1
|
Otlu B, Alexandrov LB. Evaluating topography of mutational signatures with SigProfilerTopography. Genome Biol 2025; 26:134. [PMID: 40394581 PMCID: PMC12093824 DOI: 10.1186/s13059-025-03612-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/08/2025] [Indexed: 05/22/2025] Open
Abstract
The mutations found in a cancer genome are shaped by diverse processes, each displaying a characteristic mutational signature that may be influenced by the genome's architecture. While prior analyses have evaluated the effect of topographical genomic features on mutational signatures, there has been no computational tool that can comprehensively examine this interplay. Here, we present SigProfilerTopography, a Python package that allows evaluating the effect of chromatin organization, histone modifications, transcription factor binding, DNA replication, and DNA transcription on the activities of different mutational processes. SigProfilerTopography elucidates the unique topographical characteristics of mutational signatures, unveiling their underlying biological and molecular mechanisms.
Collapse
Affiliation(s)
- Burçak Otlu
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, Ankara, 06800, Turkey
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, 92093, USA.
- Department of Bioengineering, UC San Diego, La Jolla, CA, 92093, USA.
- Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA.
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA, 92037, USA.
| |
Collapse
|
2
|
Grabski IN, Trippa L, Parmigiani G. Bayesian multi-study non-negative matrix factorization for mutational signatures. Genome Biol 2025; 26:98. [PMID: 40241112 PMCID: PMC12001700 DOI: 10.1186/s13059-025-03563-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
Mutational signatures are typically identified from tumor genome sequencing data using non-negative matrix factorization (NMF). However, existing NMF techniques only decompose a single dataset, limiting rigorous comparisons of signatures across conditions. We propose a Bayesian NMF method that jointly decomposes multiple datasets to identify signatures and their sharing pattern across conditions. We propose a fully unsupervised "discovery-only" model and a semi-supervised "recovery-discovery" model that simultaneously estimates known and novel signatures, and extend both to estimate covariate effects. We demonstrate our approach on extensive simulations, and apply our method to answer questions related to colorectal cancer and early-onset breast cancer.
Collapse
Affiliation(s)
| | - Lorenzo Trippa
- Department of Data Science, Dana Farber Cancer Institute, Boston, MA, USA
| | | |
Collapse
|
3
|
Furtado LV, Ikemura K, Benkli CY, Moncur JT, Huang RSP, Zehir A, Stellato K, Vasalos P, Sadri N, Suarez CJ. General Applicability of Existing College of American Pathologists Accreditation Requirements to Clinical Implementation of Machine Learning-Based Methods in Molecular Oncology Testing. Arch Pathol Lab Med 2025; 149:319-327. [PMID: 38871357 DOI: 10.5858/arpa.2024-0037-cp] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2024] [Indexed: 06/15/2024]
Abstract
CONTEXT.— The College of American Pathologists (CAP) accreditation requirements for clinical laboratory testing help ensure laboratories implement and maintain systems and processes that are associated with quality. Machine learning (ML)-based models share some features of conventional laboratory testing methods. Accreditation requirements that specifically address clinical laboratories' use of ML remain in the early stages of development. OBJECTIVE.— To identify relevant CAP accreditation requirements that may be applied to the clinical adoption of ML-based molecular oncology assays, and to provide examples of current and emerging ML applications in molecular oncology testing. DESIGN.— CAP accreditation checklists related to molecular pathology and general laboratory practices (Molecular Pathology, All Common and Laboratory General) were reviewed. Examples of checklist requirements that are generally applicable to validation, revalidation, quality management, infrastructure, and analytical procedures of ML-based molecular oncology assays were summarized. Instances of ML use in molecular oncology testing were assessed from literature review. RESULTS.— Components of the general CAP accreditation framework that exist for traditional molecular oncology assay validation and maintenance are also relevant for implementing ML-based tests in a clinical laboratory. Current and emerging applications of ML in molecular oncology testing include DNA methylation profiling for central nervous system tumor classification, variant calling, microsatellite instability testing, mutational signature analysis, and variant prediction from histopathology images. CONCLUSIONS.— Currently, much of the ML activity in molecular oncology is within early clinical implementation. Despite specific considerations that apply to the adoption of ML-based methods, existing CAP requirements can serve as general guidelines for the clinical implementation of ML-based assays in molecular oncology testing.
Collapse
Affiliation(s)
- Larissa V Furtado
- From the Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee (Furtado)
| | - Kenji Ikemura
- the Department of Pathology, Mass General Brigham, Boston, Massachusetts (Ikemura)
| | - Cagla Y Benkli
- the Department of Pathology, Baylor College of Medicine, Houston, Texas (Benkli)
| | - Joel T Moncur
- Office of the Director, The Joint Pathology Center, Silver Spring, Maryland (Moncur)
| | - Richard S P Huang
- Clinical Development, Foundation Medicine Inc, Cambridge, Massachusetts (Huang)
| | - Ahmet Zehir
- Precision Medicine & Biosamples, AstraZeneca, New York, New York (Zehir)
| | - Katherine Stellato
- Proficiency Testing, College of American Pathologists, Northfield, Illinois (Stellato, Vasalos)
| | - Patricia Vasalos
- Proficiency Testing, College of American Pathologists, Northfield, Illinois (Stellato, Vasalos)
| | - Navid Sadri
- the Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio (Sadri)
| | - Carlos J Suarez
- the Department of Pathology, Stanford University School of Medicine, Palo Alto, California (Suarez)
| |
Collapse
|
4
|
Puttonen M, Almusa H, Böhling T, Koljonen V, Sihto H. Whole-exome sequencing identifies distinct genomic aberrations in eccrine porocarcinomas and poromas. Orphanet J Rare Dis 2025; 20:70. [PMID: 39948683 PMCID: PMC11823087 DOI: 10.1186/s13023-025-03586-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Eccrine porocarcinoma (EPC) is a rare malignant skin tumor arising from the eccrine gland. Investigations into the genomic landscape of EPC have uncovered potential drivers of its development and progression. However, there is limited information on the discrepancies between EPC and its benign counterpart, eccrine poroma (EP). METHODS Formalin-fixed paraffin-embedded (FFPE) samples from 15 EPCs and 5 EPs were retrieved from Helsinki Biobank and Finnish Clinical Biobank Tampere. One EPC was found to be digital papillary adenocarcinoma in review of diagnoses. Whole-exome sequencing was used to conduct a comprehensive analysis to elucidate the genomic features of EPCs and EPs. RESULTS There was general heterogeneity within EPCs and EPs, with discrepancies such as exclusive TP53, NCOR1, and CDKN2A mutations in EPCs and a higher mutational load in EPCs than in EPs. Furthermore, we identified alterations in pathways associated with cell adhesion and the extracellular matrix in EPCs, while pathways associated with ketone body and amino acid metabolism were altered in EPs. The MAPK and Ras signaling pathways were enriched in genes mutated only in EPCs. CONCLUSIONS EPCs and EPs are generally heterogeneous tumor entities with a few distinct discrepancies from each other. The findings from this study emphasize the need to further verify the roles of disrupted genes and pathways in the initiation and progression of EPCs and EPs.
Collapse
Affiliation(s)
- Maya Puttonen
- Department of Pathology, University of Helsinki and Helsinki University Hospital, P.O Box 63, 00014, Helsinki, Finland.
| | - Henrikki Almusa
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Tom Böhling
- Department of Pathology, University of Helsinki and Helsinki University Hospital, P.O Box 63, 00014, Helsinki, Finland
| | - Virve Koljonen
- Department of Plastic Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Harri Sihto
- Department of Pathology, University of Helsinki and Helsinki University Hospital, P.O Box 63, 00014, Helsinki, Finland
| |
Collapse
|
5
|
Son HG, Ha DT, Xia Y, Li T, Blandin J, Oka T, Azin M, Conrad DN, Zhou C, Zeng Y, Hasegawa T, Strickley JD, Messerschmidt JL, Guennoun R, Erlich TH, Shoemaker GL, Johnson LH, Palmer KE, Fisher DE, Horn TD, Neel VA, Nazarian RM, Joh JJ, Demehri S. Commensal papillomavirus immunity preserves the homeostasis of highly mutated normal skin. Cancer Cell 2025; 43:36-48.e10. [PMID: 39672169 PMCID: PMC11732714 DOI: 10.1016/j.ccell.2024.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/12/2024] [Accepted: 11/21/2024] [Indexed: 12/15/2024]
Abstract
Immunosuppression commonly disrupts the homeostasis of mutated normal skin, leading to widespread skin dysplasia and field cancerization. However, the immune system's role in maintaining the normal state of mutated tissues remains uncertain. Herein, we demonstrate that T cell immunity to cutaneotropic papillomaviruses promotes the homeostasis of ultraviolet radiation-damaged skin. Mouse papillomavirus (MmuPV1) colonization blocks the expansion of mutant p53 clones in the epidermis in a CD8+ T cell-dependent manner. MmuPV1 activity is increased in p53-deficient keratinocytes, leading to their specific targeting by CD8+ T cells in the skin. Sun-exposed human skin containing mutant p53 clones shows increased epidermal beta-human papillomavirus (β-HPV) activity and CD8+ T cell infiltrates compared with sun-protected skin. The expansion of mutant p53 clones in premalignant skin lesions associates with β-HPV loss. Thus, immunity to commensal HPVs contributes to the homeostasis of mutated normal skin, highlighting the role of virome-immune system interactions in preserving aging human tissues.
Collapse
Affiliation(s)
- Heehwa G Son
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Dat Thinh Ha
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Yun Xia
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tiancheng Li
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jasmine Blandin
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tomonori Oka
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Marjan Azin
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Danielle N Conrad
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Can Zhou
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yuhan Zeng
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tatsuya Hasegawa
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - John D Strickley
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Medicine, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jonathan L Messerschmidt
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ranya Guennoun
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Tal H Erlich
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gregory L Shoemaker
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Medicine, University of Louisville School of Medicine, Louisville, KY, USA; Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY, USA
| | - Luke H Johnson
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Medicine, University of Louisville School of Medicine, Louisville, KY, USA; Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY, USA
| | - Kenneth E Palmer
- Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA; Center for Predictive Medicine, University of Louisville Health Sciences Center, Louisville, KY, USA
| | - David E Fisher
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Thomas D Horn
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Victor A Neel
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Rosalynn M Nazarian
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Joongho J Joh
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY, USA; Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY, USA
| | - Shadmehr Demehri
- Center for Cancer Immunology, Krantz Family Center for Cancer Research, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA; Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Chakraborty S, Guan Z, Kostrzewa CE, Shen R, Begg CB. Identifying somatic fingerprints of cancers defined by germline and environmental risk factors. Genet Epidemiol 2024; 48:455-467. [PMID: 38686586 PMCID: PMC11522022 DOI: 10.1002/gepi.22565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 01/18/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
Numerous studies over the past generation have identified germline variants that increase specific cancer risks. Simultaneously, a revolution in sequencing technology has permitted high-throughput annotations of somatic genomes characterizing individual tumors. However, examining the relationship between germline variants and somatic alteration patterns is hugely challenged by the large numbers of variants in a typical tumor, the rarity of most individual variants, and the heterogeneity of tumor somatic fingerprints. In this article, we propose statistical methodology that frames the investigation of germline-somatic relationships in an interpretable manner. The method uses meta-features embodying biological contexts of individual somatic alterations to implicitly group rare mutations. Our team has used this technique previously through a multilevel regression model to diagnose with high accuracy tumor site of origin. Herein, we further leverage topic models from computational linguistics to achieve interpretable lower-dimensional embeddings of the meta-features. We demonstrate how the method can identify distinctive somatic profiles linked to specific germline variants or environmental risk factors. We illustrate the method using The Cancer Genome Atlas whole-exome sequencing data to characterize somatic tumor fingerprints in breast cancer patients with germline BRCA1/2 mutations and in head and neck cancer patients exposed to human papillomavirus.
Collapse
Affiliation(s)
| | - Zoe Guan
- Mass General Research Institute, Boston, Massachusetts, USA
| | | | - Ronglai Shen
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Colin B Begg
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
7
|
Xue C, Miller JW, Carter SL, Huggins JH. ROBUST DISCOVERY OF MUTATIONAL SIGNATURES USING POWER POSTERIORS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619958. [PMID: 39553954 PMCID: PMC11566004 DOI: 10.1101/2024.10.23.619958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Mutational processes, such as the molecular effects of carcinogenic agents or defective DNA repair mechanisms, are known to produce different mutation types with characteristic frequency profiles, referred to as mutational signatures. Non-negative matrix factorization (NMF) has successfully been used to discover many mutational signatures, yielding novel insights into cancer etiology and targeted therapies. However, the NMF model is only a rough approximation to reality, and even small departures from this assumed model can have large negative effects on the accuracy and reliability of the results. We propose a new approach to mutational signatures analysis that improves robustness to misspecification by using a power posterior for a fully Bayesian NMF model, while employing a sparsity-inducing prior to automatically infer the number of active signatures. In extensive simulation studies, we find that our proposed approach recovers more true signatures with greater accuracy than current leading methods. On whole-genome sequencing data for six cancer types from the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium, we find that our method is able to accurately recover more signatures than the current state-of-the-art.
Collapse
Affiliation(s)
| | | | - Scott L Carter
- Dana Farber Cancer Institute, Department of Data Science
| | | |
Collapse
|
8
|
Lee D, Hua M, Wang D, Song L, Zhang T, Hua X, Yu K, Yang XR, Chanock SJ, Shi J, Landi MT, Zhu B. Pan-cancer mutational signature analysis of 111,711 targeted sequenced tumors using SATS. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.05.18.23290188. [PMID: 37425683 PMCID: PMC10327246 DOI: 10.1101/2023.05.18.23290188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Tumor mutational signatures are informative for cancer diagnosis and treatment. However, targeted sequencing, commonly used in clinical settings, lacks specialized analytical tools and a dedicated catalogue of mutational signatures. Here, we introduce SATS, a scalable mutational signature analyzer for targeted sequencing data. SATS leverages tumor mutational burdens to identify and quantify signatures in individual tumors, overcoming the challenges of sparse mutations and variable gene panels. Validations across simulated data, pseudo-targeted sequencing data, and matched whole-genome and targeted sequencing samples show that SATS can accurately detect common mutational signatures and estimate their burdens. Applying SATS to 111,711 tumors from the AACR Project GENIE, we created a pan-cancer mutational signature catalogue specific to targeted sequencing. We further validated signatures in lung, breast and colorectal cancers using an additional 16,774 independent samples. This signature catalogue is a valuable resource for estimating signature burdens in individual targeted sequenced tumors, facilitating the integration of mutational signatures with clinical data.
Collapse
|
9
|
Pancotti C, Rollo C, Codicè F, Birolo G, Fariselli P, Sanavia T. MUSE-XAE: MUtational Signature Extraction with eXplainable AutoEncoder enhances tumour types classification. Bioinformatics 2024; 40:btae320. [PMID: 38754097 PMCID: PMC11139523 DOI: 10.1093/bioinformatics/btae320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/08/2024] [Accepted: 05/15/2024] [Indexed: 05/18/2024] Open
Abstract
MOTIVATION Mutational signatures are a critical component in deciphering the genetic alterations that underlie cancer development and have become a valuable resource to understand the genomic changes during tumorigenesis. Therefore, it is essential to employ precise and accurate methods for their extraction to ensure that the underlying patterns are reliably identified and can be effectively utilized in new strategies for diagnosis, prognosis, and treatment of cancer patients. RESULTS We present MUSE-XAE, a novel method for mutational signature extraction from cancer genomes using an explainable autoencoder. Our approach employs a hybrid architecture consisting of a nonlinear encoder that can capture nonlinear interactions among features, and a linear decoder which ensures the interpretability of the active signatures. We evaluated and compared MUSE-XAE with other available tools on both synthetic and real cancer datasets and demonstrated that it achieves superior performance in terms of precision and sensitivity in recovering mutational signature profiles. MUSE-XAE extracts highly discriminative mutational signature profiles by enhancing the classification of primary tumour types and subtypes in real world settings. This approach could facilitate further research in this area, with neural networks playing a critical role in advancing our understanding of cancer genomics. AVAILABILITY AND IMPLEMENTATION MUSE-XAE software is freely available at https://github.com/compbiomed-unito/MUSE-XAE.
Collapse
Affiliation(s)
- Corrado Pancotti
- Computational Biomedicine Unit, Department of Medical Sciences, University of Torino, via Santena 19, Torino 10126, Italy
| | - Cesare Rollo
- Computational Biomedicine Unit, Department of Medical Sciences, University of Torino, via Santena 19, Torino 10126, Italy
| | - Francesco Codicè
- Computational Biomedicine Unit, Department of Medical Sciences, University of Torino, via Santena 19, Torino 10126, Italy
| | - Giovanni Birolo
- Computational Biomedicine Unit, Department of Medical Sciences, University of Torino, via Santena 19, Torino 10126, Italy
| | - Piero Fariselli
- Computational Biomedicine Unit, Department of Medical Sciences, University of Torino, via Santena 19, Torino 10126, Italy
| | - Tiziana Sanavia
- Computational Biomedicine Unit, Department of Medical Sciences, University of Torino, via Santena 19, Torino 10126, Italy
| |
Collapse
|
10
|
Park JE, Smith MA, Van Alsten SC, Walens A, Wu D, Hoadley KA, Troester MA, Love MI. Diffsig: Associating Risk Factors with Mutational Signatures. Cancer Epidemiol Biomarkers Prev 2024; 33:721-730. [PMID: 38426904 PMCID: PMC11062813 DOI: 10.1158/1055-9965.epi-23-0728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/12/2023] [Accepted: 02/28/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Somatic mutational signatures elucidate molecular vulnerabilities to therapy, and therefore detecting signatures and classifying tumors with respect to signatures has clinical value. However, identifying the etiology of the mutational signatures remains a statistical challenge, with both small sample sizes and high variability in classification algorithms posing barriers. As a result, few signatures have been strongly linked to particular risk factors. METHODS Here, we develop a statistical model, Diffsig, for estimating the association of one or more continuous or categorical risk factors with DNA mutational signatures. Diffsig takes into account the uncertainty associated with assigning signatures to samples as well as multiple risk factors' simultaneous effect on observed DNA mutations. RESULTS We applied Diffsig to breast cancer data to assess relationships between five established breast-relevant mutational signatures and etiologic variables, confirming known mechanisms of cancer development. In simulation, our model was capable of accurately estimating expected associations in a variety of contexts. CONCLUSIONS Diffsig allows researchers to quantify and perform inference on the associations of risk factors with mutational signatures. IMPACT We expect Diffsig to provide more robust associations of risk factors with signatures to lead to better understanding of the tumor development process and improved models of tumorigenesis.
Collapse
Affiliation(s)
- Ji-Eun Park
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Markia A. Smith
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Sarah C. Van Alsten
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Andrea Walens
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Di Wu
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katherine A. Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Melissa A. Troester
- Department of Pathology and Laboratory Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Michael I. Love
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
11
|
Jin H, Gulhan DC, Geiger B, Ben-Isvy D, Geng D, Ljungström V, Park PJ. Accurate and sensitive mutational signature analysis with MuSiCal. Nat Genet 2024; 56:541-552. [PMID: 38361034 PMCID: PMC10937379 DOI: 10.1038/s41588-024-01659-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 01/08/2024] [Indexed: 02/17/2024]
Abstract
Mutational signature analysis is a recent computational approach for interpreting somatic mutations in the genome. Its application to cancer data has enhanced our understanding of mutational forces driving tumorigenesis and demonstrated its potential to inform prognosis and treatment decisions. However, methodological challenges remain for discovering new signatures and assigning proper weights to existing signatures, thereby hindering broader clinical applications. Here we present Mutational Signature Calculator (MuSiCal), a rigorous analytical framework with algorithms that solve major problems in the standard workflow. Our simulation studies demonstrate that MuSiCal outperforms state-of-the-art algorithms for both signature discovery and assignment. By reanalyzing more than 2,700 cancer genomes, we provide an improved catalog of signatures and their assignments, discover nine indel signatures absent in the current catalog, resolve long-standing issues with the ambiguous 'flat' signatures and give insights into signatures with unknown etiologies. We expect MuSiCal and the improved catalog to be a step towards establishing best practices for mutational signature analysis.
Collapse
Affiliation(s)
- Hu Jin
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Doga C Gulhan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Benedikt Geiger
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Daniel Ben-Isvy
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - David Geng
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Viktor Ljungström
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Peter J Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Otlu B, Alexandrov LB. Evaluating topography of mutational signatures with SigProfilerTopography. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574683. [PMID: 38260507 PMCID: PMC10802511 DOI: 10.1101/2024.01.08.574683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The mutations found in a cancer genome are shaped by diverse processes, each displaying a characteristic mutational signature that may be influenced by the genome's architecture. While prior analyses have evaluated the effect of topographical genomic features on mutational signatures, there has been no computational tool that can comprehensively examine this interplay. Here, we present SigProfilerTopography, a Python package that allows evaluating the effect of chromatin organization, histone modifications, transcription factor binding, DNA replication, and DNA transcription on the activities of different mutational processes. SigProfilerTopography elucidates the unique topographical characteristics of mutational signatures, unveiling their underlying biological and molecular mechanisms.
Collapse
Affiliation(s)
- Burçak Otlu
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, 06800, Ankara, Turkey
| | - Ludmil B. Alexandrov
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA 92037
| |
Collapse
|
13
|
Laursen R, Maretty L, Hobolth A. Flexible model-based non-negative matrix factorization with application to mutational signatures. Stat Appl Genet Mol Biol 2024; 23:sagmb-2023-0034. [PMID: 38753402 DOI: 10.1515/sagmb-2023-0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 04/03/2024] [Indexed: 08/09/2024]
Abstract
Somatic mutations in cancer can be viewed as a mixture distribution of several mutational signatures, which can be inferred using non-negative matrix factorization (NMF). Mutational signatures have previously been parametrized using either simple mono-nucleotide interaction models or general tri-nucleotide interaction models. We describe a flexible and novel framework for identifying biologically plausible parametrizations of mutational signatures, and in particular for estimating di-nucleotide interaction models. Our novel estimation procedure is based on the expectation-maximization (EM) algorithm and regression in the log-linear quasi-Poisson model. We show that di-nucleotide interaction signatures are statistically stable and sufficiently complex to fit the mutational patterns. Di-nucleotide interaction signatures often strike the right balance between appropriately fitting the data and avoiding over-fitting. They provide a better fit to data and are biologically more plausible than mono-nucleotide interaction signatures, and the parametrization is more stable than the parameter-rich tri-nucleotide interaction signatures. We illustrate our framework in a large simulation study where we compare to state of the art methods, and show results for three data sets of somatic mutation counts from patients with cancer in the breast, Liver and urinary tract.
Collapse
Affiliation(s)
- Ragnhild Laursen
- Department of Mathematics, 1006 Aarhus University , Aarhus, Denmark
| | - Lasse Maretty
- Department of Clinical Medicine and Bioinformatics Research Center, 1006 Aarhus University , Aarhus, Denmark
| | - Asger Hobolth
- Department of Mathematics, 1006 Aarhus University , Aarhus, Denmark
| |
Collapse
|
14
|
Drummond RD, Defelicibus A, Meyenberg M, Valieris R, Dias-Neto E, Rosales RA, da Silva IT. Relating mutational signature exposures to clinical data in cancers via signeR 2.0. BMC Bioinformatics 2023; 24:439. [PMID: 37990302 PMCID: PMC10664385 DOI: 10.1186/s12859-023-05550-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/27/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Cancer is a collection of diseases caused by the deregulation of cell processes, which is triggered by somatic mutations. The search for patterns in somatic mutations, known as mutational signatures, is a growing field of study that has already become a useful tool in oncology. Several algorithms have been proposed to perform one or both the following two tasks: (1) de novo estimation of signatures and their exposures, (2) estimation of the exposures of each one of a set of pre-defined signatures. RESULTS Our group developed signeR, a Bayesian approach to both of these tasks. Here we present a new version of the software, signeR 2.0, which extends the possibilities of previous analyses to explore the relation of signature exposures to other data of clinical relevance. signeR 2.0 includes a user-friendly interface developed using the R-Shiny framework and improvements in performance. This version allows the analysis of submitted data or public TCGA data, which is embedded in the package for easy access. CONCLUSION signeR 2.0 is a valuable tool to generate and explore exposure data, both from de novo or fitting analyses and is an open-source R package available through the Bioconductor project at ( https://doi.org/10.18129/B9.bioc.signeR ).
Collapse
Affiliation(s)
- Rodrigo D Drummond
- Laboratory of Computational Biology and Bioinformatics, CIPE/A.C.Camargo Cancer Center, São Paulo, São Paulo, 01508-010, Brazil
| | - Alexandre Defelicibus
- Laboratory of Computational Biology and Bioinformatics, CIPE/A.C.Camargo Cancer Center, São Paulo, São Paulo, 01508-010, Brazil
| | - Mathilde Meyenberg
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - Renan Valieris
- Laboratory of Computational Biology and Bioinformatics, CIPE/A.C.Camargo Cancer Center, São Paulo, São Paulo, 01508-010, Brazil
| | - Emmanuel Dias-Neto
- Laboratory of Computational Biology and Bioinformatics, CIPE/A.C.Camargo Cancer Center, São Paulo, São Paulo, 01508-010, Brazil
| | - Rafael A Rosales
- Departamento de Computação e Matemática, Universidade de São Paulo, Ribeirão Preto, São Paulo, 14040-901, Brazil.
| | - Israel Tojal da Silva
- Laboratory of Computational Biology and Bioinformatics, CIPE/A.C.Camargo Cancer Center, São Paulo, São Paulo, 01508-010, Brazil.
| |
Collapse
|
15
|
Yang Y, Yang L. Somatic structural variation signatures in pediatric brain tumors. Cell Rep 2023; 42:113276. [PMID: 37851574 PMCID: PMC10748741 DOI: 10.1016/j.celrep.2023.113276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/26/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023] Open
Abstract
Brain cancer is the leading cause of cancer-related death in children. Somatic structural variations (SVs), large-scale alterations in DNA, remain poorly understood in pediatric brain tumors. Here, we detect a total of 13,199 high-confidence somatic SVs in 744 whole-genome sequences of pediatric brain tumors from the Pediatric Brain Tumor Atlas. The somatic SV occurrences have tremendous diversity among the cohort and across different tumor types. We decompose mutational signatures of clustered complex SVs, non-clustered complex SVs, and simple SVs separately to infer their mutational mechanisms. Our finding of many tumor types carrying unique sets of SV signatures suggests that distinct molecular mechanisms shape genome instability in different tumor types. The patterns of somatic SV signatures in pediatric brain tumors are substantially different from those in adult cancers. The convergence of multiple SV signatures on several major cancer driver genes implies vital roles of somatic SVs in disease progression.
Collapse
Affiliation(s)
- Yang Yang
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Lixing Yang
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA; Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA; University of Chicago Comprehensive Cancer Center, Chicago, IL 60637, USA.
| |
Collapse
|
16
|
Kahane I, Leiserson MDM, Sharan R. A mutation-level covariate model for mutational signatures. PLoS Comput Biol 2023; 19:e1011195. [PMID: 37276234 DOI: 10.1371/journal.pcbi.1011195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 05/17/2023] [Indexed: 06/07/2023] Open
Abstract
Mutational processes and their exposures in particular genomes are key to our understanding of how these genomes are shaped. However, current analyses assume that these processes are uniformly active across the genome without accounting for potential covariates such as strand or genomic region that could impact such activities. Here we suggest the first mutation-covariate models that explicitly model the effect of different covariates on the exposures of mutational processes. We apply these models to test the impact of replication strand on these processes and compare them to strand-oblivious models across a range of data sets. Our models capture replication strand specificity, point to signatures affected by it, and score better on held-out data compared to standard models that do not account for mutation-level covariate information.
Collapse
Affiliation(s)
- Itay Kahane
- School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Mark D M Leiserson
- Department of Computer Science and Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - Roded Sharan
- School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
17
|
Pelizzola M, Laursen R, Hobolth A. Model selection and robust inference of mutational signatures using Negative Binomial non-negative matrix factorization. BMC Bioinformatics 2023; 24:187. [PMID: 37158829 PMCID: PMC10165836 DOI: 10.1186/s12859-023-05304-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND The spectrum of mutations in a collection of cancer genomes can be described by a mixture of a few mutational signatures. The mutational signatures can be found using non-negative matrix factorization (NMF). To extract the mutational signatures we have to assume a distribution for the observed mutational counts and a number of mutational signatures. In most applications, the mutational counts are assumed to be Poisson distributed, and the rank is chosen by comparing the fit of several models with the same underlying distribution and different values for the rank using classical model selection procedures. However, the counts are often overdispersed, and thus the Negative Binomial distribution is more appropriate. RESULTS We propose a Negative Binomial NMF with a patient specific dispersion parameter to capture the variation across patients and derive the corresponding update rules for parameter estimation. We also introduce a novel model selection procedure inspired by cross-validation to determine the number of signatures. Using simulations, we study the influence of the distributional assumption on our method together with other classical model selection procedures. We also present a simulation study with a method comparison where we show that state-of-the-art methods are highly overestimating the number of signatures when overdispersion is present. We apply our proposed analysis on a wide range of simulated data and on two real data sets from breast and prostate cancer patients. On the real data we describe a residual analysis to investigate and validate the model choice. CONCLUSIONS With our results on simulated and real data we show that our model selection procedure is more robust at determining the correct number of signatures under model misspecification. We also show that our model selection procedure is more accurate than the available methods in the literature for finding the true number of signatures. Lastly, the residual analysis clearly emphasizes the overdispersion in the mutational count data. The code for our model selection procedure and Negative Binomial NMF is available in the R package SigMoS and can be found at https://github.com/MartaPelizzola/SigMoS .
Collapse
Affiliation(s)
- Marta Pelizzola
- Department of Mathematics, Aarhus University, Aarhus, Denmark.
| | | | - Asger Hobolth
- Department of Mathematics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
18
|
Samur MK, Roncador M, Aktas Samur A, Fulciniti M, Bazarbachi AH, Szalat R, Shammas MA, Sperling AS, Richardson PG, Magrangeas F, Minvielle S, Perrot A, Corre J, Moreau P, Thakurta A, Parmigiani G, Anderson KC, Avet-Loiseau H, Munshi NC. High-dose melphalan treatment significantly increases mutational burden at relapse in multiple myeloma. Blood 2023; 141:1724-1736. [PMID: 36603186 PMCID: PMC10273091 DOI: 10.1182/blood.2022017094] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 12/02/2022] [Accepted: 12/22/2022] [Indexed: 01/07/2023] Open
Abstract
High-dose melphalan (HDM) improves progression-free survival in multiple myeloma (MM), yet melphalan is a DNA-damaging alkylating agent; therefore, we assessed its mutational effect on surviving myeloma cells by analyzing paired MM samples collected at diagnosis and relapse in the IFM 2009 study. We performed deep whole-genome sequencing on samples from 68 patients, 43 of whom were treated with RVD (lenalidomide, bortezomib, and dexamethasone) and 25 with RVD + HDM. Although the number of mutations was similar at diagnosis in both groups (7137 vs 7230; P = .67), the HDM group had significantly more mutations at relapse (9242 vs 13 383, P = .005). No change in the frequency of copy number alterations or structural variants was observed. The newly acquired mutations were typically associated with DNA damage and double-stranded breaks and were predominantly on the transcribed strand. A machine learning model, using this unique pattern, predicted patients who would receive HDM with high sensitivity, specificity, and positive prediction value. Clonal evolution analysis showed that all patients treated with HDM had clonal selection, whereas a static progression was observed with RVD. A significantly higher percentage of mutations were subclonal in the HDM cohort. Intriguingly, patients treated with HDM who achieved complete remission (CR) had significantly more mutations at relapse yet had similar survival rates as those treated with RVD who achieved CR. This similarity could have been due to HDM relapse samples having significantly more neoantigens. Overall, our study identifies increased genomic changes associated with HDM and provides rationale to further understand clonal complexity.
Collapse
Affiliation(s)
- Mehmet Kemal Samur
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA
| | | | - Anil Aktas Samur
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Mariateresa Fulciniti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Abdul Hamid Bazarbachi
- Department of Internal Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, New York, NY
| | - Raphael Szalat
- Department of Hematology and Medical Oncology, Boston University Medical Center, Boston, MA
| | - Masood A. Shammas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Adam S. Sperling
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Paul G. Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Florence Magrangeas
- Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), INSERM, French National Centre for Scientific Research (CNRS), Angers University, and Nantes University, Nantes, France
| | - Stephane Minvielle
- Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), INSERM, French National Centre for Scientific Research (CNRS), Angers University, and Nantes University, Nantes, France
| | - Aurore Perrot
- University Cancer Center of Toulouse Institut National de la Santé, Toulouse, France
| | - Jill Corre
- University Cancer Center of Toulouse Institut National de la Santé, Toulouse, France
| | - Philippe Moreau
- Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), INSERM, French National Centre for Scientific Research (CNRS), Angers University, and Nantes University, Nantes, France
| | | | - Giovanni Parmigiani
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA
| | - Kenneth C. Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
| | - Hervé Avet-Loiseau
- University Cancer Center of Toulouse Institut National de la Santé, Toulouse, France
| | - Nikhil C. Munshi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard University, Boston, MA
- VA Boston Healthcare System, Boston, MA
| |
Collapse
|
19
|
Patterson A, Elbasir A, Tian B, Auslander N. Computational Methods Summarizing Mutational Patterns in Cancer: Promise and Limitations for Clinical Applications. Cancers (Basel) 2023; 15:1958. [PMID: 37046619 PMCID: PMC10093138 DOI: 10.3390/cancers15071958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/24/2023] [Accepted: 03/09/2023] [Indexed: 03/29/2023] Open
Abstract
Since the rise of next-generation sequencing technologies, the catalogue of mutations in cancer has been continuously expanding. To address the complexity of the cancer-genomic landscape and extract meaningful insights, numerous computational approaches have been developed over the last two decades. In this review, we survey the current leading computational methods to derive intricate mutational patterns in the context of clinical relevance. We begin with mutation signatures, explaining first how mutation signatures were developed and then examining the utility of studies using mutation signatures to correlate environmental effects on the cancer genome. Next, we examine current clinical research that employs mutation signatures and discuss the potential use cases and challenges of mutation signatures in clinical decision-making. We then examine computational studies developing tools to investigate complex patterns of mutations beyond the context of mutational signatures. We survey methods to identify cancer-driver genes, from single-driver studies to pathway and network analyses. In addition, we review methods inferring complex combinations of mutations for clinical tasks and using mutations integrated with multi-omics data to better predict cancer phenotypes. We examine the use of these tools for either discovery or prediction, including prediction of tumor origin, treatment outcomes, prognosis, and cancer typing. We further discuss the main limitations preventing widespread clinical integration of computational tools for the diagnosis and treatment of cancer. We end by proposing solutions to address these challenges using recent advances in machine learning.
Collapse
Affiliation(s)
- Andrew Patterson
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Wistar Institute, Philadelphia, PA 19104, USA
| | | | - Bin Tian
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Noam Auslander
- The Wistar Institute, Philadelphia, PA 19104, USA
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
20
|
A Biterm Topic Model for Sparse Mutation Data. Cancers (Basel) 2023; 15:cancers15051601. [PMID: 36900390 PMCID: PMC10000560 DOI: 10.3390/cancers15051601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Mutational signature analysis promises to reveal the processes that shape cancer genomes for applications in diagnosis and therapy. However, most current methods are geared toward rich mutation data that has been extracted from whole-genome or whole-exome sequencing. Methods that process sparse mutation data typically found in practice are only in the earliest stages of development. In particular, we previously developed the Mix model that clusters samples to handle data sparsity. However, the Mix model had two hyper-parameters, including the number of signatures and the number of clusters, that were very costly to learn. Therefore, we devised a new method that was several orders-of-magnitude more efficient for handling sparse data, was based on mutation co-occurrences, and imitated word co-occurrence analyses of Twitter texts. We showed that the model produced significantly improved hyper-parameter estimates that led to higher likelihoods of discovering overlooked data and had better correspondence with known signatures.
Collapse
|
21
|
Park JE, Smith MA, Van Alsten SC, Walens A, Wu D, Hoadley KA, Troester MA, Love MI. Diffsig: Associating Risk Factors With Mutational Signatures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.09.527740. [PMID: 36798154 PMCID: PMC9934616 DOI: 10.1101/2023.02.09.527740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Somatic mutational signatures elucidate molecular vulnerabilities to therapy and therefore detecting signatures and classifying tumors with respect to signatures has clinical value. However, identifying the etiology of the mutational signatures remains a statistical challenge, with both small sample sizes and high variability in classification algorithms posing barriers. As a result, few signatures have been strongly linked to particular risk factors. Here we present Diffsig, a model and R package for estimating the association of risk factors with mutational signatures, suggesting etiologies for the pre-defined mutational signatures. Diffsig is a Bayesian Dirichlet-multinomial hierarchical model that allows testing of any type of risk factor while taking into account the uncertainty associated with samples with a low number of observations. In simulation, we found that our method can accurately estimate risk factor-mutational signal associations. We applied Diffsig to breast cancer data to assess relationships between five established breast-relevant mutational signatures and etiologic variables, confirming known mechanisms of cancer development. Diffsig is implemented as an R package available at: https://github.com/jennprk/diffsig.
Collapse
|
22
|
Ansari-Pour N, Samur M, Flynt E, Gooding S, Towfic F, Stong N, Estevez MO, Mavrommatis K, Walker B, Morgan G, Munshi N, Avet-Loiseau H, Thakurta A. Whole-genome analysis identifies novel drivers and high-risk double-hit events in relapsed/refractory myeloma. Blood 2023; 141:620-633. [PMID: 36223594 PMCID: PMC10163277 DOI: 10.1182/blood.2022017010] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 11/20/2022] Open
Abstract
Large-scale analyses of genomic data from patients with newly diagnosed multiple myeloma (ndMM) have been undertaken, however, large-scale analysis of relapsed/refractory MM (rrMM) has not been performed. We hypothesize that somatic variants chronicle the therapeutic exposures and clonal structure of myeloma from ndMM to rrMM stages. We generated whole-genome sequencing (WGS) data from 418 tumors (386 patients) derived from 6 rrMM clinical trials and compared them with WGS from 198 unrelated patients with ndMM in a population-based case-control fashion. We identified significantly enriched events at the rrMM stage, including drivers (DUOX2, EZH2, TP53), biallelic inactivation (TP53), noncoding mutations in bona fide drivers (TP53BP1, BLM), copy number aberrations (CNAs; 1qGain, 17pLOH), and double-hit events (Amp1q-ISS3, 1qGain-17p loss-of-heterozygosity). Mutational signature analysis identified a subclonal defective mismatch repair signature enriched in rrMM and highly active in high mutation burden tumors, a likely feature of therapy-associated expanding subclones. Further analysis focused on the association of genomic aberrations enriched at different stages of resistance to immunomodulatory agent (IMiD)-based therapy. This analysis revealed that TP53, DUOX2, 1qGain, and 17p loss-of-heterozygosity increased in prevalence from ndMM to lenalidomide resistant (LENR) to pomalidomide resistant (POMR) stages, whereas enrichment of MAML3 along with immunoglobulin lambda (IGL) and MYC translocations distinguished POM from the LEN subgroup. Genomic drivers associated with rrMM are those that confer clonal selective advantage under therapeutic pressure. Their role in therapy evasion should be further evaluated in longitudinal patient samples, to confirm these associations with the evolution of clinical resistance and to identify molecular subsets of rrMM for the development of targeted therapies.
Collapse
Affiliation(s)
- Naser Ansari-Pour
- Medical Research Council Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- National Institute for Health and Care Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Mehmet Samur
- Dana-Farber Cancer Institute, Boston, MA
- Harvard T.H. Chan School of Public Health, Boston, MA
| | - Erin Flynt
- Translational Medicine, Bristol Myers Squibb, Summit, NJ
| | - Sarah Gooding
- Medical Research Council Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- National Institute for Health and Care Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Department of Haematology, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
| | | | | | - Maria Ortiz Estevez
- Predictive Sciences, BMS Center for Innovation and Translational Research Europe, A Bristol Myers Squibb Company, Sevilla, Spain
| | | | - Brian Walker
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology Oncology, Indiana University, Indianapolis, IN
| | - Gareth Morgan
- Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY
| | - Nikhil Munshi
- Dana-Farber Cancer Institute, Boston, MA
- VA Boston Healthcare System, West Roxbury, MA
- Harvard Medical School, Boston, MA
| | | | - Anjan Thakurta
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
- Bristol Myers Squibb, Summit, NJ
- Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Disease, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
23
|
Liu M, Wu Y, Jiang N, Boot A, Rozen S. mSigHdp: hierarchical Dirichlet process mixture modeling for mutational signature discovery. NAR Genom Bioinform 2023; 5:lqad005. [PMID: 36694663 PMCID: PMC9869330 DOI: 10.1093/nargab/lqad005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Mutational signatures are characteristic patterns of mutations caused by endogenous or exogenous mutational processes. These signatures can be discovered by analyzing mutations in large sets of samples-usually somatic mutations in tumor samples. Most programs for discovering mutational signatures are based on non-negative matrix factorization (NMF). Alternatively, signatures can be discovered using hierarchical Dirichlet process (HDP) mixture models, an approach that has been less explored. These models assign mutations to clusters and view each cluster as being generated from the signature of a particular mutational process. Here, we describe mSigHdp, an improved approach to using HDP mixture models to discover mutational signatures. We benchmarked mSigHdp and state-of-the-art NMF-based approaches on four realistic synthetic data sets. These data sets encompassed 18 cancer types. In total, they contained 3.5 × 107 single-base-substitution mutations representing 32 signatures and 6.1 × 106 small insertion and deletion mutations representing 13 signatures. For three of the four data sets, mSigHdp had the best positive predictive value for discovering mutational signatures, and for all four data sets, it had the best true positive rate. Its CPU usage was similar to that of the NMF-based approaches. Thus, mSigHdp is an important and practical addition to the set of tools available for discovering mutational signatures.
Collapse
Affiliation(s)
| | | | - Nanhai Jiang
- Programme in Cancer & Stem Cell Biology, Duke–NUS Medical School, 169857 Singapore,Centre for Computational Biology, Duke–NUS Medical School, 169857 Singapore
| | - Arnoud Boot
- Programme in Cancer & Stem Cell Biology, Duke–NUS Medical School, 169857 Singapore,Centre for Computational Biology, Duke–NUS Medical School, 169857 Singapore
| | - Steven G Rozen
- To whom correspondence should be addressed. Tel: +65 65164945;
| |
Collapse
|
24
|
Islam SA, Díaz-Gay M, Wu Y, Barnes M, Vangara R, Bergstrom EN, He Y, Vella M, Wang J, Teague JW, Clapham P, Moody S, Senkin S, Li YR, Riva L, Zhang T, Gruber AJ, Steele CD, Otlu B, Khandekar A, Abbasi A, Humphreys L, Syulyukina N, Brady SW, Alexandrov BS, Pillay N, Zhang J, Adams DJ, Martincorena I, Wedge DC, Landi MT, Brennan P, Stratton MR, Rozen SG, Alexandrov LB. Uncovering novel mutational signatures by de novo extraction with SigProfilerExtractor. CELL GENOMICS 2022; 2:None. [PMID: 36388765 PMCID: PMC9646490 DOI: 10.1016/j.xgen.2022.100179] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 04/10/2022] [Accepted: 08/31/2022] [Indexed: 12/09/2022]
Abstract
Mutational signature analysis is commonly performed in cancer genomic studies. Here, we present SigProfilerExtractor, an automated tool for de novo extraction of mutational signatures, and benchmark it against another 13 bioinformatics tools by using 34 scenarios encompassing 2,500 simulated signatures found in 60,000 synthetic genomes and 20,000 synthetic exomes. For simulations with 5% noise, reflecting high-quality datasets, SigProfilerExtractor outperforms other approaches by elucidating between 20% and 50% more true-positive signatures while yielding 5-fold less false-positive signatures. Applying SigProfilerExtractor to 4,643 whole-genome- and 19,184 whole-exome-sequenced cancers reveals four novel signatures. Two of the signatures are confirmed in independent cohorts, and one of these signatures is associated with tobacco smoking. In summary, this report provides a reference tool for analysis of mutational signatures, a comprehensive benchmarking of bioinformatics tools for extracting signatures, and several novel mutational signatures, including one putatively attributed to direct tobacco smoking mutagenesis in bladder tissues.
Collapse
Affiliation(s)
- S.M. Ashiqul Islam
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA 92037, USA
| | - Marcos Díaz-Gay
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA 92037, USA
| | - Yang Wu
- Centre for Computational Biology and Programme in Cancer & Stem Cell Biology, Duke NUS Medical School, Singapore 169857, Singapore
| | - Mark Barnes
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA 92037, USA
| | - Raviteja Vangara
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA 92037, USA
| | - Erik N. Bergstrom
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA 92037, USA
| | - Yudou He
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA 92037, USA
| | - Mike Vella
- NVIDIA Corporation, 2788 San Tomas Expressway, Santa Clara, CA 95051, USA
| | - Jingwei Wang
- Cancer, Ageing and Somatic Mutation, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Jon W. Teague
- Cancer, Ageing and Somatic Mutation, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Peter Clapham
- Cancer, Ageing and Somatic Mutation, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Sarah Moody
- Cancer, Ageing and Somatic Mutation, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Sergey Senkin
- Genetic Epidemiology Group, International Agency for Research on Cancer, Cedex 08, 69372 Lyon, France
| | - Yun Rose Li
- Departments of Radiation Oncology and Cancer Genetics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Laura Riva
- Cancer, Ageing and Somatic Mutation, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Andreas J. Gruber
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7LF, UK
- Manchester Cancer Research Centre, The University of Manchester, Manchester M20 4GJ, UK
- Department of Biology, University of Konstanz, Universitaetsstrasse 10, D-78464 Konstanz, Germany
| | - Christopher D. Steele
- Research Department of Pathology, Cancer Institute, University College London, London WC1E 6BT, UK
| | - Burçak Otlu
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA 92037, USA
| | - Azhar Khandekar
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA 92037, USA
| | - Ammal Abbasi
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA 92037, USA
| | - Laura Humphreys
- Cancer, Ageing and Somatic Mutation, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | | | - Samuel W. Brady
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Boian S. Alexandrov
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Nischalan Pillay
- Research Department of Pathology, Cancer Institute, University College London, London WC1E 6BT, UK
- Department of Cellular and Molecular Pathology, Royal National Orthopaedic Hospital NHS Trust, Stanmore, Middlesex HA7 4LP, UK
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - David J. Adams
- Cancer, Ageing and Somatic Mutation, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Iñigo Martincorena
- Cancer, Ageing and Somatic Mutation, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - David C. Wedge
- Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7LF, UK
- Manchester Cancer Research Centre, The University of Manchester, Manchester M20 4GJ, UK
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Paul Brennan
- Genetic Epidemiology Group, International Agency for Research on Cancer, Cedex 08, 69372 Lyon, France
| | - Michael R. Stratton
- Cancer, Ageing and Somatic Mutation, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Steven G. Rozen
- Centre for Computational Biology and Programme in Cancer & Stem Cell Biology, Duke NUS Medical School, Singapore 169857, Singapore
| | - Ludmil B. Alexandrov
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA 92093, USA
- Department of Bioengineering, UC San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, UC San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
25
|
Islam MA, Versypt ANF. Mathematical Modeling of Impacts of Patient Differences on COVID-19 Lung Fibrosis Outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.11.06.515367. [PMID: 36380760 DOI: 10.1101/2020.12.13.422570] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Patient-specific premorbidity, age, and sex are significant heterogeneous factors that influence the severe manifestation of lung diseases, including COVID-19 fibrosis. The renin-angiotensin system (RAS) plays a prominent role in regulating effects of these factors. Recent evidence suggests that patient-specific alteration of RAS homeostasis with premorbidity and the expression level of angiotensin converting enzyme 2 (ACE2), depending on age and sex, is correlated with lung fibrosis. However, conflicting evidence suggests decreases, increases, or no changes in RAS after SARS-CoV-2 infection. In addition, detailed mechanisms connecting the patient-specific conditions before infection to infection-induced fibrosis are still unknown. Here, a mathematical model is developed to quantify the systemic contribution of heterogeneous factors of RAS in the progression of lung fibrosis. Three submodels are connected-a RAS model, an agent-based COVID-19 in-host immune response model, and a fibrosis model-to investigate the effects of patient-group-specific factors in the systemic alteration of RAS and collagen deposition in the lung. The model results indicate cell death due to inflammatory response as a major contributor to the reduction of ACE and ACE2, whereas there are no significant changes in ACE2 dynamics due to viral-bound internalization of ACE2. Reduction of ACE reduces the homeostasis of RAS including angiotensin II (ANGII), while the decrease in ACE2 increases ANGII and results in severe lung injury and fibrosis. The model explains possible mechanisms for conflicting evidence of RAS alterations in previously published studies. Also, the results show that ACE2 variations with age and sex significantly alter RAS peptides and lead to fibrosis with around 20% additional collagen deposition from systemic RAS with slight variations depending on age and sex. This model may find further applications in patient-specific calibrations of tissue models for acute and chronic lung diseases to develop personalized treatments.
Collapse
|
26
|
Giannuzzi D, Marconato L, Fanelli A, Licenziato L, De Maria R, Rinaldi A, Rotta L, Rouquet N, Birolo G, Fariselli P, Mensah AA, Bertoni F, Aresu L. The genomic landscape of canine diffuse large B-cell lymphoma identifies distinct subtypes with clinical and therapeutic implications. Lab Anim (NY) 2022; 51:191-202. [PMID: 35726023 DOI: 10.1038/s41684-022-00998-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 05/13/2022] [Indexed: 12/13/2022]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoid neoplasm in dogs and in humans. It is characterized by a remarkable degree of clinical heterogeneity that is not completely elucidated by molecular data. This poses a major barrier to understanding the disease and its response to therapy, or when treating dogs with DLBCL within clinical trials. We performed an integrated analysis of exome (n = 77) and RNA sequencing (n = 43) data in a cohort of canine DLBCL to define the genetic landscape of this tumor. A wide range of signaling pathways and cellular processes were found in common with human DLBCL, but the frequencies of the most recurrently mutated genes (TRAF3, SETD2, POT1, TP53, MYC, FBXW7, DDX3X and TBL1XR1) differed. We developed a prognostic model integrating exonic variants and clinical and transcriptomic features to predict the outcome in dogs with DLBCL. These results comprehensively define the genetic drivers of canine DLBCL and can be prospectively utilized to identify new therapeutic opportunities.
Collapse
Affiliation(s)
- Diana Giannuzzi
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padua, Padua, Italy
| | - Laura Marconato
- Department of Veterinary Medical Science, University of Bologna, Ozzano dell'Emilia, Bologna, Italy
| | - Antonella Fanelli
- Department of Veterinary Sciences, University of Turin, Grugliasco, Turin, Italy
| | - Luca Licenziato
- Department of Veterinary Sciences, University of Turin, Grugliasco, Turin, Italy
| | - Raffaella De Maria
- Department of Veterinary Sciences, University of Turin, Grugliasco, Turin, Italy
| | - Andrea Rinaldi
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Luca Rotta
- Department of Experimental Oncology, European Institute of Oncology (IEO), Milan, Italy
| | | | - Giovanni Birolo
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Piero Fariselli
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Afua A Mensah
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland. .,Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
| | - Luca Aresu
- Department of Veterinary Sciences, University of Turin, Grugliasco, Turin, Italy.
| |
Collapse
|
27
|
Zhao Z, Yin W, Peng X, Cai Q, He B, Shi S, Peng W, Tu G, Li Y, Li D, Tao Y, Peng M, Wang X, Yu F. A Machine-Learning Approach to Developing a Predictive Signature Based on Transcriptome Profiling of Ground-Glass Opacities for Accurate Classification and Exploring the Immune Microenvironment of Early-Stage LUAD. Front Immunol 2022; 13:872387. [PMID: 35693786 PMCID: PMC9178173 DOI: 10.3389/fimmu.2022.872387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Screening for early-stage lung cancer with low-dose computed tomography is recommended for high-risk populations; consequently, the incidence of pure ground-glass opacity (pGGO) is increasing. Ground-glass opacity (GGO) is considered the appearance of early lung cancer, and there remains an unmet clinical need to understand the pathology of small GGO (<1 cm in diameter). The objective of this study was to use the transcriptome profiling of pGGO specimens <1 cm in diameter to construct a pGGO-related gene risk signature to predict the prognosis of early-stage lung adenocarcinoma (LUAD) and explore the immune microenvironment of GGO. pGGO-related differentially expressed genes (DEGs) were screened to identify prognostic marker genes with two machine learning algorithms. A 15-gene risk signature was constructed from the DEGs that were shared between the algorithms. Risk scores were calculated using the regression coefficients for the pGGO-related DEGs. Patients with Stage I/II LUAD or Stage IA LUAD and high-risk scores had a worse prognosis than patients with low-risk scores. The prognosis of high-risk patients with Stage IA LUAD was almost identical to that of patients with Stage II LUAD, suggesting that treatment strategies for patients with Stage II LUAD may be beneficial in high-risk patients with Stage IA LUAD. pGGO-related DEGs were mainly enriched in immune-related pathways. Patients with high-risk scores and high tumor mutation burden had a worse prognosis and may benefit from immunotherapy. A nomogram was constructed to facilitate the clinical application of the 15-gene risk signature. Receiver operating characteristic curves and decision curve analysis validated the predictive ability of the nomogram in patients with Stage I LUAD in the TCGA-LUAD cohort and GEO datasets.
Collapse
Affiliation(s)
- Zhenyu Zhao
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wei Yin
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiong Peng
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qidong Cai
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Boxue He
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuai Shi
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Weilin Peng
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Guangxu Tu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yunping Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, China
| | | | - Yongguang Tao
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- National Health Council (NHC) Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, China
| | - Muyun Peng
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Xiang Wang, ; Muyun Peng, ; Fenglei Yu,
| | - Xiang Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Xiang Wang, ; Muyun Peng, ; Fenglei Yu,
| | - Fenglei Yu
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Xiang Wang, ; Muyun Peng, ; Fenglei Yu,
| |
Collapse
|
28
|
Cheon H, Xing JC, Moosic KB, Ung J, Chan VW, Chung DS, Toro MF, Elghawy O, Wang JS, Hamele CE, Hardison RC, Olson TL, Tan SF, Feith DJ, Ratan A, Loughran TP. Genomic landscape of TCRαβ and TCRγδ T-large granular lymphocyte leukemia. Blood 2022; 139:3058-3072. [PMID: 35015834 PMCID: PMC9121841 DOI: 10.1182/blood.2021013164] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/18/2021] [Indexed: 11/20/2022] Open
Abstract
Large granular lymphocyte (LGL) leukemia comprises a group of rare lymphoproliferative disorders whose molecular landscape is incompletely defined. We leveraged paired whole-exome and transcriptome sequencing in the largest LGL leukemia cohort to date, which included 105 patients (93 T-cell receptor αβ [TCRαβ] T-LGL and 12 TCRγδ T-LGL). Seventy-six mutations were observed in 3 or more patients in the cohort, and out of those, STAT3, KMT2D, PIK3R1, TTN, EYS, and SULF1 mutations were shared between both subtypes. We identified ARHGAP25, ABCC9, PCDHA11, SULF1, SLC6A15, DDX59, DNMT3A, FAS, KDM6A, KMT2D, PIK3R1, STAT3, STAT5B, TET2, and TNFAIP3 as recurrently mutated putative drivers using an unbiased driver analysis approach leveraging our whole-exome cohort. Hotspot mutations in STAT3, PIK3R1, and FAS were detected, whereas truncating mutations in epigenetic modifying enzymes such as KMT2D and TET2 were observed. Moreover, STAT3 mutations co-occurred with mutations in chromatin and epigenetic modifying genes, especially KMT2D and SETD1B (P < .01 and P < .05, respectively). STAT3 was mutated in 50.5% of the patients. Most common Y640F STAT3 mutation was associated with lower absolute neutrophil count values, and N647I mutation was associated with lower hemoglobin values. Somatic activating mutations (Q160P, D170Y, L287F) in the STAT3 coiled-coil domain were characterized. STAT3-mutant patients exhibited increased mutational burden and enrichment of a mutational signature associated with increased spontaneous deamination of 5-methylcytosine. Finally, gene expression analysis revealed enrichment of interferon-γ signaling and decreased phosphatidylinositol 3-kinase-Akt signaling for STAT3-mutant patients. These findings highlight the clinical and molecular heterogeneity of this rare disorder.
Collapse
Affiliation(s)
- HeeJin Cheon
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - Jeffrey C Xing
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - Katharine B Moosic
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - Johnson Ung
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - Vivian W Chan
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - David S Chung
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - Mariella F Toro
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - Omar Elghawy
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - John S Wang
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - Cait E Hamele
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - Ross C Hardison
- Department of Biochemistry and Molecular Biology, Center for Computational Biology & Bioinformatics, The Pennsylvania State University, State College, PA
| | - Thomas L Olson
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - Su-Fern Tan
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - David J Feith
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| | - Aakrosh Ratan
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA; and
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville VA
| | - Thomas P Loughran
- Department of Medicine, University of Virginia Cancer Center, Charlottesville, VA
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
| |
Collapse
|
29
|
Li Z, Liang H, Zhang S, Luo W. A practical framework RNMF for exploring the association between mutational signatures and genes using gene cumulative contribution abundance. Cancer Med 2022; 11:4053-4069. [PMID: 35575002 PMCID: PMC9636515 DOI: 10.1002/cam4.4717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 03/04/2022] [Accepted: 03/14/2022] [Indexed: 11/17/2022] Open
Abstract
Background Mutational signatures are somatic mutation patterns enriching operational mutational processes, which can provide abundant information about the mechanism of cancer. However, understanding of the pathogenic biological processes is still limited, such as the association between mutational signatures and genes. Methods We developed a simple and practical R package called RNMF (https://github.com/zhenzhang‐li/RNMF) for mutational signature analysis, including a key model of cumulative contribution abundance (CCA), which was designed to highlight the association between mutational signatures and genes and then applying it to a meta‐analysis of 1073 individuals with esophageal squamous cell carcinoma (ESCC). Results We revealed a number of known and previously undescribed SBS or ID signatures, and we found that APOBEC signatures (SBS2* and SBS13*) were closely associated with PIK3CA mutation, especially the E545k mutation. Furthermore, we found that age signature is closely related to the frequent mutation of TP53, of which R342* is highlighted due to strongly linked to age signature. In addition, the CCA matrix image data of genes in the signatures New, SBS3*, and SBS17b* were helpful for the preliminary evaluation of shortened survival outcome. These results can be extended to estimate the distribution of mutations or features, and study the potential impact of clinical factors. Conclusions In a word, RNMF can successfully achieve the correlation analysis of mutational signatures and genes, proving a strong theoretical basis for the study of mutational processes during tumor development.
Collapse
Affiliation(s)
- Zhenzhang Li
- College of Mathematics and Systems Science, Guangdong Polytechnic Normal University, Guangzhou, China.,School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Cloud and Gene AI Research Institute, Guangzhou, China
| | - Haihua Liang
- College of Mathematics and Systems Science, Guangdong Polytechnic Normal University, Guangzhou, China
| | - Shaoan Zhang
- College of Mathematics and Systems Science, Guangdong Polytechnic Normal University, Guangzhou, China.,School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Wen Luo
- College of Mathematics and Systems Science, Guangdong Polytechnic Normal University, Guangzhou, China
| |
Collapse
|
30
|
Abdollahi S, Lin PC, Chiang JH. DiaDeL: An Accurate Deep Learning-Based Model With Mutational Signatures for Predicting Metastasis Stage and Cancer Types. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:1336-1343. [PMID: 34570707 DOI: 10.1109/tcbb.2021.3115504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Mutational signatures help identify cancer-associated genes that are being involved in tumorigenesis pathways. Hence, these pathways guide precision medicine approaches to find appropriate drugs and treatments. The pattern of mutations varies in different cancer types. Some mutations dysregulate protein function so that their accumulation is responsible for cancer development and might be associated with different cancer types. Therefore, mutations as a feature set can be used as an informative candidate to distinguish various cancer types. There are several options for demonstrating mutations. One might employ binary values to demonstrate mutation regions. Another potential method for extracting features is utilizing mutation interpreters. In this study, we investigate the trinucleotide mutational pattern of each cancer type. Moreover, we extract salient NMF-based mutational signatures across various cancer types. Then, we identify cancer-associated genes of a target cancer based on its salient signatures. We evaluate the cancer-associated genes using survival and gene expression analysis in different stages of cancer. Furthermore, we introduce DiaDeL, which is a deep learning-based binary classifier. The DiaDeL model uses mutational signatures as input features and distinct a cancer type from the others. Our proposed model outperforms six state-of-the-art methods with 0.824 and 0.88 for accuracy and AUC, respectively. The source code is available at https://github.com/sabdollahi/DiaDeL.
Collapse
|
31
|
Takenaka IKTM, Bartelli TF, Defelicibus A, Sendoya JM, Golubicki M, Robbio J, Serpa MS, Branco GP, Santos LBC, Claro LCL, Dos Santos GO, Kupper BEC, da Silva IT, Llera AS, de Mello CAL, Riechelmann RP, Dias-Neto E, Iseas S, Aguiar S, Nunes DN. Exome and Tissue-Associated Microbiota as Predictive Markers of Response to Neoadjuvant Treatment in Locally Advanced Rectal Cancer. Front Oncol 2022; 12:809441. [PMID: 35392220 PMCID: PMC8982181 DOI: 10.3389/fonc.2022.809441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
The clinical and pathological responses to multimodal neoadjuvant therapy in locally advanced rectal cancers (LARCs) remain unpredictable, and robust biomarkers are still lacking. Recent studies have shown that tumors present somatic molecular alterations related to better treatment response, and it is also clear that tumor-associated bacteria are modulators of chemotherapy and immunotherapy efficacy, therefore having implications for long-term survivorship and a good potential as the biomarkers of outcome. Here, we performed whole exome sequencing and 16S ribosomal RNA (rRNA) amplicon sequencing from 44 pre-treatment LARC biopsies from Argentinian and Brazilian patients, treated with neoadjuvant chemoradiotherapy or total neoadjuvant treatment, searching for predictive biomarkers of response (responders, n = 17; non-responders, n = 27). In general, the somatic landscape of LARC was not capable to predict a response; however, a significant enrichment in mutational signature SBS5 was observed in non-responders (p = 0.0021), as well as the co-occurrence of APC and FAT4 mutations (p < 0.05). Microbiota studies revealed a similar alpha and beta diversity of bacteria between response groups. Yet, the linear discriminant analysis (LDA) of effect size indicated an enrichment of Hungatella, Flavonifractor, and Methanosphaera (LDA score ≥3) in the pre-treatment biopsies of responders, while non-responders had a higher abundance of Enhydrobacter, Paraprevotella (LDA score ≥3) and Finegoldia (LDA score ≥4). Altogether, the evaluation of these biomarkers in pre-treatment biopsies could eventually predict a neoadjuvant treatment response, while in post-treatment samples, it could help in guiding non-operative treatment strategies.
Collapse
Affiliation(s)
| | - Thais F Bartelli
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Alexandre Defelicibus
- Laboratory of Bioinformatics and Computational Biology, International Center for Research, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Juan M Sendoya
- Laboratorio de Terapia Molecular y Celular - Genomics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mariano Golubicki
- Oncology Unit, Hospital de Gastroenterología Carlos Bonorino Udaondo, Buenos Aires, Argentina.,Clinical Oncology, Intergrupo Argentino para el Tratamiento de los Tumores Gastrointestinales (IATTGI), Buenos Aires, Argentina
| | - Juan Robbio
- Clinical Oncology, Intergrupo Argentino para el Tratamiento de los Tumores Gastrointestinales (IATTGI), Buenos Aires, Argentina
| | - Marianna S Serpa
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Gabriela P Branco
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Luana B C Santos
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Laura C L Claro
- Department of Pathology, A.C.Camargo Cancer Center, São Paulo, Brazil
| | | | - Bruna E C Kupper
- Colorectal Cancer Department, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Israel T da Silva
- Laboratory of Bioinformatics and Computational Biology, International Center for Research, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Andrea S Llera
- Laboratorio de Terapia Molecular y Celular - Genomics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina.,Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Celso A L de Mello
- Department of Clinical Oncology, A.C.Camargo Cancer Center, São Paulo, Brazil
| | | | - Emmanuel Dias-Neto
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil.,Laboratory of Neurosciences (LIM-27) Alzira Denise Hertzog Silva, Institute of Psychiatry, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Soledad Iseas
- Oncology Unit, Hospital de Gastroenterología Carlos Bonorino Udaondo, Buenos Aires, Argentina
| | - Samuel Aguiar
- Colorectal Cancer Department, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Diana Noronha Nunes
- Medical Genomics Laboratory, International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil.,National Institute of Science and Technology in Oncogenomics and Therapeutic Innovation (INCITO), São Paulo, Brazil
| |
Collapse
|
32
|
Giles Doran C, Pennington SR. Copy number alteration signatures as biomarkers in cancer: a review. Biomark Med 2022; 16:371-386. [PMID: 35195030 DOI: 10.2217/bmm-2021-0476] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Within certain cancers, extensive copy number alterations (CNAs) contribute to a complex and heterogenic genomic profile. This makes it difficult to understand and unravel the distinct molecular dynamics shaping the disease while preventing clinically effective patient stratification. CNA signature analysis represents a novel genomic stratification tool for probing this complexity, offering an intricate framework for deriving CNA patterns at the molecular level. This allows the underlying genomic mechanisms of specific cancers to be revealed, leading to the potential identification of therapeutic targets and prognostic associations. This review outlines the molecular and methodological basis of CNA signatures and focuses on recent advances highlighting their clinical utility, limitations and prospective future as novel diagnostic and prognostic cancer biomarkers.
Collapse
Affiliation(s)
- Conor Giles Doran
- UCD Conway Institute, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Stephen R Pennington
- UCD Conway Institute, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
33
|
Lee D, Wang D, Yang XR, Shi J, Landi MT, Zhu B. SUITOR: Selecting the number of mutational signatures through cross-validation. PLoS Comput Biol 2022; 18:e1009309. [PMID: 35377867 PMCID: PMC9009674 DOI: 10.1371/journal.pcbi.1009309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 04/14/2022] [Accepted: 03/09/2022] [Indexed: 11/19/2022] Open
Abstract
For de novo mutational signature analysis, the critical first step is to decide how many signatures should be expected in a cancer genomics study. An incorrect number could mislead downstream analyses. Here we present SUITOR (Selecting the nUmber of mutatIonal signaTures thrOugh cRoss-validation), an unsupervised cross-validation method that requires little assumptions and no numerical approximations to select the optimal number of signatures without overfitting the data. In vitro studies and in silico simulations demonstrated that SUITOR can correctly identify signatures, some of which were missed by other widely used methods. Applied to 2,540 whole-genome sequenced tumors across 22 cancer types, SUITOR selected signatures with the smallest prediction errors and almost all signatures of breast cancer selected by SUITOR were validated in an independent breast cancer study. SUITOR is a powerful tool to select the optimal number of mutational signatures, facilitating downstream analyses with etiological or therapeutic importance.
Collapse
Affiliation(s)
- Donghyuk Lee
- Department of Statistics, Pusan National University, Busan, Korea
| | - Difei Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Xiaohong R. Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
34
|
Fan Y, Zhou Y, Lou M, Li X, Zhu X, Yuan K. m6A Regulator-Mediated Methylation Modification Patterns and Characterisation of Tumour Microenvironment Infiltration in Non-Small Cell Lung Cancer. J Inflamm Res 2022; 15:1969-1989. [PMID: 35356071 PMCID: PMC8958726 DOI: 10.2147/jir.s356841] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- Yongfei Fan
- Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Yong Zhou
- Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Ming Lou
- Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Xinwei Li
- Department of Gastroenterology, Affiliated Cancer Hospital of Bengbu Medical College, Bengbu, People’s Republic of China
| | - Xudong Zhu
- Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
| | - Kai Yuan
- Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
- Heart and Lung Disease Laboratory, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, People’s Republic of China
- Correspondence: Kai Yuan, Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, No. 29 Xinglong Lane, Changzhou, 213003, Jiangsu Province, People’s Republic of China, Tel +86-13915890721, Email
| |
Collapse
|
35
|
Manders F, Brandsma AM, de Kanter J, Verheul M, Oka R, van Roosmalen MJ, van der Roest B, van Hoeck A, Cuppen E, van Boxtel R. MutationalPatterns: the one stop shop for the analysis of mutational processes. BMC Genomics 2022; 23:134. [PMID: 35168570 PMCID: PMC8845394 DOI: 10.1186/s12864-022-08357-3] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/01/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The collective of somatic mutations in a genome represents a record of mutational processes that have been operative in a cell. These processes can be investigated by extracting relevant mutational patterns from sequencing data. RESULTS Here, we present the next version of MutationalPatterns, an R/Bioconductor package, which allows in-depth mutational analysis of catalogues of single and double base substitutions as well as small insertions and deletions. Major features of the package include the possibility to perform regional mutation spectra analyses and the possibility to detect strand asymmetry phenomena, such as lesion segregation. On top of this, the package also contains functions to determine how likely it is that a signature can cause damaging mutations (i.e., mutations that affect protein function). This updated package supports stricter signature refitting on known signatures in order to prevent overfitting. Using simulated mutation matrices containing varied signature contributions, we showed that reliable refitting can be achieved even when only 50 mutations are present per signature. Additionally, we incorporated bootstrapped signature refitting to assess the robustness of the signature analyses. Finally, we applied the package on genome mutation data of cell lines in which we deleted specific DNA repair processes and on large cancer datasets, to show how the package can be used to generate novel biological insights. CONCLUSIONS This novel version of MutationalPatterns allows for more comprehensive analyses and visualization of mutational patterns in order to study the underlying processes. Ultimately, in-depth mutational analyses may contribute to improved biological insights in mechanisms of mutation accumulation as well as aid cancer diagnostics. MutationalPatterns is freely available at http://bioconductor.org/packages/MutationalPatterns .
Collapse
Affiliation(s)
- Freek Manders
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584CS, Utrecht, The Netherlands
- Oncode Institute, Jaarbeursplein 6, 3521 AL, Utrecht, The Netherlands
| | - Arianne M Brandsma
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584CS, Utrecht, The Netherlands
- Oncode Institute, Jaarbeursplein 6, 3521 AL, Utrecht, The Netherlands
| | - Jurrian de Kanter
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584CS, Utrecht, The Netherlands
- Oncode Institute, Jaarbeursplein 6, 3521 AL, Utrecht, The Netherlands
| | - Mark Verheul
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584CS, Utrecht, The Netherlands
- Oncode Institute, Jaarbeursplein 6, 3521 AL, Utrecht, The Netherlands
| | - Rurika Oka
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584CS, Utrecht, The Netherlands
- Oncode Institute, Jaarbeursplein 6, 3521 AL, Utrecht, The Netherlands
| | - Markus J van Roosmalen
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584CS, Utrecht, The Netherlands
- Oncode Institute, Jaarbeursplein 6, 3521 AL, Utrecht, The Netherlands
| | - Bastiaan van der Roest
- Oncode Institute, Jaarbeursplein 6, 3521 AL, Utrecht, The Netherlands
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Arne van Hoeck
- Oncode Institute, Jaarbeursplein 6, 3521 AL, Utrecht, The Netherlands
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Edwin Cuppen
- Oncode Institute, Jaarbeursplein 6, 3521 AL, Utrecht, The Netherlands
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Ruben van Boxtel
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584CS, Utrecht, The Netherlands.
- Oncode Institute, Jaarbeursplein 6, 3521 AL, Utrecht, The Netherlands.
| |
Collapse
|
36
|
Kim M, Hwang J, Kim KA, Hwang S, Lee HJ, Jung JY, Lee JG, Cha YJ, Shim HS. Genomic characteristics of invasive mucinous adenocarcinoma of the lung with multiple pulmonary sites of involvement. Mod Pathol 2022; 35:202-209. [PMID: 34290355 PMCID: PMC8786658 DOI: 10.1038/s41379-021-00872-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/13/2022]
Abstract
Invasive mucinous adenocarcinoma (IMA) of the lung frequently presents with diffuse pneumonic-type features or multifocal lesions, which are regarded as a pattern of intrapulmonary metastases. However, the genomics of multifocal IMAs have not been well studied. We performed whole exome sequencing on samples taken from 2 to 5 regions in seven patients with synchronous multifocal IMAs of the lung (24 regions total). Early initiating driver events, such as KRAS, NKX2-1, TP53, or ARID1A mutations, are clonal mutations and were present in all multifocal IMAs in each patient. The tumor mutational burden of multifocal IMAs was low (mean: 1.13/mega base), but further analyses suggested intra-tumor heterogeneity. The mutational signature analysis found that IMAs were predominantly associated with endogenous mutational process (signature 1), APOBEC activity (signatures 2 and 13), and defective DNA mismatch repair (signature 6), but not related to smoking signature. IMAs synchronously located in the bilateral lower lobes of two patients with background usual interstitial pneumonia had different mutation types, suggesting that they were double primaries. In conclusion, genomic evidence found in this study indicated the clonal intrapulmonary spread of diffuse pneumonic-type or multifocal IMAs, although they can occur in multicentric origins in the background of usual interstitial pneumonia. IMAs exhibited a heterogeneous genomic landscape despite the low somatic mutation burden. Further studies are warranted to determine the clinical significance of the genomic characteristics of IMAs in expanded cohorts.
Collapse
Affiliation(s)
- Moonsik Kim
- Department of Pathology, Kyungpook National University Chilgok Hospital, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Jinha Hwang
- Macrogen Inc., Seoul, Republic of Korea
- Department of Laboratory Medicine, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Kyung A Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sohyun Hwang
- Department of Pathology, CHA University School of Medicine, Seongnam, Republic of Korea
| | - Hye-Jeong Lee
- Department of Radiology and Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji Ye Jung
- Division of Pulmonology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Gu Lee
- Department of Thoracic and Cardiovascular Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoon Jin Cha
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyo Sup Shim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
37
|
Wu Y, Chua EHZ, Ng AWT, Boot A, Rozen SG. Accuracy of mutational signature software on correlated signatures. Sci Rep 2022; 12:390. [PMID: 35013428 PMCID: PMC8748538 DOI: 10.1038/s41598-021-04207-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/17/2021] [Indexed: 11/09/2022] Open
Abstract
Mutational signatures are characteristic patterns of mutations generated by exogenous mutagens or by endogenous mutational processes. Mutational signatures are important for research into DNA damage and repair, aging, cancer biology, genetic toxicology, and epidemiology. Unsupervised learning can infer mutational signatures from the somatic mutations in large numbers of tumors, and separating correlated signatures is a notable challenge for this task. To investigate which methods can best meet this challenge, we assessed 18 computational methods for inferring mutational signatures on 20 synthetic data sets that incorporated varying degrees of correlated activity of two common mutational signatures. Performance varied widely, and four methods noticeably outperformed the others: hdp (based on hierarchical Dirichlet processes), SigProExtractor (based on multiple non-negative matrix factorizations over resampled data), TCSM (based on an approach used in document topic analysis), and mutSpec.NMF (also based on non-negative matrix factorization). The results underscored the complexities of mutational signature extraction, including the importance and difficulty of determining the correct number of signatures and the importance of hyperparameters. Our findings indicate directions for improvement of the software and show a need for care when interpreting results from any of these methods, including the need for assessing sensitivity of the results to input parameters.
Collapse
Affiliation(s)
- Yang Wu
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Ellora Hui Zhen Chua
- Department of Biological Sciences, National University of Singapore, Singapore, 117558, Singapore
| | - Alvin Wei Tian Ng
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Arnoud Boot
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Steven G Rozen
- Programme in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.
| |
Collapse
|
38
|
Sason I, Chen Y, Leiserson MDM, Sharan R. A mixture model for signature discovery from sparse mutation data. Genome Med 2021; 13:173. [PMID: 34724984 PMCID: PMC8559697 DOI: 10.1186/s13073-021-00988-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Mutational signatures are key to understanding the processes that shape cancer genomes, yet their analysis requires relatively rich whole-genome or whole-exome mutation data. Recently, orders-of-magnitude sparser gene-panel-sequencing data have become increasingly available in the clinic. To deal with such sparse data, we suggest a novel mixture model, Mix. In application to simulated and real gene-panel sequences, Mix is shown to outperform current approaches and yield mutational signatures and patient stratifications that are in higher agreement with the literature. We further demonstrate its utility in several clinical settings, successfully predicting therapy benefit and patient groupings from MSK-IMPACT pan-cancer data. Availability: https://github.com/itaysason/Mix-MMM .
Collapse
Affiliation(s)
- Itay Sason
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Yuexi Chen
- Department of Computer Science and Center for Bioinformatics and Computational Biology, University of Maryland, College Park, 20742, MD, USA
| | - Mark D M Leiserson
- Department of Computer Science and Center for Bioinformatics and Computational Biology, University of Maryland, College Park, 20742, MD, USA
| | - Roded Sharan
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
39
|
Abbasi A, Alexandrov LB. Significance and limitations of the use of next-generation sequencing technologies for detecting mutational signatures. DNA Repair (Amst) 2021; 107:103200. [PMID: 34411908 PMCID: PMC9478565 DOI: 10.1016/j.dnarep.2021.103200] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/13/2022]
Abstract
Next generation sequencing technologies (NGS) have been critical in characterizing the genomic landscape and untangling the genetic heterogeneity of human cancer. Since its advent, NGS has played a pivotal role in identifying the patterns of somatic mutations imprinted on cancer genomes and in deciphering the signatures of the mutational processes that have generated these patterns. Mutational signatures serve as phenotypic molecular footprints of exposures to environmental factors as well as deficiency and infidelity of DNA replication and repair pathways. Since the first roadmap of mutational signatures in human cancer was generated from whole-genome and whole-exome sequencing data, there has been a growing interest to extract mutational signatures from other NGS technologies such as targeted panel sequencing, RNA sequencing, single-cell sequencing, duplex sequencing, reduced representation sequencing, and long-read sequencing. Many of these technologies have their inherent sequencing biases and produce technical artifacts that can confound the extraction of reliable and interpretable mutational signatures. In this review, we highlight the relevance, limitations, and prospects of using different NGS technologies for examining mutational patterns and for deciphering mutational signatures.
Collapse
Affiliation(s)
- Ammal Abbasi
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, 92093, USA; Department of Bioengineering, UC San Diego, La Jolla, CA, 92093, USA; Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA, 92093, USA; Department of Bioengineering, UC San Diego, La Jolla, CA, 92093, USA; Moores Cancer Center, UC San Diego, La Jolla, CA, 92037, USA.
| |
Collapse
|
40
|
Koh G, Degasperi A, Zou X, Momen S, Nik-Zainal S. Mutational signatures: emerging concepts, caveats and clinical applications. Nat Rev Cancer 2021; 21:619-637. [PMID: 34316057 DOI: 10.1038/s41568-021-00377-7] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 02/05/2023]
Abstract
Whole-genome sequencing has brought the cancer genomics community into new territory. Thanks to the sheer power provided by the thousands of mutations present in each patient's cancer, we have been able to discern generic patterns of mutations, termed 'mutational signatures', that arise during tumorigenesis. These mutational signatures provide new insights into the causes of individual cancers, revealing both endogenous and exogenous factors that have influenced cancer development. This Review brings readers up to date in a field that is expanding in computational, experimental and clinical directions. We focus on recent conceptual advances, underscoring some of the caveats associated with using the mutational signature frameworks and highlighting the latest experimental insights. We conclude by bringing attention to areas that are likely to see advancements in clinical applications.
Collapse
Affiliation(s)
- Gene Koh
- Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- MRC Cancer Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Andrea Degasperi
- Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- MRC Cancer Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Xueqing Zou
- Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- MRC Cancer Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Sophie Momen
- Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- MRC Cancer Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Serena Nik-Zainal
- Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
- MRC Cancer Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
41
|
Wojtowicz D, Hoinka J, Amgalan B, Kim YA, Przytycka TM. RepairSig: Deconvolution of DNA damage and repair contributions to the mutational landscape of cancer. Cell Syst 2021; 12:994-1003.e4. [PMID: 34375586 DOI: 10.1016/j.cels.2021.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/06/2021] [Accepted: 07/14/2021] [Indexed: 01/05/2023]
Abstract
Cancer genomes accumulate a large number of somatic mutations resulting from a combination of stochastic errors in DNA processing, cancer-related aberrations of the DNA repair machinery, or carcinogenic exposures; each mutagenic process leaves a characteristic mutational signature. A key challenge is understanding the interactions between signatures, particularly as DNA repair deficiencies often modify the effects of other mutagens. Here, we introduce RepairSig, a computational method that explicitly models additive primary mutagenic processes; non-additive secondary processes, which interact with the primary processes; and a mutation opportunity, that is, the distribution of sites across the genome that are vulnerable to damage or preferentially repaired. We demonstrate that RepairSig accurately recapitulates experimentally identified signatures, identifies autonomous signatures of deficient DNA repair processes, and explains mismatch repair deficiency in breast cancer by de novo inference of both primary and secondary signatures from patient data. RepairSig is freely available for download at https://github.com/ncbi/RepairSig.
Collapse
Affiliation(s)
- Damian Wojtowicz
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA.
| | - Jan Hoinka
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Bayarbaatar Amgalan
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Yoo-Ah Kim
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - Teresa M Przytycka
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA.
| |
Collapse
|
42
|
Lin SH, Wang Y, Hartley SW, Karyadi DM, Lee OW, Zhu B, Zhou W, Brown DW, Beilstein-Wedel E, Hazra R, Kacanek D, Chadwick EG, Marsit CJ, Poirier MC, Brummel SS, Chanock SJ, Engels EA, Machiela MJ. In-utero exposure to zidovudine-containing antiretroviral therapy and clonal hematopoiesis in HIV-exposed uninfected newborns. AIDS 2021; 35:1525-1535. [PMID: 33756513 PMCID: PMC8286286 DOI: 10.1097/qad.0000000000002894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Zidovudine (ZDV) has been extensively used in pregnant women to prevent vertical transmission of HIV but few studies have evaluated potential mutagenic effects of ZDV during fetal development. DESIGN Our study investigated clonal hematopoiesis in HIV-exposed uninfected (HEU) newborns, 94 of whom were ZDV-exposed and 91 antiretroviral therapy (ART)-unexposed and matched for potential confounding factors. METHODS Utilizing high depth sequencing and genotyping arrays, we comprehensively examined blood samples collected during the first week after birth for potential clonal hematopoiesis associated with fetal ZDV exposure, including clonal single nucleotide variants (SNVs), small insertions and deletions (indels), and large structural copy number or copy neutral alterations. RESULTS We observed no statistically significant difference in the number of SNVs and indels per person in ZDV-exposed children (adjusted ratio [95% confidence interval, CI] for expected number of mutations = 0.79 [0.50--1.22], P = 0.3), and no difference in the number of large structural alterations. Mutations in common clonal hematopoiesis driver genes were not found in the study population. Mutational signature analyses on SNVs detected no novel signatures unique to the ZDV-exposed children and the mutational profiles were similar between the two groups. CONCLUSION Our results suggest that clonal hematopoiesis at levels detectable in our study is not strongly influenced by in-utero ZDV exposure; however, additional follow-up studies are needed to further evaluate the safety and potential long-term impacts of in-utero ZDV exposure in HEU children as well as better investigate genomic aberrations occurring late in pregnancy.
Collapse
Affiliation(s)
- Shu-Hong Lin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Youjin Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Stephen W Hartley
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Danielle M Karyadi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Olivia W Lee
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Weiyin Zhou
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Derek W Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Erin Beilstein-Wedel
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Rohan Hazra
- Maternal and Pediatric Infectious Disease Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Deborah Kacanek
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Ellen G Chadwick
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Carmen J Marsit
- Departments of Environmental Health and Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Miriam C Poirier
- Carcinogen-DNA Interactions Section, Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sean S Brummel
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Eric A Engels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville
| |
Collapse
|
43
|
Kim YA, Leiserson MDM, Moorjani P, Sharan R, Wojtowicz D, Przytycka TM. Mutational Signatures: From Methods to Mechanisms. Annu Rev Biomed Data Sci 2021; 4:189-206. [PMID: 34465178 DOI: 10.1146/annurev-biodatasci-122320-120920] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mutations are the driving force of evolution, yet they underlie many diseases, in particular, cancer. They are thought to arise from a combination of stochastic errors in DNA processing, naturally occurring DNA damage (e.g., the spontaneous deamination of methylated CpG sites), replication errors, and dysregulation of DNA repair mechanisms. High-throughput sequencing has made it possible to generate large datasets to study mutational processes in health and disease. Since the emergence of the first mutational process studies in 2012, this field is gaining increasing attention and has already accumulated a host of computational approaches and biomedical applications.
Collapse
Affiliation(s)
- Yoo-Ah Kim
- National Center of Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894, USA;
| | - Mark D M Leiserson
- Department of Computer Science and Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland 20742, USA
| | - Priya Moorjani
- Department of Molecular and Cell Biology and Center for Computational Biology, University of California, Berkeley, California 94720, USA
| | - Roded Sharan
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv 69978, Israel
| | - Damian Wojtowicz
- National Center of Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894, USA;
| | - Teresa M Przytycka
- National Center of Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894, USA;
| |
Collapse
|
44
|
Prasanna A, Niranjan V. MutVis: Automated framework for analysis and visualization of mutational signatures in pathogenic bacterial strains. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2021; 91:104805. [PMID: 33689914 DOI: 10.1016/j.meegid.2021.104805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 12/17/2020] [Accepted: 03/04/2021] [Indexed: 12/01/2022]
Abstract
In recent years, mutational signature analysis has become a routine practice in cancer genomics for classification and diagnosis. Characterizing mutational signatures across species or within genomes of a bacteria helps in understanding their evolution and adaptation. However, an integrated framework for analysis and visualization of mutational signatures in bacterial genome is lacking. Hence, we aim to develop an integrated, automated, open-source and user-friendly framework called MutVis to analyze mutational signatures from bacterial whole genome next generation sequencing data. The current framework integrates various publicly available packages using Snakemake workflow management software, Python and R scripting. MutVis supports variant calling, transition (Ti) and transversion (Tv) graphical representation, generation of mutational count matrix, graphical visualization of base-pair substitution spectrum (BPSs) and mutation signatures extraction. TvTi plots provide the 6 base substitution classification for both genome and gene level. Further resolution of base pair substitution classification is provided as 96-profile BPSs plot. Mutation signatures is derived based on the characteristic pattern observed in BPSs using non-negative matrix factorization. Relative contribution of signatures is given as hierarchically clustered heatmap. This provides information on active signatures in the individual given sample and classify samples according to signature contributions. We demonstrated the MutVis framework using geographically different strains of Mycobacterium tuberculosis, downloaded from PATRIC TB-ARC Antibiotic Resistance Catalog (n = 963). The current framework can be used to study mutation biases and characteristic mutational signatures in bacterial genomes and is freely available at https://github.com/AkshathaPrasanna/MutVis.
Collapse
Affiliation(s)
- Akshatha Prasanna
- Department of Biotechnology, RV College of Engineering, Bengaluru, Karnataka 560059, India
| | - Vidya Niranjan
- Department of Biotechnology, RV College of Engineering, Bengaluru, Karnataka 560059, India..
| |
Collapse
|
45
|
Lal A, Liu K, Tibshirani R, Sidow A, Ramazzotti D. De novo mutational signature discovery in tumor genomes using SparseSignatures. PLoS Comput Biol 2021; 17:e1009119. [PMID: 34181655 PMCID: PMC8270462 DOI: 10.1371/journal.pcbi.1009119] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/09/2021] [Accepted: 05/27/2021] [Indexed: 11/18/2022] Open
Abstract
Cancer is the result of mutagenic processes that can be inferred from tumor genomes by analyzing rate spectra of point mutations, or "mutational signatures". Here we present SparseSignatures, a novel framework to extract signatures from somatic point mutation data. Our approach incorporates a user-specified background signature, employs regularization to reduce noise in non-background signatures, uses cross-validation to identify the number of signatures, and is scalable to large datasets. We show that SparseSignatures outperforms current state-of-the-art methods on simulated data using a variety of standard metrics. We then apply SparseSignatures to whole genome sequences of pancreatic and breast tumors, discovering well-differentiated signatures that are linked to known mutagenic mechanisms and are strongly associated with patient clinical features.
Collapse
Affiliation(s)
- Avantika Lal
- Department of Pathology, Stanford University, Stanford, California, United States of America
| | - Keli Liu
- Department of Statistics, Stanford University, Stanford, California, United States of America
| | - Robert Tibshirani
- Department of Statistics, Stanford University, Stanford, California, United States of America
- Department of Biomedical Data Science, Stanford University, Stanford, California, United States of America
| | - Arend Sidow
- Department of Pathology, Stanford University, Stanford, California, United States of America
- Department of Genetics, Stanford University, Stanford, California, United States of America
| | - Daniele Ramazzotti
- Department of Pathology, Stanford University, Stanford, California, United States of America
- Department of Computer Science, Stanford University, Stanford, California, United States of America
| |
Collapse
|
46
|
Buttura JR, Provisor Santos MN, Valieris R, Drummond RD, Defelicibus A, Lima JP, Calsavara VF, Freitas HC, Cordeiro de Lima VC, Fernanda Bartelli T, Wiedner M, Rosales R, Gollob KJ, Loizou J, Dias-Neto E, Nunes DN, da Silva IT. Mutational Signatures Driven by Epigenetic Determinants Enable the Stratification of Patients with Gastric Cancer for Therapeutic Intervention. Cancers (Basel) 2021; 13:490. [PMID: 33513945 PMCID: PMC7866019 DOI: 10.3390/cancers13030490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/20/2020] [Indexed: 12/30/2022] Open
Abstract
DNA mismatch repair deficiency (dMMR) is associated with the microsatellite instability (MSI) phenotype and leads to increased mutation load, which in turn may impact anti-tumor immune responses and treatment effectiveness. Various mutational signatures directly linked to dMMR have been described for primary cancers. To investigate which mutational signatures are associated with prognosis in gastric cancer, we performed a de novo extraction of mutational signatures in a cohort of 787 patients. We detected three dMMR-related signatures, one of which clearly discriminates tumors with MLH1 gene silencing caused by promoter hypermethylation (area under the curve = 98%). We then demonstrated that samples with the highest exposure of this signature share features related to better prognosis, encompassing clinical and molecular aspects and altered immune infiltrate composition. Overall, the assessment of the prognostic value and of the impact of modifications in MMR-related genes on shaping specific dMMR mutational signatures provides evidence that classification based on mutational signature exposure enables prognosis stratification.
Collapse
Affiliation(s)
- Jaqueline Ramalho Buttura
- Laboratory of Bioinformatics and Computational Biology, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (J.R.B.); (M.N.P.S.); (R.V.); (R.D.D.); (A.D.); (J.P.L.)
| | - Monize Nakamoto Provisor Santos
- Laboratory of Bioinformatics and Computational Biology, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (J.R.B.); (M.N.P.S.); (R.V.); (R.D.D.); (A.D.); (J.P.L.)
- Department of Genomics, Fleury Group, São Paulo 04344-070, Brazil
| | - Renan Valieris
- Laboratory of Bioinformatics and Computational Biology, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (J.R.B.); (M.N.P.S.); (R.V.); (R.D.D.); (A.D.); (J.P.L.)
| | - Rodrigo Duarte Drummond
- Laboratory of Bioinformatics and Computational Biology, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (J.R.B.); (M.N.P.S.); (R.V.); (R.D.D.); (A.D.); (J.P.L.)
| | - Alexandre Defelicibus
- Laboratory of Bioinformatics and Computational Biology, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (J.R.B.); (M.N.P.S.); (R.V.); (R.D.D.); (A.D.); (J.P.L.)
| | - João Paulo Lima
- Laboratory of Bioinformatics and Computational Biology, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (J.R.B.); (M.N.P.S.); (R.V.); (R.D.D.); (A.D.); (J.P.L.)
| | | | - Helano Carioca Freitas
- Medical Oncology Department, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (H.C.F.); (V.C.C.d.L.)
- Laboratory of Medical Genomics, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (T.F.B.); (E.D.-N.); (D.N.N.)
| | - Vladmir C. Cordeiro de Lima
- Medical Oncology Department, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (H.C.F.); (V.C.C.d.L.)
- Translational Immuno-Oncology Group, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil;
| | - Thais Fernanda Bartelli
- Laboratory of Medical Genomics, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (T.F.B.); (E.D.-N.); (D.N.N.)
| | - Marc Wiedner
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; (M.W.); (J.L.)
| | - Rafael Rosales
- Department of Mathematics and Computer Science, University of São Paulo, Ribeirão Preto 14049-900, Brazil;
| | - Kenneth John Gollob
- Translational Immuno-Oncology Group, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil;
| | - Joanna Loizou
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria; (M.W.); (J.L.)
- Department of Medicine, Institute of Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, 1090 Vienna, Austria
| | - Emmanuel Dias-Neto
- Laboratory of Medical Genomics, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (T.F.B.); (E.D.-N.); (D.N.N.)
- Laboratory of Neurosciences, Institute of Psychiatry, University of São Paulo, São Paulo 05403-903, Brazil
| | - Diana Noronha Nunes
- Laboratory of Medical Genomics, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (T.F.B.); (E.D.-N.); (D.N.N.)
| | - Israel Tojal da Silva
- Laboratory of Bioinformatics and Computational Biology, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (J.R.B.); (M.N.P.S.); (R.V.); (R.D.D.); (A.D.); (J.P.L.)
| |
Collapse
|
47
|
Abstract
The genome of a cancer contains somatic mutations that reflect the activities of endogenous and exogenous mutational processes, with each mutational process imprinting a characteristic mutational signature. Computational analysis of somatic mutations derived from next-generation sequencing data allows revealing the mutational signatures operative in a set of cancer genomes. In this chapter, we briefly review the concept of mutational signatures and the tools available for deciphering mutational signatures. Further, we provide a quick guide as well as an in-depth protocol for deciphering mutational signatures using the tool SigProfilerExtractor and review the results generated from an example dataset of cancer genomes.
Collapse
|
48
|
Valieris R, Amaro L, Osório CABDT, Bueno AP, Rosales Mitrowsky RA, Carraro DM, Nunes DN, Dias-Neto E, da Silva IT. Deep Learning Predicts Underlying Features on Pathology Images with Therapeutic Relevance for Breast and Gastric Cancer. Cancers (Basel) 2020; 12:cancers12123687. [PMID: 33316873 PMCID: PMC7763049 DOI: 10.3390/cancers12123687] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/22/2022] Open
Abstract
Simple Summary DNA repair deficiency (DRD) is common in many cancers. This deficiency contributes to pathogenesis of the disease, but it also presents an opportunity for therapeutic targeting. However, current DRD identification assays are not available for all patients. We propose an efficient machine learning algorithm which can predict DRD from histopathological images. The utility of our method was shown by considering the detection of homologous recombination deficiency (HRD) and mismatch repair deficiency (MMRD) in breast and gastric cancer respectively. Our findings demonstrate that machine-learning approaches can be used in advanced applications to assist therapy decisions. Abstract DNA repair deficiency (DRD) is an important driver of carcinogenesis and an efficient target for anti-tumor therapies to improve patient survival. Thus, detection of DRD in tumors is paramount. Currently, determination of DRD in tumors is dependent on wet-lab assays. Here we describe an efficient machine learning algorithm which can predict DRD from histopathological images. The utility of this algorithm is demonstrated with data obtained from 1445 cancer patients. Our method performs rather well when trained on breast cancer specimens with homologous recombination deficiency (HRD), AUC (area under curve) = 0.80. Results for an independent breast cancer cohort achieved an AUC = 0.70. The utility of our method was further shown by considering the detection of mismatch repair deficiency (MMRD) in gastric cancer, yielding an AUC = 0.81. Our results demonstrate the capacity of our learning-base system as a low-cost tool for DRD detection.
Collapse
Affiliation(s)
- Renan Valieris
- Laboratory of Computational Biology Bioinformatics, CIPE/A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (R.V.); (L.A.); (A.P.B.)
| | - Lucas Amaro
- Laboratory of Computational Biology Bioinformatics, CIPE/A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (R.V.); (L.A.); (A.P.B.)
| | | | - Adriana Passos Bueno
- Laboratory of Computational Biology Bioinformatics, CIPE/A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (R.V.); (L.A.); (A.P.B.)
- Department of Pathology, CIPE/A.C. Camargo Cancer Center, São Paulo 01525-001, Brazil;
| | | | - Dirce Maria Carraro
- Laboratory of Genomics and Molecular Biology, CIPE/A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil;
| | - Diana Noronha Nunes
- Medical Genomics Laboratory, CIPE/A.C. Camargo Cancer Center, São Paulo 01525-001, Brazil; (D.N.N.); (E.D.-N.)
| | - Emmanuel Dias-Neto
- Medical Genomics Laboratory, CIPE/A.C. Camargo Cancer Center, São Paulo 01525-001, Brazil; (D.N.N.); (E.D.-N.)
| | - Israel Tojal da Silva
- Laboratory of Computational Biology Bioinformatics, CIPE/A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (R.V.); (L.A.); (A.P.B.)
- Correspondence: ; Tel.: +55-11-2189-5000
| |
Collapse
|
49
|
Parallelized Latent Dirichlet Allocation Provides a Novel Interpretability of Mutation Signatures in Cancer Genomes. Genes (Basel) 2020; 11:genes11101127. [PMID: 32992754 PMCID: PMC7600398 DOI: 10.3390/genes11101127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 12/22/2022] Open
Abstract
Mutation signatures are defined as the distribution of specific mutations such as activity of AID/APOBEC family proteins. Previous studies have reported numerous signatures, using matrix factorization methods for mutation catalogs. Different mutation signatures are active in different tumor types; hence, signature activity varies greatly among tumor types and becomes sparse. Because of this, many previous methods require dividing mutation catalogs for each tumor type. Here, we propose parallelized latent Dirichlet allocation (PLDA), a novel Bayesian model to simultaneously predict mutation signatures with all mutation catalogs. PLDA is an extended model of latent Dirichlet allocation (LDA), which is one of the methods used for signature prediction. It has parallelized hyperparameters of Dirichlet distributions for LDA, and they represent the sparsity of signature activities for each tumor type, thus facilitating simultaneous analyses. First, we conducted a simulation experiment to compare PLDA with previous methods (including SigProfiler and SignatureAnalyzer) using artificial data and confirmed that PLDA could predict signature structures as accurately as previous methods without searching for the optimal hyperparameters. Next, we applied PLDA to PCAWG (Pan-Cancer Analysis of Whole Genomes) mutation catalogs and obtained a signature set different from the one predicted by SigProfiler. Further, we have shown that the mutation spectrum represented by the predicted signature with PLDA provides a novel interpretability through post-analyses.
Collapse
|
50
|
Hu X, Xu Z, De S. Characteristics of mutational signatures of unknown etiology. NAR Cancer 2020; 2:zcaa026. [PMID: 33015626 PMCID: PMC7520824 DOI: 10.1093/narcan/zcaa026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/01/2020] [Accepted: 09/23/2020] [Indexed: 12/25/2022] Open
Abstract
Although not all somatic mutations are cancer drivers, their mutational signatures, i.e. the patterns of genomic alterations at a genome-wide scale, provide insights into past exposure to mutagens, DNA damage and repair processes. Computational deconvolution of somatic mutation patterns and expert curation pan-cancer studies have identified a number of mutational signatures associated with point mutations, dinucleotide substitutions, insertions and deletions, and rearrangements, and have established etiologies for a subset of these signatures. However, the mechanisms underlying nearly one-third of all mutational signatures are not yet understood. The signatures with established etiology and those with hitherto unknown origin appear to have some differences in strand bias, GC content and nucleotide context diversity. It is possible that some of the hitherto ‘unknown’ signatures predominantly occur outside gene regions. While nucleotide contexts might be adequate to establish etiologies of some mutational signatures, in other cases additional features, such as broader (epi)genomic contexts, including chromatin, replication timing, processivity and local mutational patterns, may help fully understand the underlying DNA damage and repair processes. Nonetheless, remarkable progress in characterization of mutational signatures has provided fundamental insights into the biology of cancer, informed disease etiology and opened up new opportunities for cancer prevention, risk management, and therapeutic decision making.
Collapse
Affiliation(s)
- Xiaoju Hu
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Zhuxuan Xu
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Subhajyoti De
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|