1
|
Al-Rahahleh RQ, Roos WP, Saville KM, Andrews JF, Wu Z, Koczor CA, Prakash A, Sobol RW. Overexpression of the WWE domain of RNF146 modulates poly-(ADP)-ribose dynamics at sites of DNA damage. DNA Repair (Amst) 2025; 150:103845. [PMID: 40403420 DOI: 10.1016/j.dnarep.2025.103845] [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: 01/04/2024] [Revised: 04/08/2025] [Accepted: 05/06/2025] [Indexed: 05/24/2025]
Abstract
Protein poly-ADP-ribosylation (PARylation) is a post-translational modification formed by transferring successive units of ADP-ribose to target proteins to form poly-ADP-ribose (PAR) chains. PAR plays a critical role in the DNA damage response (DDR) by acting as a signaling platform to promote the recruitment of DNA repair factors to the sites of DNA damage that bind via their PAR-binding domains (PBDs). Several classes of PBD families have been identified, which recognize distinct parts of the PAR chain. Proteins encoding PBDs play an essential role in conveying the PAR-mediated signal through their interaction with PAR chains, which mediates many cellular functions, including the DDR. The WWE domain, encoded in 12 human proteins, identifies the iso-ADP-ribose moiety of the PAR chain. PARylation is a heterogeneous structure that is highly dynamic in cells. Capturing the dynamics of PARylation is essential to understanding its role in the DDR, which can be achieved by expanding the tool kit for PAR detection and tracking mediated by the unique binding capability of various sensors. We recently described the WWE domain of RNF146 as a robust genetically encoded probe, when fused to EGFP, for the detection of PAR in live cells. Expanding on this, we used structural prediction tools to evaluate all of the WWE domains encoded in human proteins, evaluating each as molecular PAR probes in live cells. We demonstrate unique PAR dynamics when tracked by WWE-encoded PAR binding domains, in addition to an engineered macrodomain, that can be exploited for modulation of the PAR-dependent DNA damage response.
Collapse
Affiliation(s)
- Rasha Q Al-Rahahleh
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, USA; Department of Pharmacology & Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Wynand P Roos
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
| | - Kate M Saville
- Department of Pharmacology & Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Joel F Andrews
- Department of Pharmacology & Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Zhijin Wu
- Department of Biostatistics, Brown University, Providence, RI 02912, USA
| | - Christopher A Koczor
- Department of Pharmacology & Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Aishwarya Prakash
- Department of Biochemistry and Molecular Biology & Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Robert W Sobol
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, USA; Department of Pharmacology & Mitchell Cancer Institute, College of Medicine, University of South Alabama, Mobile, AL 36604, USA.
| |
Collapse
|
2
|
Lin MT, Christenson ES, Pallavajjala A, Eshleman JR. Highly sensitive and specific markers for detection of mismatch repair deficiency by next-generation sequencing. Am J Clin Pathol 2025:aqaf026. [PMID: 40318191 DOI: 10.1093/ajcp/aqaf026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 03/16/2025] [Indexed: 05/07/2025] Open
Abstract
OBJECTIVE To identify exonic markers that could improve analytic performance characteristics of next-generation sequencing (NGS) in detecting mismatch repair deficiency (dMMR) using colorectal cancer (CRC) as a model. METHODS Coding sequences of a target NGS panel (~1.13 megabase) were compared between dMMR CRC and mismatch repair-proficient (pMMR) CRC in a training cohort (41 dMMR CRCs and 213 pMMR CRCs) and a validation cohort (33 dMMR CRCs and 307 pMMR CRCs) with documented mismatch repair status by immunohistochemical and/or microsatellite instability assays. RESULTS The dMMR CRC cases showed significantly higher insertion/deletion (indel) mutations within exonic homopolymers (homo-indels), occurring predominantly within longer repeats of 5 to 10 nucleotides (92%, P < .0001), rather than shorter repeats of 2 to 4 nucleotides seen in pMMR CRC (62%). Homo-indels in dMMR CRC were not random. Hotspot loci were consistent between the training and validation cohorts. The dMMR defined by indels within homopolymers of 5 or more nucleotides, homopolymers of 7 or more nucleotides, or a panel of hotspots all showed 100% sensitivity and specificity with a range of cutoffs. CONCLUSIONS We propose that this approach allows one to identify highly sensitive and specific markers for detecting dMMR CRC by NGS alone. Further studies are warranted to test whether these markers are applicable to non-CRC neoplasms.
Collapse
Affiliation(s)
- Ming-Tseh Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Eric S Christenson
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Aparna Pallavajjala
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| | - James R Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| |
Collapse
|
3
|
Koh GCC, Nanda AS, Rinaldi G, Boushaki S, Degasperi A, Badja C, Pregnall AM, Zhao SJ, Chmelova L, Black D, Heskin L, Dias J, Young J, Memari Y, Shooter S, Czarnecki J, Brown MA, Davies HR, Zou X, Nik-Zainal S. A redefined InDel taxonomy provides insights into mutational signatures. Nat Genet 2025; 57:1132-1141. [PMID: 40210680 DOI: 10.1038/s41588-025-02152-y] [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: 02/14/2024] [Accepted: 03/04/2025] [Indexed: 04/12/2025]
Abstract
Despite their deleterious effects, small insertions and deletions (InDels) have received far less attention than substitutions. Here we generated isogenic CRISPR-edited human cellular models of postreplicative repair dysfunction (PRRd), including individual and combined gene edits of DNA mismatch repair (MMR) and replicative polymerases (Pol ε and Pol δ). Unique, diverse InDel mutational footprints were revealed. However, the prevailing InDel classification framework was unable to discriminate these InDel signatures from background mutagenesis and from each other. To address this, we developed an alternative InDel classification system that considers flanking sequences and informative motifs (for example, longer homopolymers), enabling unambiguous InDel classification into 89 subtypes. Through focused characterization of seven tumor types from the 100,000 Genomes Project, we uncovered 37 InDel signatures; 27 were new. In addition to unveiling previously hidden biological insights, we also developed PRRDetect-a highly specific classifier of PRRd status in tumors, with potential implications for immunotherapies.
Collapse
Affiliation(s)
- Gene Ching Chiek Koh
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia
| | - Arjun Scott Nanda
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Giuseppe Rinaldi
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Soraya Boushaki
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Andrea Degasperi
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Cherif Badja
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Andrew Marcel Pregnall
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Salome Jingchen Zhao
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Lucia Chmelova
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Daniella Black
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Laura Heskin
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - João Dias
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Jamie Young
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Yasin Memari
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Scott Shooter
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Jan Czarnecki
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Matthew Arthur Brown
- Genomics England, Queen Mary University of London, Dawson Hall, Charterhouse Square, London, UK
| | - Helen Ruth Davies
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Xueqing Zou
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Serena Nik-Zainal
- Department of Genomic Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
4
|
Shen J, Ge L, Jiang G, Meng Y, Ma W. Pharmacokinetic Study of HRO761 in Rats by Liquid Chromatography Combined With Electrospray Ionization Tandem Mass Spectrometry. Biomed Chromatogr 2025; 39:e70033. [PMID: 39995302 DOI: 10.1002/bmc.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/17/2025] [Accepted: 02/09/2025] [Indexed: 02/26/2025]
Abstract
A simple and sensitive liquid chromatography tandem mass spectrometry method was established and validated for determination of HRO761 in rat plasma. After prepared by protein precipitation with acetonitrile, HRO761 and internal standard were separated on a Waters BEH C18 column using acetonitrile containing 0.1% formic acid and 0.1% formic acid in water as mobile phase by gradient elution. The method showed excellent linearity over the range of 5-5000 ng/mL with acceptable intraday and interday precision, accuracy, matrix effect, and recovery. The stability assay indicated that HRO761 was stable during the sample acquisition, preparation, and storage. The method was applied to pharmacokinetic study of HRO761 in rats. The result suggested that after intravenous administration at dose of 1 mg/kg, HRO761 was quickly eliminated from the plasma with the elimination half-life of 1.9 h. After oral administration at doses of 5, 10, and 20 mg/kg, HRO761 was quickly absorbed into plasma and reach the peak concentration (Cmax) of 2598.1-9379.2 ng/mL at 1.0-4.0 h. The exposure increased proportionally with the dose. The oral bioavailability was 79.0%-99.1% over the range of 5-20 mg/kg. This study provides useful information for its further development in clinic.
Collapse
Affiliation(s)
- Jie Shen
- Pharmacy Department, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Liye Ge
- Pharmacy Department, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Guowei Jiang
- Pharmacy Department, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yongjun Meng
- Pharmacy Department, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Weina Ma
- Pharmacy Department, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
5
|
Khalili S, Mohseninia A, Liu C, Banister CE, Heine P, Khazan M, Morrison SE, Gokare P, Cowley GS, Weir BA, Pocalyko D, Bachman KE, Buckhaults PJ. Comprehensive genomic dependency landscape of a human colon cancer organoid. Commun Biol 2025; 8:436. [PMID: 40082551 PMCID: PMC11906589 DOI: 10.1038/s42003-025-07822-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025] Open
Abstract
Identifying genetic dependencies in human colon cancer could help identify effective treatment strategies. Genome-wide CRISPR-Cas9 dropout screens have the potential to reveal genetic dependencies, some of which could be exploited as therapeutic targets using existing drugs. In this study, we comprehensively characterized genetic dependencies present in a colon cancer organoid avatar, and validated tumor-specific selectivity of select pharmacologic agents. We conducted a genome-wide CRISPR dropout screen to elucidate the genetic dependencies that interacted with select driver somatic mutations. We found distinct genetic dependencies that interacted with WNT, MAPK, PI3K, TP53, and mismatch repair pathways and validated targets that could be exploited as treatments for this specific subtype of colon cancer. These findings demonstrate the utility of functional genomic screening in the context of personalized medicine.
Collapse
Affiliation(s)
| | | | | | | | - Paige Heine
- University of South Carolina, Columbia, SC, US
| | | | | | - Prashanth Gokare
- Janssen Research and Development, LLC Spring House, Spring House, PA, US
| | - Glenn S Cowley
- Janssen Research and Development, LLC Cambridge, Cambridge, MA, US
| | - Barbara A Weir
- Janssen Research and Development, LLC Cambridge, Cambridge, MA, US
| | - David Pocalyko
- Janssen Research and Development, LLC Spring House, Spring House, PA, US
| | - Kurtis E Bachman
- Janssen Research and Development, LLC Spring House, Spring House, PA, US
| | | |
Collapse
|
6
|
Zhou B, Wang Y, Ding L, Tian X, Sun W, Zhang W, Liu YH. A novel algorithm for the detection of microsatellite instability in endometrial cancer using next‑generation sequencing data. Oncol Lett 2025; 29:86. [PMID: 39664615 PMCID: PMC11632413 DOI: 10.3892/ol.2024.14832] [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: 07/02/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
The molecular-based detection of microsatellite instability (MSI) in endometrial cancer is complex, due to the low sensitivity of PCR and a lack of standardization in next-generation sequencing (NGS) methods. In the present study, sequenced data were obtained from an NGS panel following the addition of five commonly used microsatellite loci. Subsequently, a novel algorithm, namely MSIPeak, was developed for data analysis. Results of the present study demonstrated that MSI data obtained using MSIPeak were presented in a peak, using a threshold of 1.10 to distinguish stable and unstable loci. MSIPeak was further validated using synthetic DNA samples and endometrial cancer tissue and the results were compared with the immunohistochemical analysis-determined mismatch repair status. The PCR results demonstrated a 3-base-pair (bp) deletion in synthetic DNA samples, compared with 1- and 2-bp deletion controls. Results obtained using MSIPeak demonstrated notable differences in peak profiles and positive scores in synthetic DNA samples with 1-, 2- and 3-bp deletions, compared with controls. Thus, the results of the present study demonstrated that NGS-based MSI detection exhibited a higher sensitivity compared with PCR. In addition, NGS-based MSI detection exhibited higher levels of repeatability and applicability compared with other MSI-NGS-based methods, such as MSISensor2 and MANTIS. Collectively, the results of the present study highlighted that the combination of MSIPeak and NGS exhibits potential in the detection of cancer.
Collapse
Affiliation(s)
- Bing Zhou
- Department of Pathology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Yu Wang
- Department of Pathology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Lu Ding
- Department of Gynecology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Xiaolei Tian
- Department of Pathology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Wu Sun
- Department of Pathology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Wei Zhang
- Department of Pathology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Yin-Hua Liu
- Department of Pathology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| |
Collapse
|
7
|
Yurgelun MB, Rhees J, Papadopoulos N, Vogelstein B, Boland CR. Taming Lynch Syndrome: The Remarkable Power of Prevention for One Family. Gastroenterology 2025; 168:195-199. [PMID: 39038760 DOI: 10.1053/j.gastro.2024.06.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/24/2024]
Affiliation(s)
- Matthew B Yurgelun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jennifer Rhees
- Department of Medicine, UCSD School of Medicine, La Jolla, California
| | | | - Bert Vogelstein
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - C Richard Boland
- Department of Medicine, UCSD School of Medicine, La Jolla, California
| |
Collapse
|
8
|
Canchi Sistla H, Talluri S, Rajagopal T, Venkatabalasubramanian S, Rao Dunna N. Genomic instability in ovarian cancer: Through the lens of single nucleotide polymorphisms. Clin Chim Acta 2025; 565:119992. [PMID: 39395774 DOI: 10.1016/j.cca.2024.119992] [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: 08/06/2024] [Revised: 10/04/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024]
Abstract
Ovarian cancer (OC) is the deadliest gynecological malignancy among all female reproductive cancers. It is characterized by high mortality rate and poor prognosis. Genomic instability caused by mutations, single nucleotide polymorphisms (SNPs), copy number variations (CNVs), microsatellite instability (MSI), and chromosomal instability (CIN) are associated with OC predisposition. SNPs, which are highly prevalent in the general population, show a greater relative risk contribution, particularly in sporadic cancers. Understanding OC etiology in terms of genetic basis can increase the use of molecular diagnostics and provide promising approaches for designing novel treatment modalities. This will help deliver personalized medicine to OC patients, which may soon be within reach. Given the pivotal impact of SNPs in cancers, the primary emphasis of this review is to shed light on their prevalence in key caretaker genes that closely monitor genomic integrity, viz., DNA damage response, repair, cell cycle checkpoints, telomerase maintenance, and apoptosis and their clinical implications in OC. We highlight the current challenges faced in different SNP-based studies. Various computational methods and bioinformatic tools employed to predict the functional impact of SNPs have also been comprehensively reviewed concerning OC research. Overall, this review identifies that variants in the DDR and HRR pathways are the most studied, implying their critical role in the disease. Conversely, variants in other pathways, such as NHEJ, MMR, cell cycle, apoptosis, telomere maintenance, and PARP genes, have been explored the least.
Collapse
Affiliation(s)
- Harshavardhani Canchi Sistla
- Cancer Genomics Laboratory, Department of Biotechnology, School of Chemical and Biotechnology, SASTRA- Deemed University, Thanjavur 613 401, India
| | - Srikanth Talluri
- Dana Farber Cancer Institute, Boston, MA 02215, USA; Veterans Administration Boston Healthcare System, West Roxbury, MA 02132, USA
| | | | - Sivaramakrishnan Venkatabalasubramanian
- Department of Genetic Engineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur Campus, Chennai 603 203, India
| | - Nageswara Rao Dunna
- Cancer Genomics Laboratory, Department of Biotechnology, School of Chemical and Biotechnology, SASTRA- Deemed University, Thanjavur 613 401, India.
| |
Collapse
|
9
|
Unger M, Loeffler CML, Žigutytė L, Sainath S, Lenz T, Vibert J, Mock A, Fröhling S, Graham TA, Carrero ZI, Kather JN. Deep Learning for Biomarker Discovery in Cancer Genomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631471. [PMID: 39829845 PMCID: PMC11741323 DOI: 10.1101/2025.01.06.631471] [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: 01/22/2025]
Abstract
Background Genomic data is essential for clinical decision-making in precision oncology. Bioinformatic algorithms are widely used to analyze next-generation sequencing (NGS) data, but they face two major challenges. First, these pipelines are highly complex, involving multiple steps and the integration of various tools. Second, they generate features that are human-interpretable but often result in information loss by focusing only on predefined genetic properties. This limitation restricts the full potential of NGS data in biomarker extraction and slows the discovery of new biomarkers in precision oncology. Methods We propose an end-to-end deep learning (DL) approach for analyzing NGS data. Specifically, we developed a multiple instance learning DL framework that integrates somatic mutation sequences to predict two compound biomarkers: microsatellite instability (MSI) and homologous recombination deficiency (HRD). To achieve this, we utilized data from 3,184 cancer patients obtained from two public databases: The Cancer Genome Atlas (TCGA) and the Clinical Proteome Tumor Analysis Consortium (CPTAC). Results Our proposed deep learning method demonstrated high accuracy in identifying clinically relevant biomarkers. For predicting MSI status, the model achieved an accuracy of 0.98, a sensitivity of 0.95, and a specificity of 1.00 on an external validation cohort. For predicting HRD status, the model achieved an accuracy of 0.80, a sensitivity of 0.75, and a specificity of 0.86. Furthermore, the deep learning approach significantly outperformed traditional machine learning methods in both tasks (MSI accuracy, p-value = 5.11×10-18; HRD accuracy, p-value = 1.07×10-10). Using explainability techniques, we demonstrated that the model's predictions are based on biologically meaningful features, aligning with key DNA damage repair mutation signatures. Conclusion We demonstrate that deep learning can identify patterns in unfiltered somatic mutations without the need for manual feature extraction. This approach enhances the detection of actionable targets and paves the way for developing NGS-based biomarkers using minimally processed data.
Collapse
Affiliation(s)
- Michaela Unger
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
| | - Chiara M L Loeffler
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
- Medical Department 1, University Hospital and Faculty of Medicine Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Laura Žigutytė
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
| | - Srividhya Sainath
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
| | - Tim Lenz
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
| | - Julien Vibert
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | - Andreas Mock
- Institute of Pathology, Ludwig-Maximilians-University München, Munich, Germany
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Fröhling
- Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany
- Division of Translational Precision Medicine, Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Trevor A Graham
- Centre for Evolution and Cancer, Institute of Cancer Research, London, UK
| | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, University of Technology Dresden, Dresden, Germany
- Medical Department 1, University Hospital and Faculty of Medicine Carl Gustav Carus, University of Technology Dresden, Dresden, Germany
- National Center for Tumor Diseases Dresden (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
10
|
Tian Z, Yang L, Yang R, Yang W. The prognostic and immunomodulatory role of the MMR system in patients with stomach adenocarcinoma. Sci Rep 2025; 15:180. [PMID: 39748125 PMCID: PMC11695722 DOI: 10.1038/s41598-024-84613-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025] Open
Abstract
The mismatch repair (MMR) system plays a crucial role in the maintenance of DNA replication fidelity and genomic stability. The clinical value of the MMR molecular marker as an immunotherapy for advanced solid tumors has been documented. However, this therapy is not effective in some patients. This study aimed to develop an MMR-related molecular prognostic model for identifying appropriate populations of stomach adenocarcinoma (STAD) for better treatment outcome. The MMR genes expression data were downloaded from TCGA and CCLE databases. The expression of four MMR genes, construction of a prognostic risk model, and assessment of immune infiltration in STAD were performed using Xiantao online tool. GEPIA2 was used to explore the association of MMR genes expression with clinical stage and overall survival. The frequency and prognostic value of MMR genes in STAD were conducted on the cBioPortal. The MLH1 co-expression network was established based on the LinkedOmics database. This study found that the expression of MSH2, MSH6 and PMS2 was up-regulated in STAD tissues. Moreover, differential MMR genetic expression levels were not significantly correlated with the clinical stages of STAD. Besides, no significant difference in PFS or OS was observed in STAD patients with or without MMR genetic alteration. Moreover, MLH1 and MSH2 were used to establish a prognostic risk model. The immune infiltration levels of most immune cells were upregulated in the high-risk group with elevated expression of PDCD1 and low TMB score. Finally, we found that MLH1 was an independent predictor of STAD prognosis among the four MMR genes. An MMR-related prognostic model for STAD was constructed based on genes. This model provides a new therapeutic concept for the diagnosis and treatment of STAD.
Collapse
Affiliation(s)
- Zhihui Tian
- Department of Gastroenterology, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan City, 030013, Shanxi Province, China.
- Department of Gastroenterology, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital, Shanxi Medical University, No.3, Staff New Street, Xinghualing District, Taiyuan City, 030000, Shanxi Province, China.
| | - Lili Yang
- Department of Intensive Care Unit, Shanxi Hospital Affiliated to Cancer Hospital, Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Shanxi Medical University, Taiyuan City, 030013, Shanxi Province, China
| | - Rong Yang
- Department of Gastroenterology, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan City, 030013, Shanxi Province, China.
- Department of Gastroenterology, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital, Shanxi Medical University, No.3, Staff New Street, Xinghualing District, Taiyuan City, 030000, Shanxi Province, China.
| | - Wenhui Yang
- Department of Gastroenterology, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan City, 030013, Shanxi Province, China.
- Department of Gastroenterology, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital, Shanxi Medical University, No.3, Staff New Street, Xinghualing District, Taiyuan City, 030000, Shanxi Province, China.
| |
Collapse
|
11
|
Guo H, Luo Y, Fu Z, Wang D. Indocyanine green fluorescence imaging for lymph node detection and long-term clinical outcomes in colorectal cancer surgery: A systematic review and meta-analysis. World J Surg 2024; 48:2818-2830. [PMID: 39542857 DOI: 10.1002/wjs.12412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND The indocyanine green fluorescence imaging (ICG-FI) technique is increasingly being used in laparoscopic colorectal surgery for lymph node mapping. However, there is no definitive standard regarding whether the application of this technique can significantly increase the detection rate of metastatic lymph nodes and improve long-term prognosis. METHODS PubMed, Embase, Web of Science, and Cochrane Library databases were searched to include studies including ICG-FI in laparoscopic colorectal surgery. Data on the detection rate of lymph nodes, metastatic rate of ICG-positive nodes, and long-term clinical outcomes were extracted following inclusion criteria. RESULTS Eighteen studies with a total of 1552 patients 922 with ICG-guided laparoscopic and 630 without ICG technique were finally included. Clinical stage II/III colorectal tumors were the most commonly studies types. The patients using the ICG-FI technique had more harvested lymph nodes (weighted mean: 23.5 vs. 18.9; WMD = 4.6; p < 0.00001) during dissection but a lower metastasis rate of ICG-positive nodes (61/218 [28%] vs. 96/333 [28.9%]; OR = 1.45; p = 0.08). Compared with conventional laparoscopic colorectal surgery, additional ICG-FI technique did not improve the 3 year overall survival rate (272/289 [94.1%] vs. 269/289 [93.1%]; OR = 1.19; p = 0.61), relapse-free survival (246/289 [85.1%] vs. 249/289 [86.2%]; OR = 0.92; p = 0.72), and local recurrence rate (22/289 [7.6%] vs. 28/289 [9.7%]; OR = 0.77; p = 0.38). The overall detection rate of sentinel lymph nodes, lymph flow, and metastatic rate of ICG-positive nodes with the help of ICG-FI were 86.8%, 89.9%, and 22.8%, respectively. No patients experienced major adverse events during ICG injection preoperatively or postoperatively. CONCLUSIONS Indocyanine green fluorescence imaging-guided procedure, compared to conventional laparoscopic dissection, can assist in obtaining a greater number of harvested lymph nodes and metastatic lymph nodes, however, it did not significantly improve the long-term clinical outcomes. LEVEL OF EVIDENCE Level III systematic review of randomized control and nonrandomized studies.
Collapse
Affiliation(s)
- Hong Guo
- Department of Emergency, Zigong Fourth People's Hospital, Zigong, China
| | - Yun Luo
- Department of Emergency, Zigong Fourth People's Hospital, Zigong, China
| | - Zhaojun Fu
- Department of Emergency, Zigong Fourth People's Hospital, Zigong, China
| | - Dengchao Wang
- Department of General Surgery, Zigong Fourth People's Hospital, Zigong, China
| |
Collapse
|
12
|
Miyashita K, Shioi S, Kajitani T, Koi Y, Shimokawa M, Makiyama A, Oda S, Esaki T. More subtle microsatellite instability better predicts fluorouracil insensitivity in colorectal cancer patients. Sci Rep 2024; 14:27257. [PMID: 39516234 PMCID: PMC11549419 DOI: 10.1038/s41598-024-77770-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Microsatellite instability (MSI) is now widely used as an indispensable biomarker. However, the relationship between MSI-H (high) and defective DNA mismatch repair (MMR) is not as straightforward as has been expected. Genome-edited cells carrying Lynch syndrome mutations do not exhibit drastic MSI typical in MSI-H (i.e. Type B) but more subtle MSI (i.e. Type A). In this study, we explored a connection between Type A MSI and 5-fluorouracil (5-FU) resistance in colorectal cancer patients. Using our precision and high-resolution MSI assay technique, tumour microsatellites were analysed in 30 colorectal cancer patients treated with FOLFOX or CAPOX. Among 30 tumours, eleven (37%) were judged as Type A MSI-positive. In Type A MSI+ tumours, the patient response to fluoropyrimidine and oxaliplatin was significantly poor (Fisher's exact test, p = 0.021). Accordingly, median PFS and OS were significantly poor in Type A+ patients (log-rank test, p < 0.001/p = 0.009). Type A MSI was an independent predictor of patient prognosis in this pilot cohort (Cox regression analysis, p = 0.003). Thus, more subtle Type A MSI better predicts fluoropyrimidine insensitivity in colorectal cancer patients, which may shed light on a hitherto overlooked connection between the MSI phenotypes and drug resistance in human cancer.
Collapse
Affiliation(s)
- Kaname Miyashita
- Cancer Genetics Laboratory, Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, 811-1395, Japan
- Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, Japan
- Department of Hematology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Seijiro Shioi
- Cancer Genetics Laboratory, Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, 811-1395, Japan
- Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Tatsuhiro Kajitani
- Department of Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yumiko Koi
- Department of Breast Oncology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Mototsugu Shimokawa
- Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, Japan
- Cancer Biostatistics Laboratory, Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, Japan
| | | | - Shinya Oda
- Cancer Genetics Laboratory, Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, 811-1395, Japan.
- Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, Japan.
| | - Taito Esaki
- Clinical Research Institute, NHO Kyushu Cancer Center, Fukuoka, Japan
- Department of Gastrointestinal and Medical Oncology, NHO Kyushu Cancer Center, Fukuoka, Japan
| |
Collapse
|
13
|
Chen H, Jin Z, Dai X, Zhu J, Chen G. The diagnostic value of histogram analysis of DWI and DKI for the mismatch repair status of rectal adenocarcinoma. Heliyon 2024; 10:e37526. [PMID: 39309916 PMCID: PMC11416531 DOI: 10.1016/j.heliyon.2024.e37526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/14/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Objectives To compare the diagnostic value of histogram analysis derived from diffusion weighted imaging (DWI) and diffusion kurtosis imaging (DKI) in differentiating the mismatch repair (MMR) status of rectal adenocarcinoma. Methods DWI and DKI were performed in 124 patients with rectal adenocarcinoma, which were divided into deficient mismatch repair (dMMR) group and proficient mismatch repair (pMMR) group. The patients' general clinical information, pathology and image characteristics were compared. The histogram analysis of apparent diffusion coefficient (ADC), diffusion kurtosis (K) and diffusion coefficient (D)derived from DWI and DKI at b values of 1000 and 2000 s/mm2 were calculated. The diagnostic efficacy of quantitative parameters for MMR in rectal adenocarcinoma was compared. Results The mean, 50th, 75th and 90th in ADC quantitative parameters of dMMR group were lower when the b value was 2000 s/mm2 (all P < 0.05). With b value of 1000 s/mm2, the 10th, 25th, and 50th in the dMMR group were lower, and the skewness was higher (all P < 0.05). D values (10th, 25th and 50th) derived from DKI quantitative parameters were lower in the dMMR group. The K values (75th, 90th and Kskewness) were higher in the dMMR group, while Kkurtosis was lower (all P < 0.05). The results of multivariate logistic regression analysis showed that ADC75th(b = 2000 s/mm2), ADCskewness (b = 1000 s/mm2) and Kskewness were the statistical significant parameters (P = 0.014, 0.036 and 0.002, respectively), and the AUC values were 0.713, 0.818 and 0.835, respectively. Conclusion Histogram analysis derived from DWI and DKI can be good predictor of MMR. Kskewness is the strongest independent factor for predicting MMR.
Collapse
Affiliation(s)
- Hao Chen
- Department of Medical Imaging, Anqing Municipal Hospital, Anqing, China
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhicheng Jin
- Department of Nuclear Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoxiao Dai
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Juan Zhu
- Department of Medical Imaging, Anqing Municipal Hospital, Anqing, China
| | - Guangqiang Chen
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
14
|
Ghimire B, Kurlberg G, Falk P, Singh Y, Wettergren Y. Epigenetic differences in the tumor suppressor genes MLH1 and p16INK4a between Nepalese and Swedish patients with colorectal cancer. Innov Surg Sci 2024; 9:153-163. [PMID: 39309195 PMCID: PMC11416036 DOI: 10.1515/iss-2023-0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/02/2024] [Indexed: 09/25/2024] Open
Abstract
Objectives Colorectal cancer (CRC) is one of the most prevalent cancer types worldwide, exhibiting significant variance in incidence rates across different ethnicities and geographical regions. Notably, there is a rising incidence of CRC among younger adults, particularly evident in advanced stages, with a more pronounced trend observed in developing nations. Epigenetic alterations potentially play a role in the early onset of CRC and could elucidate interpopulation disparities. This study aimed to examine DNA methylation levels in the tumor suppressor genes MLH1 and p16INK4a, comparing Nepalese and Swedish patients with CRC. Methods Patients who underwent CRC surgery at Tribhuvan University Teaching Hospital, Nepal (n=39), and Sahlgrenska University Hospital, Sweden (n=39) were included. Demographic and clinicopathological data were analyzed, and pyrosequencing was employed to determine methylation levels in the MLH1 promoter region and the first exon of p16INK4a in tumor tissues and adjacent mucosa located 10 cm from the tumor site. Subsequently, methylation status was compared between Nepalese and Swedish patients and correlated with clinicopathological parameters. Results Nepalese and Swedish patients displayed equal levels of MLH1 and p16INK4a methylation in tumors, but Nepalese patients exhibited a significantly higher level of MLH1 methylation in mucosa compared to Swedish patients (p=0.0008). Moreover, a greater proportion of Nepalese patients showed MLH1 methylation in mucosa compared to Swedish patients (31 vs. 2.6 %). Aberrant methylation of p16INK4a was also observed in the mucosa of Nepalese patients, characterized by high methylation at specific sites rather than uniform methylation across CpG sites. There were no significant differences in methylation levels based on tumor location among Nepalese patients, whereas Swedish patients exhibited higher methylation in right- compared to left-sided colon tumors. Swedish patients showed an increase in p16INK4a methylation in tumors with advancing age. Conclusions Nepalese and Swedish patients displayed equal levels of MLH1 and p16INK4a methylation in tumors. In contrast, Nepalese patients had a higher level of MLH1 methylation as well as aberrant methylation of p16INK4a in mucosa compared to Swedish patients. These epigenetic differences may be linked to environmental and lifestyle factors. Ongoing research will further explore whether hypermethylation in the mucosa of Nepalese patients is associated with tumorigenesis and its potential utility in screening high-risk patients or predicting recurrence.
Collapse
Affiliation(s)
- Bikal Ghimire
- Department of GI and General Surgery, Maharajgung Medical Campus, Institute of Medicine, Tribhuvan University, Kathmandu, Nepal
| | - Göran Kurlberg
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter Falk
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Yogendra Singh
- Department of GI and General Surgery, Maharajgung Medical Campus, Institute of Medicine, Tribhuvan University, Kathmandu, Nepal
| | - Yvonne Wettergren
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
15
|
Brandaleone L, Dal Buono A, Gabbiadini R, Marcozzi G, Polverini D, Carvello M, Spinelli A, Hassan C, Repici A, Bezzio C, Armuzzi A. Hereditary Colorectal Cancer Syndromes and Inflammatory Bowel Diseases: Risk Management and Surveillance Strategies. Cancers (Basel) 2024; 16:2967. [PMID: 39272825 PMCID: PMC11394661 DOI: 10.3390/cancers16172967] [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: 07/10/2024] [Revised: 08/15/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Background and aims: Hereditary colorectal cancer syndromes (HCCS), including familial adenomatous polyposis (FAP) and Lynch syndrome (LS), are the two most important high-risk conditions for colorectal cancer (CRC). Inflammatory bowel disease (IBD) increases the risk by two to six times compared with that in the general population. The intersection of these two conditions has rarely been documented in literature. We aimed to summarize the prevalence, pathogenesis, and current evidence-based management of IBD and HCCS and the underlying molecular mechanisms of accelerated carcinogenesis due to combined inflammation and genetic predisposition. Methods: PubMed and Scopus were searched until June 2024 to identify relevant studies investigating the epidemiology, pathogenesis, and management of IBD and coexisting hereditary CRC syndromes. Results: Co-occurrence of IBD and hereditary CRC syndromes is exceptionally uncommon. Individuals with LS and IBD tend to develop CRC at a younger age than those without IBD, with patients with ulcerative colitis facing particularly elevated risks. The interaction between mismatch deficiency and chronic inflammation requires further investigation.
Collapse
Affiliation(s)
- Luca Brandaleone
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Arianna Dal Buono
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Roberto Gabbiadini
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Giacomo Marcozzi
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Davide Polverini
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Michele Carvello
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
- Colon and Rectal Surgery Division, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Antonino Spinelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
- Colon and Rectal Surgery Division, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Cesare Hassan
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
- Endoscopy Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Alessandro Repici
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
- Endoscopy Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Cristina Bezzio
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Alessandro Armuzzi
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| |
Collapse
|
16
|
Kulchak Rahm A, Wolfinger T, Salvati ZM, Schneider JL, Cragun D. Development, Evaluation, and User Testing of a Decision-Making Toolkit to Promote Organizations to Implement Universal Tumor Screening for Lynch Syndrome. Public Health Genomics 2024; 27:136-149. [PMID: 39159623 DOI: 10.1159/000540943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION The Implementing Universal Lynch Syndrome Screening (IMPULSS) study explained institutional variation in universal tumor screening (UTS) with the goal of identifying ways to aid organizational decision-makers in implementing and optimizing Lynch syndrome UTS programs. METHODS After applying the Consolidated Framework for Implementation Research (CFIR 1.0) to analyze interviews with 66 stakeholders across 9 healthcare systems to develop a toolkit for implementation, we adapted the International Patient Decision Aid Standards (IPDAS) to assess toolkit potential to aid decision-making consistent with organizational values. We then conducted user testing with two experienced and four non-experienced implementers of UTS to improve the content and functionality of the toolkit and assess its acceptability and appropriateness. RESULTS Toolkit components were organized to address findings related to CFIR 1.0 constructs of evidence strength and quality, relative advantage, cost, engaging, planning, executing, and reflecting and evaluating. A home page was added to direct users to different sections based on whether they are deciding to implement UTS, planning for implementation, improving an existing UTS program, or considering a different approach to identify patients with Lynch syndrome. Upon initial evaluation, 31 of 64 IPDAS criteria were met by the original toolkit. All users rated the toolkit as acceptable and appropriate for assisting organizational decision-making and identified multiple areas for improvement. Numerous iterative changes were made to the toolkit, resulting in meeting 17 of the previously unmet IPDAS criteria. CONCLUSION We demonstrate the rigorous development of a toolkit guided by the CFIR and show how user testing helped improve the toolkit to ensure it is acceptable, appropriate, and meets most IPDAS criteria relevant to organizational values-based decision-making.
Collapse
Affiliation(s)
| | | | - Zachary M Salvati
- Department of Genomic Health, Geisinger, Danville, Pennsylvania, USA
| | | | | |
Collapse
|
17
|
Fu X, Huang J, Zhu J, Fan X, Wang C, Deng W, Tan X, Chen Z, Cai Y, Lin H, Wang G, Zhang N, Zhu Y, Chen J, Zhan H, Huang S, Fang Y, Li Y, Huang Y. Prognosis and immunotherapy efficacy in dMMR&MSS colorectal cancer patients and an MSI status predicting model. Int J Cancer 2024; 155:766-775. [PMID: 38594805 DOI: 10.1002/ijc.34946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/07/2024] [Accepted: 03/15/2024] [Indexed: 04/11/2024]
Abstract
The inconsistency between mismatch repair (MMR) protein immunohistochemistry (IHC) and microsatellite instability PCR (MSI-PCR) methods has been widely reported. We aim to investigate the prognosis and the effect of immunotherapy in dMMR by IHC but MSS by MSI-PCR (dMMR&MSS) colorectal cancer (CRC) patients. A microsatellite instability (MSI) predicting model was established to help find dMMR&MSS patients. MMR and MSI states were detected by the IHC and MSI-PCR in 1622 CRC patients (ZS6Y-1 cohort). Logistic regression analysis was used to screen clinical features to construct an MSI-predicting nomogram. We propose a new nomogram-based assay to find patients with dMMR&MSS, in which the MSI-PCR assay only detects dMMR patients with MSS predictive results. We applied the new strategy to a random cohort of 248 CRC patients (ZS6Y-2 cohort). The consistency of MMR IHC and MSI-PCR in the ZS6Y-1 cohort was 95.7% (1553/1622). Both pMMR&MSS and dMMR&MSS groups experienced significantly shorter overall survival (OS) than those in dMMR by IHC and MSI-H by MSI-PCR (dMMR&MSI-H) group (hazard ratio [HR] = 2.429, 95% confidence interval [CI]: 1.89-3.116, p < .01; HR = 21.96, 95% CI: 7.24-66.61, p < .01). The dMMR&MSS group experienced shorter OS than the pMMR&MSS group, but the difference did not reach significance (log rank test, p = .0686). In the immunotherapy group, the progression-free survival of dMMR&MSS patients was significantly shorter than that of dMMR&MSI-H patients (HR = 13.83, 95% CI: 1.508-126.8, p < .05). The ZS6Y-MSI-Pre nomogram (C-index = 0.816, 95% CI: 0.792-0.841, already online) found 66% (2/3) dMMR&MSS patients in the ZS6Y-2 cohort. There are significant differences in OS and immunotherapy effect between dMMR&MSI-H and dMMR&MSS patients. Our prediction model provides an economical way to screen dMMR&MSS patients.
Collapse
Affiliation(s)
- Xinhui Fu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinglin Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Junling Zhu
- Department of Pathology, The First People's Hospital of Kashgar, Kashgar, China
| | - Xinjuan Fan
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chao Wang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weihao Deng
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoli Tan
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiting Chen
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yacheng Cai
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hanjie Lin
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guannan Wang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ning Zhang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongmin Zhu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ji Chen
- Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huanmiao Zhan
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuhui Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yongzhen Fang
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuhua Li
- Department of Pathology, The First People's Hospital of Kashgar, Kashgar, China
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Jonchère V, Montémont H, Le Scanf E, Siret A, Letourneur Q, Tubacher E, Battail C, Fall A, Labreche K, Renault V, Ratovomanana T, Buhard O, Jolly A, Le Rouzic P, Feys C, Despras E, Zouali H, Nicolle R, Cervera P, Svrcek M, Bourgoin P, Blanché H, Boland A, Lefèvre J, Parc Y, Touat M, Bielle F, Arzur D, Cueff G, Le Jossic-Corcos C, Quéré G, Dujardin G, Blondel M, Le Maréchal C, Cohen R, André T, Coulet F, de la Grange P, de Reyniès A, Fléjou JF, Renaud F, Alentorn A, Corcos L, Deleuze JF, Collura A, Duval A. Microsatellite instability at U2AF-binding polypyrimidic tract sites perturbs alternative splicing during colorectal cancer initiation. Genome Biol 2024; 25:210. [PMID: 39107855 PMCID: PMC11304650 DOI: 10.1186/s13059-024-03340-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Microsatellite instability (MSI) due to mismatch repair deficiency (dMMR) is common in colorectal cancer (CRC). These cancers are associated with somatic coding events, but the noncoding pathophysiological impact of this genomic instability is yet poorly understood. Here, we perform an analysis of coding and noncoding MSI events at the different steps of colorectal tumorigenesis using whole exome sequencing and search for associated splicing events via RNA sequencing at the bulk-tumor and single-cell levels. RESULTS Our results demonstrate that MSI leads to hundreds of noncoding DNA mutations, notably at polypyrimidine U2AF RNA-binding sites which are endowed with cis-activity in splicing, while higher frequency of exon skipping events are observed in the mRNAs of MSI compared to non-MSI CRC. At the DNA level, these noncoding MSI mutations occur very early prior to cell transformation in the dMMR colonic crypt, accounting for only a fraction of the exon skipping in MSI CRC. At the RNA level, the aberrant exon skipping signature is likely to impair colonic cell differentiation in MSI CRC affecting the expression of alternative exons encoding protein isoforms governing cell fate, while also targeting constitutive exons, making dMMR cells immunogenic in early stage before the onset of coding mutations. This signature is characterized by its similarity to the oncogenic U2AF1-S34F splicing mutation observed in several other non-MSI cancer. CONCLUSIONS Overall, these findings provide evidence that a very early RNA splicing signature partly driven by MSI impairs cell differentiation and promotes MSI CRC initiation, far before coding mutations which accumulate later during MSI tumorigenesis.
Collapse
Affiliation(s)
- Vincent Jonchère
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | - Hugo Montémont
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | - Enora Le Scanf
- INSERM, UMR 1078, Université de Brest, Génétique Génomique Fonctionnelle Et Biotechnologies, Etablissement Français du Sang, F-29200, Brest, France
- CHU de Brest, Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, F-29200, France
| | - Aurélie Siret
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | - Quentin Letourneur
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | - Emmanuel Tubacher
- Laboratory for Genomics, Foundation Jean Dausset-CEPH (Centre d'Etude du Polymorphisme Humain), Paris, France
| | - Christophe Battail
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057, Evry, France
| | - Assane Fall
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | - Karim Labreche
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | - Victor Renault
- Laboratory for Genomics, Foundation Jean Dausset-CEPH (Centre d'Etude du Polymorphisme Humain), Paris, France
| | - Toky Ratovomanana
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | - Olivier Buhard
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | | | - Philippe Le Rouzic
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | - Cody Feys
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | - Emmanuelle Despras
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | - Habib Zouali
- Laboratory for Genomics, Foundation Jean Dausset-CEPH (Centre d'Etude du Polymorphisme Humain), Paris, France
| | - Rémy Nicolle
- Programme "Cartes d'Identité Des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Pascale Cervera
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
- Department of Pathology, Sorbonne Université, AP-HP.Sorbonne UniversitéHôpital Saint-Antoine, 47-83 Boulevard de L'hôpital, 75012, Paris, France
| | - Magali Svrcek
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
- Department of Pathology, Sorbonne Université, AP-HP.Sorbonne UniversitéHôpital Saint-Antoine, 47-83 Boulevard de L'hôpital, 75012, Paris, France
| | - Pierre Bourgoin
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
- Department of Pathology, Sorbonne Université, AP-HP.Sorbonne UniversitéHôpital Saint-Antoine, 47-83 Boulevard de L'hôpital, 75012, Paris, France
| | - Hélène Blanché
- Laboratory for Genomics, Foundation Jean Dausset-CEPH (Centre d'Etude du Polymorphisme Humain), Paris, France
| | - Anne Boland
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057, Evry, France
| | - Jérémie Lefèvre
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
- Department of Digestive Surgery, Sorbonne Université, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Yann Parc
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
- Department of Digestive Surgery, Sorbonne Université, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Mehdi Touat
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
- Sorbonne Université, Inserm, CNRS, UMR S 1127 and SIRIC CURAMUS, Institut du Cerveau Et de La Moelle Épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2 Mazarin, Paris, France
| | - Franck Bielle
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau Et de La Moelle Épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neuropathologie Laboratoire Escourolle, Paris, France
| | - Danielle Arzur
- INSERM, UMR 1078, Université de Brest, Génétique Génomique Fonctionnelle Et Biotechnologies, Etablissement Français du Sang, F-29200, Brest, France
- CHU de Brest, Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, F-29200, France
| | - Gwennina Cueff
- INSERM, UMR 1078, Université de Brest, Génétique Génomique Fonctionnelle Et Biotechnologies, Etablissement Français du Sang, F-29200, Brest, France
- CHU de Brest, Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, F-29200, France
| | - Catherine Le Jossic-Corcos
- INSERM, UMR 1078, Université de Brest, Génétique Génomique Fonctionnelle Et Biotechnologies, Etablissement Français du Sang, F-29200, Brest, France
- CHU de Brest, Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, F-29200, France
| | - Gaël Quéré
- INSERM, UMR 1078, Université de Brest, Génétique Génomique Fonctionnelle Et Biotechnologies, Etablissement Français du Sang, F-29200, Brest, France
- CHU de Brest, Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, F-29200, France
| | - Gwendal Dujardin
- INSERM, UMR 1078, Université de Brest, Génétique Génomique Fonctionnelle Et Biotechnologies, Etablissement Français du Sang, F-29200, Brest, France
- CHU de Brest, Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, F-29200, France
| | - Marc Blondel
- INSERM, UMR 1078, Université de Brest, Génétique Génomique Fonctionnelle Et Biotechnologies, Etablissement Français du Sang, F-29200, Brest, France
- CHU de Brest, Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, F-29200, France
| | - Cédric Le Maréchal
- INSERM, UMR 1078, Université de Brest, Génétique Génomique Fonctionnelle Et Biotechnologies, Etablissement Français du Sang, F-29200, Brest, France
- CHU de Brest, Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, F-29200, France
| | - Romain Cohen
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
- Department of Medical Oncology, Sorbonne Université, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Thierry André
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
- Department of Medical Oncology, Sorbonne Université, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Florence Coulet
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
- Genetics Department, AP-HP.Sorbonne Université, Paris, France
| | | | - Aurélien de Reyniès
- Programme "Cartes d'Identité Des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Jean-François Fléjou
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
- Department of Pathology, Sorbonne Université, AP-HP.Sorbonne UniversitéHôpital Saint-Antoine, 47-83 Boulevard de L'hôpital, 75012, Paris, France
| | - Florence Renaud
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | - Agusti Alentorn
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | - Laurent Corcos
- INSERM, UMR 1078, Université de Brest, Génétique Génomique Fonctionnelle Et Biotechnologies, Etablissement Français du Sang, F-29200, Brest, France
- CHU de Brest, Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, F-29200, France
| | - Jean-François Deleuze
- Laboratory for Genomics, Foundation Jean Dausset-CEPH (Centre d'Etude du Polymorphisme Humain), Paris, France
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057, Evry, France
| | - Ada Collura
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France
| | - Alex Duval
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité Des Microsatellites Et Cancer, Equipe Labellisée Par La Ligue Nationale Contre Le Cancer, 75012, Paris, France.
- Genetics Department, AP-HP.Sorbonne Université, Paris, France.
| |
Collapse
|
19
|
Choi JW, Lee JO, Lee S. Detecting microsatellite instability by length comparison of microsatellites in the 3' untranslated region with RNA-seq. Brief Bioinform 2024; 25:bbae423. [PMID: 39210504 PMCID: PMC11361843 DOI: 10.1093/bib/bbae423] [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: 04/30/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Microsatellite instability (MSI), a phenomenon caused by deoxyribonucleic acid (DNA) mismatch repair system deficiencies, is an important biomarker in cancer research and clinical diagnostics. MSI detection often involves next-generation sequencing data, with many studies focusing on DNA. Here, we introduce a novel approach by measuring microsatellite lengths directly from ribonucleic acid sequencing (RNA-seq) data and comparing its distribution to detect MSI. Our findings reveal distinct instability patterns between MSI-high (MSI-H) and microsatellite stable samples, indicating the efficacy of RNA-based MSI detection. Additionally, microsatellites in the 3'-untranslated regions showed the greatest predictive value for MSI detection. Notably, this efficacy extends to detecting MSI-H samples even in tumors not commonly associated with MSI. Our approach highlights the utility of RNA-seq data in MSI detection, facilitating more precise diagnostics through the integration of various biological data.
Collapse
Affiliation(s)
- Jin-Wook Choi
- Department of Health Science and Technology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, 08826 Seoul, Republic of Korea
| | - Jin-Ok Lee
- Department of Health Science and Technology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, 08826 Seoul, Republic of Korea
| | - Sejoon Lee
- Department of Health Science and Technology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, 08826 Seoul, Republic of Korea
- Department of Pathology and Translational Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82 Gumi-ro 173beon-gil, Bundang-gu, 13620 Seongnam, Republic of Korea
- Precision Medicine Center, Seoul National University Bundang Hospital, 82 Gumi-ro, Bundang-gu, 13620 Seongnam, Republic of Korea
- Department of Genomic Medicine, Seoul National University Bundang Hospital, 82 Gumi-ro, Bundang-gu, 13620 Seongnam, Republic of Korea
| |
Collapse
|
20
|
Anthony H, Seoighe C. Performance assessment of computational tools to detect microsatellite instability. Brief Bioinform 2024; 25:bbae390. [PMID: 39129364 PMCID: PMC11317526 DOI: 10.1093/bib/bbae390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/26/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024] Open
Abstract
Microsatellite instability (MSI) is a phenomenon seen in several cancer types, which can be used as a biomarker to help guide immune checkpoint inhibitor treatment. To facilitate this, researchers have developed computational tools to categorize samples as having high microsatellite instability, or as being microsatellite stable using next-generation sequencing data. Most of these tools were published with unclear scope and usage, and they have yet to be independently benchmarked. To address these issues, we assessed the performance of eight leading MSI tools across several unique datasets that encompass a wide variety of sequencing methods. While we were able to replicate the original findings of each tool on whole exome sequencing data, most tools had worse receiver operating characteristic and precision-recall area under the curve values on whole genome sequencing data. We also found that they lacked agreement with one another and with commercial MSI software on gene panel data, and that optimal threshold cut-offs vary by sequencing type. Lastly, we tested tools made specifically for RNA sequencing data and found they were outperformed by tools designed for use with DNA sequencing data. Out of all, two tools (MSIsensor2, MANTIS) performed well across nearly all datasets, but when all datasets were combined, their precision decreased. Our results caution that MSI tools can have much lower performance on datasets other than those on which they were originally evaluated, and in the case of RNA sequencing tools, can even perform poorly on the type of data for which they were created.
Collapse
Affiliation(s)
- Harrison Anthony
- School of Mathematical and Statistical Sciences, University of Galway, Galway H91 TK33, Ireland
- The SFI Centre for Research Training in Genomics Data Science, Galway D02 FX65, Ireland
| | - Cathal Seoighe
- School of Mathematical and Statistical Sciences, University of Galway, Galway H91 TK33, Ireland
- The SFI Centre for Research Training in Genomics Data Science, Galway D02 FX65, Ireland
| |
Collapse
|
21
|
Bowen CM, Demarest K, Vilar E, Shah PD. Novel Cancer Prevention Strategies in Individuals With Hereditary Cancer Syndromes: Focus on BRCA1, BRCA2, and Lynch Syndrome. Am Soc Clin Oncol Educ Book 2024; 44:e433576. [PMID: 38913968 DOI: 10.1200/edbk_433576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Germline pathogenic variants (PVs) in the BRCA1 and BRCA2 genes confer elevated risks of breast, ovarian, and other cancers. Lynch syndrome (LS) is associated with increased risks of multiple cancer types including colorectal and uterine cancers. Current cancer risk mitigation strategies have focused on pharmacologic risk reduction, enhanced surveillance, and preventive surgeries. While these approaches can be effective, they stand to be improved on because of either limited efficacy or undesirable impact on quality of life. The current review summarizes ongoing investigational efforts in cancer risk prevention strategies for patients with germline PVs in BRCA1, BRCA2, or LS-associated genes. These efforts span radiation, surgery, and pharmacology including vaccine strategies. Understanding the molecular events involved in the premalignant to malignant transformation in high-risk individuals may ultimately contribute significantly to novel prevention strategies.
Collapse
Affiliation(s)
- Charles M Bowen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Payal D Shah
- Perelman Center for Advanced Medicine, Abramson Cancer Center, Philadelphia, PA
| |
Collapse
|
22
|
Beech C, Hechtman JF. Molecular Approach to Colorectal Carcinoma: Current Evidence and Clinical Application. Clin Lab Med 2024; 44:221-238. [PMID: 38821642 DOI: 10.1016/j.cll.2023.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Colorectal carcinoma is one of the most common cancer types in men and women, responsible for both the third highest incidence of new cancer cases and the third highest cause of cancer deaths. In the last several decades, the molecular mechanisms surrounding colorectal carcinoma's tumorigenesis have become clearer through research, providing new avenues for diagnostic testing and novel approaches to therapeutics. Laboratories are tasked with providing the most current information to help guide clinical decisions. In this review, we summarize the current knowledge surrounding colorectal carcinoma tumorigenesis and highlight clinically relevant molecular testing.
Collapse
Affiliation(s)
- Cameron Beech
- Department of Pathology, Yale New Haven Hospital, New Haven, CT, USA
| | - Jaclyn F Hechtman
- Molecular and GI Pathologist, NeoGenomics Laboratories, Fort Myers, FL, USA.
| |
Collapse
|
23
|
Dong F. Pan-Cancer Molecular Biomarkers: A Paradigm Shift in Diagnostic Pathology. Clin Lab Med 2024; 44:325-337. [PMID: 38821647 DOI: 10.1016/j.cll.2023.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
The rapid adoption of next-generation sequencing in clinical oncology has enabled the detection of molecular biomarkers shared between multiple tumor types. These pan-cancer biomarkers include sequence-altering mutations, copy number changes, gene rearrangements, and mutational signatures and have been demonstrated to predict response to targeted therapy. This article reviews issues surrounding current and emerging pan-cancer molecular biomarkers in clinical oncology: technological advances that enable the broad detection of cancer mutations across hundreds of genes, the spectrum of driver and passenger mutations derived from human cancer genomes, and implications for patient care now and in the near future.
Collapse
Affiliation(s)
- Fei Dong
- Department of Pathology, Stanford University School of Medicine, 3375 Hillview Ave, Palo Alto, CA 94304, USA.
| |
Collapse
|
24
|
Ferretti S, Hamon J, de Kanter R, Scheufler C, Andraos-Rey R, Barbe S, Bechter E, Blank J, Bordas V, Dammassa E, Decker A, Di Nanni N, Dourdoigne M, Gavioli E, Hattenberger M, Heuser A, Hemmerlin C, Hinrichs J, Kerr G, Laborde L, Jaco I, Núñez EJ, Martus HJ, Quadt C, Reschke M, Romanet V, Schaeffer F, Schoepfer J, Schrapp M, Strang R, Voshol H, Wartmann M, Welly S, Zécri F, Hofmann F, Möbitz H, Cortés-Cros M. Discovery of WRN inhibitor HRO761 with synthetic lethality in MSI cancers. Nature 2024; 629:443-449. [PMID: 38658754 PMCID: PMC11078746 DOI: 10.1038/s41586-024-07350-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 03/26/2024] [Indexed: 04/26/2024]
Abstract
The Werner syndrome RecQ helicase WRN was identified as a synthetic lethal target in cancer cells with microsatellite instability (MSI) by several genetic screens1-6. Despite advances in treatment with immune checkpoint inhibitors7-10, there is an unmet need in the treatment of MSI cancers11-14. Here we report the structural, biochemical, cellular and pharmacological characterization of the clinical-stage WRN helicase inhibitor HRO761, which was identified through an innovative hit-finding and lead-optimization strategy. HRO761 is a potent, selective, allosteric WRN inhibitor that binds at the interface of the D1 and D2 helicase domains, locking WRN in an inactive conformation. Pharmacological inhibition by HRO761 recapitulated the phenotype observed by WRN genetic suppression, leading to DNA damage and inhibition of tumour cell growth selectively in MSI cells in a p53-independent manner. Moreover, HRO761 led to WRN degradation in MSI cells but not in microsatellite-stable cells. Oral treatment with HRO761 resulted in dose-dependent in vivo DNA damage induction and tumour growth inhibition in MSI cell- and patient-derived xenograft models. These findings represent preclinical pharmacological validation of WRN as a therapeutic target in MSI cancers. A clinical trial with HRO761 (NCT05838768) is ongoing to assess the safety, tolerability and preliminary anti-tumour activity in patients with MSI colorectal cancer and other MSI solid tumours.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jutta Blank
- Novartis BioMedical Research, Basel, Switzerland
| | | | | | | | | | | | - Elena Gavioli
- Novartis BioMedical Research, Basel, Switzerland
- Novartis Pharma AG, Basel, Switzerland
| | | | - Alisa Heuser
- Novartis BioMedical Research, Basel, Switzerland
| | | | | | - Grainne Kerr
- Novartis BioMedical Research, Basel, Switzerland
| | | | - Isabel Jaco
- Novartis BioMedical Research, Basel, Switzerland
| | - Eloísa Jiménez Núñez
- Novartis BioMedical Research, Basel, Switzerland
- Pierre Fabre Laboratories, Toulouse, France
| | | | | | | | | | | | | | | | - Ross Strang
- Novartis BioMedical Research, Basel, Switzerland
| | - Hans Voshol
- Novartis BioMedical Research, Basel, Switzerland
| | | | - Sarah Welly
- Novartis BioMedical Research, Basel, Switzerland
| | | | - Francesco Hofmann
- Novartis BioMedical Research, Basel, Switzerland
- Pierre Fabre Laboratories, Toulouse, France
| | | | | |
Collapse
|
25
|
Ioffe D, McSweeny M, Hall MJ. Precision Medicine in the Era of Genetic Testing: Microsatellite Instability Evolved. Clin Colon Rectal Surg 2024; 37:157-171. [PMID: 38617845 PMCID: PMC11007599 DOI: 10.1055/s-0043-1770385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The recognized importance of microsatellite instability (MSI) in cancer has evolved considerably in the past 30 years. From its beginnings as a molecular predictor for Lynch syndrome, MSI first transitioned to a universal screening test in all colorectal and endometrial cancers, substantially increasing the identification of patients with Lynch syndrome among cancer patients. More recently, MSI has been shown to be a powerful biomarker of response to immune checkpoint blockade therapy across a diversity of tumor types, and in 2017 was granted Food and Drug Administration approval as the first tumor histology-agnostic biomarker for a cancer therapy. Focusing on colorectal cancer specifically, immune checkpoint blockade therapy has been shown to be highly effective in the treatment of both MSI-high (MSI-H) colon and rectal cancer, with data increasingly suggesting an early role for immune checkpoint blockade therapy in MSI-H colorectal tumors in the neoadjuvant setting, with the potential to avoid more toxic and morbid approaches using traditional chemotherapy, radiation therapy, and surgery. The success of MSI as an immune checkpoint blockade target has inspired ongoing vigorous research to identify new similar targets for immune checkpoint blockade therapy that may help to one day expand the reach of this revolutionary cancer therapy to a wider swath of patients and indications.
Collapse
Affiliation(s)
- Dina Ioffe
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Michelle McSweeny
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Michael J. Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
26
|
Wilbur HC, Le DT, Agarwal P. Immunotherapy of MSI Cancer: Facts and Hopes. Clin Cancer Res 2024; 30:1438-1447. [PMID: 38015720 DOI: 10.1158/1078-0432.ccr-21-1935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/14/2023] [Accepted: 11/06/2023] [Indexed: 11/30/2023]
Abstract
Microsatellite instability (MSI) is a tumor molecular phenotype that evolves from loss of function in the mismatch repair (MMR) proteins through deleterious germline mutations, epigenetic inactivation, or somatic biallelic mutations. This phenotype is characterized by genomic hyper-mutability, increased neoantigen expression, and a favorable, immune-rich tumor microenvironment. These features confer a greater likelihood of response to treatment with the class of agents known as immune checkpoint inhibitors (ICI) and, potentially, other immune-based therapeutics. MSI as a predictive biomarker for response to treatment with ICIs ultimately led to the first tissue-agnostic approval of pembrolizumab for advanced, previously treated MSI or deficient MMR (dMMR) tumors. Nevertheless, response to ICIs in dMMR/MSI tumors is not universal. Identifying predictors of response and elucidating mechanisms of immune escape will be crucial to continued successful treatment of this subset. In this review, we aim to describe the pathogenesis and key immunologic features of dMMR/MSI tumors, provide a brief overview of the currently approved treatments, and discuss promising novel immune-based therapeutics currently under investigation.
Collapse
Affiliation(s)
- H Catherine Wilbur
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Dung T Le
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Parul Agarwal
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
27
|
Lynch A, Bradford S, Burkard ME. The reckoning of chromosomal instability: past, present, future. Chromosome Res 2024; 32:2. [PMID: 38367036 DOI: 10.1007/s10577-024-09746-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 01/11/2024] [Accepted: 01/27/2024] [Indexed: 02/19/2024]
Abstract
Quantitative measures of CIN are crucial to our understanding of its role in cancer. Technological advances have changed the way CIN is quantified, offering increased accuracy and insight. Here, we review measures of CIN through its rise as a field, discuss considerations for its measurement, and look forward to future quantification of CIN.
Collapse
Affiliation(s)
- Andrew Lynch
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, USA
- Division of Hematology/Oncology, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Shermineh Bradford
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, USA
- Division of Hematology/Oncology, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Mark E Burkard
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI, USA.
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI, USA.
- Division of Hematology/Oncology, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
28
|
Naumov SS, Krakhmal NV, Tashireva LA, Vtorushin SV. [Expression of immune checkpoints PD-L1, CTLA4, LAG3 in the microenvironment of colon adenocarcinoma depending on MMR status]. Arkh Patol 2024; 86:6-13. [PMID: 38591901 DOI: 10.17116/patol2024860216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
OBJECTIVE Study of the features of expression of immune checkpoint proteins PD-L1, CTLA4 and LAG3 in the microenvironment of colon adenocarcinoma depending on MMR status. MATERIAL AND METHODS The study group consisted of 32 patients with a morphologically confirmed diagnosis of colon cancer; all of them underwent surgical treatment in the form of hemicolonectomy or resection. The work assessed samples of tumor tissue obtained as a result of surgery, the study was carried out in 3 stages: morphological examination of histological slides of colon tumors at the light-optical level, immunohistochemistry examination of tumor samples to determine the dMMR/pMMR status of carcinoma using a panel of antibodies to proteins of the unpaired nucleotide repair system MLH1, MSH2, MSH6 and PMS2, multiplex analysis of PD-L1, CTLA4, LAG3, CD3+, CD8+, CD163+ markers using the Vectra 3.0.3 tissue scanning system (Perkin Elmer, USA). RESULTS Significant differences in the expression of PD-L1, CTLA4, LAG3 in the area of the invasive tumor margin were revealed between the dMMR and pMMR groups of colon adenocarcinomas in patients comparable in clinical and morphological characteristics and treatment. In the group of tumors with dMMR status, an increase in the expression of all studied markers was noted. The number of CD3+ TILs was also significantly higher in the invasive margin of tumors with dMMR status. Similarly, in this group of colon carcinomas, a large number of CD163+ macrophages were noted both in the center and in the invasive margin zone. No statistically significant differences were found in the expression of immune checkpoints and the composition of TILs in the central zone of tumors with different MMR status. CONCLUSION A study using multiplex immunohistochemical analysis showed that MMR-deficient colon adenocarcinomas are characterized by more pronounced immune infiltration and increased expression of immune checkpoints in microenvironmental cells, mainly in the area of invasive tumor growth. The data obtained may be important for understanding the mechanisms of immune-mediated control of tumor growth and the choice of immunotherapy tactics depending on MMR status.
Collapse
Affiliation(s)
- S S Naumov
- Siberian State Medical University, Tomsk, Russia
| | - N V Krakhmal
- Siberian State Medical University, Tomsk, Russia
- Cancer Research Institute - Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - L A Tashireva
- Cancer Research Institute - Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| | - S V Vtorushin
- Siberian State Medical University, Tomsk, Russia
- Cancer Research Institute - Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
29
|
Reynolds T, Riddick G, Meyers G, Gordon M, Flores Monar GV, Moon D, Moon C. Results Obtained from a Pivotal Validation Trial of a Microsatellite Analysis (MSA) Assay for Bladder Cancer Detection through a Statistical Approach Using a Four-Stage Pipeline of Modern Machine Learning Techniques. Int J Mol Sci 2023; 25:472. [PMID: 38203643 PMCID: PMC10778918 DOI: 10.3390/ijms25010472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/10/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Several studies have shown that microsatellite changes can be profiled in urine for the detection of bladder cancer. The use of microsatellite analysis (MSA) for bladder cancer detection requires a comprehensive analysis of as many as 15 to 20 markers, based on the amplification and interpretations of many individual MSA markers, and it can be technically challenging. Here, to develop fast, more efficient, standardized, and less costly MSA for the detection of bladder cancer, we developed three multiplex-polymerase-chain-reaction-(PCR)-based MSA assays, all of which were analyzed via a genetic analyzer. First, we selected 16 MSA markers based on 9 selected publications. Based on samples from Johns Hopkins University (the JHU sample, the first set sample), we developed an MSA based on triplet, three-tube-based multiplex PCR (a Triplet MSA assay). The discovery, validation, and translation of biomarkers for the early detection of cancer are the primary focuses of the Early Detection Research Network (EDRN), an initiative of the National Cancer Institute (NCI). A prospective study sponsored by the EDRN was undertaken to determine the efficacy of a novel set of MSA markers for the early detection of bladder cancer. This work and data analysis were performed through a collaboration between academics and industry partners. In the current study, we undertook a re-analysis of the primary data from the Compass study to enhance the predictive power of the dataset in bladder cancer diagnosis. Using a four-stage pipeline of modern machine learning techniques, including outlier removal with a nonlinear model, correcting for majority/minority class imbalance, feature engineering, and the use of a model-derived variable importance measure to select predictors, we were able to increase the utility of the original dataset to predict the occurrence of bladder cancer. The results of this analysis showed an increase in accuracy (85%), sensitivity (82%), and specificity (83%) compared to the original analysis. The re-analysis of the EDRN study results using machine learning statistical analysis proved to achieve an appropriate level of accuracy, sensitivity, and specificity to support the use of the MSA for bladder cancer detection and monitoring. This assay can be a significant addition to the tools urologists use to both detect primary bladder cancers and monitor recurrent bladder cancer.
Collapse
Affiliation(s)
- Thomas Reynolds
- NEXT Bio-Research Services, LLC, 11601 Ironbridge Road, Suite 101, Chester, VA 23831, USA; (T.R.); (G.M.)
| | - Gregory Riddick
- NEXT Bio-Research Services, LLC, 11601 Ironbridge Road, Suite 101, Chester, VA 23831, USA; (T.R.); (G.M.)
| | - Gregory Meyers
- NEXT Bio-Research Services, LLC, 11601 Ironbridge Road, Suite 101, Chester, VA 23831, USA; (T.R.); (G.M.)
| | - Maxie Gordon
- HJM Cancer Research Foundation Corporation, 10606 Candlewick Road, Lutherville, MD 21093, USA; (M.G.)
- BCD Innovations USA, 10606 Candlewick Road, Lutherville, MD 21093, USA
| | | | - David Moon
- HJM Cancer Research Foundation Corporation, 10606 Candlewick Road, Lutherville, MD 21093, USA; (M.G.)
| | - Chulso Moon
- HJM Cancer Research Foundation Corporation, 10606 Candlewick Road, Lutherville, MD 21093, USA; (M.G.)
- BCD Innovations USA, 10606 Candlewick Road, Lutherville, MD 21093, USA
- Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins Medical Institution, Cancer Research Building II, 5M3, 1550 Orleans Street, Baltimore, MD 21205, USA
| |
Collapse
|
30
|
Reynolds T, Bertsche K, Moon D, Moon C. Qualification of the Microsatellite Instability Analysis (MSA) for Bladder Cancer Detection: The Technical Challenges of Concordance Analysis. Int J Mol Sci 2023; 25:209. [PMID: 38203379 PMCID: PMC10779061 DOI: 10.3390/ijms25010209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/27/2023] [Accepted: 12/07/2023] [Indexed: 01/12/2024] Open
Abstract
Bladder cancer (here we refer to transitional carcinoma of bladder) is a major cause of morbidity and mortality in the Western world, and recent understanding of its etiology, the molecular characteristics associated with its progression, renders bladder cancer an ideal candidate for screening. Cystoscopy is invasive and sometimes carries unwanted complications, but it is the gold standard for the detection of bladder cancer. Urine cytology, while the most commonly used test as an initial screening tool, is of limited value due to its low sensitivity, particularly for low-grade tumors. Several new "molecular assays" for the diagnosis of urothelial cancer have been developed over the last two decades. Here, we have established our new bladder cancer test based on an assay established for the Early Detection Research Network (EDRN) study. As a part of the study, a quality control CLIA/College of American Pathology (CAP) accredited laboratory, (QA Lab), University of Maryland Baltimore Biomarker Reference Laboratory (UMB-BRL), performed quality assurance analysis. Quality assurance measures included a concordance study between the testing laboratory (AIBioTech), also CLIA/CAP accredited, and the QA lab to ensure that the assay was performed and the results were analyzed in a consistent manner. Therefore, following the technical transfer and training of the microsatellite analysis assay to the UMB-BRL and prior to the initiation of analysis of the clinical samples by the testing lab, a series of qualification studies were performed. This report details the steps taken to ensure qualification of the assay and illustrates the technical challenges facing biomarker validation of this kind.
Collapse
Affiliation(s)
- Thomas Reynolds
- NEXT Bio-Research Services, LLC, 11601 Ironbridge Road, Suite 101, Chester, VI 23831, USA
| | - Katie Bertsche
- NEXT Bio-Research Services, LLC, 11601 Ironbridge Road, Suite 101, Chester, VI 23831, USA
| | - David Moon
- HJM Cancer Research Foundation Corporation, 10606 Candlewick Road, Lutherville, MD 21093, USA
| | - Chulso Moon
- HJM Cancer Research Foundation Corporation, 10606 Candlewick Road, Lutherville, MD 21093, USA
- BCD Innovations USA, 10606 Candlewick Road, Lutherville, MD 21093, USA
- Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins Medical Institution, Cancer Research Building II, 5M3, 1550 Orleans Street, Baltimore, MD 21205, USA
| |
Collapse
|
31
|
Bacher JW, Udho EB, Strauss EE, Vyazunova I, Gallinger S, Buchanan DD, Pai RK, Templeton AS, Storts DR, Eshleman JR, Halberg RB. A Highly Sensitive Pan-Cancer Test for Microsatellite Instability. J Mol Diagn 2023; 25:806-826. [PMID: 37544360 PMCID: PMC10629437 DOI: 10.1016/j.jmoldx.2023.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 08/08/2023] Open
Abstract
Microsatellite instability (MSI) is an evolving biomarker for cancer detection and treatment. MSI was first used to identify patients with Lynch syndrome, a hereditary form of colorectal cancer (CRC), but has recently become indispensable in predicting patient response to immunotherapy. To address the need for pan-cancer MSI detection, a new multiplex assay was developed that uses novel long mononucleotide repeat (LMR) markers to improve sensitivity. A total of 469 tumor samples from 20 different cancer types, including 319 from patients with Lynch syndrome, were tested for MSI using the new LMR MSI Analysis System. Results were validated by using deficient mismatch repair (dMMR) status according to immunohistochemistry as the reference standard and compared versus the Promega pentaplex MSI panel. The sensitivity of the LMR panel for detection of dMMR status by immunohistochemistry was 99% for CRC and 96% for non-CRC. The overall percent agreement between the LMR and Promega pentaplex panels was 99% for CRC and 89% for non-CRC tumors. An increased number of unstable markers and the larger size shifts observed in dMMR tumors using the LMR panel increased confidence in MSI determinations. The LMR MSI Analysis System expands the spectrum of cancer types in which MSI can be accurately detected.
Collapse
Affiliation(s)
- Jeffery W Bacher
- R&D Clinical Diagnostics, Promega Corporation, Madison, Wisconsin; Department of Medicine, University of Wisconsin, Madison, Wisconsin.
| | - Eshwar B Udho
- R&D Clinical Diagnostics, Promega Corporation, Madison, Wisconsin
| | | | - Irina Vyazunova
- R&D Clinical Diagnostics, Promega Corporation, Madison, Wisconsin
| | - Steven Gallinger
- Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia; University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, Victoria, Australia; Genomic Medicine and Family Cancer Clinic, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Rish K Pai
- Health Science Research, Mayo Clinic, Scottsdale, Arizona
| | | | - Douglas R Storts
- R&D Clinical Diagnostics, Promega Corporation, Madison, Wisconsin
| | - James R Eshleman
- School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Richard B Halberg
- Department of Medicine, University of Wisconsin, Madison, Wisconsin; Department of Oncology, McArdle Laboratory of Cancer Research, University of Wisconsin, Madison, Wisconsin; University of Wisconsin Carbone Cancer Center, Madison, Wisconsin.
| |
Collapse
|
32
|
Reynolds T, Gordon M, Monar GVF, Moon D, Moon C. Development of Multiplex Polymerase Chain Reaction (PCR)-Based MSA Assay for Bladder Cancer Detection. Int J Mol Sci 2023; 24:13651. [PMID: 37686456 PMCID: PMC10488090 DOI: 10.3390/ijms241713651] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/18/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Several studies have shown that microsatellite changes can be profiled in the urine to detect bladder cancer. Microsatellite analysis (MSA) of bladder cancer detection requires a comprehensive analysis of up to 15-20 markers based on amplifying and interpreting many individual MSA markers, which can be technically challenging. To develop fast, efficient, standardized, and less costly MSA to detect bladder cancer, we developed three multiplex polymerase chain reaction (PCR) based MSA assays, all of which were analyzed by a genetic analyzer. First, we selected 16 MSA markers based on nine publications. We developed MSA assays based on triplet or three-tube-based multiplex PCR (Triplet MSA assay) using samples from Johns Hopkins University (JHU Sample, first set of samples). In the second set of samples (samples from six cancer patients and fourteen healthy individuals), our Triplet Assay with 15 MSA markers correctly predicted all 6/6 cancer samples to be cancerous and 14/14 healthy samples to be healthy. Although we could improve our report with more clinical information from patient samples and an increased number of cancer patients, our overall results suggest that our Triplet MSA Assay combined with a genetic analyzer is a potentially time- and cost-effective genetic assay for bladder cancer detection and has potential use as a dependable assay in patient care.
Collapse
Affiliation(s)
- Thomas Reynolds
- NEXT Bio-Research Services, LLC, 11601 Ironbridge Road, Suite 101, Chester, VA 23831, USA
| | - Maxie Gordon
- HJM Cancer Research Foundation Corporation, 10606 Candlewick Road, Lutherville, MD 21093, USA
- BCD Innovations USA, 10606 Candlewick Road, Lutherville, MD 21093, USA
| | | | - David Moon
- HJM Cancer Research Foundation Corporation, 10606 Candlewick Road, Lutherville, MD 21093, USA
| | - Chulso Moon
- HJM Cancer Research Foundation Corporation, 10606 Candlewick Road, Lutherville, MD 21093, USA
- BCD Innovations USA, 10606 Candlewick Road, Lutherville, MD 21093, USA
- Department of Otolaryngology-Head and Neck Surgery, The Johns Hopkins Medical Institution, Cancer Research Building II, 5M3, 1550 Orleans Street, Baltimore, MD 21205, USA
| |
Collapse
|
33
|
Guan J, Li GM. DNA mismatch repair in cancer immunotherapy. NAR Cancer 2023; 5:zcad031. [PMID: 37325548 PMCID: PMC10262306 DOI: 10.1093/narcan/zcad031] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/08/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023] Open
Abstract
Tumors defective in DNA mismatch repair (dMMR) exhibit microsatellite instability (MSI). Currently, patients with dMMR tumors are benefitted from anti-PD-1/PDL1-based immune checkpoint inhibitor (ICI) therapy. Over the past several years, great progress has been made in understanding the mechanisms by which dMMR tumors respond to ICI, including the identification of mutator phenotype-generated neoantigens, cytosolic DNA-mediated activation of the cGAS-STING pathway, type-I interferon signaling and high tumor-infiltration of lymphocytes in dMMR tumors. Although ICI therapy shows great clinical benefits, ∼50% of dMMR tumors are eventually not responsive. Here we review the discovery, development and molecular basis of dMMR-mediated immunotherapy, as well as tumor resistant problems and potential therapeutic interventions to overcome the resistance.
Collapse
Affiliation(s)
- Junhong Guan
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Guo-Min Li
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
34
|
Boland CR, Koi M, Hawn MT, Carethers JM, Yurgelun MB. Serendipity Strikes: How Pursuing Novel Hypotheses Shifted the Paradigm Regarding the Genetic Basis of Colorectal Cancer and Changed Cancer Therapy. Dig Dis Sci 2023; 68:3504-3513. [PMID: 37402979 PMCID: PMC11262588 DOI: 10.1007/s10620-023-08006-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2023] [Indexed: 07/06/2023]
Abstract
In this installment of the "Paradigm Shifts in Perspective" series, the authors, all scientists who have been involved in colorectal cancer (CRC) research for most or all of their careers, have watched the field develop from early pathological descriptions of tumor formation to the current understanding of tumor pathogenesis that informs personalized therapies. We outline how our understanding of the pathogenetic basis of CRC began with seemingly isolated discoveries-initially with the mutations in RAS and the APC gene, the latter of which was initially found in the context of intestinal polyposis, to the more complex process of multistep carcinogenesis, to the chase for tumor suppressor genes, which led to the unexpected discovery of microsatellite instability (MSI). These discoveries enabled the authors to better understand how the DNA mismatch repair (MMR) system not only recognizes DNA damage but also responds to damage by DNA repair or by triggering apoptosis in the injured cell. This work served, in part, to link the earlier findings on the pathogenesis of CRC to the development of immune checkpoint inhibitors, which has been transformative-and curative-for certain types of CRCs and other cancers as well. These discoveries also highlight the circuitous routes that scientific progress takes, which can include thoughtful hypothesis testing and at other times recognizing the importance of seemingly serendipitous observations that substantially change the flow and direction of the discovery process. What has happened over the past 37 years was not predictable when this journey began, but it does speak to the power of careful scientific experimentation, following the facts, perseverance in the face of opposition, and the willingness to think outside of established paradigms.
Collapse
Affiliation(s)
| | | | - Mary T Hawn
- Department of Surgery, Stanford University School of Medicine, CJ Huang Bldg, Palo Alto, CA, 94306, USA
| | | | - Matthew B Yurgelun
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
35
|
Desai H, Ofori S, Boatner L, Yu F, Villanueva M, Ung N, Nesvizhskii AI, Backus K. Multi-omic stratification of the missense variant cysteinome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.12.553095. [PMID: 37645963 PMCID: PMC10461992 DOI: 10.1101/2023.08.12.553095] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Cancer genomes are rife with genetic variants; one key outcome of this variation is gain-ofcysteine, which is the most frequently acquired amino acid due to missense variants in COSMIC. Acquired cysteines are both driver mutations and sites targeted by precision therapies. However, despite their ubiquity, nearly all acquired cysteines remain uncharacterized. Here, we pair cysteine chemoproteomics-a technique that enables proteome-wide pinpointing of functional, redox sensitive, and potentially druggable residues-with genomics to reveal the hidden landscape of cysteine acquisition. For both cancer and healthy genomes, we find that cysteine acquisition is a ubiquitous consequence of genetic variation that is further elevated in the context of decreased DNA repair. Our chemoproteogenomics platform integrates chemoproteomic, whole exome, and RNA-seq data, with a customized 2-stage false discovery rate (FDR) error controlled proteomic search, further enhanced with a user-friendly FragPipe interface. Integration of CADD predictions of deleteriousness revealed marked enrichment for likely damaging variants that result in acquisition of cysteine. By deploying chemoproteogenomics across eleven cell lines, we identify 116 gain-of-cysteines, of which 10 were liganded by electrophilic druglike molecules. Reference cysteines proximal to missense variants were also found to be pervasive, 791 in total, supporting heretofore untapped opportunities for proteoform-specific chemical probe development campaigns. As chemoproteogenomics is further distinguished by sample-matched combinatorial variant databases and compatible with redox proteomics and small molecule screening, we expect widespread utility in guiding proteoform-specific biology and therapeutic discovery.
Collapse
Affiliation(s)
- Heta Desai
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, 90095, USA
| | - Samuel Ofori
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Lisa Boatner
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, 90095, USA
| | - Fengchao Yu
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Miranda Villanueva
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, 90095, USA
| | - Nicholas Ung
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, 90095, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, 90095, USA
- DOE Institute for Genomics and Proteomics, UCLA, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, 90095, USA
| | - Alexey I Nesvizhskii
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Keriann Backus
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, 90095, USA
- DOE Institute for Genomics and Proteomics, UCLA, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, 90095, USA
| |
Collapse
|
36
|
Ascrizzi S, Arillotta GM, Grillone K, Caridà G, Signorelli S, Ali A, Romeo C, Tassone P, Tagliaferri P. Lynch Syndrome Biopathology and Treatment: The Potential Role of microRNAs in Clinical Practice. Cancers (Basel) 2023; 15:3930. [PMID: 37568746 PMCID: PMC10417124 DOI: 10.3390/cancers15153930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Lynch syndrome (LS), also known as Hereditary Non-Polyposis Colorectal Cancer (HNPCC), is an autosomal dominant cancer syndrome which causes about 2-3% of cases of colorectal carcinoma. The development of LS is due to the genetic and epigenetic inactivation of genes involved in the DNA mismatch repair (MMR) system, causing an epiphenomenon known as microsatellite instability (MSI). Despite the fact that the genetics of the vast majority of MSI-positive (MSI+) cancers can be explained, the etiology of this specific subset is still poorly understood. As a possible new mechanism, it has been recently demonstrated that the overexpression of certain microRNAs (miRNAs, miRs), such as miR-155, miR-21, miR-137, can induce MSI or modulate the expression of the genes involved in LS pathogenesis. MiRNAs are small RNA molecules that regulate gene expression at the post-transcriptional level by playing a critical role in the modulation of key oncogenic pathways. Increasing evidence of the link between MSI and miRNAs in LS prompted a deeper investigation into the miRNome involved in these diseases. In this regard, in this study, we discuss the emerging role of miRNAs as crucial players in the onset and progression of LS as well as their potential use as disease biomarkers and therapeutic targets in the current view of precision medicine.
Collapse
Affiliation(s)
- Serena Ascrizzi
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Grazia Maria Arillotta
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Katia Grillone
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Giulio Caridà
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Stefania Signorelli
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Asad Ali
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Caterina Romeo
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
- Medical Oncology and Translational Medical Oncology Units, University Hospital Renato Dulbecco, 88100 Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy; (S.A.); (G.M.A.); (K.G.); (G.C.); (S.S.); (A.A.); (C.R.); (P.T.)
- Medical Oncology and Translational Medical Oncology Units, University Hospital Renato Dulbecco, 88100 Catanzaro, Italy
| |
Collapse
|
37
|
Ratovomanana T, Nicolle R, Cohen R, Diehl A, Siret A, Letourneur Q, Buhard O, Perrier A, Guillerm E, Coulet F, Cervera P, Benusiglio P, Labrèche K, Colle R, Collura A, Despras E, Le Rouzic P, Renaud F, Cros J, Alentorn A, Touat M, Ayadi M, Bourgoin P, Prunier C, Tournigand C, Fouchardière CDL, Tougeron D, Jonchère V, Bennouna J, de Reynies A, Fléjou JF, Svrcek M, André T, Duval A. Prediction of response to immune checkpoint blockade in patients with metastatic colorectal cancer with microsatellite instability. Ann Oncol 2023; 34:703-713. [PMID: 37269904 DOI: 10.1016/j.annonc.2023.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND Mismatch repair-deficient (dMMR) tumors displaying microsatellite instability (MSI) represent a paradigm for the success of immune checkpoint inhibitor (ICI)-based immunotherapy, particularly in patients with metastatic colorectal cancer (mCRC). However, a proportion of patients with dMMR/MSI mCRC exhibit resistance to ICI. Identification of tools predicting MSI mCRC patient response to ICI is required for the design of future strategies further improving this therapy. PATIENTS AND METHODS We combined high-throughput DNA and RNA sequencing of tumors from 116 patients with MSI mCRC treated with anti-programmed cell death protein 1 ± anti-cytotoxic T-lymphocyte-associated protein 4 of the NIPICOL phase II trial (C1, NCT03350126, discovery set) and the ImmunoMSI prospective cohort (C2, validation set). The DNA/RNA predictors whose status was significantly associated with ICI status of response in C1 were subsequently validated in C2. Primary endpoint was progression-free survival by immune RECIST (iRECIST) (iPFS). RESULTS Analyses showed no impact of previously suggested DNA/RNA indicators of resistance to ICI, e.g. MSIsensor score, tumor mutational burden, or specific cellular and molecular tumoral contingents. By contrast, iPFS under ICI was shown in C1 and C2 to depend both on a multiplex MSI signature involving the mutations of 19 microsatellites hazard ratio cohort C2 (HRC2) = 3.63; 95% confidence interval (CI) 1.65-7.99; P = 1.4 × 10-3] and the expression of a set of 182 RNA markers with a non-epithelial transforming growth factor beta (TGFB)-related desmoplastic orientation (HRC2 = 1.75; 95% CI 1.03-2.98; P = 0.035). Both DNA and RNA signatures were independently predictive of iPFS. CONCLUSIONS iPFS in patients with MSI mCRC can be predicted by simply analyzing the mutational status of DNA microsatellite-containing genes in epithelial tumor cells together with non-epithelial TGFB-related desmoplastic RNA markers.
Collapse
Affiliation(s)
- T Ratovomanana
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris
| | - R Nicolle
- Université Paris Cité, Centre de Recherche sur l'Inflammation (CRI), INSERM, U1149, CNRS, ERL 8252, Paris; GERCOR, Groupe Coopérateur Multidisciplinaire en Oncologie, Paris
| | - R Cohen
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris; GERCOR, Groupe Coopérateur Multidisciplinaire en Oncologie, Paris; Departments of Medical Oncology
| | - A Diehl
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris
| | - A Siret
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris
| | - Q Letourneur
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris
| | - O Buhard
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris
| | - A Perrier
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris; Molecular Biology and Medical Genetics, Sorbonne Université, AP-HP, Hospital Pitié-Salpêtrière, Paris
| | - E Guillerm
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris; Molecular Biology and Medical Genetics, Sorbonne Université, AP-HP, Hospital Pitié-Salpêtrière, Paris
| | - F Coulet
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris; Molecular Biology and Medical Genetics, Sorbonne Université, AP-HP, Hospital Pitié-Salpêtrière, Paris
| | - P Cervera
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris
| | - P Benusiglio
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris; Molecular Biology and Medical Genetics, Sorbonne Université, AP-HP, Hospital Pitié-Salpêtrière, Paris
| | - K Labrèche
- CinBioS, MS 37 PASS Production de données en Sciences de la vie et de la Santé, INSERM, Sorbonne Université et SIRIC CURAMUS, Paris
| | - R Colle
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris; GERCOR, Groupe Coopérateur Multidisciplinaire en Oncologie, Paris; Departments of Medical Oncology
| | - A Collura
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris
| | - E Despras
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris
| | - P Le Rouzic
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris
| | - F Renaud
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris
| | - J Cros
- Department of Pathology, Beaujon Hospital, AP-HP, Clichy
| | - A Alentorn
- Service de Neurologie 2-Mazarin, Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, 47-83 boulevard de l'Hôpital, Paris
| | - M Touat
- Service de Neurologie 2-Mazarin, Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, 47-83 boulevard de l'Hôpital, Paris
| | - M Ayadi
- Programme "Cartes d'Identité des Tumeurs", Ligue Nationale Contre le Cancer, Paris
| | - P Bourgoin
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris; Department of Pathology, Sorbonne Université, AP-HP, Hôpital Saint-Antoine, Paris
| | - C Prunier
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Signalisation TGFB, plasticité cellulaire et Cancer, Paris
| | - C Tournigand
- Department of Medical Oncology, Hôpital Henri-Mondor, APHP, Université Paris Est Creteil, INSERM U955, Créteil
| | | | - D Tougeron
- ProDicET, UR 24144, University of Poitiers and Hepato-Gastroenterology Department, Poitiers University Hospital, Poitiers
| | - V Jonchère
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris
| | - J Bennouna
- Centre De Recherche En Cancérologie Et Immunologie Nantes-Angers (CRCINA), INSERM, Université d'Angers, Université De Nantes, Nantes
| | - A de Reynies
- Cartes d'Identité des Tumeurs Program, Ligue Nationale Contre Cancer, Paris, France
| | - J-F Fléjou
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris; Department of Pathology, Sorbonne Université, AP-HP, Hôpital Saint-Antoine, Paris
| | - M Svrcek
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris; Department of Pathology, Sorbonne Université, AP-HP, Hôpital Saint-Antoine, Paris
| | - T André
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris; GERCOR, Groupe Coopérateur Multidisciplinaire en Oncologie, Paris; Departments of Medical Oncology
| | - A Duval
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris; Molecular Biology and Medical Genetics, Sorbonne Université, AP-HP, Hospital Pitié-Salpêtrière, Paris.
| |
Collapse
|
38
|
Vuković Đerfi K, Salar A, Cacev T, Kapitanović S. EMAST Type of Microsatellite Instability-A Distinct Entity or Blurred Overlap between Stable and MSI Tumors. Genes (Basel) 2023; 14:1474. [PMID: 37510378 PMCID: PMC10380056 DOI: 10.3390/genes14071474] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Microsatellite instability (MSI) represents an accumulation of frameshifts in short tandem repeats, microsatellites, across the genome due to defective DNA mismatch repair (dMMR). MSI has been associated with distinct clinical, histological, and molecular features of tumors and has proven its prognostic and therapeutic value in different types of cancer. Recently, another type of microsatellite instability named elevated microsatellite alterations at selected tetranucleotide repeats (EMAST) has been reported across many different tumors. EMAST tumors have been associated with chronic inflammation, higher tumor stage, and poor prognosis. Nevertheless, the clinical significance of EMAST and its relation to MSI remains unclear. It has been proposed that EMAST arises as a result of isolated MSH3 dysfunction or as a secondary event in MSI tumors. Even though previous studies have associated EMAST with MSI-low phenotype in tumors, recent studies show a certain degree of overlap between EMAST and MSI-high tumors. However, even in stable tumors, (MSS) frameshifts in microsatellites can be detected as a purely stochastic event, raising the question of whether EMAST truly represents a distinct type of microsatellite instability. Moreover, a significant fraction of patients with MSI tumors do not respond to immunotherapy and it can be speculated that in these tumors, EMAST might act as a modifying factor.
Collapse
Affiliation(s)
- Kristina Vuković Đerfi
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Rudjer Boskovic Institute, Bijenicka cesta 54, 10000 Zagreb, Croatia
| | - Anamarija Salar
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Rudjer Boskovic Institute, Bijenicka cesta 54, 10000 Zagreb, Croatia
| | - Tamara Cacev
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Rudjer Boskovic Institute, Bijenicka cesta 54, 10000 Zagreb, Croatia
| | - Sanja Kapitanović
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Rudjer Boskovic Institute, Bijenicka cesta 54, 10000 Zagreb, Croatia
| |
Collapse
|
39
|
Rantanen P, Keränen A, Barot S, Ghazi S, Liljegren A, Nordenvall C, Lindblom A, Lindforss U. The prognostic significance of microsatellite instability in colorectal cancer: a Swedish multi-center study. Int J Colorectal Dis 2023; 38:197. [PMID: 37458848 PMCID: PMC10352163 DOI: 10.1007/s00384-023-04480-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/27/2023] [Indexed: 07/20/2023]
Abstract
PURPOSE About 10 to 15% of patients with sporadic colorectal cancer display mutations in DNA mismatch repair (MMR) genes shown as microsatellite instability (MSI). Previous reports of colorectal cancer (CRC) indicate a better prognosis for patients with MSI tumors compared to patients with microsatellite stable (MSS) tumors. In this study, our aim was to investigate whether MSI is an independent prognostic factor in CRC. PATIENTS AND METHODS Patients with stage I-III colorectal cancer and subject to curative surgery during 2002-2006 in the Swedish low-risk colorectal cancer study group cohort were eligible for inclusion. Deficient MMR (dMMR) status was analyzed by immunohistochemistry (IHC) and/or by MSI testing with polymerase chain reaction (PCR). Prognostic follow-up and treatment data were retrieved from patient records. Statistical analyses to assess MSI-status and prognosis were done using logistic regression and survival analyses using the Kaplan-Meier method and Cox regression hazards models adjusted for age, sex, stage, comorbidity, and tumor location. RESULTS In total, 463 patients were included, MSI high tumors were present in 66 patients (14%), and the remaining 397 were MSS/MSI low. Within 6 years, distant recurrences were present in 9.1% and 20.2% (P = 0.049), and death occurred in 25.8% and 31.5% in MSI and MSS patients, respectively. There was no statistically significant difference in overall mortality (HR 0.80, 95% CI 0.46-1.38), relapse-free survival (HR 0.82, 95% CI 0.50-1.36), or cancer-specific mortality (HR 1.60, 95% CI 0.73-3.51). CONCLUSION Despite distant metastases being less common in patients with MSI, there was no association between MSI and overall, relapse-free, or cancer-specific survival.
Collapse
Affiliation(s)
- Petri Rantanen
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
| | - Anne Keränen
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Shabane Barot
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet Stockholm, Sweden
| | - Sam Ghazi
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Annelie Liljegren
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Caroline Nordenvall
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Annika Lindblom
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Genetics, Karolinska Institutet, Stockholm, Sweden
| | - Ulrik Lindforss
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
40
|
Mestrallet G, Brown M, Bozkus CC, Bhardwaj N. Immune escape and resistance to immunotherapy in mismatch repair deficient tumors. Front Immunol 2023; 14:1210164. [PMID: 37492581 PMCID: PMC10363668 DOI: 10.3389/fimmu.2023.1210164] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/21/2023] [Indexed: 07/27/2023] Open
Abstract
Up to 30% of colorectal, endometrial and gastric cancers have a deficiency in mismatch repair (MMR) protein expression due to either germline or epigenetic inactivation. Patients with Lynch Syndrome who inherit an inactive MMR allele have an up to 80% risk for developing a mismatch repair deficient (MMRd) cancer. Due to an inability to repair DNA, MMRd tumors present with genomic instability in microsatellite regions (MS). Tumors with high MS instability (MSI-H) are characterized by an increased frequency of insertion/deletions (indels) that can encode novel neoantigens if they occur in coding regions. The high tumor antigen burden for MMRd cancers is accompanied by an inflamed tumor microenvironment (TME) that contributes to the clinical effectiveness of anti-PD-1 therapy in this patient population. However, between 40 and 70% of MMRd cancer patients do not respond to treatment with PD-1 blockade, suggesting that tumor-intrinsic and -extrinsic resistance mechanisms may affect the success of checkpoint blockade. Immune evasion mechanisms that occur during early tumorigenesis and persist through cancer development may provide a window into resistance pathways that limit the effectiveness of anti-PD-1 therapy. Here, we review the mechanisms of immune escape in MMRd tumors during development and checkpoint blockade treatment, including T cell dysregulation and myeloid cell-mediated immunosuppression in the TME. Finally, we discuss the development of new therapeutic approaches to tackle resistance in MMRd tumors, including cancer vaccines, therapies targeting immunosuppressive myeloid programs, and immune checkpoint combination strategies.
Collapse
Affiliation(s)
- Guillaume Mestrallet
- Division of Hematology and Oncology, Hess Center for Science & Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Matthew Brown
- Division of Hematology and Oncology, Hess Center for Science & Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Cansu Cimen Bozkus
- Division of Hematology and Oncology, Hess Center for Science & Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nina Bhardwaj
- Division of Hematology and Oncology, Hess Center for Science & Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Extramural member, Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
| |
Collapse
|
41
|
Fan X, Bai Q, Shi C, Xiao Y, Wang X. External quality assessment for the molecular detection of microsatellite instability in China, 2021-2022. Expert Rev Mol Diagn 2023; 23:1037-1043. [PMID: 37682059 DOI: 10.1080/14737159.2023.2257133] [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: 07/07/2023] [Revised: 08/14/2023] [Accepted: 08/27/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Microsatellite instability (MSI) analysis of tumors informs Lynch syndrome testing, therapeutic choice, and prognosis. The status of MSI is mainly detected by polymerase chain reaction coupled with capillary electrophoresis. However, there are various assays with different detection loci and the obtained results may vary. The objective of this study was to evaluate the concordance among different assays and the performance among different laboratories. METHODS External quality assessment (EQA) for the detection of MSI was performed in 2021 and 2022. Each sample panel consisted of five samples, including microsatellite-stable and MSI tumor tissues. The sample panels were coded at random, and the returned results were compared and scored. RESULTS The fully validated sample panels showed appropriate applicability with commercially available assays. There were eight false-negative results in 2021 and five false results (two false-positives and three false-negatives) in 2022. Among the participating laboratories, in 2021, 20 (74.07%) provided completely correct results; in 2022, 38 (92.68%) obtained an optimal score. CONCLUSION The molecular detection of MSI in China exhibited an improvement in a 2-year EQA study. Participation in EQA program is an efficient way of assessing the performance of laboratories and improving their ability.
Collapse
Affiliation(s)
- Xiaoyu Fan
- Department of Molecular Biology, Shanghai Center for Clinical Laboratory, Shanghai, P.R. China
| | - Qianming Bai
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Chunli Shi
- Department of Molecular Biology, Shanghai Center for Clinical Laboratory, Shanghai, P.R. China
| | - Yanqun Xiao
- Department of Molecular Biology, Shanghai Center for Clinical Laboratory, Shanghai, P.R. China
| | - Xueliang Wang
- Department of Molecular Biology, Shanghai Center for Clinical Laboratory, Shanghai, P.R. China
- Department of Molecular Diagnostic Innovation Technology, Shanghai Academy of Experimental Medicine, Shanghai, P.R. China
| |
Collapse
|
42
|
Sadee W, Wang D, Hartmann K, Toland AE. Pharmacogenomics: Driving Personalized Medicine. Pharmacol Rev 2023; 75:789-814. [PMID: 36927888 PMCID: PMC10289244 DOI: 10.1124/pharmrev.122.000810] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
Personalized medicine tailors therapies, disease prevention, and health maintenance to the individual, with pharmacogenomics serving as a key tool to improve outcomes and prevent adverse effects. Advances in genomics have transformed pharmacogenetics, traditionally focused on single gene-drug pairs, into pharmacogenomics, encompassing all "-omics" fields (e.g., proteomics, transcriptomics, metabolomics, and metagenomics). This review summarizes basic genomics principles relevant to translation into therapies, assessing pharmacogenomics' central role in converging diverse elements of personalized medicine. We discuss genetic variations in pharmacogenes (drug-metabolizing enzymes, drug transporters, and receptors), their clinical relevance as biomarkers, and the legacy of decades of research in pharmacogenetics. All types of therapies, including proteins, nucleic acids, viruses, cells, genes, and irradiation, can benefit from genomics, expanding the role of pharmacogenomics across medicine. Food and Drug Administration approvals of personalized therapeutics involving biomarkers increase rapidly, demonstrating the growing impact of pharmacogenomics. A beacon for all therapeutic approaches, molecularly targeted cancer therapies highlight trends in drug discovery and clinical applications. To account for human complexity, multicomponent biomarker panels encompassing genetic, personal, and environmental factors can guide diagnosis and therapies, increasingly involving artificial intelligence to cope with extreme data complexities. However, clinical application encounters substantial hurdles, such as unknown validity across ethnic groups, underlying bias in health care, and real-world validation. This review address the underlying science and technologies germane to pharmacogenomics and personalized medicine, integrated with economic, ethical, and regulatory issues, providing insights into the current status and future direction of health care. SIGNIFICANCE STATEMENT: Personalized medicine aims to optimize health care for the individual patients with use of predictive biomarkers to improve outcomes and prevent adverse effects. Pharmacogenomics drives biomarker discovery and guides the development of targeted therapeutics. This review addresses basic principles and current trends in pharmacogenomics, with large-scale data repositories accelerating medical advances. The impact of pharmacogenomics is discussed, along with hurdles impeding broad clinical implementation, in the context of clinical care, ethics, economics, and regulatory affairs.
Collapse
Affiliation(s)
- Wolfgang Sadee
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus Ohio (W.S., A.E.T.); Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida (D.W.); Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania (K.H.); Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California (W.S.); and Aether Therapeutics, Austin, Texas (W.S.)
| | - Danxin Wang
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus Ohio (W.S., A.E.T.); Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida (D.W.); Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania (K.H.); Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California (W.S.); and Aether Therapeutics, Austin, Texas (W.S.)
| | - Katherine Hartmann
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus Ohio (W.S., A.E.T.); Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida (D.W.); Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania (K.H.); Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California (W.S.); and Aether Therapeutics, Austin, Texas (W.S.)
| | - Amanda Ewart Toland
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus Ohio (W.S., A.E.T.); Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida (D.W.); Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania (K.H.); Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California (W.S.); and Aether Therapeutics, Austin, Texas (W.S.)
| |
Collapse
|
43
|
Shechter S, Ya'ar Bar S, Khattib H, Gage MJ, Avni D. Riok1, A Novel Potential Target in MSI-High p53 Mutant Colorectal Cancer Cells. Molecules 2023; 28:molecules28114452. [PMID: 37298928 DOI: 10.3390/molecules28114452] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/23/2023] [Accepted: 05/27/2023] [Indexed: 06/12/2023] Open
Abstract
The vulnerabilities of cancer cells constitute a promising strategy for drug therapeutics. This paper integrates proteomics, bioinformatics, and cell genotype together with in vitro cell proliferation assays to identify key biological processes and potential novel kinases that could account, at least in part, for the clinical differences observed in colorectal cancer (CRC) patients. This study started by focusing on CRC cell lines stratified by their microsatellite (MS) state and p53 genotype. It shows that cell-cycle checkpoint, metabolism of proteins and RNA, signal transduction, and WNT signaling processes are significantly more active in MSI-High p53-WT cell lines. Conversely, MSI-High cell lines with a mutant (Mut) p53 gene showed hyperactivation of cell signaling, DNA repair, and immune-system processes. Several kinases were linked to these phenotypes, from which RIOK1 was selected for additional exploration. We also included the KRAS genotype in our analysis. Our results showed that RIOK1's inhibition in CRC MSI-High cell lines was dependent on both the p53 and KRAS genotypes. Explicitly, Nintedanib showed relatively low cytotoxicity in MSI-High with both mutant p53 and KRAS (HCT-15) but no inhibition in p53 and KRAS WT (SW48) MSI-High cells. This trend was flipped in CRC MSI-High bearing opposite p53-KRAS genotypes (e.g., p53-Mut KRAS-WT or p53-WT KRAS-Mut), where observed cytotoxicity was more extensive compared to the p53-KRAS WT-WT or Mut-Mut cells, with HCT 116 (KRAS-Mut and p53-WT) being the most sensitive to RIOK1 inhibition. These results highlight the potential of our in silico computational approach to identify novel kinases in CRC sub-MSI-High populations as well as the importance of clinical genomics in determining drug potency.
Collapse
Affiliation(s)
- Sharon Shechter
- Department of Chemistry, University of Massachusetts Lowell, Lowell, MA 01854-2874, USA
| | - Sapir Ya'ar Bar
- Department of Natural Compound, Nutrition, and Health, MIGAL Galilee Research Institute, Kiryat Shmona 1101600, Israel
| | - Hamdan Khattib
- Department of Natural Compound, Nutrition, and Health, MIGAL Galilee Research Institute, Kiryat Shmona 1101600, Israel
| | - Matthew J Gage
- Department of Chemistry, University of Massachusetts Lowell, Lowell, MA 01854-2874, USA
| | - Dorit Avni
- Department of Natural Compound, Nutrition, and Health, MIGAL Galilee Research Institute, Kiryat Shmona 1101600, Israel
| |
Collapse
|
44
|
Kavun A, Veselovsky E, Lebedeva A, Belova E, Kuznetsova O, Yakushina V, Grigoreva T, Mileyko V, Fedyanin M, Ivanov M. Microsatellite Instability: A Review of Molecular Epidemiology and Implications for Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2023; 15:cancers15082288. [PMID: 37190216 DOI: 10.3390/cancers15082288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Microsatellite instability (MSI) is one of the most important molecular characteristics of a tumor, which occurs among various tumor types. In this review article, we examine the molecular characteristics of MSI tumors, both sporadic and Lynch-associated. We also overview the risks of developing hereditary forms of cancer and potential mechanisms of tumor development in patients with Lynch syndrome. Additionally, we summarize the results of major clinical studies on the efficacy of immune checkpoint inhibitors for MSI tumors and discuss the predictive role of MSI in the context of chemotherapy and checkpoint inhibitors. Finally, we briefly discuss some of the underlying mechanisms causing therapy resistance in patients treated with immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Egor Veselovsky
- OncoAtlas LLC, 119049 Moscow, Russia
- Department of Evolutionary Genetics of Development, Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, 119334 Moscow, Russia
| | | | - Ekaterina Belova
- OncoAtlas LLC, 119049 Moscow, Russia
- Faculty of Physics, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Olesya Kuznetsova
- OncoAtlas LLC, 119049 Moscow, Russia
- N.N. Blokhin Russian Cancer Research Center, 115478 Moscow, Russia
| | - Valentina Yakushina
- OncoAtlas LLC, 119049 Moscow, Russia
- Laboratory of Epigenetics, Research Centre for Medical Genetics, 115522 Moscow, Russia
| | - Tatiana Grigoreva
- OncoAtlas LLC, 119049 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia
| | | | - Mikhail Fedyanin
- N.N. Blokhin Russian Cancer Research Center, 115478 Moscow, Russia
- State Budgetary Institution of Health Care of the City of Moscow "Moscow Multidisciplinary Clinical Center" "Kommunarka" of the Department of Health of the City of Moscow, 142770 Moscow, Russia
- Federal State Budgetary Institution "National Medical and Surgical Center named after N.I. Pirogov" of the Ministry of Health of the Russian Federation, 105203 Moscow, Russia
| | - Maxim Ivanov
- OncoAtlas LLC, 119049 Moscow, Russia
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| |
Collapse
|
45
|
Shimozaki K, Nakayama I, Hirota T, Yamaguchi K. Current Strategy to Treat Immunogenic Gastrointestinal Cancers: Perspectives for a New Era. Cells 2023; 12:1049. [PMID: 37048122 PMCID: PMC10093684 DOI: 10.3390/cells12071049] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/15/2023] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
Since pembrolizumab, an anti-programmed death-1 (PD-1) antibody, showed a dramatic response to immunogenic cancers with microsatellite instability-high (MSI-H) and/or deficient mismatch repair (dMMR) in the pilot clinical trial KEYNOTE-016, subsequent studies have confirmed durable responses of anti-PD-1 inhibitors for MSI-H/dMMR solid tumors. As immunotherapy is described as a "game changer," the therapeutic landscape for MSI-H/dMMR solid tumors including gastrointestinal cancers has changed considerably in the last decade. An MSI/MMR status has been established as the predictive biomarker for immune checkpoint blockades, playing an indispensable role in the clinical practice of patients with MSI-H/dMMR tumors. Immunotherapy is also now investigated for locally advanced MSI-H/dMMR gastrointestinal cancers. Despite this great success, a few populations with MSI-H/dMMR gastrointestinal cancers do not respond to immunotherapy, possibly due to the existence of intrinsic or acquired resistance mechanisms. Clarifying the underlying mechanisms of resistance remains a future task, whereas attempts to overcome resistance and improve the efficacy of immunotherapy are currently ongoing. Herein, we review recent clinical trials with special attention to MSI-H/dMMR gastrointestinal cancers together with basic/translational findings, which provide their rationale, and discuss perspectives for the further therapeutic development of treatment in this field.
Collapse
Affiliation(s)
- Keitaro Shimozaki
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo 135-0063, Japan
- Department of Gastroenterology and Hepatology, Division of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Izuma Nakayama
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo 135-0063, Japan
| | - Toru Hirota
- Department of Experimental Pathology, Cancer Institute of the Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Kensei Yamaguchi
- Department of Gastrointestinal Oncology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo 135-0063, Japan
| |
Collapse
|
46
|
The Importance of Immunohistochemical Heterogeneous Expression of MMR Protein in Patients with Colorectal Cancer in Stage II and III of the Disease. Medicina (B Aires) 2023; 59:medicina59030489. [PMID: 36984490 PMCID: PMC10051778 DOI: 10.3390/medicina59030489] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Background and objectives: In patients with colorectal cancer (CRC), heterogeneous expression of Mismatch repair (MMR) proteins can manifest itself in several different forms and is not such a rare phenomenon. Therefore, it is very important to recognize the nuclear expression of MMR proteins of different MMR status in order to avoid false positive or false negative results. The aim of this study was to determine the frequency and distribution of heterogeneous expression of MMR proteins in patients with stages II and III of the disease as well as its association with clinical, demographic and pathological characteristics of CRC in relation to proficient and deficient expression of MMR proteins. Material and Methods: The study included 104 cases of colorectal cancer obtained from surgical colectomy material in stages II and III of the disease. Results: From a total of 104 patients with colorectal cancer, immunohistochemical analysis of the expression of all four MMR proteins showed that heterogeneous expression of MMR proteins (as well as deficient immunoreactivity of tumor cells) was present in 12 cases, while proficient expression of MMR proteins was detected in 80 tumors. Conclusions: Our study showed that the only independent predictors of the loss of MMR protein expression were younger patient age and right-sided anatomical location of the tumor. The study also established the existence of heterogeneous expression of MMR proteins in a non-negligible percentage of CRCs (11.5%), where heterogeneous nuclear expression of MMR proteins was described in several different forms.
Collapse
|
47
|
Kansikas M, Vähätalo L, Kantelinen J, Kasela M, Putula J, Døhlen A, Paloviita P, Kärkkäinen E, Lahti N, Arnez P, Kilpinen S, Alcala-Repo B, Pylvänäinen K, Pöyhönen M, Peltomäki P, Järvinen HJ, Seppälä TT, Renkonen-Sinisalo L, Lepistö A, Mecklin JP, Nyström M. Tumor-independent Detection of Inherited Mismatch Repair Deficiency for the Diagnosis of Lynch Syndrome with High Specificity and Sensitivity. CANCER RESEARCH COMMUNICATIONS 2023; 3:361-370. [PMID: 36875157 PMCID: PMC9979712 DOI: 10.1158/2767-9764.crc-22-0384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/20/2022] [Accepted: 02/07/2023] [Indexed: 06/18/2023]
Abstract
UNLABELLED Lynch syndrome (LS) is the most common hereditary cancer syndrome. Early diagnosis improves prognosis and reduces health care costs, through existing cancer surveillance methods. The problem is finding and diagnosing the cancer predisposing genetic condition. The current workup involves a complex array of tests that combines family cancer history and clinical phenotypes with tumor characteristics and sequencing data, followed by a challenging task to interpret the found variant(s). On the basis of the knowledge that an inherited mismatch repair (MMR) deficiency is a hallmark of LS, we have developed and validated a functional MMR test, DiagMMR, that detects inherited MMR deficiency directly from healthy tissue without need of tumor and variant information. The validation included 119 skin biopsies collected from clinically pathogenic MMR variant carriers (MSH2, MSH6) and controls, and was followed by a small clinical pilot study. The repair reaction was performed on proteins extracted from primary fibroblasts and the interpretation was based on the MMR capability of the sample in relation to cutoff, which distinguishes MMR proficient (non-LS) from MMR deficient (LS) function. The results were compared with the reference standard (germline NGS). The test was shown to have exceptional specificity (100%) with high sensitivity (89%) and accuracy (97%). The ability to efficiently distinguish LS carriers from controls was further shown with a high area under the receiving operating characteristic (AUROC) value (0.97). This test offers an excellent tool for detecting inherited MMR deficiency linked to MSH2 or MSH6 and can be used alone or with conventional tests to recognize genetically predisposed individuals. SIGNIFICANCE Clinical validation of DiagMMR shows high accuracy in distinguishing individuals with hereditary MSH2 or MSH6 MMR deficiency (i.e., LS). The method presented overcomes challenges faced by the complexity of current methods and can be used alone or with conventional tests to improve the ability to recognize genetically predisposed individuals.
Collapse
Affiliation(s)
- Minttu Kansikas
- LS CancerDiag Ltd., Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Laura Vähätalo
- LS CancerDiag Ltd., Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Jukka Kantelinen
- LS CancerDiag Ltd., Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Mariann Kasela
- LS CancerDiag Ltd., Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Jaana Putula
- LS CancerDiag Ltd., Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Anni Døhlen
- LS CancerDiag Ltd., Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Pauliina Paloviita
- LS CancerDiag Ltd., Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Emmi Kärkkäinen
- LS CancerDiag Ltd., Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Niklas Lahti
- LS CancerDiag Ltd., Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Philippe Arnez
- LS CancerDiag Ltd., Helsinki, Finland
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Sami Kilpinen
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | | | - Kirsi Pylvänäinen
- Department of Education and Science, Nova Hospital, Central Finland Health Care District, Jyväskylä, Finland
| | - Minna Pöyhönen
- Department of Genetics, HUSLAB, Helsinki University Hospital Diagnostic Center, Helsinki, Finland
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Applied Tumor Genomics, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Päivi Peltomäki
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | | | - Toni T. Seppälä
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Applied Tumor Genomics, Research Programs Unit, University of Helsinki, Helsinki, Finland
- Faculty of Medicine and Medical Technology, University of Tampere, Tampere, Finland
- Department of Gastroenterology and Alimentary Tract Surgery, Tampere University Hospital, Tampere, Finland
| | - Laura Renkonen-Sinisalo
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Applied Tumor Genomics, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Anna Lepistö
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Applied Tumor Genomics, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Jukka-Pekka Mecklin
- Department of Education and Science, Nova Hospital, Central Finland Health Care District, Jyväskylä, Finland
- Faculty of Sports and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Minna Nyström
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
48
|
Calluori S, Stark R, Pearson BL. Gene-Environment Interactions in Repeat Expansion Diseases: Mechanisms of Environmentally Induced Repeat Instability. Biomedicines 2023; 11:515. [PMID: 36831049 PMCID: PMC9953593 DOI: 10.3390/biomedicines11020515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Short tandem repeats (STRs) are units of 1-6 base pairs that occur in tandem repetition to form a repeat tract. STRs exhibit repeat instability, which generates expansions or contractions of the repeat tract. Over 50 diseases, primarily affecting the central nervous system and muscles, are characterized by repeat instability. Longer repeat tracts are typically associated with earlier age of onset and increased disease severity. Environmental exposures are suspected to play a role in the pathogenesis of repeat expansion diseases. Here, we review the current knowledge of mechanisms of environmentally induced repeat instability in repeat expansion diseases. The current evidence demonstrates that environmental factors modulate repeat instability via DNA damage and induction of DNA repair pathways, with distinct mechanisms for repeat expansion and contraction. Of particular note, oxidative stress is a key mediator of environmentally induced repeat instability. The preliminary evidence suggests epigenetic modifications as potential mediators of environmentally induced repeat instability. Future research incorporating an array of environmental exposures, new human cohorts, and improved model systems, with a continued focus on cell-types, tissues, and critical windows, will aid in identifying mechanisms of environmentally induced repeat instability. Identifying environmental modulators of repeat instability and their mechanisms of action will inform preventions, therapies, and public health measures.
Collapse
Affiliation(s)
- Stephanie Calluori
- Department of Environmental Health Sciences, Mailman School of Public Health Columbia University, New York, NY 10032, USA
- Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, USA
| | - Rebecca Stark
- Department of Environmental Health Sciences, Mailman School of Public Health Columbia University, New York, NY 10032, USA
| | - Brandon L. Pearson
- Department of Environmental Health Sciences, Mailman School of Public Health Columbia University, New York, NY 10032, USA
| |
Collapse
|
49
|
Rider SD, Damewood FJ, Gadgil RY, Hitch DC, Alhawach V, Shrestha R, Shanahan M, Zavada N, Leffak M. Suppressors of Break-Induced Replication in Human Cells. Genes (Basel) 2023; 14:genes14020398. [PMID: 36833325 PMCID: PMC9956954 DOI: 10.3390/genes14020398] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/23/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Short tandem DNA repeats are drivers of genome instability. To identify suppressors of break-induced mutagenesis human cells, unbiased genetic screens were conducted using a lentiviral shRNA library. The recipient cells possessed fragile non-B DNA that could induce DNA double-strand breaks (DSBs), integrated at an ectopic chromosomal site adjacent to a thymidine kinase marker gene. Mutagenesis of the thymidine kinase gene rendered cells resistant to the nucleoside analog ganciclovir (GCV). The screen identified genes that have established roles in DNA replication and repair, chromatin modification, responses to ionizing radiation, and genes encoding proteins enriched at replication forks. Novel loci implicated in BIR included olfactory receptors, the G0S2 oncogene/tumor suppressor axis, the EIF3H-METTL3 translational regulator, and the SUDS3 subunit of the Sin3A corepressor. Consistent with a role in suppressing BIR, siRNA knockdown of selected candidates increased the frequency of the GCVr phenotype and increased DNA rearrangements near the ectopic non-B DNA. Inverse PCR and DNA sequence analyses showed that hits identified in the screen increased genome instability. Further analysis quantitated repeat-induced hypermutagenesis at the ectopic site and showed that knockdown of a primary hit, COPS2, induced mutagenic hotspots, remodeled the replication fork, and increased nonallelic chromosome template switches.
Collapse
|
50
|
Sultana H, Aamir MN, Madni A, Rehman MU, Shafiq A, Shirazi JH, Hassan S, Sumaira. Polymeric Nanogel for Oral Delivery of the Chemotherapeutic Agent: Fabrication and Evaluation Alongside Toxicological Studies and Histopathological Examination. AAPS PharmSciTech 2023; 24:43. [PMID: 36702971 DOI: 10.1208/s12249-023-02499-7] [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: 08/16/2022] [Accepted: 12/27/2022] [Indexed: 01/27/2023] Open
Abstract
Nanogel has attracted considerable attention as one of the most versatile drug delivery systems, especially for site-specific and/or time-controlled delivery of the chemotherapeutic agent. The main objective of this study was to prepare the polymeric nanogel characterized by Fourier transform infrared spectroscopy, x-ray diffraction, thermogravimetric analysis, differential scanning, and oral acute toxicity. Free radical polymerization was done for the fabrication of polymeric nanogel. Fourier transform infrared spectroscopy was used to confirm the successful free radical polymerization. Various techniques such as x-ray diffraction, differential scanning calorimetric, and thermogravimetric analysis measurement were used to investigate the thermal behavior and crystallinity of developed nanogel. Parameters such as swelling, drug loading, and in vitro drug release is enhanced as polymers and monomers concentrations increase while these parameters decrease in case of increasing crosslinker concentration. The oral biocompatibility results of developed nanogel exhibited no toxicity in rabbits. Histopathological changes were observed between empty and loaded group. The nanosized gel offers a specific surface area which increases the stability of loaded drug (oxaliplatin) and bioavailability of the drug (oxaliplatin) as compared to the conventional drug delivery systems.
Collapse
Affiliation(s)
- Humaira Sultana
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Bahawalpur, Pakistan
| | - Muhammad Naeem Aamir
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Bahawalpur, Pakistan. .,School of Pharmacy, Newcastle University, Newcastle Upon Tyne, NE1 7RU, UK.
| | - Asadullah Madni
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Bahawalpur, Pakistan
| | - Masood Ur Rehman
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, 45320, Pakistan
| | - Afifa Shafiq
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Bahawalpur, Pakistan
| | - Jafir Hussain Shirazi
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Bahawalpur, Pakistan
| | - Sidra Hassan
- Bahawalpur College of Pharmacy, Bahawalpur Medical and Dental College, Bahawalpur, Pakistan
| | - Sumaira
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, 63100, Bahawalpur, Pakistan
| |
Collapse
|