathology, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology, Receptors, Androgen metabolism, SOXB1 Transcription Factors metabolism, Transcription, Genetic drug effects, Up-Regulation drug effects, Xenograft Model Antitumor Assays, Androgen Receptor Antagonists pharmacology, Gene Expression Regulation, Neoplastic drug effects, Neuroendocrine Tumors genetics, Prostatic Neoplasms genetics, Trypsin Inhibitor, Kazal Pancreatic metabolism
- Abstract
Emergence of an aggressive androgen receptor (AR)-independent neuroendocrine prostate cancer (NEPC) after androgen-deprivation therapy (ADT) is well-known. Nevertheless, the majority of advanced-stage prostate cancer patients, including those with SPINK1-positive subtype, are treated with AR-antagonists. Here, we show AR and its corepressor, REST, function as transcriptional-repressors of SPINK1, and AR-antagonists alleviate this repression leading to SPINK1 upregulation. Increased SOX2 expression during NE-transdifferentiation transactivates SPINK1, a critical-player for maintenance of NE-phenotype. SPINK1 elicits epithelial-mesenchymal-transition, stemness and cellular-plasticity. Conversely, pharmacological Casein Kinase-1 inhibition stabilizes REST, which in cooperation with AR causes SPINK1 transcriptional-repression and impedes SPINK1-mediated oncogenesis. Elevated levels of SPINK1 and NEPC markers are observed in the tumors of AR-antagonists treated mice, and in a subset of NEPC patients, implicating a plausible role of SPINK1 in treatment-related NEPC. Collectively, our findings provide an explanation for the paradoxical clinical-outcomes after ADT, possibly due to SPINK1 upregulation, and offers a strategy for adjuvant therapies.
- Published
- 2020
- Full Text
- View/download PDF
45. PARP Inhibition Suppresses GR-MYCN-CDK5-RB1-E2F1 Signaling and Neuroendocrine Differentiation in Castration-Resistant Prostate Cancer.
- Author
-
Liu B, Li L, Yang G, Geng C, Luo Y, Wu W, Manyam GC, Korentzelos D, Park S, Tang Z, Wu C, Dong Z, Sigouros M, Sboner A, Beltran H, Chen Y, Corn PG, Tetzlaff MT, Troncoso P, Broom B, and Thompson TC
- Subjects
- Animals, Benzamides, Bridged Bicyclo Compounds, Heterocyclic administration & dosage, Cell Line, Tumor, Cyclic N-Oxides, Cyclin-Dependent Kinase 5 genetics, Cyclin-Dependent Kinase 5 metabolism, E2F1 Transcription Factor genetics, E2F1 Transcription Factor metabolism, Gene Expression Regulation, Neoplastic drug effects, Humans, Indolizines, Male, Mice, Nude, N-Myc Proto-Oncogene Protein genetics, N-Myc Proto-Oncogene Protein metabolism, Neuroendocrine Tumors genetics, Neuroendocrine Tumors metabolism, Nitriles, Phenylthiohydantoin administration & dosage, Phenylthiohydantoin analogs & derivatives, Phthalazines administration & dosage, Piperazines administration & dosage, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant metabolism, Proteins metabolism, Pyridinium Compounds administration & dosage, Receptors, Glucocorticoid genetics, Receptors, Glucocorticoid metabolism, Retinoblastoma Binding Proteins genetics, Retinoblastoma Binding Proteins metabolism, Signal Transduction drug effects, Signal Transduction genetics, Treatment Outcome, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Xenograft Model Antitumor Assays methods, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Neuroendocrine Tumors drug therapy, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Prostatic Neoplasms, Castration-Resistant drug therapy, Proteins genetics
- Abstract
Purpose: In this study, we addressed the underlying mechanisms for the association between enzalutamide (ENZ) treatment and neuroendocrine prostate cancer (NEPC), and the critical involvement of MYCN, and loss of RB1 function in neuroendocrine differentiation (NED) of prostatic epithelial cells, and the development of NEPC. We further sought to determine whether PARP inhibition could suppress NEPC, and to identify molecular determinants of this therapeutic activity., Experimental Design: We used a novel prostate cancer patient-derived xenograft (PDX) treatment model, prostatic adenocarcinoma and NEPC cell lines, an NEPC organoid line, and NEPC xenograft models to address the mechanistic basis of ENZ-induced NED, and to analyze suppression of NED and NEPC growth by PARP inhibition., Results: We identified an ENZ treatment-associated glucocorticoid receptor (GR)-MYCN-CDK5-RB1-E2F1 signaling pathway that drives NED in prostatic adenocarcinoma PDX and cell line models. Mechanistically, long-term ENZ treatment transcriptionally upregulates signaling of the GR-MYCN axis, leading to CDK5R1 and CDK5R2 upregulation, Rb1 phosphorylation, and N-Myc-mediated and E2F1-mediated NED gene expression. Importantly, olaparib (OLA) or talazoparib (TALA) suppressed these activities, and the combination of OLA and dinaciclib (DINA), an inhibitor of CDK2 and CDK5, which also inhibits Rb1 phosphorylation, suppressed NED and significantly improved therapeutic efficiency in NEPC cells in vitro and in NEPC tumors in vivo ., Conclusions: The results of our study indicate an important role of GR-MYCN-CDK5R1/2-RB1-NED signaling in ENZ-induced and PARP inhibitor-suppressed NEPC. We also demonstrated efficacy for OLA+DINA combination therapy in NEPC xenograft models., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
46. Clinical features of neuroendocrine prostate cancer.
- Author
-
Conteduca V, Oromendia C, Eng KW, Bareja R, Sigouros M, Molina A, Faltas BM, Sboner A, Mosquera JM, Elemento O, Nanus DM, Tagawa ST, Ballman KV, and Beltran H
- Subjects
- Adenocarcinoma mortality, Adenocarcinoma pathology, Aged, Biopsy, Carcinoma, Small Cell mortality, Carcinoma, Small Cell pathology, Cohort Studies, Disease Progression, Humans, Male, Middle Aged, Neoplasm Metastasis, Prognosis, Retrospective Studies, Survival Analysis, Treatment Outcome, Carcinoma, Neuroendocrine diagnosis, Carcinoma, Neuroendocrine mortality, Carcinoma, Neuroendocrine pathology, Carcinoma, Neuroendocrine therapy, Prostatic Neoplasms diagnosis, Prostatic Neoplasms mortality, Prostatic Neoplasms pathology, Prostatic Neoplasms therapy
- Abstract
Background: Neuroendocrine prostate cancer (NEPC) is an aggressive variant of prostate cancer that may arise de novo or in patients previously treated with hormonal therapies for prostate adenocarcinoma as a mechanism of resistance. Despite being important to recognise, the clinical features of NEPC are poorly defined and could help guide when to perform a biopsy to look for NEPC histologic transformation., Methods: We reviewed baseline, treatment and outcome data of 87 patients with metastatic prostate cancer and tumour biopsy confirming NEPC histology. Forty-seven (54.0%) NEPC cases presented de novo, and 40 (46.0%) were therapy-related (t-NEPC). Thirty-six (41.4%) were classified as pure small-cell carcinoma, and 51 (58.6%) demonstrated mixed features with both small-cell carcinoma and adenocarcinoma present. Genomic data were available for 47 patients., Results: The median age at time of NEPC was 68.1 years, median prostate-specific antigen (PSA) was 1.20 ng/ml (0.14 ng/mL small-cell carcinoma, 1.55 ng/mL mixed carcinoma) and sites of metastases included bone (72.6%), lymph node (47.0%), and viscera (65.5%). Median time from adenocarcinoma to t-NEPC diagnosis was 39.7 months (range, 24.5-93.8) with a median of two lines of prior systemic therapy. Platinum chemotherapy was used to treat 57.5% of patients, with a median progression-free survival of 3.9 months. Small-cell carcinoma was associated with worse overall survival (OS) than mixed histology (8.9 months from NEPC diagnosis versus 26.1 months, P < 0.001). Median OS of de novo NEPC was shorter than that of t-NEPC (16.8 months from prostate cancer diagnosis versus 53.5 months, P = 0.043). An average PSA rise per month of ≤0.7 ng/ml before t-NEPC; elevated lactate dehydrogenase levels, RB1 and TP53 loss and liver metastases were poor prognostic features., Conclusions: We describe the clinical features of a cohort of patients with NEPC. These characteristics may inform future diagnostic strategies., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
47. Delta-like protein 3 expression and therapeutic targeting in neuroendocrine prostate cancer.
- Author
-
Puca L, Gavyert K, Sailer V, Conteduca V, Dardenne E, Sigouros M, Isse K, Kearney M, Vosoughi A, Fernandez L, Pan H, Motanagh S, Hess J, Donoghue AJ, Sboner A, Wang Y, Dittamore R, Rickman D, Nanus DM, Tagawa ST, Elemento O, Mosquera JM, Saunders L, and Beltran H
- Subjects
- Aged, Animals, Antibodies, Monoclonal, Humanized pharmacology, Antibodies, Monoclonal, Humanized therapeutic use, Benzodiazepinones pharmacology, Benzodiazepinones therapeutic use, Carcinoma, Neuroendocrine pathology, Cell Line, Tumor, Gene Expression Regulation, Neoplastic drug effects, Genetic Heterogeneity, Humans, Immunoconjugates pharmacology, Immunoconjugates therapeutic use, Intracellular Signaling Peptides and Proteins metabolism, Male, Membrane Proteins metabolism, Mice, Neoplastic Cells, Circulating metabolism, Neoplastic Cells, Circulating pathology, Prostatic Neoplasms pathology, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant pathology, Time Factors, Treatment Outcome, Carcinoma, Neuroendocrine drug therapy, Carcinoma, Neuroendocrine genetics, Intracellular Signaling Peptides and Proteins genetics, Membrane Proteins genetics, Molecular Targeted Therapy, Prostatic Neoplasms drug therapy, Prostatic Neoplasms genetics
- Abstract
Histologic transformation to small cell neuroendocrine prostate cancer occurs in a subset of patients with advanced prostate cancer as a mechanism of treatment resistance. Rovalpituzumab tesirine (SC16LD6.5) is an antibody-drug conjugate that targets delta-like protein 3 (DLL3) and was initially developed for small cell lung cancer. We found that DLL3 is expressed in most of the castration-resistant neuroendocrine prostate cancer (CRPC-NE) (36 of 47, 76.6%) and in a subset of castration-resistant prostate adenocarcinomas (7 of 56, 12.5%). It shows minimal to no expression in localized prostate cancer (1 of 194) and benign prostate (0 of 103). DLL3 expression correlates with neuroendocrine marker expression, RB1 loss, and aggressive clinical features. DLL3 in circulating tumor cells was concordant with matched metastatic biopsy (87%). Treatment of DLL3-expressing prostate cancer xenografts with a single dose of SC16LD6.5 resulted in complete and durable responses, whereas DLL3-negative models were insensitive. We highlight a patient with neuroendocrine prostate cancer with a meaningful clinical and radiologic response to SC16LD6.5 when treated on a phase 1 trial. Overall, our findings indicate that DLL3 is preferentially expressed in CRPC-NE and provide rationale for targeting DLL3 in patients with DLL3-positive metastatic prostate cancer., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2019
- Full Text
- View/download PDF
48. Biallelic tumour suppressor loss and DNA repair defects in de novo small-cell prostate carcinoma.
- Author
-
Chedgy EC, Vandekerkhove G, Herberts C, Annala M, Donoghue AJ, Sigouros M, Ritch E, Struss W, Konomura S, Liew J, Parimi S, Vergidis J, Hurtado-Coll A, Sboner A, Fazli L, Beltran H, Chi KN, and Wyatt AW
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma mortality, Adenocarcinoma pathology, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carboplatin therapeutic use, Carcinoma, Small Cell drug therapy, Carcinoma, Small Cell mortality, Carcinoma, Small Cell pathology, Cisplatin therapeutic use, Databases, Factual, Etoposide pharmacology, Gene Expression Profiling, Genetic Predisposition to Disease, Humans, Male, Middle Aged, Neoplasms, Complex and Mixed drug therapy, Neoplasms, Complex and Mixed mortality, Neoplasms, Complex and Mixed pathology, Phenotype, Prostatic Neoplasms drug therapy, Prostatic Neoplasms mortality, Prostatic Neoplasms pathology, Retrospective Studies, Time Factors, Treatment Outcome, Adenocarcinoma genetics, Biomarkers, Tumor genetics, Carcinoma, Small Cell genetics, DNA Repair, Genes, Tumor Suppressor, Genomic Instability, Neoplasms, Complex and Mixed genetics, Prostatic Neoplasms genetics
- Abstract
Small-cell prostate carcinoma (SCPC) is an aggressive malignancy that is managed similarly to small-cell lung cancer. SCPC can evolve from prostate adenocarcinoma in response to androgen deprivation therapy, but, in rare cases, is present at initial cancer diagnosis. The molecular aetiology of de novo SCPC is incompletely understood, owing to the scarcity of tumour tissue and the short life-expectancy of patients. Through a retrospective search of our regional oncology pharmacy database, we identified 18 patients diagnosed with de novo SCPC between 2004 and 2017. Ten patients had pure SCPC pathology, and the remainder had some admixed adenocarcinoma foci, but all were treated with first-line platinum-based chemotherapy. The median overall survival was 28 months. We performed targeted DNA sequencing, whole exome sequencing and mRNA profiling on formalin-fixed paraffin-embedded archival tumour tissue. We observed frequent biallelic deletion and/or mutation of the tumour suppressor genes TP53, RB1, and PTEN, similarly to what was found in treatment-related SCPC. Indeed, at the RNA level, pure de novo SCPC closely resembled treatment-related SCPC. However, five patients had biallelic loss of DNA repair genes, including BRCA1, BRCA2, ATM, and MSH2/6, potentially underlying the high genomic instability of this rare disease variant. Two patients with pure de novo SCPC harboured ETS gene rearrangements involving androgen-driven promoters, consistent with the evolution of de novo SCPC from an androgen-driven ancestor. Overall, our results reveal a highly aggressive molecular landscape that underlies this unusual pathological variant, and suggest opportunities for targeted therapy strategies in a disease with few treatment options. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd., (Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
49. Patient derived organoids to model rare prostate cancer phenotypes.
- Author
-
Puca L, Bareja R, Prandi D, Shaw R, Benelli M, Karthaus WR, Hess J, Sigouros M, Donoghue A, Kossai M, Gao D, Cyrta J, Sailer V, Vosoughi A, Pauli C, Churakova Y, Cheung C, Deonarine LD, McNary TJ, Rosati R, Tagawa ST, Nanus DM, Mosquera JM, Sawyers CL, Chen Y, Inghirami G, Rao RA, Grandori C, Elemento O, Sboner A, Demichelis F, Rubin MA, and Beltran H
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Line, Tumor, Cell Survival drug effects, Cell Survival genetics, Epigenomics methods, Gene Expression Profiling methods, Genomics methods, Humans, Male, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Neuroendocrine Tumors drug therapy, Neuroendocrine Tumors pathology, Organoids pathology, Phenotype, Prostate pathology, Prostatic Neoplasms drug therapy, Prostatic Neoplasms pathology, Xenograft Model Antitumor Assays, Neuroendocrine Tumors genetics, Organoids metabolism, Prostate metabolism, Prostatic Neoplasms genetics
- Abstract
A major hurdle in the study of rare tumors is a lack of existing preclinical models. Neuroendocrine prostate cancer is an uncommon and aggressive histologic variant of prostate cancer that may arise de novo or as a mechanism of treatment resistance in patients with pre-existing castration-resistant prostate cancer. There are few available models to study neuroendocrine prostate cancer. Here, we report the generation and characterization of tumor organoids derived from needle biopsies of metastatic lesions from four patients. We demonstrate genomic, transcriptomic, and epigenomic concordance between organoids and their corresponding patient tumors. We utilize these organoids to understand the biologic role of the epigenetic modifier EZH2 in driving molecular programs associated with neuroendocrine prostate cancer progression. High-throughput organoid drug screening nominated single agents and drug combinations suggesting repurposing opportunities. This proof of principle study represents a strategy for the study of rare cancer phenotypes.
- Published
- 2018
- Full Text
- View/download PDF
50. BRCA2-Associated Prostate Cancer in a Patient With Spinal and Bulbar Muscular Atrophy.
- Author
-
Conteduca V, Sigouros M, Sboner A, Pritchard CC, and Beltran H
- Abstract
Competing Interests: AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/po/author-center. Vincenza Conteduca No relationship to disclose Michael Sigouros No relationship to disclose Andrea Sboner No relationship to disclose Colin C. Pritchard No relationship to disclose Himisha Beltran Consulting or Advisory Role: Janssen Oncology, Sanofi, Genzyme, GlaxoSmithKline, AbbVie
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.