1
|
Zhao Y, Lan T, Zhong G, Hagen J, Pan H, Chung WK, Shen Y. A probabilistic graphical model for estimating selection coefficients of nonsynonymous variants from human population sequence data. Nat Commun 2025; 16:4670. [PMID: 40393980 DOI: 10.1038/s41467-025-59937-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/06/2025] [Indexed: 05/22/2025] Open
Abstract
Accurately predicting the effect of missense variants is important in discovering disease risk genes and clinical genetic diagnostics. Commonly used computational methods predict pathogenicity, which does not capture the quantitative impact on fitness in humans. We develop a method, MisFit, to estimate missense fitness effect using a graphical model. MisFit jointly models the effect at a molecular level ( d ) and a population level (selection coefficient, s ), assuming that in the same gene, missense variants with similar d have similar s . We train it by maximizing probability of observed allele counts in 236,017 individuals of European ancestry. We show that s is informative in predicting allele frequency across ancestries and consistent with the fraction of de novo mutations in sites under strong selection. Further, s outperforms previous methods in prioritizing de novo missense variants in individuals with neurodevelopmental disorders. In conclusion, MisFit accurately predicts s and yields new insights from genomic data.
Collapse
Affiliation(s)
- Yige Zhao
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- The Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, New York, NY, USA
| | - Tian Lan
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Guojie Zhong
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- The Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, New York, NY, USA
| | - Jake Hagen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hongbing Pan
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
| | - Wendy K Chung
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA.
- JP Sulzberger Columbia Genome Center, Columbia University, New York, NY, USA.
| |
Collapse
|
2
|
Varga MJ, Richardson ME, Chamberlin A. Structural biology in variant interpretation: Perspectives and practices from two studies. Am J Hum Genet 2025; 112:984-992. [PMID: 40233741 DOI: 10.1016/j.ajhg.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
Structural biology offers a powerful lens through which to assess genetic variants by providing insights into their impact on clinically relevant protein structure and function. Due to the availability of new, user-friendly, web-based tools, structural analyses by wider audiences have become more mainstream. These new tools, including AlphaMissense and AlphaFold, have recently been in the limelight due to their initial success and projected future promise; however, the intricacies and limitations of using these tools still need to be disseminated to the more general audience that is likely to use them in variant analysis. Here, we expound on frameworks applying structural biology to variant interpretation by examining two accompanying articles. To this end, we explore the nuances of choosing the correct protein model, compare and contrast various structural approaches, and highlight both the advantages and limitations of employing structural biology in variant interpretation. Using two articles published in this issue of The American Journal of Human Genetics as a baseline, we focus on case studies in TP53 and BRCA1 to illuminate gene-specific differences in the applications of structural information, which illustrate the complexities inherent in this field. Additionally, we discuss the implications of recent advancements, such as AlphaFold, and provide practical guidance for researchers navigating variant interpretation using structural biology.
Collapse
|
3
|
Jaber S, Eldawra E, Rakotopare J, Simeonova I, Lejour V, Gabriel M, Cañeque T, Volochtchouk V, Licaj M, Fajac A, Rodriguez R, Morillon A, Bardot B, Toledo F. Oncogenic and teratogenic effects of Trp53Y217C, an inflammation-prone mouse model of the human hotspot mutant TP53Y220C. eLife 2025; 13:RP102434. [PMID: 40223808 PMCID: PMC11996178 DOI: 10.7554/elife.102434] [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] [Indexed: 04/15/2025] Open
Abstract
Missense 'hotspot' mutations localized in six p53 codons account for 20% of TP53 mutations in human cancers. Hotspot p53 mutants have lost the tumor suppressive functions of the wildtype protein, but whether and how they may gain additional functions promoting tumorigenesis remain controversial. Here, we generated Trp53Y217C, a mouse model of the human hotspot mutant TP53Y220C. DNA damage responses were lost in Trp53Y217C/Y217C (Trp53YC/YC) cells, and Trp53YC/YC fibroblasts exhibited increased chromosome instability compared to Trp53-/- cells. Furthermore, Trp53YC/YC male mice died earlier than Trp53-/- males, with more aggressive thymic lymphomas. This correlated with an increased expression of inflammation-related genes in Trp53YC/YC thymic cells compared to Trp53-/- cells. Surprisingly, we recovered only one Trp53YC/YC female for 22 Trp53YC/YC males at weaning, a skewed distribution explained by a high frequency of Trp53YC/YC female embryos with exencephaly and the death of most Trp53YC/YC female neonates. Strikingly, however, when we treated pregnant females with the anti-inflammatory drug supformin (LCC-12), we observed a fivefold increase in the proportion of viable Trp53YC/YC weaned females in their progeny. Together, these data suggest that the p53Y217C mutation not only abrogates wildtype p53 functions but also promotes inflammation, with oncogenic effects in males and teratogenic effects in females.
Collapse
Affiliation(s)
- Sara Jaber
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Eliana Eldawra
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Jeanne Rakotopare
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Iva Simeonova
- Chromatin Dynamics, Institut Curie, CNRS UMR3664, Sorbonne University, PSL UniversityParisFrance
| | - Vincent Lejour
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Marc Gabriel
- Non Coding RNA, Epigenetic and Genome Fluidity, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Tatiana Cañeque
- Chemical Biology, Institut Curie, CNRS UMR3666, INSERM U1143, PSL UniversityParisFrance
| | - Vitalina Volochtchouk
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Monika Licaj
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Anne Fajac
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Raphaël Rodriguez
- Chemical Biology, Institut Curie, CNRS UMR3666, INSERM U1143, PSL UniversityParisFrance
| | - Antonin Morillon
- Non Coding RNA, Epigenetic and Genome Fluidity, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Boris Bardot
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
- Signaling and Neural Crest Development, Institut Curie, CNRS UMR3347, INSERM U1021, Université Paris-Saclay, PSL UniversityOrsayFrance
| | - Franck Toledo
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
- Hematopoietic and Leukemic Development, Centre de Recherche Saint-Antoine, INSERM UMRS938, Sorbonne UniversityParisFrance
| |
Collapse
|
4
|
Zhao Y, Lan T, Zhong G, Hagen J, Pan H, Chung WK, Shen Y. A probabilistic graphical model for estimating selection coefficient of nonsynonymous variants from human population sequence data. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2023.12.11.23299809. [PMID: 38168397 PMCID: PMC10760286 DOI: 10.1101/2023.12.11.23299809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Accurately predicting the effect of missense variants is important in discovering disease risk genes and clinical genetic diagnostics. Commonly used computational methods predict pathogenicity, which does not capture the quantitative impact on fitness in humans. We developed a method, MisFit, to estimate missense fitness effect using a graphical model. MisFit jointly models the effect at a molecular level (𝑑) and a population level (selection coefficient, 𝑠), assuming that in the same gene, missense variants with similar 𝑑 have similar 𝑠. We trained it by maximizing probability of observed allele counts in 236,017 European individuals. We show that 𝑠 is informative in predicting allele frequency across ancestries and consistent with the fraction of de novo mutations in sites under strong selection. Further, 𝑠 outperforms previous methods in prioritizing de novo missense variants in individuals with neurodevelopmental disorders. In conclusion, MisFit accurately predicts 𝑠 and yields new insights from genomic data.
Collapse
Affiliation(s)
- Yige Zhao
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032
- The Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, New York, NY 10032
| | - Tian Lan
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032
| | - Guojie Zhong
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032
- The Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, New York, NY 10032
| | - Jake Hagen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032
- . Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115
| | - Hongbing Pan
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY 10032
| | - Wendy K. Chung
- . Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115
| | - Yufeng Shen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY 10032
- JP Sulzberger Columbia Genome Center, Columbia University, New York, NY 10032
| |
Collapse
|
5
|
Urano T, Yokota E, Iwai M, Yukawa T, Naomoto Y, Takigawa N, Fujiwara H, Akiyama T, Haisa M, Fukazawa T, Yamatsuji T. Establishment and characterization of novel patient-derived esophageal tumoroids with long-term cultivability. Hum Cell 2025; 38:72. [PMID: 40108093 DOI: 10.1007/s13577-025-01206-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Esophageal cancer is an aggressive and fatal disease classified into two distinct histologic types: esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). To develop novel therapeutic strategies, it is important to establish preclinical models of esophageal carcinoma. In this study, we successfully established three types of human esophageal cancer organoids (tumoroids) from surgical specimens for long-term culture. Two of the tumoroids were derived from ESCC and one from EAC, which arose from Barrett's esophagus. Whole-exome sequencing revealed that the tumoroids inherited genetic mutations from the parental tumors and patient-derived tumor xenografts closely mimicked the pathology of the original esophageal cancers. In addition to whole-exome analysis, copy number and immunohistochemical analyses demonstrated HER2 expression and amplification as well as HER3 expression and mutation in esophageal tumoroids derived from adenocarcinoma in Barrett's esophagus. HER2-targeting antibody-drug conjugates (ADCs), trastuzumab deruxtecan (T-DXd), and patritumab deruxtecan (P-DXd) effectively suppressed the viability of the tumoroids. Therefore, the establishment of esophageal tumoroids with long-term cultivability makes it possible to obtain reproducible basic data, including drug sensitivity studies, which are important for the development of personalized therapies and treatment strategies.
Collapse
Affiliation(s)
- Takashi Urano
- Department of General Surgery, Kawasaki Medical School, Okayama, 700-8505, Japan
| | - Etsuko Yokota
- Department of General Surgery, Kawasaki Medical School, Okayama, 700-8505, Japan
| | - Miki Iwai
- General Medical Center Research Unit, Kawasaki Medical School, Okayama, 700-8505, Japan
| | - Takuro Yukawa
- Department of General Surgery, Kawasaki Medical School, Okayama, 700-8505, Japan
| | - Yoshio Naomoto
- Department of General Surgery, Kawasaki Medical School, Okayama, 700-8505, Japan
| | - Nagio Takigawa
- General Medical Center Research Unit, Kawasaki Medical School, Okayama, 700-8505, Japan
- Department of General Internal Medicine 4, Kawasaki Medical School, Okayama, 700-8505, Japan
| | - Hideyo Fujiwara
- Department of Pathology, Kawasaki Medical School, Okayama, 700-8505, Japan
| | - Takashi Akiyama
- Department of Pathology, Kawasaki Medical School, Okayama, 700-8505, Japan
| | - Minoru Haisa
- Kawasaki Medical School General Medical Center, Okayama, 700-8505, Japan
| | - Takuya Fukazawa
- Department of General Surgery, Kawasaki Medical School, Okayama, 700-8505, Japan.
- General Medical Center Research Unit, Kawasaki Medical School, Okayama, 700-8505, Japan.
| | - Tomoki Yamatsuji
- Department of General Surgery, Kawasaki Medical School, Okayama, 700-8505, Japan
| |
Collapse
|
6
|
Lock IC, Leisenring NH, Floyd W, Xu ES, Luo L, Ma Y, Mansell EC, Cardona DM, Lee CL, Kirsch DG. Mis-splicing drives loss of function of p53E224D point mutation. PLoS One 2025; 20:e0318856. [PMID: 40043089 PMCID: PMC11882087 DOI: 10.1371/journal.pone.0318856] [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: 09/03/2024] [Accepted: 01/22/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND The tumor suppressor p53 (Trp53), also known as p53, is the most commonly mutated gene in cancer. Canonical p53 DNA damage response pathways are well characterized and classically thought to underlie the tumor suppressive effect of p53. Challenging this dogma, mouse models have revealed that p53-driven apoptosis and cell cycle arrest are dispensable for tumor suppression. Here, we investigated the inverse context of a p53 mutation predicted to drive the expression of canonical targets but is detected in human cancer. METHODS We established a novel mouse model with a single base pair mutation (GAG>GAT, p53E221D) in the DNA-Binding domain that has wild-type function in screening assays, but is paradoxically found in human cancer in Li-Fraumeni syndrome. Using mouse p53E221D and the analogous human p53E224D mutants, we evaluated expression, transcriptional activation, and tumor suppression in vitro and in vivo. RESULTS Expression of human p53E224D from cDNA translated to a fully functional p53 protein. However, p53E221D/E221D RNA transcribed from the endogenous locus is mis-spliced resulting in nonsense-mediated decay. Moreover, fibroblasts derived from p53E221D/E221D mice do not express a detectable protein product. Mice homozygous for p53E221D exhibited increased tumor penetrance and decreased life expectancy compared to p53WT/WT animals. CONCLUSIONS Mouse p53E221D and human p53E224D mutations lead to splice variation and a biologically relevant p53 loss of function in vitro and in vivo.
Collapse
Affiliation(s)
- Ian C. Lock
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Nathan H. Leisenring
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Warren Floyd
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Radiation Oncology, Maryland Anderson Cancer Center, Houston, Texas, United States of America
| | - Eric S. Xu
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Erin C. Mansell
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Diana M. Cardona
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Chang-Lung Lee
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - David G. Kirsch
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Narasimhamurthy M, Le A, Boruah N, Moses R, Kelly G, Bleiweiss I, Maxwell KN, Nayak A. Clinicopathologic Features of Breast Tumors in Germline TP53 Variant-Associated Li-Fraumeni Syndrome. Am J Surg Pathol 2025; 49:195-205. [PMID: 39629784 DOI: 10.1097/pas.0000000000002338] [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: 02/20/2025]
Abstract
We present one of the largest cohorts of TP53 -pathogenic germline variants (PGVs) associated with patients with Li-Fraumeni syndrome (n = 82) with breast tumors (19 to 76 y; median age: 35). Most had missense variants (77%), followed by large gene rearrangements (LGRs; 12%), truncating (6%), and splice-site (5%) variants. Twenty-one unique germline missense variants were found, with hotspots at codons 175, 181, 245, 248, 273, 334, and 337. Of 100 total breast tumors, 63% were invasive (mostly ductal), 30% pure ductal carcinoma in situ, 4% fibroepithelial lesions, and 3% with unknown histology. Unlike BRCA -associated tumors, approximately half of the breast cancers exhibited HER2 -positivity, of which ~50% showed estrogen receptor coexpression. Pathology slides were available for review for 61 tumors (44 patients), and no significant correlation between the type of TP53 PGVs and histologic features was noted. High p53 immunohistochemistry expression (>50%) was seen in 67% of tumors tested (mostly missense variant). Null pattern (<1% cells) was seen in 2 (LGR and splicing variants carriers). Surprisingly, 2 tumors from patients with an LGR and 1 tumor from a patient with a truncating variant showed p53 overexpression (>50%). The subset of patients with the Brazilian p.R337H variant presented at a higher age than those with non-p.R337H variant (46 vs 35 y) though statistically insignificant ( P = 0.071) due to an imbalance in the sample size, and were uniquely negative for HER2 -overexpressing tumors. To conclude, breast cancer in carriers of TP53 PGVs has some unique clinicopathological features that suggest differential mechanisms of tumor formation. p53 immunohistochemistry cannot be used as a surrogate marker to identify germline TP53 -mutated breast cancers.
Collapse
Affiliation(s)
| | - Anh Le
- Department of Medicine, Division of Hematology/Oncology
| | | | - Renyta Moses
- Department of Medicine, Division of Hematology/Oncology
| | - Gregory Kelly
- Department of Medicine, Division of Hematology/Oncology
| | | | - Kara N Maxwell
- Department of Medicine, Division of Hematology/Oncology, University of Pennsylvania Health System
| | - Anupma Nayak
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Health System, Philadelphia, PA
| |
Collapse
|
8
|
Butera A, Amelio I. Deciphering the significance of p53 mutant proteins. Trends Cell Biol 2025; 35:258-268. [PMID: 38960851 DOI: 10.1016/j.tcb.2024.06.003] [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: 04/29/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 07/05/2024]
Abstract
Mutations in the p53 gene compromise its role as guardian of genomic integrity, yielding predominantly missense p53 mutant proteins. The gain-of-function hypothesis has long suggested that these mutant proteins acquire new oncogenic properties; however, recent studies challenge this notion, indicating that targeting these mutants may not impact the fitness of cancer cells. Mounting evidence indicates that tumorigenesis involves a cooperative interplay between driver mutations and cellular state, influenced by developmental stage, external insults, and tissue damage. Consistently, the behavior and properties of p53 mutants are altered by the context. This article aims to provide a balanced summary of the evolving evidence regarding the contribution of p53 mutants in the biology of cancer while contemplating alternative frameworks to decipher the complexity of p53 mutants within their physiological contexts.
Collapse
Affiliation(s)
- Alessio Butera
- Chair of Systems Toxicology, University of Konstanz, Konstanz, Germany
| | - Ivano Amelio
- Chair of Systems Toxicology, University of Konstanz, Konstanz, Germany.
| |
Collapse
|
9
|
Cai WR, Sun XG, Yu Y, Wang X, Cao XC, Liu XF. Unveiling the prognostic value of ARID3A in breast cancer through bioinformatic analysis. Heliyon 2025; 11:e42024. [PMID: 40028521 PMCID: PMC11868939 DOI: 10.1016/j.heliyon.2025.e42024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/19/2024] [Accepted: 01/15/2025] [Indexed: 03/05/2025] Open
Abstract
Objective Identifying reliable prognostic markers for breast cancer is crucial for improving survival rates and reducing mortality. Recent studies highlight the AT-rich interactive domain-containing protein (ARID) family, particularly ARID3A, as influential in cancer progression, though its specific role in breast cancer remains unclear. This study investigates ARID3A's expression, prognostic relevance, clinicopathological correlations, co-expression profiles, and protein-protein interactions in breast cancer. Methods ARID3A mRNA and protein expression levels were analyzed using UALCAN, GEPIA databases, and immunohistochemistry from our hospital samples. Clinical prognostic parameters and survival data were examined through bioinformatics tools, including GEPIA, Bc-GenExMiner, and BEST. Subtype-specific expression and co-expression, particularly with REXO1, were evaluated using LinkedOmics, TIMER, and bc-GenExMiner. Functional enrichment analysis was conducted via LinkedOmics. Protein-protein interactions (PPI) were established using GeneMANIA and STRING, with validation through molecular docking using Cluspro. Results Elevated ARID3A expression was associated with poor prognosis in breast cancer, particularly in Luminal and HER2-positive subtypes. A positive correlation with REXO1 was identified, and enrichment analysis emphasized ARID3A's involvement in immune-related pathways, such as "interferon gamma production" and "primary immunodeficiency." PPI network and docking studies identified TP53 as a potential binding partner, suggesting a novel interaction influencing tumor progression. Conclusion These findings indicate that ARID3A may serve as a prognostic biomarker and therapeutic target in breast cancer, providing insights into its involvement in oncogenic pathways and interactions, particularly with TP53, that may drive cancer development and progression.
Collapse
Affiliation(s)
- Wen-Run Cai
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Xu-Gang Sun
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yue Yu
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Xin Wang
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Xu-Chen Cao
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Xiao-Feng Liu
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| |
Collapse
|
10
|
Colegrove HL, Monnat RJ, Feder AF. Epithelial competition determines gene therapy potential to suppress Fanconi Anemia oral cancer risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.26.640284. [PMID: 40060430 PMCID: PMC11888451 DOI: 10.1101/2025.02.26.640284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Fanconi Anemia (FA) is a heritable syndrome characterized by DNA damage repair deficits, frequent malformations and a significantly elevated risk of bone marrow failure, leukemia, and mucosal head and neck squamous cell carcinomas (HNSCC). Hematopoietic stem cell gene therapy can prevent marrow failure and lower leukemia risk, but mucosal gene therapy to lower HNSCC risk remains untested. Major knowledge gaps include an incomplete understanding of how rapidly gene-corrected cellular lineages could spread through the oral epithelium, and which delivery parameters are critical for ensuring efficient gene correction. To answer these questions, we extended an agent-based model of the oral epithelium to include the delivery of gene correction in situ to FA cells and the competitive dynamics between cellular lineages with and without gene correction. We found that only gene-corrected lineages with substantial proliferative advantages (probability of resisting displacement out of the basal layer ≥ 0.1) could spread on clinically relevant timelines, and that these lineages were initially at high risk of loss in the generations following correction. Delivering gene correction to many cells minimizes the risk of loss, while delivery to many distinct locations within a tissue maximizes the rate of spread. To determine the impact of mucosal gene therapy in preventing the clonal expansion of pre-cancerous mutations, we compared the expected burden of T P 53 mutations in simulated tissue sections with and without gene correction. We found that when FA cells have elevated genome instability or a T P 53 -dependent proliferative advantage, gene correction can substantially reduce the accumulation of pro-tumorigenic mutations. This model illustrates the power of computational frameworks to identify critical determinants of therapeutic success to enable experimental optimization and support novel and effective gene therapy applications.
Collapse
Affiliation(s)
| | - Raymond J Monnat
- Department of Genome Sciences, University of Washington, Seattle, WA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
- Department of Bioengineering, University of Washington, Seattle, WA
| | - Alison F Feder
- Department of Genome Sciences, University of Washington, Seattle, WA
- Herbold Computational Biology Program, Fred Hutch Cancer Center, Seattle, WA
| |
Collapse
|
11
|
Joerger AC, Stiewe T, Soussi T. TP53: the unluckiest of genes? Cell Death Differ 2025; 32:219-224. [PMID: 39443700 PMCID: PMC11803090 DOI: 10.1038/s41418-024-01391-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/22/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
The transcription factor p53 plays a key role in the cellular defense against cancer development. It is inactivated in virtually every tumor, and in every second tumor this inactivation is due to a mutation in the TP53 gene. In this perspective, we show that this diverse mutational spectrum is unique among all other cancer-associated proteins and discuss what drives the selection of TP53 mutations in cancer. We highlight that several factors conspire to make the p53 protein particularly vulnerable to inactivation by the mutations that constantly plague our genome. It appears that the TP53 gene has emerged as a victim of its own evolutionary past that shaped its structure and function towards a pluripotent tumor suppressor, but came with an increased structural fragility of its DNA-binding domain. TP53 loss of function - with associated dominant-negative effects - is the main mechanism that will impair TP53 tumor suppressive function, regardless of whether a neomorphic phenotype is associated with some of these variants.
Collapse
Affiliation(s)
- Andreas C Joerger
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany.
- Structural Genomics Consortium (SGC), Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany.
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps University, Marburg, Germany.
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany.
| | - Thierry Soussi
- Equipe « Hematopoietic and Leukemic Development », Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, SIRIC CURAMUS, Paris, France.
- Dept. of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Clinical Genetics, Uppsala University Hospital, Uppsala, Sweden.
| |
Collapse
|
12
|
Fortuno C, Llinares-Burguet I, Canson DM, de la Hoya M, Bueno-Martínez E, Sanoguera-Miralles L, Caldes S, James PA, Velasco-Sampedro EA, Spurdle AB. Exploring the role of splicing in TP53 variant pathogenicity through predictions and minigene assays. Hum Genomics 2025; 19:2. [PMID: 39780207 PMCID: PMC11715486 DOI: 10.1186/s40246-024-00714-5] [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: 11/15/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND TP53 variant classification benefits from the availability of large-scale functional data for missense variants generated using cDNA-based assays. However, absence of comprehensive splicing assay data for TP53 confounds the classification of the subset of predicted missense and synonymous variants that are also predicted to alter splicing. Our study aimed to generate and apply splicing assay data for a prioritised group of 59 TP53 predicted missense or synonymous variants that are also predicted to affect splicing by either SpliceAI or MaxEntScan. METHODS We conducted splicing analyses using a minigene construct containing TP53 exons 2 to 9 transfected into human breast cancer SKBR3 cells, and compared results against different splice prediction methods, including correlation with the SpliceAI-10k calculator. We additionally applied the splicing results for TP53 variant classification using an approach consistent with the ClinGen Sequence Variant Interpretation Splicing Subgroup recommendations. RESULTS Aberrant transcript profile consistent with loss of function, and for which a PVS1 (RNA) code would be assigned, was observed for 42 (71%) of prioritised variants, of which aberrant transcript expression was over 50% for 26 variants, and over 80% for 15 variants. Data supported the use of SpliceAI ≥ 0.2 cutoff for predicted splicing impact of TP53 variants. Prediction of aberration types using SpliceAI-10k calculator generally aligned with the corresponding assay results, though maximum SpliceAI score did not accurately predict level of aberrant expression. Application of the observed splicing results was used to reclassify 27/59 (46%) test variants as (likely) pathogenic or (likely) benign. CONCLUSIONS In conclusion, this study enhances the integration of splicing predictions and provides splicing assay data for exonic variants to support TP53 germline classification.
Collapse
Affiliation(s)
- Cristina Fortuno
- Population Health Program, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Inés Llinares-Burguet
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Consejo Superior de Investigaciones Científicas - Universidad de Valladolid (CSIC-UVa), 47003, Valladolid, Spain
| | - Daffodil M Canson
- Population Health Program, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Miguel de la Hoya
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), Madrid, Spain
| | - Elena Bueno-Martínez
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Consejo Superior de Investigaciones Científicas - Universidad de Valladolid (CSIC-UVa), 47003, Valladolid, Spain
| | - Lara Sanoguera-Miralles
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Consejo Superior de Investigaciones Científicas - Universidad de Valladolid (CSIC-UVa), 47003, Valladolid, Spain
| | - Sonsoles Caldes
- Population Health Program, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Paul A James
- Parkville Familial Cancer Centre, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Eladio A Velasco-Sampedro
- Splicing and Genetic Susceptibility to Cancer, Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), Consejo Superior de Investigaciones Científicas - Universidad de Valladolid (CSIC-UVa), 47003, Valladolid, Spain
| | - Amanda B Spurdle
- Population Health Program, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia.
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4006, Australia.
| |
Collapse
|
13
|
Liu Y, Elmas A, Huang KL. Mutation impact on mRNA versus protein expression across human cancers. Gigascience 2025; 14:giae113. [PMID: 39775839 PMCID: PMC11702362 DOI: 10.1093/gigascience/giae113] [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: 05/17/2024] [Revised: 09/13/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Cancer mutations are often assumed to alter proteins, thus promoting tumorigenesis. However, how mutations affect protein expression-in addition to gene expression-has rarely been systematically investigated. This is significant as mRNA and protein levels frequently show only moderate correlation, driven by factors such as translation efficiency and protein degradation. Proteogenomic datasets from large tumor cohorts provide an opportunity to systematically analyze the effects of somatic mutations on mRNA and protein abundance and identify mutations with distinct impacts on these molecular levels. RESULTS We conduct a comprehensive analysis of mutation impacts on mRNA- and protein-level expressions of 953 cancer cases with paired genomics and global proteomic profiling across 6 cancer types. Protein-level impacts are validated for 47.2% of the somatic expression quantitative trait loci (seQTLs), including CDH1 and MSH3 truncations, as well as other mutations from likely "long-tail" driver genes. Devising a statistical pipeline for identifying somatic protein-specific QTLs (spsQTLs), we reveal several gene mutations, including NF1 and MAP2K4 truncations and TP53 missenses showing disproportional influence on protein abundance not readily explained by transcriptomics. Cross-validating with data from massively parallel assays of variant effects (MAVE), TP53 missenses associated with high tumor TP53 proteins are more likely to be experimentally confirmed as functional. CONCLUSION This study reveals that somatic mutations can exhibit distinct impacts on mRNA and protein levels, underscoring the necessity of integrating proteogenomic data to comprehensively identify functionally significant cancer mutations. These insights provide a framework for prioritizing mutations for further functional validation and therapeutic targeting.
Collapse
Affiliation(s)
- Yuqi Liu
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Abdulkadir Elmas
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kuan-lin Huang
- Department of Genetics and Genomic Sciences, Department of Artificial Intelligence and Human Health, Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
14
|
Müntnich LJ, Dutzmann CM, Großhennig A, Härter V, Keymling M, Mastronuzzi A, Montellier E, Nees J, Palmaers NE, Penkert J, Pfister SM, Ripperger T, Schott S, Silchmüller F, Hainaut P, Kratz CP. Cancer risk in carriers of TP53 germline variants grouped into different functional categories. JNCI Cancer Spectr 2025; 9:pkaf008. [PMID: 39873732 PMCID: PMC11879075 DOI: 10.1093/jncics/pkaf008] [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: 11/07/2024] [Revised: 01/03/2025] [Accepted: 01/17/2025] [Indexed: 01/30/2025] Open
Abstract
Li-Fraumeni syndrome is a cancer predisposition syndrome caused by pathogenic TP53 germline variants; it is associated with a high lifelong cancer risk. We analyzed the German Li-Fraumeni syndrome registry, which contains data on 304 individuals. Cancer phenotypes were correlated with variants grouped according to their ability to transactivate target genes in a yeast assay using a traditional (nonfunctional, partially functional) and a novel (clusters A, B, and C) classification of variants into different groups. Partially functional and cluster B or C variants were enriched in patients who did not meet clinical testing criteria. Time to first malignancy was longer in carriers of partially functional variants (hazard ratio = 0.38, 95% CI = 0.22 to 0.66). Variants grouped within cluster B (hazard ratio = 0.45, 95% CI = 0.28 to 0.71) or C (hazard ratio = 0.34, 95% CI = 0.19 to 0.62) were associated with later cancer onset than NULL variants. These findings can be used to risk-stratify patients and inform care.
Collapse
Affiliation(s)
- Lucas John Müntnich
- Pediatric Hematology and Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Christina M Dutzmann
- Pediatric Hematology and Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Anika Großhennig
- Institute of Biostatistics, Hannover Medical School, 30625 Hannover, Germany
| | - Valentina Härter
- Pediatric Hematology and Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Myriam Keymling
- Division of Radiology, German Cancer Research Center, 30625 Heidelberg, Germany
| | - Angela Mastronuzzi
- Department of Paediatric Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy
| | - Emilie Montellier
- Université Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, 38700 Grenoble, France
| | - Juliane Nees
- Department of Obstetrics and Gynecology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Natalie E Palmaers
- Pediatric Hematology and Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Judith Penkert
- Pediatric Hematology and Oncology, Hannover Medical School, 30625 Hannover, Germany
- Department of Human Genetics, Hannover Medical School, 30625 Hannover, Germany
| | - Stefan M Pfister
- Hopp Children’s Cancer Center Heidelberg and Division of Pediatric Neurooncology, German Cancer Research Center, and German Cancer Consortium, Department of Pediatric Hematology and Oncology, Heidelberg University Hospital and National Center for Tumor Diseases, 69120 Heidelberg, Germany
| | - Tim Ripperger
- Department of Human Genetics, Hannover Medical School, 30625 Hannover, Germany
| | - Sarah Schott
- Department of Obstetrics and Gynecology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Farina Silchmüller
- Pediatric Hematology and Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Pierre Hainaut
- Université Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, 38700 Grenoble, France
| | - Christian P Kratz
- Pediatric Hematology and Oncology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
15
|
Funk JS, Klimovich M, Drangenstein D, Pielhoop O, Hunold P, Borowek A, Noeparast M, Pavlakis E, Neumann M, Balourdas DI, Kochhan K, Merle N, Bullwinkel I, Wanzel M, Elmshäuser S, Teply-Szymanski J, Nist A, Procida T, Bartkuhn M, Humpert K, Mernberger M, Savai R, Soussi T, Joerger AC, Stiewe T. Deep CRISPR mutagenesis characterizes the functional diversity of TP53 mutations. Nat Genet 2025; 57:140-153. [PMID: 39774325 PMCID: PMC11735402 DOI: 10.1038/s41588-024-02039-4] [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: 03/17/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025]
Abstract
The mutational landscape of TP53, a tumor suppressor mutated in about half of all cancers, includes over 2,000 known missense mutations. To fully leverage TP53 mutation status for personalized medicine, a thorough understanding of the functional diversity of these mutations is essential. We conducted a deep mutational scan using saturation genome editing with CRISPR-mediated homology-directed repair to engineer 9,225 TP53 variants in cancer cells. This high-resolution approach, covering 94.5% of all cancer-associated TP53 missense mutations, precisely mapped the impact of individual mutations on tumor cell fitness, surpassing previous deep mutational scan studies in distinguishing benign from pathogenic variants. Our results revealed even subtle loss-of-function phenotypes and identified promising mutants for pharmacological reactivation. Moreover, we uncovered the roles of splicing alterations and nonsense-mediated messenger RNA decay in mutation-driven TP53 dysfunction. These findings underscore the power of saturation genome editing in advancing clinical TP53 variant interpretation for genetic counseling and personalized cancer therapy.
Collapse
Affiliation(s)
- Julianne S Funk
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Maria Klimovich
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | | | - Ole Pielhoop
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Pascal Hunold
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Anna Borowek
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Maxim Noeparast
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | | | - Michelle Neumann
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Dimitrios-Ilias Balourdas
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences and Structural Genomics Consortium (SGC), Frankfurt am Main, Germany
| | - Katharina Kochhan
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Nastasja Merle
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Imke Bullwinkel
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Michael Wanzel
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | | | - Julia Teply-Szymanski
- Institute of Pathology, Philipps-University, Marburg University Hospital, Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps-University, Marburg, Germany
| | - Tara Procida
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Marek Bartkuhn
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Biomedical Informatics and Systems Medicine, Justus-Liebig-University, Giessen, Germany
| | - Katharina Humpert
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
- Bioinformatics Core Facility, Philipps-University, Marburg, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany
| | - Rajkumar Savai
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany
- Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Lung Microenvironmental Niche in Cancerogenesis, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thierry Soussi
- Centre de Recherche Saint-Antoine UMRS_938, INSERM, Sorbonne Université, Paris, France
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Clinical Genetics, Uppsala University Hospital, Uppsala, Sweden
| | - Andreas C Joerger
- Institute of Pharmaceutical Chemistry, Goethe University, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences and Structural Genomics Consortium (SGC), Frankfurt am Main, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, Marburg, Germany.
- Genomics Core Facility, Philipps-University, Marburg, Germany.
- Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany.
- Bioinformatics Core Facility, Philipps-University, Marburg, Germany.
- Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany.
| |
Collapse
|
16
|
Cirincione A, Simpson D, Yan W, McNulty R, Ravisankar P, Solley SC, Yan J, Lim F, Farley EK, Singh M, Adamson B. A benchmarked, high-efficiency prime editing platform for multiplexed dropout screening. Nat Methods 2025; 22:92-101. [PMID: 39562753 PMCID: PMC11725502 DOI: 10.1038/s41592-024-02502-4] [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: 03/12/2024] [Accepted: 10/11/2024] [Indexed: 11/21/2024]
Abstract
Prime editing installs precise edits into the genome with minimal unwanted byproducts, but low and variable editing efficiencies have complicated application of the approach to high-throughput functional genomics. Here we assembled a prime editing platform capable of high-efficiency substitution editing suitable for functional interrogation of small genetic variants. We benchmarked this platform for pooled, loss-of-function screening using a library of ~240,000 engineered prime editing guide RNAs (epegRNAs) targeting ~17,000 codons with 1-3 bp substitutions. Comparing the abundance of these epegRNAs across screen samples identified negative selection phenotypes for 7,996 nonsense mutations targeted to 1,149 essential genes and for synonymous mutations that disrupted splice site motifs at 3' exon boundaries. Rigorous evaluation of codon-matched controls demonstrated that these phenotypes were highly specific to the intended edit. Altogether, we established a prime editing approach for multiplexed, functional characterization of genetic variants with simple readouts.
Collapse
Affiliation(s)
- Ann Cirincione
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Danny Simpson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Weihao Yan
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Ryan McNulty
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Purnima Ravisankar
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Sabrina C Solley
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Jun Yan
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Fabian Lim
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA, USA
- Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Emma K Farley
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Mona Singh
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Britt Adamson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
17
|
Pavlova S, Malcikova J, Radova L, Bonfiglio S, Cowland JB, Brieghel C, Andersen MK, Karypidou M, Biderman B, Doubek M, Lazarian G, Rapado I, Vynck M, Porret NA, Andres M, Rosenberg D, Sahar D, Martínez‐Laperche C, Buño I, Hindley A, Donaldson D, Sánchez JB, García‐Marco JA, Serrano‐Alcalá A, Ferrer‐Lores B, Fernández‐Rodriguez C, Bellosillo B, Stilgenbauer S, Tausch E, Nikdin H, Quinn F, Atkinson E, van de Corput L, Yildiz C, Bilbao‐Sieyro C, Florido Y, Thiede C, Schuster C, Stoj A, Czekalska S, Chatzidimitriou A, Laidou S, Bidet A, Dussiau C, Nollet F, Piras G, Monne M, Smirnova S, Nikitin E, Sloma I, Claudel A, Largeaud L, Ysebaert L, Valk PJM, Christian A, Walewska R, Oscier D, Sebastião M, da Silva MG, Galieni P, Angelini M, Rossi D, Spina V, Matos S, Martins V, Stokłosa T, Pepek M, Baliakas P, Andreu R, Luna I, Kahre T, Murumets Ü, Pikousova T, Kurucova T, Laird S, Ward D, Alcoceba M, Balanzategui A, Scarfo L, Gandini F, Zapparoli E, Blanco A, Abrisqueta P, Rodríguez‐Vicente AE, Benito R, Bravetti C, Davi F, Gameiro P, Martinez‐Lopez J, Tazón‐Vega B, Baran‐Marszak F, Davis Z, Catherwood M, Sudarikov A, Rosenquist R, Niemann CU, Stamatopoulos K, Ghia P, Pospisilova S. Detection of clinically relevant variants in the TP53 gene below 10% allelic frequency: A multicenter study by ERIC, the European Research Initiative on CLL. Hemasphere 2025; 9:e70065. [PMID: 39840379 PMCID: PMC11746920 DOI: 10.1002/hem3.70065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/04/2024] [Accepted: 11/13/2024] [Indexed: 01/23/2025] Open
Abstract
In chronic lymphocytic leukemia, the reliability of next-generation sequencing (NGS) to detect TP53 variants ≤10% allelic frequency (low-VAF) is debated. We tested the ability to detect 23 such variants in 41 different laboratories using their NGS method of choice. The sensitivity was 85.6%, 94.5%, and 94.8% at 1%, 2%, and 3% VAF cut-off, respectively. While only one false positive (FP) result was reported at >2% VAF, it was more challenging to distinguish true variants <2% VAF from background noise (37 FPs reported by 9 laboratories). The impact of low-VAF variants on time-to-second-treatment (TTST) and overall survival (OS) was investigated in a series of 1092 patients. Among patients not treated with targeted agents, patients with low-VAF TP53 variants had shorter TTST and OS versus wt-TP53 patients, and the relative risk of second-line treatment or death increased continuously with increasing VAF. Targeted therapy in ≥2 line diminished the difference in OS between patients with low-VAF TP53 variants and wt-TP53 patients, while patients with high-VAF TP53 variants had inferior OS compared to wild type-TP53 cases. Altogether, NGS-based approaches are technically capable of detecting low-VAF variants. No strict threshold can be suggested from a technical standpoint, laboratories reporting TP53 mutations should participate in a standardized validation set-up. Finally, whereas low-VAF variants affected outcomes in patients receiving chemoimmunotherapy, their impact on those treated with novel therapies remains undetermined. Our results pave the way for the harmonized and accurate TP53 assessment, which is indispensable for elucidating the role of TP53 mutations in targeted treatment.
Collapse
Affiliation(s)
- Sarka Pavlova
- Department of Internal Medicine, Hematology and Oncology, and Institute of Medical Genetics and Genomics, University Hospital Brno and Medical FacultyMasaryk UniversityBrnoCzech Republic
- Centre for Molecular Medicine, Central European Institute of Technology (CEITEC)Masaryk UniversityBrnoCzech Republic
| | - Jitka Malcikova
- Department of Internal Medicine, Hematology and Oncology, and Institute of Medical Genetics and Genomics, University Hospital Brno and Medical FacultyMasaryk UniversityBrnoCzech Republic
- Centre for Molecular Medicine, Central European Institute of Technology (CEITEC)Masaryk UniversityBrnoCzech Republic
| | - Lenka Radova
- Centre for Molecular Medicine, Central European Institute of Technology (CEITEC)Masaryk UniversityBrnoCzech Republic
| | - Silvia Bonfiglio
- B‐Cell Neoplasia Unit and Strategic Research Program on CLLIRCCS Ospedale San RaffaeleMilanItaly
- Center for Omics SciencesIRCCS Ospedale San RaffaeleMilanItaly
| | - Jack B. Cowland
- Department of Clinical Genetics, Centre of Diagnostic InvestigationsCopenhagen University Hospital ‐ RigshospitaletCopenhagenDenmark
| | - Christian Brieghel
- Department of HematologyCopenhagen University Hospital ‐ RigshospitaletCopenhagenDenmark
| | - Mette K. Andersen
- Department of Clinical Genetics, Centre of Diagnostic InvestigationsCopenhagen University Hospital ‐ RigshospitaletCopenhagenDenmark
| | - Maria Karypidou
- Institute of Applied BiosciencesCentre for Research and Technology HellasThessalonikiGreece
| | - Bella Biderman
- National Medical Research Center for HematologyMoscowRussia
| | - Michael Doubek
- Department of Internal Medicine, Hematology and Oncology, and Institute of Medical Genetics and Genomics, University Hospital Brno and Medical FacultyMasaryk UniversityBrnoCzech Republic
- Centre for Molecular Medicine, Central European Institute of Technology (CEITEC)Masaryk UniversityBrnoCzech Republic
| | - Gregory Lazarian
- Hematology laboratoryHUPSSD, Hôpital Avicenne, APHPBobignyFrance
- INSERM U978Université Sorbonne Paris NordBobignyFrance
| | - Inmaculada Rapado
- Department of HematologyHospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Complutense University, CNIO, CIBERONCMadridSpain
| | - Matthijs Vynck
- Department of Laboratory MedicineAZ Sint‐Jan HospitalBrugesBelgium
| | - Naomi A. Porret
- Department of Hematology and Central Hematology Laboratory, InselspitalBern University Hospital, University of BernBernSwitzerland
| | - Martin Andres
- Department of Hematology and Central Hematology Laboratory, InselspitalBern University Hospital, University of BernBernSwitzerland
| | | | - Dvora Sahar
- Hematology LaboratoryRambam Medical CenterHaifaIsrael
| | - Carolina Martínez‐Laperche
- Department of HematologyGregorio Marañón General University Hospital, Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
| | - Ismael Buño
- Department of HematologyGregorio Marañón General University Hospital, Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Genomics UnitGregorio Marañón General University Hospital, Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Department of Cell Biology, Medical SchoolComplutense University of MadridMadridSpain
| | | | | | - Julio B. Sánchez
- Molecular Cytogenetics Unit, Hematology DepartmentHospital Universitario Puerta de Hierro‐MajadahondaMadridSpain
| | - José A. García‐Marco
- Molecular Cytogenetics Unit, Hematology DepartmentHospital Universitario Puerta de Hierro‐MajadahondaMadridSpain
| | | | | | | | - Beatriz Bellosillo
- Pathology DepartmentHospital del Mar, IMIM (Hospital del Mar Medical Research Institute)BarcelonaSpain
| | | | - Eugen Tausch
- Department of Internal Medicine IIIUlm UniversityUlmGermany
| | - Hero Nikdin
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
- Department of Clinical Genetics and GenomicsKarolinska University HospitalStockholmSweden
| | - Fiona Quinn
- Cancer Molecular Diagnostics DepartmentCentre for Laboratory Medicine and Molecular Pathology, St. James HospitalDublinIreland
| | - Emer Atkinson
- Cancer Molecular Diagnostics DepartmentCentre for Laboratory Medicine and Molecular Pathology, St. James HospitalDublinIreland
| | - Lisette van de Corput
- Central Diagnostic LaboratoryUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Cafer Yildiz
- Central Diagnostic LaboratoryUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Cristina Bilbao‐Sieyro
- Servicio de HematologíaHospital Universitario de Gran Canaria Dr. Negrín, Departamento de Morfología de La Universidad de Las Palmas de Gran CanariaGran CanariaSpain
| | - Yanira Florido
- Servicio de HematologíaHospital Universitario de Gran Canaria Dr. Negrín, Departamento de Morfología de La Universidad de Las Palmas de Gran CanariaGran CanariaSpain
| | | | | | - Anastazja Stoj
- Department of Hematological Diagnostics and GeneticsUniversity Hospital in KrakowKrakowPoland
| | - Sylwia Czekalska
- Department of Hematological Diagnostics and GeneticsUniversity Hospital in KrakowKrakowPoland
| | | | - Stamatia Laidou
- Institute of Applied BiosciencesCentre for Research and Technology HellasThessalonikiGreece
| | - Audrey Bidet
- Laboratoire d'Hématologie BiologiqueCHU BordeauxBordeauxFrance
| | - Charles Dussiau
- Laboratoire d'Hématologie BiologiqueCHU BordeauxBordeauxFrance
| | - Friedel Nollet
- Department of Laboratory MedicineAZ Sint‐Jan HospitalBrugesBelgium
| | - Giovanna Piras
- Laboratorio specialistico UOC ematologiaOspedale San FrancescoASL NuoroItaly
| | - Maria Monne
- Laboratorio specialistico UOC ematologiaOspedale San FrancescoASL NuoroItaly
| | | | - Eugene Nikitin
- Outpatient department of Hematology, Oncology and Chemotherapy, Botkin Hospital, and Department of Hematology and TransfusiologyRussian Medical Academy of Continuous Professional EducationMoscowRussia
| | - Ivan Sloma
- Univ Paris Est Creteil, INSERM, IMRBCreteilFrance
- Departement d'Hematologie et Immunologie BiologiqueAP‐HP, Hopital Henri MondorCreteilFrance
| | - Alexis Claudel
- Univ Paris Est Creteil, INSERM, IMRBCreteilFrance
- Departement d'Hematologie et Immunologie BiologiqueAP‐HP, Hopital Henri MondorCreteilFrance
| | | | - Loïc Ysebaert
- Institut Universitaire de Cancérologie de ToulouseToulouseFrance
| | - Peter J. M. Valk
- Department of Hematology, Erasmus MC Cancer InstituteUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Amy Christian
- Molecular PathologyUniversity Hospitals DorsetBournemouthUK
| | | | - David Oscier
- Molecular PathologyUniversity Hospitals DorsetBournemouthUK
| | - Marta Sebastião
- Laboratório Hemato‐OncologiaInstituto Português de Oncologia de LisboaLisbonPortugal
| | | | - Piero Galieni
- UOC HematologyMazzoni Hospital‐Ascoli PicenoAscoli PicenoItaly
| | - Mario Angelini
- UOC HematologyMazzoni Hospital‐Ascoli PicenoAscoli PicenoItaly
| | - Davide Rossi
- Institute of Oncology Research and OncologyInstitute of Southern SwitzerlandBellinzonaSwitzerland
| | - Valeria Spina
- Laboratorio di Diagnostica Molecolare, Servizio di Genetica Medica EOLABEnte Ospedaliero CantonaleBellinzonaSwitzerland
| | - Sónia Matos
- Genomed‐Diagnósticos de Medicina MoleculariMM ‐ Instituto de Medicina Molecular, Faculdade de MedicinaLisboaPortugal
| | - Vânia Martins
- Genomed‐Diagnósticos de Medicina MoleculariMM ‐ Instituto de Medicina Molecular, Faculdade de MedicinaLisboaPortugal
| | - Tomasz Stokłosa
- Department of Tumor Biology and GeneticsMedical University of WarsawWarsawPoland
| | - Monika Pepek
- Department of Tumor Biology and GeneticsMedical University of WarsawWarsawPoland
| | - Panagiotis Baliakas
- Department of Immunology, Genetics and PathologyUppsala UniversityUppsalaSweden
| | - Rafa Andreu
- Hematology DepartmentHospital Universitari i Politècnic la FeValenciaSpain
| | - Irene Luna
- Hematology DepartmentHospital Universitari i Politècnic la FeValenciaSpain
| | - Tiina Kahre
- Department of Laboratory Genetics, Genetics and Personalized ClinicTartu University HospitalTartuEstonia
- Genetics and Personalized Medicine Clinic, Institute of Clinical MedicineTartu UniversityTartuEstonia
| | - Ülle Murumets
- Department of Laboratory Genetics, Genetics and Personalized ClinicTartu University HospitalTartuEstonia
| | - Tereza Pikousova
- Centre for Molecular Medicine, Central European Institute of Technology (CEITEC)Masaryk UniversityBrnoCzech Republic
| | - Terezia Kurucova
- Centre for Molecular Medicine, Central European Institute of Technology (CEITEC)Masaryk UniversityBrnoCzech Republic
| | - Sophie Laird
- Wessex Genomics Laboratory ServiceSalisbury NHS Foundation TrustSalisburyUK
| | - Daniel Ward
- Wessex Genomics Laboratory ServiceSalisbury NHS Foundation TrustSalisburyUK
| | - Miguel Alcoceba
- Department of HematologyUniversity Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Center of Salamanca‐IBMCC (USAL‐CSIC)SalamancaSpain
| | - Ana Balanzategui
- Department of HematologyUniversity Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Center of Salamanca‐IBMCC (USAL‐CSIC)SalamancaSpain
| | - Lydia Scarfo
- B‐Cell Neoplasia Unit and Strategic Research Program on CLLIRCCS Ospedale San RaffaeleMilanItaly
- Medical SchoolUniversità Vita‐Salute San RaffaeleMilanItaly
| | - Francesca Gandini
- B‐Cell Neoplasia Unit and Strategic Research Program on CLLIRCCS Ospedale San RaffaeleMilanItaly
- Medical SchoolUniversità Vita‐Salute San RaffaeleMilanItaly
| | | | - Adoración Blanco
- Department of HematologyHospital Universitari Vall d'Hebron (HUVH)BarcelonaSpain
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO)BarcelonaSpain
- Department of MedicineUniversitat Autònoma de Barcelona (UAB)BarcelonaSpain
| | - Pau Abrisqueta
- Department of HematologyHospital Universitari Vall d'Hebron (HUVH)BarcelonaSpain
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO)BarcelonaSpain
- Department of MedicineUniversitat Autònoma de Barcelona (UAB)BarcelonaSpain
| | - Ana E. Rodríguez‐Vicente
- Oncohematology Research Group, Institute of Biomedical Research of Salamanca (IBSAL)Cancer Research Centre (IBMCC, USAL‐CSIC) and University of SalamancaSalamancaSpain
- Department of Human Anatomy and Histology, Faculty of MedicineUniversity of SalamancaSalamancaSpain
| | - Rocío Benito
- Oncohematology Research Group, Institute of Biomedical Research of Salamanca (IBSAL)Cancer Research Centre (IBMCC, USAL‐CSIC) and University of SalamancaSalamancaSpain
| | - Clotilde Bravetti
- Department of Biological HematologySorbonne Université, AP‐HP, Pitié‐Salpêtrière HospitalParisFrance
| | - Frédéric Davi
- Department of Biological HematologySorbonne Université, AP‐HP, Pitié‐Salpêtrière HospitalParisFrance
| | - Paula Gameiro
- Laboratório Hemato‐OncologiaInstituto Português de Oncologia de LisboaLisbonPortugal
| | - Joaquin Martinez‐Lopez
- Department of HematologyHospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Complutense University, CNIO, CIBERONCMadridSpain
| | - Bárbara Tazón‐Vega
- Department of HematologyHospital Universitari Vall d'Hebron (HUVH)BarcelonaSpain
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO)BarcelonaSpain
- Department of MedicineUniversitat Autònoma de Barcelona (UAB)BarcelonaSpain
| | - Fanny Baran‐Marszak
- Hematology laboratoryHUPSSD, Hôpital Avicenne, APHPBobignyFrance
- INSERM U978Université Sorbonne Paris NordBobignyFrance
| | - Zadie Davis
- Molecular PathologyUniversity Hospitals DorsetBournemouthUK
| | | | | | - Richard Rosenquist
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
- Department of Clinical Genetics and GenomicsKarolinska University HospitalStockholmSweden
| | - Carsten U. Niemann
- Department of HematologyCopenhagen University Hospital ‐ RigshospitaletCopenhagenDenmark
| | - Kostas Stamatopoulos
- Institute of Applied BiosciencesCentre for Research and Technology HellasThessalonikiGreece
| | - Paolo Ghia
- B‐Cell Neoplasia Unit and Strategic Research Program on CLLIRCCS Ospedale San RaffaeleMilanItaly
- Medical SchoolUniversità Vita‐Salute San RaffaeleMilanItaly
| | - Sarka Pospisilova
- Department of Internal Medicine, Hematology and Oncology, and Institute of Medical Genetics and Genomics, University Hospital Brno and Medical FacultyMasaryk UniversityBrnoCzech Republic
- Centre for Molecular Medicine, Central European Institute of Technology (CEITEC)Masaryk UniversityBrnoCzech Republic
| |
Collapse
|
18
|
Montellier E, Lemonnier N, Penkert J, Freycon C, Blanchet S, Amadou A, Chuffart F, Fischer NW, Achatz MI, Levine AJ, Goudie C, Malkin D, Bougeard G, Kratz CP, Hainaut P. Clustering of TP53 variants into functional classes correlates with cancer risk and identifies different phenotypes of Li-Fraumeni syndrome. iScience 2024; 27:111296. [PMID: 39634561 PMCID: PMC11615613 DOI: 10.1016/j.isci.2024.111296] [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: 06/11/2024] [Revised: 09/05/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024] Open
Abstract
Li-Fraumeni syndrome (LFS) is a heterogeneous predisposition to an individually variable spectrum of cancers caused by pathogenic TP53 germline variants. We used a clustering method to assign TP53 missense variants to classes based on their functional activities in experimental assays assessing biological p53 functions. Correlations with LFS phenotypes were analyzed using the public germline TP53 mutation database and validated in three LFS clinical cohorts. Class A carriers recapitulated all phenotypic traits of fully penetrant LFS, whereas class B carriers showed a slightly less penetrant form dominated by specific cancers, consistent with the notion that these classes identify variants with distinct functional properties. Class C displayed a lower lifetime cancer risk associated with attenuated LFS features, consistent with the notion that these variants have hypomorphic features. Class D carriers showed low lifetime cancer risks inconsistent with LFS definitions. This classification of TP53 variants provides insights into structural/functional features causing pathogenicity.
Collapse
Affiliation(s)
- Emilie Montellier
- University Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, F38000 Grenoble, France
| | - Nathanaël Lemonnier
- University Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, F38000 Grenoble, France
| | - Judith Penkert
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Claire Freycon
- University Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, F38000 Grenoble, France
- Department of Pediatric Hematology-Oncology, Grenoble Alpes University Hospital, Grenoble, France
- Department of Pediatrics, Division of Hematology-Oncology, Montreal Children’s Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Sandrine Blanchet
- University Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, F38000 Grenoble, France
| | - Amina Amadou
- University Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, F38000 Grenoble, France
- Department of Prevention Cancer Environment, Centre Léon Bérard, Lyon, France
| | - Florent Chuffart
- University Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, F38000 Grenoble, France
| | - Nicholas W. Fischer
- Genetics and Genome Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | - Arnold J. Levine
- Simons Center for Systems Biology, Institute for Advanced Study, Princeton, NJ, USA
| | - Catherine Goudie
- Department of Pediatrics, Division of Hematology-Oncology, Montreal Children’s Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - David Malkin
- Genetics and Genome Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Division of Hematology/Oncology, The Hospital for Sick Children, Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Gaëlle Bougeard
- University Rouen Normandie, Inserm U1245, Normandie University, CHU Rouen, Department of Genetics, F-76000 Rouen, France
| | - Christian P. Kratz
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Pierre Hainaut
- University Grenoble Alpes, Inserm 1209, CNRS 5309, Institute for Advanced Biosciences, F38000 Grenoble, France
| |
Collapse
|
19
|
Patil MR, Bihari A. Role of artificial intelligence in cancer detection using protein p53: A Review. Mol Biol Rep 2024; 52:46. [PMID: 39658610 DOI: 10.1007/s11033-024-10051-4] [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: 08/21/2024] [Accepted: 10/22/2024] [Indexed: 12/12/2024]
Abstract
Normal cell development and prevention of tumor formation rely on the tumor-suppressor protein p53. This crucial protein is produced from the Tp53 gene, which encodes the p53 protein. The p53 protein plays a vital role in regulating cell growth, DNA repair, and apoptosis (programmed cell death), thereby maintaining the integrity of the genome and preventing the formation of tumors. Since p53 was discovered 43 years ago, many researchers have clarified its functions in the development of tumors. With the support of the protein p53 and targeted artificial intelligence modeling, it will be possible to detect cancer and tumor activity at an early stage. This will open up new research opportunities. In this review article, a comprehensive analysis was conducted on different machine learning techniques utilized in conjunction with the protein p53 to predict and speculate cancer. The study examined the types of data incorporated and evaluated the performance of these techniques. The aim was to provide a thorough understanding of the effectiveness of machine learning in predicting and speculating cancer using the protein p53.
Collapse
Affiliation(s)
- Manisha R Patil
- School of Computer Science Engineering and Information System, Vellore Institute of Technology, Vellore, Tamil Nadu, India
| | - Anand Bihari
- Department of Computational Intelligence, School of Computer Science and Engineering, Vellore Institute of Technology, Vellore, Tamil Nadu, India.
| |
Collapse
|
20
|
Hwang SH, Baek SH, Lee MJ, Kook Y, Bae SJ, Ahn SG, Jeong J. Clinical Relevance of TP53 Mutation and Its Characteristics in Breast Cancer with Long-Term Follow-Up Date. Cancers (Basel) 2024; 16:3899. [PMID: 39682089 DOI: 10.3390/cancers16233899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND The TP53 mutation is one of the most frequently identified mutations in human cancers and is typically associated with a poor prognosis. However, there are conflicting findings regarding its impact. We aimed to clarify the clinical relevance of TP53 mutations across all breast cancer subtypes and treatments utilizing long-term follow-up data. METHODS We retrospectively identified the data of breast cancer patients who underwent TP53 mutation testing. Stratified log-rank tests and Cox regression analysis were performed to compare oncologic outcomes based on TP53 mutation status and the characteristics of these mutations, including types and locations. Mutations in exons 5-9 were identified using polymerase chain reaction-denaturing high-performance liquid chromatography (PCR-DHPLC) and direct sequencing. RESULTS Between January 2007 and December 2015, 650 breast cancer patients underwent TP53 mutation testing in Gangnam Severance Hospital. The TP53 mutations were identified in 172 patients (26.5%), with 34 (19.8%) exhibiting missense hotspot mutations. Patients with TP53 mutations (TP53-mutated group) had worse prognosis, demonstrated by a 10-year recurrence-free survival (RFS) rate of 83.5% compared to 86.6% in patients without mutations (HR, 1.67; p = 0.026) and a 10-year overall survival (OS) rate of 88.1% versus 91.0% (HR, 3.02; p = 0.003). However, subgroup analyses within the TP53-mutated group did not reveal significant differences in oncologic outcomes based on mutation types and locations. CONCLUSIONS Our findings establish that TP53 mutations are linked to poorer oncologic outcomes in breast cancer across all subtypes. Yet, within the TP53-mutated group, the specific characteristics of TP53 mutations do not influence oncologic outcomes.
Collapse
Affiliation(s)
- Seung Hyun Hwang
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Breast and Thyroid Surgery, Sam Hospital, Anyang 14030, Republic of Korea
| | - Seung Ho Baek
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Min Ji Lee
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Yoonwon Kook
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Soong June Bae
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Sung Gwe Ahn
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Joon Jeong
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| |
Collapse
|
21
|
Nguyen E, Poli M, Durrant MG, Kang B, Katrekar D, Li DB, Bartie LJ, Thomas AW, King SH, Brixi G, Sullivan J, Ng MY, Lewis A, Lou A, Ermon S, Baccus SA, Hernandez-Boussard T, Ré C, Hsu PD, Hie BL. Sequence modeling and design from molecular to genome scale with Evo. Science 2024; 386:eado9336. [PMID: 39541441 PMCID: PMC12057570 DOI: 10.1126/science.ado9336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024]
Abstract
The genome is a sequence that encodes the DNA, RNA, and proteins that orchestrate an organism's function. We present Evo, a long-context genomic foundation model with a frontier architecture trained on millions of prokaryotic and phage genomes, and report scaling laws on DNA to complement observations in language and vision. Evo generalizes across DNA, RNA, and proteins, enabling zero-shot function prediction competitive with domain-specific language models and the generation of functional CRISPR-Cas and transposon systems, representing the first examples of protein-RNA and protein-DNA codesign with a language model. Evo also learns how small mutations affect whole-organism fitness and generates megabase-scale sequences with plausible genomic architecture. These prediction and generation capabilities span molecular to genomic scales of complexity, advancing our understanding and control of biology.
Collapse
Affiliation(s)
- Eric Nguyen
- Arc Institute, Palo Alto, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Michael Poli
- Department of Computer Science, Stanford University, Stanford, CA, USA
- TogetherAI, San Francisco, CA, USA
| | | | - Brian Kang
- Arc Institute, Palo Alto, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - David B. Li
- Arc Institute, Palo Alto, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - Armin W. Thomas
- Stanford Data Science, Stanford University, Stanford, CA, USA
| | - Samuel H. King
- Arc Institute, Palo Alto, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Garyk Brixi
- Arc Institute, Palo Alto, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Madelena Y. Ng
- Stanford Center for Biomedical Informatics Research, Stanford, CA, USA
| | - Ashley Lewis
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Aaron Lou
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Stefano Ermon
- Department of Computer Science, Stanford University, Stanford, CA, USA
- CZ Biohub, San Francisco, CA, USA
| | | | | | - Christopher Ré
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Patrick D. Hsu
- Arc Institute, Palo Alto, CA, USA
- Department of Bioengineering and Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Brian L. Hie
- Arc Institute, Palo Alto, CA, USA
- Stanford Data Science, Stanford University, Stanford, CA, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
22
|
Rocca V, Lo Feudo E, Dinatolo F, Lavano SM, Bilotta A, Amato R, D’Antona L, Trapasso F, Baudi F, Colao E, Perrotti N, Paduano F, Iuliano R. Germline Variant Spectrum in Southern Italian High-Risk Hereditary Breast Cancer Patients: Insights from Multi-Gene Panel Testing. Curr Issues Mol Biol 2024; 46:13003-13020. [PMID: 39590369 PMCID: PMC11592649 DOI: 10.3390/cimb46110775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Hereditary breast cancer accounts for 5-10% of all cases, with pathogenic variants in BRCA1/2 and other susceptibility genes playing a crucial role. This study elucidates the prevalence and spectrum of germline variants in 13 cancer predisposition genes among high-risk hereditary breast cancer patients from Southern Italy. We employed next-generation sequencing (NGS) to analyze 254 individuals selected through genetic counseling. Pathogenic or likely pathogenic variants were identified in 13% (34/254) of patients, with 54% of these variants occurring in non-BRCA1/2 genes. Notably, we observed a recurrent BRCA1 c.4964_4982del founder mutation, underscoring the importance of population-specific genetic screening. The spectrum of variants extended beyond BRCA1/2 to include PALB2, ATM, TP53, CHEK2, and RAD51C, highlighting the genetic heterogeneity of breast cancer susceptibility. Variants of uncertain significance were detected in 20% of patients, emphasizing the ongoing challenge of variant interpretation in the era of multi-gene panel testing. These findings not only enhance our understanding of the genetic landscape of breast cancer in Southern Italy but also provide a foundation for developing more targeted, population-specific approaches to genetic testing and counseling, ultimately contributing to the advancement of precision medicine in oncology.
Collapse
Affiliation(s)
- Valentina Rocca
- Medical Genetics Unit, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy; (V.R.); (E.L.F.); (F.D.); (S.M.L.); (A.B.); (R.A.); (F.T.); (F.B.); (E.C.); (N.P.)
- Department of Clinical and Experimental Medicine, Campus S. Venuta, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
| | - Elisa Lo Feudo
- Medical Genetics Unit, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy; (V.R.); (E.L.F.); (F.D.); (S.M.L.); (A.B.); (R.A.); (F.T.); (F.B.); (E.C.); (N.P.)
- Department of Clinical and Experimental Medicine, Campus S. Venuta, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
| | - Francesca Dinatolo
- Medical Genetics Unit, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy; (V.R.); (E.L.F.); (F.D.); (S.M.L.); (A.B.); (R.A.); (F.T.); (F.B.); (E.C.); (N.P.)
| | - Serena Marianna Lavano
- Medical Genetics Unit, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy; (V.R.); (E.L.F.); (F.D.); (S.M.L.); (A.B.); (R.A.); (F.T.); (F.B.); (E.C.); (N.P.)
| | - Anna Bilotta
- Medical Genetics Unit, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy; (V.R.); (E.L.F.); (F.D.); (S.M.L.); (A.B.); (R.A.); (F.T.); (F.B.); (E.C.); (N.P.)
| | - Rosario Amato
- Medical Genetics Unit, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy; (V.R.); (E.L.F.); (F.D.); (S.M.L.); (A.B.); (R.A.); (F.T.); (F.B.); (E.C.); (N.P.)
- Department of Health Sciences, Campus S. Venuta, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Lucia D’Antona
- Department of Health Sciences, Campus S. Venuta, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Francesco Trapasso
- Medical Genetics Unit, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy; (V.R.); (E.L.F.); (F.D.); (S.M.L.); (A.B.); (R.A.); (F.T.); (F.B.); (E.C.); (N.P.)
- Department of Clinical and Experimental Medicine, Campus S. Venuta, University Magna Græcia of Catanzaro, 88100 Catanzaro, Italy
| | - Francesco Baudi
- Medical Genetics Unit, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy; (V.R.); (E.L.F.); (F.D.); (S.M.L.); (A.B.); (R.A.); (F.T.); (F.B.); (E.C.); (N.P.)
- Department of Health Sciences, Campus S. Venuta, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Emma Colao
- Medical Genetics Unit, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy; (V.R.); (E.L.F.); (F.D.); (S.M.L.); (A.B.); (R.A.); (F.T.); (F.B.); (E.C.); (N.P.)
| | - Nicola Perrotti
- Medical Genetics Unit, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy; (V.R.); (E.L.F.); (F.D.); (S.M.L.); (A.B.); (R.A.); (F.T.); (F.B.); (E.C.); (N.P.)
- Department of Health Sciences, Campus S. Venuta, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Francesco Paduano
- Stem Cells and Medical Genetics Units, Biomedical Section, Tecnologica Research Institute and Marrelli Health, 88900 Crotone, Italy
| | - Rodolfo Iuliano
- Medical Genetics Unit, Renato Dulbecco University Hospital, 88100 Catanzaro, Italy; (V.R.); (E.L.F.); (F.D.); (S.M.L.); (A.B.); (R.A.); (F.T.); (F.B.); (E.C.); (N.P.)
- Department of Health Sciences, Campus S. Venuta, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| |
Collapse
|
23
|
Zhang L, Abro B, Campbell A, Ding Y. TP53 mutations in myeloid neoplasms: implications for accurate laboratory detection, diagnosis, and treatment. Lab Med 2024; 55:686-699. [PMID: 39001691 PMCID: PMC11532620 DOI: 10.1093/labmed/lmae048] [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: 11/05/2024] Open
Abstract
Genetic alterations that affect the function of p53 tumor suppressor have been extensively investigated in myeloid neoplasms, revealing their significant impact on disease progression, treatment response, and patient outcomes. The identification and characterization of TP53 mutations play pivotal roles in subclassifying myeloid neoplasms and guiding treatment decisions. Starting with the presentation of a typical case, this review highlights the complicated nature of genetic alterations involving TP53 and provides a comprehensive analysis of TP53 mutations and other alterations in myeloid neoplasms. Currently available methods used in clinical laboratories to identify TP53 mutations are discussed, focusing on the importance of establishing a robust testing protocol within clinical laboratories to ensure the delivery of accurate and reliable results. The treatment implications of TP53 mutations in myeloid neoplasms and clinical trial options are reviewed. Ultimately, we hope that this review provides valuable insights into the patterns of TP53 alterations in myeloid neoplasms and offers guidance to establish practical laboratory testing protocols to support the best practices of precision oncology.
Collapse
Affiliation(s)
- Linsheng Zhang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, US
| | - Brooj Abro
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, US
| | - Andrew Campbell
- Department of Laboratory Medicine, Geisinger Medical Center, Danville, PA, US
| | - Yi Ding
- Department of Laboratory Medicine, Geisinger Medical Center, Danville, PA, US
| |
Collapse
|
24
|
Arnon J, Zick A, Maoz M, Salaymeh N, Gugenheim A, Marouani M, Mor E, Hamburger T, Saadi N, Elia A, Ganz G, Fahham D, Meirovitz A, Kadouri L, Meiner V, Yablonski-Peretz T, Shkedi-Rafid S. Clinical and genetic characteristics of carriers of the TP53 c.541C > T, p.Arg181Cys pathogenic variant causing hereditary cancer in patients of Arab-Muslim descent. Fam Cancer 2024; 23:531-542. [PMID: 38743206 PMCID: PMC11512851 DOI: 10.1007/s10689-024-00391-2] [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: 02/23/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024]
Abstract
TP53 pathogenic variants cause Li-Fraumeni syndrome (LFS), with some variants causing an attenuated phenotype. Herein, we describe the clinical phenotype and genetic characteristics of carriers of NM_000546.6 (TP53): c.541C > T, (p.Arg181Cys) treated at Hadassah Medical Center. We retrospectively examined our genetic databases to identify all carriers of TP53 p.Arg181Cys. We reached out to carriers and their relatives and collected clinical and demographic data, lifestyle factors, carcinogenic exposures as well as additional blood samples for genetic testing and whole exome sequencing. Between 2005 and 2022 a total of 2875 cancer patients underwent genetic testing using genetic panels, whole exome sequencing or targeted TP53 assays. A total of 30 cancer patients, all of Arab-Muslim descent, were found to be carriers of TP53 p.Arg181Cys, the majority from Jerusalem and Hebron, two of which were homozygous for the variant. Carriers were from 24 distinct families of them, 15 families (62.5%) met updated Chompret criteria for LFS. Median age of diagnosis was 35 years-old (range 1-69) with cancers characteristic of LFS (16 Breast cancer; 6 primary CNS tumors; 3 sarcomas) including 4 children with choroid plexus carcinoma, medulloblastoma, or glioblastoma. A total of 21 healthy carriers of TP53 p.Arg181Cys were identified at a median age of 39 years-old (range 2-54)-19 relatives and 2 additional pediatric non-cancer patients, in which the finding was incidental. We report a shared haplotype of 350kb among carriers, limited co-morbidities and low BMI in both cancer patients and healthy carriers. There were no demographic factors or carcinogenic exposures unique to carriers who developed malignancy. Upon exome analysis no other known pathogenic variants in cancer predisposing genes were identified. TP53 p.Arg181Cys is a founder pathogenic variant predominant to the Arab-Muslim population in Jerusalem and Hebron, causing attenuated-LFS. We suggest strict surveillance in established carriers and encourage referral to genetic testing for all cancer patients of Arab-Muslim descent in this region with LFS-associated malignancies as well as family members of established carriers.
Collapse
Affiliation(s)
- Johnathan Arnon
- Sharett Institute of Oncology, Hadassah University Medical Center, Jerusalem, Israel.
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Aviad Zick
- Sharett Institute of Oncology, Hadassah University Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Myriam Maoz
- Sharett Institute of Oncology, Hadassah University Medical Center, Jerusalem, Israel
| | - Nada Salaymeh
- Sharett Institute of Oncology, Hadassah University Medical Center, Jerusalem, Israel
| | - Ahinoam Gugenheim
- Sharett Institute of Oncology, Hadassah University Medical Center, Jerusalem, Israel
| | - MazalTov Marouani
- Sharett Institute of Oncology, Hadassah University Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eden Mor
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tamar Hamburger
- Sharett Institute of Oncology, Hadassah University Medical Center, Jerusalem, Israel
| | - Nagam Saadi
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Anna Elia
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Pathology, Hadassah University Medical Center, Jerusalem, Israel
| | - Gael Ganz
- Department of Genetics, Hadassah University Medical Center, Jerusalem, Israel
| | - Duha Fahham
- Department of Genetics, Hadassah University Medical Center, Jerusalem, Israel
| | - Amichay Meirovitz
- Sharett Institute of Oncology, Hadassah University Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Luna Kadouri
- Sharett Institute of Oncology, Hadassah University Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Vardiella Meiner
- Department of Genetics, Hadassah University Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tamar Yablonski-Peretz
- Sharett Institute of Oncology, Hadassah University Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shiri Shkedi-Rafid
- Department of Genetics, Hadassah University Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
25
|
Puzo CJ, Hager KM, Rinder HM, Weinberg OK, Siddon AJ. Overall survival in TP53-mutated AML and MDS. Ann Hematol 2024:10.1007/s00277-024-06054-7. [PMID: 39443370 DOI: 10.1007/s00277-024-06054-7] [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: 08/08/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
TP53 mutations in patients with AML and MDS frequently portend a poor prognosis, related to both p53 allele status and blast count. In 2022, the ICC and WHO released updated guidelines for classifying p53-mutated AML/MDS. The characteristics of p53 mutations, their associated co-mutations, and their effects on overall survival (OS) are not known in the context of these new guidelines. A retrospective chart review was undertaken for all patients with AML or MDS and at least one TP53 mutation detected on next generation sequencing (NGS) at Yale New Haven Hospital from 2015 to 2023. All patients (N = 210) met criteria for one of the 5 diagnostic classes based on WHO and ICC guidelines. Kaplan-Meier curves with associated log-rank testing and Cox proportional hazards model quantified the effects of clinical and molecular data on survival. Multi-hit pathogenic mutations were related to poorer OS in MDS but not AML using either the WHO (p = .02) or the ICC (p = .01) diagnostic criteria. The most significant predictors of OS in the sample overall were platelet count < 50 K (HR: 2.01, 95% CI [1.47, 2.75], p < .001) and TP53 VAF ≤ 40% (HR: 0.68, 95% CI[0.50, 0.91], p = .01). Blast count ranges, complex karyotype, and p53 mutation type or location, showed no association with OS. In our cohort defined by the 2022 ICC and WHO criteria, VAF and thrombocytopenia, rather than blast count or p53 mutation features, significantly predicted OS. These results speak to each criteria's ability to identify cases of similarly aggressive disease biology and prognosis.
Collapse
Affiliation(s)
| | - Karl M Hager
- Department of Laboratory Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Henry M Rinder
- Department of Laboratory Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Olga K Weinberg
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexa J Siddon
- Department of Laboratory Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
26
|
Qayoom H, Haq BU, Sofi S, Jan N, Jan A, Mir MA. Targeting mutant p53: a key player in breast cancer pathogenesis and beyond. Cell Commun Signal 2024; 22:484. [PMID: 39390510 PMCID: PMC11466041 DOI: 10.1186/s12964-024-01863-9] [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: 03/14/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
The p53 mutation is the most common genetic mutation associated with human neoplasia. TP53 missense mutations, which frequently arise early in breast cancer, are present in over thirty percent of breast tumors. In breast cancer, p53 mutations are linked to a more aggressive course of the disease and worse overall survival rates. TP53 mutations are mostly seen in triple-negative breast cancer, a very diverse kind of the disease. The majority of TP53 mutations originate in the replacement of individual amino acids within the p53 protein's core domain, giving rise to a variety of variations referred to as "mutant p53s." In addition to gaining carcinogenic qualities through gain-of-function pathways, these mutants lose the typical tumor-suppressive features of p53 to variable degrees. The gain-of-function impact of stabilized mutant p53 causes tumor-specific dependency and resistance to therapy. P53 is a prospective target for cancer therapy because of its tumor-suppressive qualities and the numerous alterations that it experiences in tumors. Phenotypic abnormalities in breast cancer, notably poorly differentiated basal-like tumors are frequently linked to high-grade tumors. By comparing data from cell and animal models with clinical outcomes in breast cancer, this study investigates the molecular mechanisms that convert gene alterations into the pathogenic consequences of mutant p53's tumorigenic activity. The study delves into current and novel treatment approaches aimed at targeting p53 mutations, taking into account the similarities and differences in p53 regulatory mechanisms between mutant and wild-type forms, as well.
Collapse
Affiliation(s)
- Hina Qayoom
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir Srinagar, Kashmir Srinagar, J&K, 190006, India
| | - Burhan Ul Haq
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir Srinagar, Kashmir Srinagar, J&K, 190006, India
| | - Shazia Sofi
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir Srinagar, Kashmir Srinagar, J&K, 190006, India
| | - Nusrat Jan
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir Srinagar, Kashmir Srinagar, J&K, 190006, India
| | - Asma Jan
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir Srinagar, Kashmir Srinagar, J&K, 190006, India
| | - Manzoor A Mir
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir Srinagar, Kashmir Srinagar, J&K, 190006, India.
| |
Collapse
|
27
|
Noeraparast M, Krajina K, Pichler R, Niedersüß‐Beke D, Shariat SF, Grünwald V, Ahyai S, Pichler M. FGFR3 alterations in bladder cancer: Sensitivity and resistance to targeted therapies. Cancer Commun (Lond) 2024; 44:1189-1208. [PMID: 39161208 PMCID: PMC11483561 DOI: 10.1002/cac2.12602] [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: 04/07/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024] Open
Abstract
In this review, we revisit the pivotal role of fibroblast growth factor receptor 3 (FGFR3) in bladder cancer (BLCA), underscoring its prevalence in both non-muscle-invasive and muscle-invasive forms of the disease. FGFR3 mutations in up to half of BLCAs play a well-established role in tumorigenesis, shaping distinct tumor initiation patterns and impacting the tumor microenvironment (TME). Emphasizing the importance of considering epithelial-mesenchymal transition profile and TME status, we revisit their relevance in predicting responses to immune checkpoint inhibitors in FGFR3-mutated BLCAs. This writing highlights the initially promising yet transient efficacy of the FGFR inhibitor Erdafitinib on FGFR3-mutated BLCA, stressing the pressing need to unravel resistance mechanisms and identify co-targets for future combinatorial studies. A thorough analysis of recent preclinical and clinical evidence reveals resistance mechanisms, including secondary mutations, epigenetic alterations in pathway effectors, phenotypic heterogeneity, and population-specific variations within FGFR3 mutational status. Lastly, we discuss the potential of combinatorial treatments and concepts like synthetic lethality for discovering more effective targeted therapies against FGFR3-mutated BLCA.
Collapse
Affiliation(s)
- Maxim Noeraparast
- Translational OncologyII. Med Clinics Hematology and OncologyAugsburgGermany
| | - Katarina Krajina
- Translational OncologyII. Med Clinics Hematology and OncologyAugsburgGermany
| | - Renate Pichler
- Department of UrologyMedical University of InnsbruckInnsbruckAustria
| | | | | | - Viktor Grünwald
- Interdisciplinary Genitourinary OncologyClinic for Urology, Clinic for Medical OncologyUniversity Hospital Essen, Hufelandstraße 55EssenGermany
| | - Sascha Ahyai
- Department of UrologyMedical University of GrazGrazAustria
| | - Martin Pichler
- Translational OncologyII. Med Clinics Hematology and OncologyAugsburgGermany
| |
Collapse
|
28
|
Shorthouse D, Lister H, Freeman GS, Hall BA. Understanding large scale sequencing datasets through changes to protein folding. Brief Funct Genomics 2024; 23:517-524. [PMID: 38521964 PMCID: PMC11428155 DOI: 10.1093/bfgp/elae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/25/2024] Open
Abstract
The expansion of high-quality, low-cost sequencing has created an enormous opportunity to understand how genetic variants alter cellular behaviour in disease. The high diversity of mutations observed has however drawn a spotlight onto the need for predictive modelling of mutational effects on phenotype from variants of uncertain significance. This is particularly important in the clinic due to the potential value in guiding clinical diagnosis and patient treatment. Recent computational modelling has highlighted the importance of mutation induced protein misfolding as a common mechanism for loss of protein or domain function, aided by developments in methods that make large computational screens tractable. Here we review recent applications of this approach to different genes, and how they have enabled and supported subsequent studies. We further discuss developments in the approach and the role for the approach in light of increasingly high throughput experimental approaches.
Collapse
Affiliation(s)
- David Shorthouse
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Harris Lister
- Department of Medical Physics and Biomedical Engineering, Malet Place Engineering Building, University College London, Gower Street, London WC1E 6BT, UK
| | - Gemma S Freeman
- Department of Medical Physics and Biomedical Engineering, Malet Place Engineering Building, University College London, Gower Street, London WC1E 6BT, UK
| | - Benjamin A Hall
- Department of Medical Physics and Biomedical Engineering, Malet Place Engineering Building, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
29
|
Demajo S, Ramis-Zaldivar JE, Muiños F, Grau ML, Andrianova M, López-Bigas N, González-Pérez A. Identification of Clonal Hematopoiesis Driver Mutations through In Silico Saturation Mutagenesis. Cancer Discov 2024; 14:1717-1731. [PMID: 38722595 PMCID: PMC11372364 DOI: 10.1158/2159-8290.cd-23-1416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/05/2024] [Accepted: 05/07/2024] [Indexed: 05/21/2024]
Abstract
Clonal hematopoiesis (CH) is a phenomenon of clonal expansion of hematopoietic stem cells driven by somatic mutations affecting certain genes. Recently, CH has been linked to the development of hematologic malignancies, cardiovascular diseases, and other conditions. Although the most frequently mutated CH driver genes have been identified, a systematic landscape of the mutations capable of initiating this phenomenon is still lacking. In this study, we trained machine learning models for 12 of the most recurrent CH genes to identify their driver mutations. These models outperform expert-curated rules based on prior knowledge of the function of these genes. Moreover, their application to identify CH driver mutations across almost half a million donors of the UK Biobank reproduces known associations between CH driver mutations and age, and the prevalence of several diseases and conditions. We thus propose that these models support the accurate identification of CH across healthy individuals. Significance: We developed and validated gene-specific machine learning models to identify CH driver mutations, showing their advantage with respect to expert-curated rules. These models can support the identification and clinical interpretation of CH mutations in newly sequenced individuals. See related commentary by Arends and Jaiswal, p. 1581.
Collapse
Affiliation(s)
- Santiago Demajo
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
| | - Joan E. Ramis-Zaldivar
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
| | - Ferran Muiños
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
| | - Miguel L. Grau
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Maria Andrianova
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Núria López-Bigas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
- University Pompeu Fabra, Barcelona, Spain.
| | - Abel González-Pérez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Centro de Investigación Biomédica en Red en Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
- University Pompeu Fabra, Barcelona, Spain.
| |
Collapse
|
30
|
Ortaköylü MY, Özdemir Sİ, Dinçaslan H, Taçyıldız N, Ateş U, Ünal AE, Soydal Ç, Fitoz ÖS, Karabulut HG, Ruhi HI, Ünal EC. De novo germline TP53 mutation in a pediatric patient with Li-Fraumeni syndrome and diffuse peritoneal mesothelioma. Pediatr Blood Cancer 2024; 71:e31071. [PMID: 38773723 DOI: 10.1002/pbc.31071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/24/2024]
Affiliation(s)
- Melek Yaman Ortaköylü
- Division of Pediatric Oncology, Department of Pediatrics, Ankara University, Ankara, Turkey
| | - Sonay İncesoy Özdemir
- Division of Pediatric Oncology, Department of Pediatrics, Ankara University, Ankara, Turkey
| | - Handan Dinçaslan
- Division of Pediatric Oncology, Department of Pediatrics, Ankara University, Ankara, Turkey
| | - Nurdan Taçyıldız
- Division of Pediatric Oncology, Department of Pediatrics, Ankara University, Ankara, Turkey
| | - Ufuk Ateş
- Department of Pediatric Surgery, Ankara University, Ankara, Turkey
| | - Ali Ekrem Ünal
- Department of General Surgery, Ankara University, Ankara, Turkey
| | - Çiğdem Soydal
- Department of Nuclear Medicine, Ankara University, Ankara, Turkey
| | - Ömer Suat Fitoz
- Division of Pediatric Radiology, Radiology Department, Ankara University, Ankara, Turkey
| | | | | | - Emel Cabi Ünal
- Division of Pediatric Oncology, Department of Pediatrics, Ankara University, Ankara, Turkey
| |
Collapse
|
31
|
Carrasco-Tenezaca F, Moreira-Dinzey J, Manrai PA, Bearse M, Burela S, Podany P, Singh K, Pareja F, Zheng J, Muscato NE, Liang Y, Zhan H, Krishnamurti U, Dolezal D, Wang J, Harigopal M. Breast Carcinoma With Tubulopapillary Features Has a Distinct Immunophenotypic and Molecular Signature: A Report of Two Tumors and Literature Review. Int J Surg Pathol 2024; 32:1037-1045. [PMID: 37908113 DOI: 10.1177/10668969231209780] [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: 11/02/2023]
Abstract
Breast carcinoma with tubulopapillary features is a newly described entity associated with poor prognosis with only 14 tumors reported in the literature. We report 2 additional tumors and identify novel immunohistochemical and molecular features of the tumor. The first tumor was from a 72-year-old woman with nonmetastatic breast carcinoma and the second was from a 32-year-old woman with metastatic breast carcinoma who received neoadjuvant therapy. Both tumors had high-grade nuclear features with a distinctive morphology characterized by infiltrating open glands with intratubular papillary and micropapillary projections in >90% of the invasive carcinoma. In addition to the usual predictors of aggressive behavior, both tumors showed a high expression of p16 and SOX10, which has not been previously described. Targeted tumor sequencing revealed pathogenic variants of TP53 in both tumors, in agreement with previous reports. Prior studies have shown a correlation between p16 and SOX10 expression with high-grade features and worse prognosis; typically seen in triple-negative carcinomas as demonstrated in both of our tumors. However, not all reported tumors of breast carcinoma with tubulopapillary features have demonstrated a triple-negative profile as there are a few reports of tumors with estrogen receptor and/or human epidermal growth factor 2 expression. Due to their distinct morphologic and molecular characteristics, breast carcinoma with tubulopapillary features may represent a new breast cancer histologic subtype.
Collapse
Affiliation(s)
| | | | - Padmini A Manrai
- Department of Pathology, Yale New Haven Hospital, New Haven, CT, USA
| | - Mayara Bearse
- Department of Pathology, Yale New Haven Hospital, New Haven, CT, USA
| | | | - Peter Podany
- Department of Pathology, Yale New Haven Hospital, New Haven, CT, USA
| | - Kamaljeet Singh
- Pathology and Laboratory Medicine, Brown University Warren Alpert Medical School, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Fresia Pareja
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Nicole E Muscato
- Department of Pathology, Lawrence and Memorial Hospital, New London, CT, USA
| | - Yuanxin Liang
- Department of Pathology, Yale New Haven Hospital, New Haven, CT, USA
| | - Haiying Zhan
- Department of Pathology, Yale New Haven Hospital, New Haven, CT, USA
| | - Uma Krishnamurti
- Department of Pathology, Yale New Haven Hospital, New Haven, CT, USA
| | - Darin Dolezal
- Department of Pathology, Yale New Haven Hospital, New Haven, CT, USA
| | - Jianhui Wang
- Department of Pathology, Yale New Haven Hospital, New Haven, CT, USA
| | - Malini Harigopal
- Department of Pathology, Yale New Haven Hospital, New Haven, CT, USA
| |
Collapse
|
32
|
Padigepati SR, Stafford DA, Tan CA, Silvis MR, Jamieson K, Keyser A, Nunez PAC, Nicoludis JM, Manders T, Fresard L, Kobayashi Y, Araya CL, Aradhya S, Johnson B, Nykamp K, Reuter JA. Scalable approaches for generating, validating and incorporating data from high-throughput functional assays to improve clinical variant classification. Hum Genet 2024; 143:995-1004. [PMID: 39085601 PMCID: PMC11303574 DOI: 10.1007/s00439-024-02691-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/12/2024] [Indexed: 08/02/2024]
Abstract
As the adoption and scope of genetic testing continue to expand, interpreting the clinical significance of DNA sequence variants at scale remains a formidable challenge, with a high proportion classified as variants of uncertain significance (VUSs). Genetic testing laboratories have historically relied, in part, on functional data from academic literature to support variant classification. High-throughput functional assays or multiplex assays of variant effect (MAVEs), designed to assess the effects of DNA variants on protein stability and function, represent an important and increasingly available source of evidence for variant classification, but their potential is just beginning to be realized in clinical lab settings. Here, we describe a framework for generating, validating and incorporating data from MAVEs into a semi-quantitative variant classification method applied to clinical genetic testing. Using single-cell gene expression measurements, cellular evidence models were built to assess the effects of DNA variation in 44 genes of clinical interest. This framework was also applied to models for an additional 22 genes with previously published MAVE datasets. In total, modeling data was incorporated from 24 genes into our variant classification method. These data contributed evidence for classifying 4043 observed variants in over 57,000 individuals. Genetic testing laboratories are uniquely positioned to generate, analyze, validate, and incorporate evidence from high-throughput functional data and ultimately enable the use of these data to provide definitive clinical variant classifications for more patients.
Collapse
Affiliation(s)
| | | | | | - Melanie R Silvis
- Invitae Corporation, San Francisco, CA, 94103, USA
- Epic Bio, South San Francisco, CA, 94080, USA
| | - Kirsty Jamieson
- Invitae Corporation, San Francisco, CA, 94103, USA
- Epic Bio, South San Francisco, CA, 94080, USA
| | - Andrew Keyser
- Invitae Corporation, San Francisco, CA, 94103, USA
- Calico Life Sciences, South San Francisco, CA, 94080, USA
| | | | - John M Nicoludis
- Invitae Corporation, San Francisco, CA, 94103, USA
- Department of Structural Biology, Genentech, South San Francisco, CA, 94080, USA
| | - Toby Manders
- Invitae Corporation, San Francisco, CA, 94103, USA
| | | | | | - Carlos L Araya
- Invitae Corporation, San Francisco, CA, 94103, USA
- Tapanti.org, Santa Barbara, CA, 93108, USA
| | | | - Britt Johnson
- Invitae Corporation, San Francisco, CA, 94103, USA
- GeneDx, Stamford, CT, 06902, USA
| | - Keith Nykamp
- Invitae Corporation, San Francisco, CA, 94103, USA.
| | | |
Collapse
|
33
|
Scatolini M, Grisanti S, Tomaiuolo P, Grosso E, Basile V, Cosentini D, Puglisi S, Laganà M, Perotti P, Saba L, Rossini E, Palermo F, Sigala S, Volante M, Berruti A, Terzolo M. Germline NGS targeted analysis in adult patients with sporadic adrenocortical carcinoma. Eur J Cancer 2024; 205:114088. [PMID: 38714106 DOI: 10.1016/j.ejca.2024.114088] [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: 12/13/2023] [Revised: 04/11/2024] [Accepted: 04/21/2024] [Indexed: 05/09/2024]
Abstract
BACKGROUND Adrenocortical carcinoma (ACC) is a rare cancer that arises sporadically or due to hereditary syndromes. Data on germline variants (GVs) in sporadic ACC are limited. Our aim was to characterize GVs of genes potentially related to adrenal diseases in 150 adult patients with sporadic ACC. METHODS This was a retrospective analysis of stage I-IV ACC patients with sporadic ACC from two reference centers for ACC in Italy. Patients were included in the analysis if they had confirmed diagnosis of ACC, a frozen peripheral blood sample and complete clinical and follow-up data. Next generation sequencing technology was used to analyze the prevalence of GVs in a custom panel of 17 genes belonging to either cancer-predisposition genes or adrenocortical-differentiation genes categories. RESULTS We identified 18 GVs based on their frequency, enrichment and predicted functional characteristics. We found six pathogenic (P) or likely pathogenic (LP) variants in ARMC5, CTNNB1, MSH2, PDE11A and TP53 genes; and twelve variants lacking evidence of pathogenicity. New unique P/LP variants were identified in TP53 (p.G105D) and, for the first time, in ARMC5 (p.P731R). The presence of P/LP GVs was associated with reduced survival outcomes and had a significant and independent impact on both progression-free survival and overall survival. CONCLUSIONS GVs were present in 6.7 % of patients with sporadic ACC, and we identified novel variants of ARMC5 and TP53. These findings may improve understanding of ACC pathogenesis and enable genetic counseling of patients and their families.
Collapse
Affiliation(s)
- Maria Scatolini
- Molecular Oncology Laboratory, Fondazione Edo ed Elvo Tempia, 13875 Ponderano, BI, Italy
| | - Salvatore Grisanti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili, 25123 Brescia, Italy
| | - Pasquale Tomaiuolo
- Molecular Oncology Laboratory, Fondazione Edo ed Elvo Tempia, 13875 Ponderano, BI, Italy; Internal Medicine, Department of Clinical and Biological Sciences, S. Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Italy
| | - Enrico Grosso
- Molecular Oncology Laboratory, Fondazione Edo ed Elvo Tempia, 13875 Ponderano, BI, Italy
| | - Vittoria Basile
- Internal Medicine, Department of Clinical and Biological Sciences, S. Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Italy
| | - Deborah Cosentini
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili, 25123 Brescia, Italy
| | - Soraya Puglisi
- Internal Medicine, Department of Clinical and Biological Sciences, S. Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Italy.
| | - Marta Laganà
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili, 25123 Brescia, Italy
| | - Paola Perotti
- Internal Medicine, Department of Clinical and Biological Sciences, S. Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Italy
| | - Laura Saba
- Internal Medicine, Department of Clinical and Biological Sciences, S. Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Italy
| | - Elisa Rossini
- Department of Molecular & Translational Medicine, Section of Pharmacology, University of Brescia, 25123 Brescia, Italy
| | - Flavia Palermo
- Molecular Oncology Laboratory, Fondazione Edo ed Elvo Tempia, 13875 Ponderano, BI, Italy
| | - Sandra Sigala
- Department of Molecular & Translational Medicine, Section of Pharmacology, University of Brescia, 25123 Brescia, Italy
| | - Marco Volante
- Pathology Unit, Oncology department, University of Turin, San Luigi Gonzaga University Hospital, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Alfredo Berruti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili, 25123 Brescia, Italy
| | - Massimo Terzolo
- Internal Medicine, Department of Clinical and Biological Sciences, S. Luigi Gonzaga Hospital, University of Turin, 10043 Orbassano, Italy
| |
Collapse
|
34
|
Kovács Z, Sugimura H, György TA, Osvath E, Manirakiza F, Gurzu S. Bioinformatic Identification of TP53 Gene Mutation Hotspots in Colorectal Cancer. Int J Mol Sci 2024; 25:6612. [PMID: 38928318 PMCID: PMC11203433 DOI: 10.3390/ijms25126612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Mutations and inactivation of the TP53 gene are frequently observed in various types of malignancies. Precise knowledge of the genetic structure and detection of mutation hotspots are crucial, as these indicate a high probability of developing cancer. The aim of our study was to perform the bioinformatic detection of mutation hotspots in the TP53 gene in patients diagnosed with malignant colon neoplasms using self-developed software (version 1). We compared TP53 gene sequences from 50 healthy individuals with those from 50 patients diagnosed with colorectal carcinoma. Of the 50 samples from cancer patients, the most frequent mutations were observed in exons 5 and 8 (12 mutations per exon) and gene sequences of 12 samples, which differed from those of the 50 samples from healthy individuals. Based on our results, the distribution of mutations in the TP53 gene structure was not even across different exons. By comparing the gene sequences of healthy individuals with those of colon cancer samples, we conclude that structural changes occurring in similar gene regions are not associated with increases in susceptibility to malignancies in every case, namely, that the pathological mechanism is multifactorial.
Collapse
Affiliation(s)
- Zsolt Kovács
- Department of Pathology, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania; (Z.K.); (T.A.G.); (E.O.)
- Research Center of Oncopathology and Translational Research (CCOMT), 540139 Targu Mures, Romania
| | | | - Tamás Attila György
- Department of Pathology, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania; (Z.K.); (T.A.G.); (E.O.)
- Research Center of Oncopathology and Translational Research (CCOMT), 540139 Targu Mures, Romania
| | - Eva Osvath
- Department of Pathology, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania; (Z.K.); (T.A.G.); (E.O.)
- Research Center of Oncopathology and Translational Research (CCOMT), 540139 Targu Mures, Romania
- Department of Oncology, Clinical County Hospital, 540140 Targu Mures, Romania
| | - Felix Manirakiza
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Kigali P.O. Box 3286, Rwanda;
| | - Simona Gurzu
- Department of Pathology, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania; (Z.K.); (T.A.G.); (E.O.)
- Research Center of Oncopathology and Translational Research (CCOMT), 540139 Targu Mures, Romania
- Romanian Academy of Medical Sciences, 030167 București, Romania
| |
Collapse
|
35
|
Guo Y, Wu H, Wiesmüller L, Chen M. Canonical and non-canonical functions of p53 isoforms: potentiating the complexity of tumor development and therapy resistance. Cell Death Dis 2024; 15:412. [PMID: 38866752 PMCID: PMC11169513 DOI: 10.1038/s41419-024-06783-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024]
Abstract
Full-length p53 (p53α) plays a pivotal role in maintaining genomic integrity and preventing tumor development. Over the years, p53 was found to exist in various isoforms, which are generated through alternative splicing, alternative initiation of translation, and internal ribosome entry site. p53 isoforms, either C-terminally altered or N-terminally truncated, exhibit distinct biological roles compared to p53α, and have significant implications for tumor development and therapy resistance. Due to a lack of part and/or complete C- or N-terminal domains, ectopic expression of some p53 isoforms failed to induce expression of canonical transcriptional targets of p53α like CDKN1A or MDM2, even though they may bind their promoters. Yet, p53 isoforms like Δ40p53α still activate subsets of targets including MDM2 and BAX. Furthermore, certain p53 isoforms transactivate even novel targets compared to p53α. More recently, non-canonical functions of p53α in DNA repair and of different isoforms in DNA replication unrelated to transcriptional activities were discovered, amplifying the potential of p53 as a master regulator of physiological and tumor suppressor functions in human cells. Both regarding canonical and non-canonical functions, alternative p53 isoforms frequently exert dominant negative effects on p53α and its partners, which is modified by the relative isoform levels. Underlying mechanisms include hetero-oligomerization, changes in subcellular localization, and aggregation. These processes ultimately influence the net activities of p53α and give rise to diverse cellular outcomes. Biological roles of p53 isoforms have implications for tumor development and cancer therapy resistance. Dysregulated expression of isoforms has been observed in various cancer types and is associated with different clinical outcomes. In conclusion, p53 isoforms have expanded our understanding of the complex regulatory network involving p53 in tumors. Unraveling the mechanisms underlying the biological roles of p53 isoforms provides new avenues for studies aiming at a better understanding of tumor development and developing therapeutic interventions to overcome resistance.
Collapse
Affiliation(s)
- Yitian Guo
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China.
| | - Hang Wu
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Lisa Wiesmüller
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| | - Ming Chen
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China.
| |
Collapse
|
36
|
Ea V, Berthozat C, Dreyfus H, Legrand C, Rousselet E, Peysselon M, Baudet L, Martinez G, Coutton C, Bidart M. BRCA1 Intragenic Duplication Combined with a Likely Pathogenic TP53 Variant in a Patient with Triple-Negative Breast Cancer: Clinical Risk and Management. Int J Mol Sci 2024; 25:6274. [PMID: 38892462 PMCID: PMC11173113 DOI: 10.3390/ijms25116274] [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: 04/17/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
For patients with hereditary breast and ovarian cancer, the probability of carrying two pathogenic variants (PVs) in dominant cancer-predisposing genes is rare. Using targeted next-generation sequencing (NGS), we investigated a 49-year-old Caucasian woman who developed a highly aggressive breast tumor. Our analyses identified an intragenic germline heterozygous duplication in BRCA1 with an additional likely PV in the TP53 gene. The BRCA1 variant was confirmed by multiplex ligation probe amplification (MLPA), and genomic breakpoints were characterized at the nucleotide level (c.135-2578_442-1104dup). mRNA extracted from lymphocytes was amplified by RT-PCR and then Sanger sequenced, revealing a tandem duplication r.135_441dup; p.(Gln148Ilefs*20). This duplication results in the synthesis of a truncated and, most likely, nonfunctional protein. Following functional studies, the TP53 exon 5 c.472C > T; p.(Arg158Cys) missense variant was classified as likely pathogenic by the Li-Fraumeni Syndrome (LFS) working group. This type of unexpected association will be increasingly identified in the future, with the switch from targeted BRCA sequencing to hereditary breast and ovarian cancer (HBOC) panel sequencing, raising the question of how these patients should be managed. It is therefore important to record and investigate these rare double-heterozygous genotypes.
Collapse
Affiliation(s)
- Vuthy Ea
- UM Génétique Moléculaire: Maladies Héréditaires et Oncologie, University Hospital Grenoble Alpes, 38000 Grenoble, France;
- INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble Alpes University, 38000 Grenoble, France; (G.M.); (C.C.)
| | - Claudine Berthozat
- Department of Medical Oncology, Cancer and Blood Diseases, Grenoble Alpes University Hospital, 38000 Grenoble, France;
| | - Hélène Dreyfus
- Genetic Service, Department of Genetics and Procreation, University Hospital Grenoble Alpes, 38000 Grenoble, France; (H.D.); (C.L.); (E.R.); (M.P.); (L.B.)
| | - Clémentine Legrand
- Genetic Service, Department of Genetics and Procreation, University Hospital Grenoble Alpes, 38000 Grenoble, France; (H.D.); (C.L.); (E.R.); (M.P.); (L.B.)
| | - Estelle Rousselet
- Genetic Service, Department of Genetics and Procreation, University Hospital Grenoble Alpes, 38000 Grenoble, France; (H.D.); (C.L.); (E.R.); (M.P.); (L.B.)
| | - Magalie Peysselon
- Genetic Service, Department of Genetics and Procreation, University Hospital Grenoble Alpes, 38000 Grenoble, France; (H.D.); (C.L.); (E.R.); (M.P.); (L.B.)
| | - Laura Baudet
- Genetic Service, Department of Genetics and Procreation, University Hospital Grenoble Alpes, 38000 Grenoble, France; (H.D.); (C.L.); (E.R.); (M.P.); (L.B.)
| | - Guillaume Martinez
- INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble Alpes University, 38000 Grenoble, France; (G.M.); (C.C.)
- UM de Génétique Chromosomique, University Hospital Grenoble Alpes, 38000 Grenoble, France
| | - Charles Coutton
- INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble Alpes University, 38000 Grenoble, France; (G.M.); (C.C.)
- UM de Génétique Chromosomique, University Hospital Grenoble Alpes, 38000 Grenoble, France
| | - Marie Bidart
- UM Génétique Moléculaire: Maladies Héréditaires et Oncologie, University Hospital Grenoble Alpes, 38000 Grenoble, France;
- INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble Alpes University, 38000 Grenoble, France; (G.M.); (C.C.)
| |
Collapse
|
37
|
K AR, Arumugam S, Muninathan N, Baskar K, S D, D DR. P53 Gene as a Promising Biomarker and Potential Target for the Early Diagnosis of Reproductive Cancers. Cureus 2024; 16:e60125. [PMID: 38864057 PMCID: PMC11165294 DOI: 10.7759/cureus.60125] [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: 04/04/2024] [Accepted: 05/10/2024] [Indexed: 06/13/2024] Open
Abstract
One of the crucial aspects of cancer research is diagnosis with specificity and accuracy. Early cancer detection mostly helps make appropriate decisions regarding treatment and metastasis. The well-studied transcription factor tumor suppressor protein p53 is essential for maintaining genetic integrity. p53 is a key tumor suppressor that recognizes the carcinogenic biological pathways and eradicates them by apoptosis. A wide range of carcinomas, especially gynecological such as ovarian, cervical, and endometrial cancers, frequently undergo TP53 gene mutations. This study evaluates the potential of the p53 gene as a biological marker for the diagnosis of reproductive system neoplasms. Immunohistochemistry of p53 is rapid, easy to accomplish, cost-effective, and preferred by pathologists as a surrogate for the analysis of TP53 mutation. Thus, this review lays a groundwork for future efforts to develop techniques using p53 for the early diagnosis of cancer.
Collapse
Affiliation(s)
- Aswathi R K
- Medical Biochemistry, Meenakshi Academy of Higher Education and Research, Chennai, IND
| | - Suresh Arumugam
- Central Research Laboratory, Meenakshi Medical College Hospital and Research Institute, Kanchipuram, IND
| | - Natrajan Muninathan
- Central Research Laboratory, Meenakshi Medical College Hospital and Research Institute, Kanchipuram, IND
| | - Kuppusamy Baskar
- Central Research Laboratory, Meenakshi Medical College Hospital and Research Institute, Kanchipuram, IND
| | - Deepthi S
- Research and Development, Meenakshi Academy of Higher Education and Research, Chennai, IND
| | - Dinesh Roy D
- Centre for Advanced Genetic Studies, Genetika, Thiruvananthapuram, IND
| |
Collapse
|
38
|
Pereira MP, Herrity E, Kim DDH. TP53-mutated acute myeloid leukemia and myelodysplastic syndrome: biology, treatment challenges, and upcoming approaches. Ann Hematol 2024; 103:1049-1067. [PMID: 37770618 DOI: 10.1007/s00277-023-05462-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/17/2023] [Indexed: 09/30/2023]
Abstract
Improved understanding of TP53 biology and the clinicopathological features of TP53-mutated myeloid neoplasms has led to the recognition of TP53-mutated acute myeloid leukemia/myelodysplastic syndrome (TP53m AML/MDS) as a unique entity, characterized by dismal outcomes following conventional therapies. Several clinical trials have investigated combinations of emerging therapies for these patients with the poorest molecular prognosis among myeloid neoplasms. Although some emerging therapies have shown improvement in overall response rates, this has not translated into better overall survival, hence the notion that p53 remains an elusive target. New therapeutic strategies, including novel targeted therapies, immune checkpoint inhibitors, and monoclonal antibodies, represent a shift away from cytotoxic and hypomethylating-based therapies, towards approaches combining non-immune and novel immune therapeutic strategies. The triple combination of azacitidine and venetoclax with either magrolimab or eprenetapopt have demonstrated safety in early trials, with phase III trials currently underway, and promising interim clinical results. This review compiles background on TP53 biology, available and emerging therapies along with their mechanisms of action for the TP53m disease entity, current treatment challenges, and recently published data and status of ongoing clinical trials for TP53m AML/MDS.
Collapse
Affiliation(s)
- Mariana Pinto Pereira
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada
| | - Elizabeth Herrity
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada
| | - Dennis D H Kim
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada.
- Leukemia Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
- Department of Hematology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
39
|
Cirincione A, Simpson D, Ravisankar P, Solley SC, Yan J, Singh M, Adamson B. A benchmarked, high-efficiency prime editing platform for multiplexed dropout screening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.25.585978. [PMID: 38585933 PMCID: PMC10996517 DOI: 10.1101/2024.03.25.585978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Prime editing installs precise edits into the genome with minimal unwanted byproducts, but low and variable editing efficiencies have complicated application of the approach to high-throughput functional genomics. Leveraging several recent advances, we assembled a prime editing platform capable of high-efficiency substitution editing across a set of engineered prime editing guide RNAs (epegRNAs) and corresponding target sequences (80% median intended editing). Then, using a custom library of 240,000 epegRNAs targeting >17,000 codons with 175 different substitution types, we benchmarked our platform for functional interrogation of small substitution variants (1-3 nucleotides) targeted to essential genes. Resulting data identified negative growth phenotypes for nonsense mutations targeted to ~8,000 codons, and comparing those phenotypes to results from controls demonstrated high specificity. We also observed phenotypes for synonymous mutations that disrupted splice site motifs at 3' exon boundaries. Altogether, we establish and benchmark a high-throughput prime editing approach for functional characterization of genetic variants with simple readouts from multiplexed experiments.
Collapse
Affiliation(s)
- Ann Cirincione
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Danny Simpson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Purnima Ravisankar
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Present address: Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Sabrina C Solley
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Jun Yan
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Mona Singh
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Department of Computer Science, Princeton University, Princeton, NJ 08544, USA
| | - Britt Adamson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
40
|
Abida W, Hahn AW, Shore N, Agarwal N, Sieber P, Smith MR, Dorff T, Monk P, Rettig M, Patel R, Page A, Duff M, Xu R, Wang J, Barkund S, Pankov A, Wang A, Junttila M, Multani PS, Daemen A, Maneval EC, Logothetis CJ, Morris MJ. Phase I Study of ORIC-101, a Glucocorticoid Receptor Antagonist, in Combination with Enzalutamide in Patients with Metastatic Castration-resistant Prostate Cancer Progressing on Enzalutamide. Clin Cancer Res 2024; 30:1111-1120. [PMID: 38226958 PMCID: PMC10947849 DOI: 10.1158/1078-0432.ccr-23-3508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/12/2023] [Accepted: 01/10/2024] [Indexed: 01/17/2024]
Abstract
PURPOSE Increased glucocorticoid receptor (GR) signaling is a proposed compensatory mechanism of resistance to androgen receptor (AR) inhibition in metastatic castration-resistant prostate cancer (mCRPC). ORIC-101 is a potent and selective orally-bioavailable GR antagonist. PATIENTS AND METHODS Safety, pharmacokinetic/pharmacodynamic, and antitumor activity of ORIC-101 in combination with enzalutamide were studied in patients with mCRPC progressing on enzalutamide. ORIC-101 doses ranging from 80 to 240 mg once daily were tested in combination with enzalutamide 160 mg once daily. Pharmacokinetics/pharmacodynamics was assessed after a single dose and at steady state. Disease control rate (DCR) at 12 weeks was evaluated at the recommended phase 2 dose (RP2D). RESULTS A total of 41 patients were enrolled. There were no dose-limiting toxicities and the RP2D was selected as 240 mg of ORIC-101 and 160 mg of enzalutamide daily. At the RP2D, the most common treatment-related adverse events were fatigue (38.7%), nausea (29.0%), decreased appetite (19.4%), and constipation (12.9%). Pharmacokinetic/pharmacodynamic data confirmed ORIC-101 achieved exposures necessary for GR target engagement. Overall, for 31 patients treated at the RP2D, there was insufficient clinical benefit based on DCR (25.8%; 80% confidence interval: 15.65-38.52) which did not meet the prespecified target rate, leading to termination of the study. Exploratory subgroup analyses based on baseline GR expression, presence of AR resistance variants, and molecular features of aggressive variant prostate cancer suggested possible benefit in patients with high GR expression and no other resistance markers, although this would require confirmation. CONCLUSIONS Although the combination of ORIC-101 and enzalutamide demonstrated an acceptable tolerability profile, GR target inhibition with ORIC-101 did not produce clinical benefit in men with metastatic prostate cancer resistant to enzalutamide.
Collapse
Affiliation(s)
- Wassim Abida
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrew W Hahn
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | | | | | | | - Paul Monk
- The Ohio State University, Arthur James Cancer Hospital, Columbus, OH
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Michael J Morris
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
41
|
Timofeev O, Giron P, Lawo S, Pichler M, Noeparast M. ERK pathway agonism for cancer therapy: evidence, insights, and a target discovery framework. NPJ Precis Oncol 2024; 8:70. [PMID: 38485987 PMCID: PMC10940698 DOI: 10.1038/s41698-024-00554-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/16/2024] [Indexed: 03/18/2024] Open
Abstract
At least 40% of human cancers are associated with aberrant ERK pathway activity (ERKp). Inhibitors targeting various effectors within the ERKp have been developed and explored for over two decades. Conversely, a substantial body of evidence suggests that both normal human cells and, notably to a greater extent, cancer cells exhibit susceptibility to hyperactivation of ERKp. However, this vulnerability of cancer cells remains relatively unexplored. In this review, we reexamine the evidence on the selective lethality of highly elevated ERKp activity in human cancer cells of varying backgrounds. We synthesize the insights proposed for harnessing this vulnerability of ERK-associated cancers for therapeutical approaches and contextualize these insights within established pharmacological cancer-targeting models. Moreover, we compile the intriguing preclinical findings of ERK pathway agonism in diverse cancer models. Lastly, we present a conceptual framework for target discovery regarding ERKp agonism, emphasizing the utilization of mutual exclusivity among oncogenes to develop novel targeted therapies for precision oncology.
Collapse
Affiliation(s)
- Oleg Timofeev
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University, 35043, Marburg, Germany
| | - Philippe Giron
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research group Genetics, Reproduction and Development, Centre for Medical Genetics, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Steffen Lawo
- CRISPR Screening Core Facility, Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Martin Pichler
- Translational Oncology, II. Med Clinics Hematology and Oncology, 86156, Augsburg, Germany
| | - Maxim Noeparast
- Translational Oncology, II. Med Clinics Hematology and Oncology, 86156, Augsburg, Germany.
| |
Collapse
|
42
|
Wu R, Li D, Zhang S, Wang J, Chen K, Tuo Z, Miyamoto A, Yoo KH, Wei W, Zhang C, Feng D, Han P. A pan-cancer analysis of the oncogenic and immunological roles of transglutaminase 1 (TGM1) in human cancer. J Cancer Res Clin Oncol 2024; 150:123. [PMID: 38472489 PMCID: PMC10933153 DOI: 10.1007/s00432-024-05640-6] [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: 09/01/2023] [Accepted: 02/02/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND There is currently a limited number of studies on transglutaminase type 1 (TGM1) in tumors. The objective of this study is to perform a comprehensive analysis across various types of cancer to determine the prognostic significance of TGM1 in tumors and investigate its role in the immune environment. METHOD Pan-cancer and mutational data were retrieved from the TCGA database and analyzed using R (version 3.6.4) and its associated software package. The expression difference and prognosis of TGM1 were examined, along with its correlation with tumor heterogeneity, stemness, mutation landscape, and RNA modification. Additionally, the relationship between TGM1 expression and tumor immunity was investigated using the TIMER method. RESULTS TGM1 is expressed differently in various tumors and normal samples and is associated with the overall survival and progression-free time of KIRC, ACC, SKCM, LIHC, and STES. In LICH, we found a negative correlation between TGM1 expression and 6 indicators of tumor stemness. The mutation frequencies of BLCA, LIHC, and KIRC were 1.7%, 0.3%, and 0.3% respectively. In BLCA and BRCA, there was a significant correlation between TGM1 expression and the infiltration of CD4 + T cells, CD8 + T cells, neutrophils, and dendritic cells. CONCLUSION TGM1 has the potential to serve as both a prognostic marker and a drug target.
Collapse
Affiliation(s)
- Ruicheng Wu
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dengxiong Li
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuxia Zhang
- Research Core Facilities, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kai Chen
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Akira Miyamoto
- Department of Rehabilitation, West Kyushu University, Fukuoka, Japan
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, Seoul, South Korea
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chi Zhang
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
| | - Ping Han
- Department of Urology, Institute of Urology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
43
|
Gould SI, Wuest AN, Dong K, Johnson GA, Hsu A, Narendra VK, Atwa O, Levine SS, Liu DR, Sánchez Rivera FJ. High-throughput evaluation of genetic variants with prime editing sensor libraries. Nat Biotechnol 2024:10.1038/s41587-024-02172-9. [PMID: 38472508 DOI: 10.1038/s41587-024-02172-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/09/2024] [Indexed: 03/14/2024]
Abstract
Tumor genomes often harbor a complex spectrum of single nucleotide alterations and chromosomal rearrangements that can perturb protein function. Prime editing has been applied to install and evaluate genetic variants, but previous approaches have been limited by the variable efficiency of prime editing guide RNAs. Here we present a high-throughput prime editing sensor strategy that couples prime editing guide RNAs with synthetic versions of their cognate target sites to quantitatively assess the functional impact of endogenous genetic variants. We screen over 1,000 endogenous cancer-associated variants of TP53-the most frequently mutated gene in cancer-to identify alleles that impact p53 function in mechanistically diverse ways. We find that certain endogenous TP53 variants, particularly those in the p53 oligomerization domain, display opposite phenotypes in exogenous overexpression systems. Our results emphasize the physiological importance of gene dosage in shaping native protein stoichiometry and protein-protein interactions, and establish a framework for studying genetic variants in their endogenous sequence context at scale.
Collapse
Affiliation(s)
- Samuel I Gould
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alexandra N Wuest
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kexin Dong
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- University of Chinese Academy of Sciences, Beijing, China
| | - Grace A Johnson
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alvin Hsu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Varun K Narendra
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ondine Atwa
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Stuart S Levine
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Francisco J Sánchez Rivera
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
44
|
Mateoiu C, Palicelli A, Maloberti T, De Biase D, De Leo A, Lindh M, Bohlin KS, Stolnicu S. Primary vulvar adenocarcinoma of intestinal type: Report of two cases showing molecular similarity with colorectal adenocarcinoma. Pathol Res Pract 2024; 255:155181. [PMID: 38340583 DOI: 10.1016/j.prp.2024.155181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/30/2023] [Accepted: 01/27/2024] [Indexed: 02/12/2024]
Abstract
Primary vulvar adenocarcinoma is a particularly rare tumor with poorly understood histogenesis and unclear clinical characteristics and prognosis. Vulvar adenocarcinoma of intestinal type (VAIt) is a very uncommon subtype of primary vulvar adenocarcinoma and only 27 cases have been described in the literature in the past. Of these cases, two have been described as human papillomavirus (HPV)-associated VAIt. The current report presents two additional cases of primary VAIt showing variants in the KRAS, TP53, and DPYD genes and no evidence of HPV DNA by real-time polymerase chain reaction (RT-PCR). Next-generation sequencing (NGS) revealed TP53 pathogenic variants in both cases, but only one case had aberrant p53 protein immunohistochemical characteristics. KRAS and DPYD mutations were identified separately in the two cases. Due to their capacity to imitate the spread of more prevalent gastrointestinal carcinomas, these tumors may present diagnostic issues. Additional cases can contribute to a better understanding of the pathophysiology and prognosis of VAIt.
Collapse
Affiliation(s)
- Claudia Mateoiu
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Andrea Palicelli
- S.C. di Anat Patol Azienda USL-IRCCS, Ospedale S. Maria Nuova, di Reggio Emilia, Italy
| | - Thais Maloberti
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Italy
| | - Dario De Biase
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, Italy; Department of Pharmacy and Biotechnology, University of Bologna, Italy
| | - Antonio De Leo
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Italy
| | - Magnus Lindh
- Department of Infectious Diseases, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Katja Stenström Bohlin
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Simona Stolnicu
- Department of Pathology, University of Medicine, Pharmacy, Sciences and Technology "George E Palade" of Targu Mures, Targu Mures, Romania
| |
Collapse
|
45
|
Naeimzadeh Y, Tajbakhsh A, Fallahi J. Understanding the prion-like behavior of mutant p53 proteins in triple-negative breast cancer pathogenesis: The current therapeutic strategies and future directions. Heliyon 2024; 10:e26260. [PMID: 38390040 PMCID: PMC10881377 DOI: 10.1016/j.heliyon.2024.e26260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/20/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
Breast cancer (BC) is viewed as a significant public health issue and is the primary cause of cancer-related deaths among women worldwide. Triple-negative breast cancer (TNBC) is a particularly aggressive subtype that predominantly affects young premenopausal women. The tumor suppressor p53 playsa vital role in the cellular response to DNA damage, and its loss or mutations are commonly present in many cancers, including BC. Recent evidence suggests that mutant p53 proteins can aggregate and form prion-like structures, which may contribute to the pathogenesis of different types of malignancies, such as BC. This review provides an overview of BC molecular subtypes, the epidemiology of TNBC, and the role of p53 in BC development. We also discuss the potential implications of prion-like aggregation in BC and highlight future research directions. Moreover, a comprehensive analysis of the current therapeutic approaches targeting p53 aggregates in BC treatment is presented. Strategies including small molecules, chaperone inhibitors, immunotherapy, CRISPR-Cas9, and siRNA are discussed, along with their potential benefits and drawbacks. The use of these approaches to inhibit p53 aggregation and degradation represents a promising target for cancer therapy. Future investigations into the efficacy of these approaches against various p53 mutations or binding to non-p53 proteins should be conducted to develop more effective and personalized therapies for BC treatment.
Collapse
Affiliation(s)
- Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, 7133654361, Iran
| |
Collapse
|
46
|
Richau CS, Scherer NDM, Matta BP, de Armas EM, de Barros Moreira FC, Bergmann A, Pereira Chaves CB, Boroni M, dos Santos ACE, Moreira MAM. BRCA1, BRCA2, and TP53 germline and somatic variants and clinicopathological characteristics of Brazilian patients with epithelial ovarian cancer. Cancer Med 2024; 13:e6729. [PMID: 38308422 PMCID: PMC10905552 DOI: 10.1002/cam4.6729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/20/2023] [Accepted: 11/07/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Approximately 3/4 of ovarian cancers are diagnosed in advanced stages, with the high-grade epithelial ovarian carcinoma (EOC) accounting for 90% of the cases. EOC present high genomic instability and somatic loss-of-function variants in genes associated with homologous recombination mutational repair pathway (HR), such as BRCA1 and BRCA2, and in TP53. The identification of germline variants in HR genes in EOC is relevant for treatment of platinum resistant tumors and relapsed tumors with therapies based in synthetic lethality such as PARP inhibitors. Patients with somatic variants in HR genes may also benefit from these therapies. In this work was analyzed the frequency of somatic variants in BRCA1, BRCA2, and TP53 in an EOC cohort of Brazilian patients, estimating the proportion of variants in tumoral tissue and their association with progression-free survival and overall survival. METHODS The study was conducted with paired blood/tumor samples from 56 patients. Germline and tumoral sequences of BRCA1, BRCA2, and TP53 were obtained by massive parallel sequencing. The HaplotypeCaller method was used for calling germline variants, and somatic variants were called with Mutect2. RESULTS A total of 26 germline variants were found, and seven patients presented germline pathogenic or likely pathogenic variants in BRCA1 or BRCA2. The analysis of tumoral tissue identified 52 somatic variants in 41 patients, being 43 somatic variants affecting or likely affecting protein functionality. Survival analyses showed that tumor staging was associated with overall survival (OS), while the presence of somatic mutation in TP53 was not associated with OS or progression-free survival. CONCLUSION Frequency of pathogenic or likely pathogenic germline variants in BRCA1 and BRCA2 (12.5%) was lower in comparison with other studies. TP53 was the most altered gene in tumors, with 62.5% presenting likely non-functional or non-functional somatic variants, while eight 14.2% presented likely non-functional or non-functional somatic variants in BRCA1 or BRCA2.
Collapse
Affiliation(s)
| | | | - Bruna Palma Matta
- Tumoral Genetics and Virology ProgramInstituto Nacional de CâncerRio de JaneiroBrazil
- Present address:
Hospital BP ‐ A Beneficência Portuguesa de São PauloSão PauloBrazil
| | | | | | - Anke Bergmann
- Clinical EpidemiologyInstituto Nacional de CâncerRio de JaneiroBrazil
| | | | - Mariana Boroni
- Bioinformatics and Computational Biology LaboratoryInstituto Nacional de CâncerRio de JaneiroBrazil
| | | | | |
Collapse
|
47
|
Tuval A, Strandgren C, Heldin A, Palomar-Siles M, Wiman KG. Pharmacological reactivation of p53 in the era of precision anticancer medicine. Nat Rev Clin Oncol 2024; 21:106-120. [PMID: 38102383 DOI: 10.1038/s41571-023-00842-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2023] [Indexed: 12/17/2023]
Abstract
p53, which is encoded by the most frequently mutated gene in cancer, TP53, is an attractive target for novel cancer therapies. Despite major challenges associated with this approach, several compounds that either augment the activity of wild-type p53 or restore all, or some, of the wild-type functions to p53 mutants are currently being explored. In wild-type TP53 cancer cells, p53 function is often abrogated by overexpression of the negative regulator MDM2, and agents that disrupt p53-MDM2 binding can trigger a robust p53 response, albeit potentially with induction of p53 activity in non-malignant cells. In TP53-mutant cancer cells, compounds that promote the refolding of missense mutant p53 or the translational readthrough of nonsense mutant TP53 might elicit potent cell death. Some of these compounds have been, or are being, tested in clinical trials involving patients with various types of cancer. Nonetheless, no p53-targeting drug has so far been approved for clinical use. Advances in our understanding of p53 biology provide some clues as to the underlying reasons for the variable clinical activity of p53-restoring therapies seen thus far. In this Review, we discuss the intricate interactions between p53 and its cellular and microenvironmental contexts and factors that can influence p53's activity. We also propose several strategies for improving the clinical efficacy of these agents through the complex perspective of p53 functionality.
Collapse
Affiliation(s)
- Amos Tuval
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm, Sweden
| | | | - Angelos Heldin
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm, Sweden
| | | | - Klas G Wiman
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm, Sweden.
| |
Collapse
|
48
|
Attardi LD, Boutelle AM. Targeting p53 gain-of-function activity in cancer therapy: a cautionary tale. Cell Death Differ 2024; 31:133-135. [PMID: 38151545 PMCID: PMC10850540 DOI: 10.1038/s41418-023-01253-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/08/2023] [Accepted: 12/15/2023] [Indexed: 12/29/2023] Open
Affiliation(s)
- Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Anthony M Boutelle
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
49
|
Triantafyllidis CP, Barberis A, Hartley F, Cuervo AM, Gjerga E, Charlton P, van Bijsterveldt L, Rodriguez JS, Buffa FM. A machine learning and directed network optimization approach to uncover TP53 regulatory patterns. iScience 2023; 26:108291. [PMID: 38047081 PMCID: PMC10692668 DOI: 10.1016/j.isci.2023.108291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/21/2023] [Accepted: 10/18/2023] [Indexed: 12/05/2023] Open
Abstract
TP53, the Guardian of the Genome, is the most frequently mutated gene in human cancers and the functional characterization of its regulation is fundamental. To address this we employ two strategies: machine learning to predict the mutation status of TP53 from transcriptomic data, and directed regulatory networks to reconstruct the effect of mutations on the transcipt levels of TP53 targets. Using data from established databases (Cancer Cell Line Encyclopedia, The Cancer Genome Atlas), machine learning could predict the mutation status, but not resolve different mutations. On the contrary, directed network optimization allowed to infer the TP53 regulatory profile across: (1) mutations, (2) irradiation in lung cancer, and (3) hypoxia in breast cancer, and we could observe differential regulatory profiles dictated by (1) mutation type, (2) deleterious consequences of the mutation, (3) known hotspots, (4) protein changes, (5) stress condition (irradiation/hypoxia). This is an important first step toward using regulatory networks for the characterization of the functional consequences of mutations, and could be extended to other perturbations, with implications for drug design and precision medicine.
Collapse
Affiliation(s)
- Charalampos P. Triantafyllidis
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, UK
- Department of Epidemiology & Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Alessandro Barberis
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Fiona Hartley
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Ana Miar Cuervo
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Enio Gjerga
- Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany
| | - Philip Charlton
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, UK
| | | | - Julio Saez Rodriguez
- Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany
| | - Francesca M. Buffa
- Department of Oncology, Medical Sciences Division, University of Oxford, Oxford, UK
- Department of Computing Sciences, BIDSA, Bocconi University, Milan, Italy
| |
Collapse
|
50
|
Notin P, Kollasch AW, Ritter D, van Niekerk L, Paul S, Spinner H, Rollins N, Shaw A, Weitzman R, Frazer J, Dias M, Franceschi D, Orenbuch R, Gal Y, Marks DS. ProteinGym: Large-Scale Benchmarks for Protein Design and Fitness Prediction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.07.570727. [PMID: 38106144 PMCID: PMC10723403 DOI: 10.1101/2023.12.07.570727] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Predicting the effects of mutations in proteins is critical to many applications, from understanding genetic disease to designing novel proteins that can address our most pressing challenges in climate, agriculture and healthcare. Despite a surge in machine learning-based protein models to tackle these questions, an assessment of their respective benefits is challenging due to the use of distinct, often contrived, experimental datasets, and the variable performance of models across different protein families. Addressing these challenges requires scale. To that end we introduce ProteinGym, a large-scale and holistic set of benchmarks specifically designed for protein fitness prediction and design. It encompasses both a broad collection of over 250 standardized deep mutational scanning assays, spanning millions of mutated sequences, as well as curated clinical datasets providing high-quality expert annotations about mutation effects. We devise a robust evaluation framework that combines metrics for both fitness prediction and design, factors in known limitations of the underlying experimental methods, and covers both zero-shot and supervised settings. We report the performance of a diverse set of over 70 high-performing models from various subfields (eg., alignment-based, inverse folding) into a unified benchmark suite. We open source the corresponding codebase, datasets, MSAs, structures, model predictions and develop a user-friendly website that facilitates data access and analysis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ada Shaw
- Applied Mathematics, Harvard University
| | | | | | - Mafalda Dias
- Centre for Genomic Regulation, Universitat Pompeu Fabra
| | | | | | - Yarin Gal
- Computer Science, University of Oxford
| | | |
Collapse
|