1
|
Singh A, Malhotra L, Mishra A, Kundral S, Tiwari PK, Kumar S, Gururao H, Kaur P, Ethayathulla AS. The R337C mutation in the p53 oligomerization domain affects the regulatory domain and its ability to bind response elements: Evidence based on structural and biophysical studies. Arch Biochem Biophys 2025; 768:110381. [PMID: 40064360 DOI: 10.1016/j.abb.2025.110381] [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: 10/01/2024] [Revised: 02/07/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
The homotetrameric form of p53 is critical for performing essential functions like maintaining genomic stability and preventing uncontrolled cell proliferation. In part, these crucial functions are mediated by the p53 C-terminal region (CTR) containing the tetramerization/oligomerization domain (TD/OD) and regulatory domain (RD), responsible for maintaining the protein's oligomeric state and regulating its function. Mutations in the tetramerization domain reduce the transactivation potential and alter the transactivation specificity of p53. This study investigates the effect of high-frequency tetramerization missense mutation p53R337C on protein stability, oligomeric state, and its ability to bind the DNA response elements. For the first time using CD and FTIR spectroscopy, we have shown that the p53 regulatory domain (residues 363-393) and oligomerization domain (residues 327-355) possess a characteristic alpha helix secondary structure, which is enhanced upon binding to DNA, implicating stabilization of the domain. The mutation R337C in the OD impacts the secondary and tertiary structure of p53 CTR, leading to the loss of secondary structure and the formation of unstable tetramers, as shown by CD and DSC thermal studies. Surprisingly, the secondary structure of mutant p53 CTR partially stabilized upon binding to the DNA sequence. Our data suggests that the unstable p53R337C tetramer exhibits weaker binding to the DNA promoter sequence with decreased transcription activity, consistent with previous cell-based assays. Our study conclude that the loss of salt-bridge interactions between Arg337 and Asp352 in the intra-dimer of p53 leads to the formation of unstable tetramers, and the DNA-binding ability of the regulatory domain.
Collapse
Affiliation(s)
- Alankrita Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Lakshay Malhotra
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India; Department of Biochemistry, Sri Venkateswara College, University of Delhi, New Delhi, 110021, India
| | - Abhay Mishra
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Simran Kundral
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Pawan Kumar Tiwari
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Hariprasad Gururao
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | | |
Collapse
|
2
|
Richman LP, Waller BF, Lovitch SB, Jambhekar A. Detection of TP53 mutations by immunohistochemistry in acute myeloid leukemia varies with interpreter expertise and mutation status. Am J Clin Pathol 2025:aqaf047. [PMID: 40414698 DOI: 10.1093/ajcp/aqaf047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 04/14/2025] [Indexed: 05/27/2025] Open
Abstract
OBJECTIVE TP53 mutations, including missense and inactivating (frameshift, splice site, and nonsense) mutations, occur in approximately 10% of myeloid neoplasms and confer adverse outcomes. Classification of myeloid neoplasms by World Health Organization and International Consensus Classification standards recognizes the importance of early detection of TP53 mutations. p53 immunohistochemistry (IHC) is a widely accessible method used to detect mutations; however, previous studies have demonstrated variable accuracy, especially for inactivating TP53 mutations. Recently, sequencing using targeted panels has seen increased use. Although highly accurate, sequencing is resource intensive and not universally available. METHODS Using 134 bone marrow samples from patients with acute myeloid leukemia evaluated for TP53 mutation by sequencing, we assessed the concordance of p53 IHC with sequencing as well as the interrater-reliability for IHC intensity and percent positivity. RESULTS Consistent with previous studies, we found that p53 IHC was strongly specific and modestly sensitive for missense mutations and that overall performance improved with dedicated hematopathology training. We also found that IHC performed poorly for inactivating mutations and was even variable between cases harboring identical amino acid changes. Low predicted transcriptional activity of p53 missense proteins correlated with a mutant pattern of IHC staining. The status of the second allele and variant allele frequency also affected the accuracy of p53 IHC as a surrogate for TP53 allele status. CONCLUSION Cases of acute myeloid leukemia with TP53 mutations predicted to have low transcriptional activity showed reduced overall survival. Our results demonstrate limited practical utility of p53 IHC for accurate evaluation of TP53 mutation status because of multifactorial confounders.
Collapse
Affiliation(s)
- Lee P Richman
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, United States
| | - Brianna F Waller
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, United States
| | - Scott B Lovitch
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, United States
| | - Ashwini Jambhekar
- Department of Systems Biology, Harvard Medical School, Boston, MA, United States
- Ludwig Center at Harvard, Boston, MA, United States
| |
Collapse
|
3
|
Achatz MI, Villani A, Bertuch AA, Bougeard G, Chang VY, Doria AS, Gallinger B, Godley LA, Greer MLC, Kamihara J, Khincha PP, Kohlmann WK, Kratz CP, MacFarland SP, Maese LD, Maxwell KN, Mitchell SG, Nakano Y, Pfister SM, Wasserman JD, Woodward ER, Garber JE, Malkin D. Update on Cancer Screening Recommendations for Individuals with Li-Fraumeni Syndrome. Clin Cancer Res 2025; 31:1831-1840. [PMID: 40072304 DOI: 10.1158/1078-0432.ccr-24-3301] [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: 10/04/2024] [Revised: 01/05/2025] [Accepted: 03/11/2025] [Indexed: 05/16/2025]
Abstract
Li-Fraumeni syndrome (LFS) is an autosomal dominant cancer predisposition condition characterized by a high lifetime risk for a wide spectrum of malignancies associated with germline pathogenic/likely pathogenic variants in the TP53 tumor suppressor gene. Secondary malignant neoplasms are particularly common. Early cancer detection through surveillance enables early intervention and leads to improved clinical outcomes with reduced tumor-related mortality and treatment-related morbidity. Since the 2017 publication of LFS tumor surveillance guidelines from the inaugural American Association for Cancer Research Childhood Cancer Predisposition Workshop, understanding the genotype-phenotype relationships in LFS has evolved, and adaptations of the guidelines have been implemented in institutions worldwide. The "Toronto Protocol" remains the current standard for lifelong surveillance; however, as outlined in this perspective, modifications should be considered about the use of certain modalities to target organs in an age-dependent manner. The Working Group's recommendations have also been extended to include a more detailed outline for surveillance in the adult TP53 pathogenic/likely pathogenic variant carrier population, based on the recognition that early education of both practitioners and patients on what to expect after the transition from childhood/adolescence to young adulthood is important in preparing them for changes in surveillance strategies. In this perspective, we provide an up-to-date clinical overview of LFS and present our updated consensus tumor surveillance recommendations from the 2023 American Association for Cancer Research Childhood Cancer Predisposition Workshop.
Collapse
Affiliation(s)
| | - Anita Villani
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | | | - Gaëlle Bougeard
- Department of Genetics, Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Rouen, France
| | - Vivian Y Chang
- Pediatric Hematology-Oncology, University of California, Los Angeles, Los Angeles, California
| | - Andrea S Doria
- Department of Diagnostic and Interventional Radiology, The Hospital for Sick Children, University of Toronto, Toronto, Canada
- Department of Medical Imaging, University of Toronto, Toronto, Canada
| | - Bailey Gallinger
- Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, The University of Toronto, Toronto, Canada
| | - Lucy A Godley
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Mary-Louise C Greer
- Department of Diagnostic and Interventional Radiology, The Hospital for Sick Children, University of Toronto, Toronto, Canada
- Department of Medical Imaging, University of Toronto, Toronto, Canada
| | | | - Payal P Khincha
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Wendy K Kohlmann
- VA Medical Center, National TeleOncology Program, Clinical Cancer Genetics Service, Durham, North Carolina
- University of Utah Huntsman Cancer Institute, Salt Lake City, Utah
| | - Christian P Kratz
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Suzanne P MacFarland
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Luke D Maese
- University of Utah-Huntsman Cancer Institute, Primary Children's Hospital, Salt Lake City, Utah
| | - Kara N Maxwell
- Department of Medicine-Hematology/Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Medicine Service, Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania
| | - Sarah G Mitchell
- Department of Pediatric Hematology/Oncology, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia
| | - Yoshiko Nakano
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Stefan M Pfister
- Division Pediatric Neurooncology, Hopp Children's Cancer Center Heidelberg (KiTZ), German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg University Hospital and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Jonathan D Wasserman
- Division of Endocrinology, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Emma R Woodward
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester Centre for Genomic Medicine, Manchester, United Kingdom
| | - Judy E Garber
- Division of Cancer Genetics and Prevention, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David Malkin
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| |
Collapse
|
4
|
Holoubek A, Strachotová D, Wolfová K, Otevřelova P, Belejová S, Röselová P, Benda A, Brodská B, Herman P. Correlation of p53 oligomeric status and its subcellular localization in the presence of the AML-associated NPM mutant. PLoS One 2025; 20:e0322096. [PMID: 40334261 PMCID: PMC12058200 DOI: 10.1371/journal.pone.0322096] [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: 11/01/2024] [Accepted: 03/17/2025] [Indexed: 05/09/2025] Open
Abstract
Tumor suppressor p53 is a key player in the cell response to DNA damage that suffers by frequent inactivating aberrations. Some of them disturb p53 oligomerization and influence cell decision between proliferation, growth arrest and apoptosis. Active p53 resides mostly in the nucleus, degradation occurs in the cytoplasm. Acute myeloid leukemia (AML)-related mutation of NPM (NPMmut) induces massive mislocalization of p53 to the cytoplasm, which might be related to leukemia initiation. Since both proteins interact and execute their function as oligomers, we investigated the role of perturbed p53 oligomerization in the p53 mislocalization process in live cells by FLIM (fluorescence lifetime imaging microscopy), fluorescence anisotropy imaging (FAIM), fluorescence cross-correlation spectroscopy (FCCS) and immunochemical methods. On a set of fluorescently labeled p53 variants, monomeric R337G and L344P, dimeric L344A, and multimeric D352G and A353S, we correlated their cellular localization, oligomerization and interaction with NPMmut. Interplay between nuclear export signal (NES) and nuclear localization signal (NLS) of p53 was investigated as well. While NLS was found critical for the nuclear p53 localization, NES plays less significant role. We observed cytoplasmic translocation only for multimeric A353S variant with sufficient stability and strong interaction with NPMmut. Less stable multimer D352G and L344A dimer were not translocated, monomeric p53 variants always resided in the nucleus independently of the presence of NPMmut and NES intactness. Oligomeric state of NPMmut is not required for p53 translocation, which happens also in the presence of the nonoligomerizing NPMmut variant. The prominent structural and functional role of the R337 residue is shown.
Collapse
Affiliation(s)
- Aleš Holoubek
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Dita Strachotová
- Faculty of Mathematics and Physics, Institute of Physics, Charles University, Prague, Czech Republic
| | - Kateřina Wolfová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Petra Otevřelova
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Sára Belejová
- Faculty of Mathematics and Physics, Institute of Physics, Charles University, Prague, Czech Republic
| | - Pavla Röselová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Aleš Benda
- Imaging Methods Core Facility at BIOCEV, Faculty of Science, Charles University, Vestec, Czech Republic
| | - Barbora Brodská
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Petr Herman
- Faculty of Mathematics and Physics, Institute of Physics, Charles University, Prague, Czech Republic
| |
Collapse
|
5
|
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
|
6
|
Rotenberg N, Fortuno C, Varga MJ, Chamberlin AC, Ramadane-Morchadi L, Feng BJ, de la Hoya M, Richardson ME, Spurdle AB. Integration of protein stability and AlphaMissense scores improves bioinformatic impact prediction for p53 missense and in-frame amino acid deletion variants. Am J Hum Genet 2025; 112:1003-1014. [PMID: 40233742 DOI: 10.1016/j.ajhg.2025.01.012] [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: 05/28/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 04/17/2025] Open
Abstract
The clinical classification of germline missense variants and single-amino-acid deletions is challenging. The BayesDel and Align-GVGD bioinformatic prediction tools currently used for ClinGen TP53 variant curation expert panel (VCEP) classification do not directly capture changes in protein folding stability, measured using computed destabilization energies (ΔΔG scores). The AlphaMissense tool recently developed by Google DeepMind to predict pathogenicity for all human proteome missense variants is trained in part using AlphaFold2 architecture. Our study investigated whether protein folding stability and/or AlphaMissense scores could improve impact prediction for p53 missense and single-amino-acid deletion variants. ΔΔG scores were calculated for missense variants using FoldX and for single-amino-acid deletions using an AlphaFold2/RosettaRelax protocol. Residue surface exposure was categorized using relative solvent accessibility (RSA) measures. The predictive values of ΔΔG scores, AlphaMissense, BayesDel, and Align-GVGD were examined using Boruta and binary logistic regression based on functionally defined reference sets. The likelihood ratio (LR) toward pathogenicity was estimated and used to refine optimal categories for predicting variant pathogenicity for different RSA values. We showed that current VCEP predictive approaches for missense variants were improved by integrating ΔΔG scores ≥2.5 kcal/mol for partially buried and buried residues, but better performance was achieved using AlphaMissense with ΔΔG and RSA. For deletion variants, ΔΔG scores ≥4.8 Rosetta energy unit (REU) in buried residues outperformed currently used predictive approaches. Future TP53 VCEP specifications for p53 missense impact prediction may consider AlphaMissense, ΔΔG score, and RSA combined for substitution variants and ΔΔG score alone for deletion variants.
Collapse
Affiliation(s)
- Nitsan Rotenberg
- Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia; University of Queensland, Brisbane, QLD, Australia
| | - Cristina Fortuno
- Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia
| | | | | | - Lobna Ramadane-Morchadi
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | - Bing-Jian Feng
- University of Utah Department of Dermatology, Salt Lake City, UT, USA; University of Utah Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Miguel de la Hoya
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, IdISSC (Instituto de Investigación Sanitaria del Hospital Clínico San Carlos), 28040 Madrid, Spain
| | | | - Amanda B Spurdle
- Molecular Cancer Epidemiology Laboratory, QIMR Berghofer MRI, Herston, QLD 4006, Australia; University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
7
|
Osbourne R, Thayer KM. Structural and mechanistic diversity in p53-mediated regulation of organismal longevity across taxonomical orders. PLoS Comput Biol 2025; 21:e1012382. [PMID: 40315252 PMCID: PMC12068700 DOI: 10.1371/journal.pcbi.1012382] [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: 08/04/2024] [Revised: 05/12/2025] [Accepted: 04/21/2025] [Indexed: 05/04/2025] Open
Abstract
The link between p53 tumor suppressive functions and organismal lifespan is multifaceted. Its DNA-repair mechanism is longevity-enhancing while its role in cellular senescence pathways induces pro-aging phenotypes. To understand how p53 may regulate organismal lifespan, cross-species genotype-phenotype (GP) studies of the p53 DNA-binding domain (DBD) have been used to assess the correlation of amino acid changes to lifespan. Amino acid changes in non-DNA-binding regions such as the transactivation (TAD), proline-rich (PRD), regulatory (REG), and tetramerization (TET) are largely unexplored. In addition, existing GP correlation tools such as SigniSite do not account for phylogenetic relationships between aligned sequences in correlating genotypic differences to phenotypes such as lifespan. To identify phylogenetically significant, longevity-correlated residues in full-length p53 alignments, we developed a Python- and R-based workflow, Relative Evolutionary Scoring (RES). While RES-predicted longevity-associated residues (RPLARs) are concentrated primarily in the DBD, the PRD, TET, and REG domains also house RPLARs. While yeast functional assay enrichment reveals that RPLARs may be dispensable for p53-mediated transactivation, PEPPI and Rosetta-based protein-protein interaction prediction suggests a role for RPLARs in p53 stability and interaction interfaces of tumor suppressive protein-protein complexes. With experimental validation of the RPLARs' roles in p53 stability, transactivation, and involvement in senescence-regulatory pathways, we can gain crucial insights into mechanisms underlying dysregulated tumor suppression and accelerated aging.
Collapse
Affiliation(s)
- Romani Osbourne
- Department of Molecular Biology & Biochemistry, Wesleyan University, Middletown, Connecticut, United States of America
- College of Integrative Sciences, Wesleyan University, Middletown, Connecticut, United States of America
| | - Kelly M. Thayer
- College of Integrative Sciences, Wesleyan University, Middletown, Connecticut, United States of America
| |
Collapse
|
8
|
Li L, Muftuoglu M, Ayoub E, Lv J, Basyal M, Bidikian A, Zhao R, Prashant M, Mak PY, Kesharwani RK, Nishida Y, Issa GC, Varadarajan N, Carter B, Andreeff M. Somatic TP53 Mutations Drive T and NK Cell Dysfunction in AML and Can be Rescued by Reactivating Wild Type p53. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.11.25325281. [PMID: 40297420 PMCID: PMC12036377 DOI: 10.1101/2025.04.11.25325281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Therapeutic advances in immunotherapy have significantly improved outcomes in lymphomas and myelomas, yet patients with TP53-mutant acute myeloid leukemia (AML) continue to be challenged. While TP53 mutations in leukemic blasts have been extensively characterized, their incidence and impact within immune cells remain largely unexplored. Here, using single-cell multi-omics and integrated phenotypic analyses, we identify TP53 mutations in T and NK cells from AML patients. Notably, T cells harboring monoallelic TP53 mutations exhibited elevated proliferative markers yet showed reduced cytotoxic capacity and increased expression of inhibitory receptors, including PD-1, TIGIT, and TIM-3. To investigate the functional consequences of p53-mutant immune cells, we engineered CAR-T cells carrying clinically relevant p53 mutations (Y220C and R175H). These mutant p53 CAR-T cells exhibited a pronounced exhaustion phenotype, with diminished cytokine secretion and impaired tumor cytolysis both in vitro and in PDX mouse models. Crucially, restoring mutant p53 to a wild-type conformation using a targeted small-molecule reactivator rescued CAR-T functionality, reduced exhaustion marker expression, and prolonged survival in AML PDX mouse models, revealing a direct mechanistic link between TP53 mutations in T cells and therapeutic resistance. Our findings establish TP53-mutant T cells as a previously unrecognized driver of immune escape in AML, highlighting the importance of immune-cell genotyping and p53 reactivation strategies. By demonstrating that mutant p53 can be selectively corrected to restore T-cell function, this study opens new avenues for immunotherapeutic intervention in TP53-mutant AML.
Collapse
|
9
|
Hylton-McComas HM, Cordes A, Floros KV, Faber A, Drapkin BJ, Miles WO. Myc family proteins: Molecular drivers of tumorigenesis and resistance in neuroendocrine tumors. Biochim Biophys Acta Rev Cancer 2025; 1880:189332. [PMID: 40280500 DOI: 10.1016/j.bbcan.2025.189332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Neuroendocrine cancers are a diverse and poorly understood collection of malignancies derived from neuroendocrine cells throughout the body. These cancers uniquely exhibit properties of both the nervous and endocrine systems. Only a limited number of genetic driver mutations have been identified in neuroendocrine cancers, however the mechanisms of how these genetic aberrations alter tumor biology remain elusive. Recent studies have implicated the MYC family of transcription factors as important oncogenic factors in neuroendocrine tumors. We take a systematic approach to understand the roles of the MYC family (c-MYC, n-MYC, l-MYC) in the tumorigenesis of neuroendocrine cancers of the lung, GI tract, pancreas, kidney, prostate, pediatric neuroblastoma, and adrenal glands. Reflecting the complexity of neuroendocrine cancers, we highlight the roles of the MYC family in deregulating the cell cycle and transcriptional networks, invoking cellular plasticity, affecting proliferation capacity, aiding in chromatin remodeling, angiogenesis, metabolic changes, and resistance mechanisms. Depicting the diversity of neuroendocrine cancers, we suggest new approaches in understanding the underlying tumorigenic processes of neuroendocrine cancers from the perspective of MYC.
Collapse
Affiliation(s)
- Hannah M Hylton-McComas
- Department of Cancer Biology and Genetics, The Ohio State University, 460 West 12(th) Avenue, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, The Ohio State University, 460 West 12(th) Avenue, Columbus, OH 43210, USA
| | - Alyssa Cordes
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Konstantinos V Floros
- VCU Philips Institute, Virginia Commonwealth University School of Dentistry and Massey Comprehensive Cancer Center, Richmond, VA 23298, USA; Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Anthony Faber
- VCU Philips Institute, Virginia Commonwealth University School of Dentistry and Massey Comprehensive Cancer Center, Richmond, VA 23298, USA; Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Benjamin J Drapkin
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wayne O Miles
- Department of Cancer Biology and Genetics, The Ohio State University, 460 West 12(th) Avenue, Columbus, OH 43210, USA; The Ohio State University Comprehensive Cancer Center, The Ohio State University, 460 West 12(th) Avenue, Columbus, OH 43210, USA.
| |
Collapse
|
10
|
Silva JL, de Andrade GC, Petronilho EC, de Sousa GDS, Mota MF, Quarti J, Guedes-da-Silva FH, Ferretti GDS, Rangel LP, Vieira TCRG, Marques MA, de Oliveira GAP. Phase Separation and Prion-Like Aggregation of p53 Family Tumor Suppressors: From Protein Evolution to Cancer Treatment. J Neurochem 2025; 169:e70055. [PMID: 40178008 DOI: 10.1111/jnc.70055] [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: 01/20/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/05/2025]
Abstract
Biomolecular condensates, formed through phase separation (PS), are essential in various physiological processes, but they can also transition into amyloid-like structures, contributing to diseases like cancer and neurodegenerative disorders. This review centers on the tumor suppressor protein p53 and its paralogs, p63 and p73, which play significant roles in cancer biology. Mutations in the TP53 gene, present in over half of all malignant tumors, disrupt the function of p53 and contribute to cancer progression. Mutant p53 not only misfolds but also forms biomolecular condensates and amyloid-like aggregates, like the toxic amyloids seen in neurodegenerative diseases. These amyloid-like structures, characteristic of mutant p53, might be associated with its gain of function (GoF) in cancer. Recent in vitro and in cell studies demonstrate that mutant p53 can exert a prion-like effect on its paralogs, p63 and p73, which typically do not form amyloids under physiological conditions. Heparin inhibits the prion-like effect of mutant p53 on p63 and p73. These findings underscore the critical role of mutant p53 in promoting the aggregation of p63 and p73, and likely of other transcription factors, suggesting new therapeutic targets. The amyloid-like aggregation of mutant p53 is an excellent candidate target for cancer, as evidenced by recent studies. By understanding the phase transitions and amyloid formation of mutant p53, innovative diagnostic and treatment strategies have been explored to reveal and disrupt these processes, offering hope for improved cancer therapies.
Collapse
Affiliation(s)
- Jerson L Silva
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Center for Structural Biology and Bioimaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Guilherme C de Andrade
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elaine C Petronilho
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gileno Dos S de Sousa
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michelle F Mota
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julia Quarti
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Francisca H Guedes-da-Silva
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giulia D S Ferretti
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana P Rangel
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tuane C R G Vieira
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mayra A Marques
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guilherme A P de Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis, National Institute of Science and Technology for Structural Biology and Bioimaging, National Center of Nuclear Magnetic Resonance Jiri Jonas, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
Huan X, Li J, Chu Z, Zhang H, Cheng L, Lun P, Du X, Chen X, Jiao Q, Jiang H. Dysregulation of Iron Homeostasis Mediated by FTH Increases Ferroptosis Sensitivity in TP53-Mutant Glioblastoma. Neurosci Bull 2025; 41:569-582. [PMID: 39666195 PMCID: PMC11978602 DOI: 10.1007/s12264-024-01322-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/19/2024] [Indexed: 12/13/2024] Open
Abstract
Iron metabolism is a critical factor in tumorigenesis and development. Although TP53 mutations are prevalent in glioblastoma (GBM), the mechanisms by which TP53 regulates iron metabolism remain elusive. We reveal an imbalance iron homeostasis in GBM via TCGA database analysis. TP53 mutations disrupted iron homeostasis in GBM, characterized by elevated total iron levels and reduced ferritin (FTH). The gain-of-function effect triggered by TP53 mutations upregulates itchy E3 ubiquitin-protein ligase (ITCH) protein expression in astrocytes, leading to FTH degradation and an increase in free iron levels. TP53-mut astrocytes were more tolerant to the high iron environment induced by exogenous ferric ammonium citrate (FAC), but the increase in intracellular free iron made them more sensitive to Erastin-induced ferroptosis. Interestingly, we found that Erastin combined with FAC treatment significantly increased ferroptosis. These findings provide new insights for drug development and therapeutic modalities for GBM patients with TP53 mutations from iron metabolism perspectives.
Collapse
Affiliation(s)
- Xuejie Huan
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Jiangang Li
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Zhaobin Chu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Hongliang Zhang
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Lei Cheng
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Peng Lun
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, China.
- Qingdao Key Laboratory of Neurorehabilitation, University of Health and Rehabilitation Sciences, Qingdao, 266113, China.
| |
Collapse
|
12
|
Ruzzo A, Graziano F, Palladino S, Fischer NW, Catalano V, Giordani P, Malkin D, Tamburrano T, Patriti A, Petrelli F, Sarti D, Chiari R. Clinical impact of TP53 functional mutations in patients with metastatic colorectal cancer treated with bevacizumab and chemotherapy. Oncologist 2025; 30:oyae277. [PMID: 39436921 PMCID: PMC11954512 DOI: 10.1093/oncolo/oyae277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Clinical and experimental studies indicate that the tumor protein p53 (TP53) gene loss of function due to missense mutations (MMs) may confer sensitivity to anti-angiogenics. This effect seems to be linked to cross-talk mechanisms among TP53, vascular endothelial growth factor (VEGF), and VEGF receptors. We investigated whether specific TP53 MMs are associated with clinical outcomes of patients with metastatic colorectal cancer (mCRC) treated with first-line chemotherapy plus Bevacizumab. The study focused on KRAS-mutated, liver-only mCRC cases as a homogeneous subgroup that may represent a relevant setting for exploring this association. MATERIALS AND METHODS MMs were identified on primary tumors. MMs were classified by mutant-specific residual transcriptional activity scores (TP53RTAS) as transcriptionally inactive (TP53inactive = TP53RTAS 0%) or active (TP53active = TP53RTAS ≥ 1%) and used for stratifying patients in progression-free survival (PFS), response rate, and overall survival (OS) analyses. RESULTS The study population consisted of 62 patients. MMs were found in 39 cases (62%) with 16 having TP53inactive and 23 TP53active MMs. Patients with TP53inactive MMs showed better PFS in comparison with the remaining groups (wild-type and TP53active). This effect was retained in the multivariate model. A similar clinical impact was observed in the OS analysis. There was a significant difference in the overall response rate and rate of post-treatment resection of liver metastases between the TP53inactive and the wild-type or TP53active MMs cases. CONCLUSIONS Specific TP53 MMs may identify sub-groups of patients who benefit from Bevacizumab-based systemic therapy and these findings could lead to novel tailored treatment strategies in this setting.
Collapse
Affiliation(s)
- Annamaria Ruzzo
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy
| | | | - Silvia Palladino
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy
| | - Nicholas W Fischer
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | | | | | - David Malkin
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, M5G 0A4, Canada
- Division of Hematology-Oncology, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
- Department of Pediatrics, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | | | | | | | | | - Rita Chiari
- Medical Oncology Unit, AST1, 61121 Pesaro, Italy
| |
Collapse
|
13
|
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
|
14
|
Zhao T, Iafrate AJ, Wu CL. Primary urethral adenocarcinoma harbors recurrent KRAS and EGFR alterations. Hum Pathol 2025; 157:105771. [PMID: 40194726 DOI: 10.1016/j.humpath.2025.105771] [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: 02/07/2025] [Revised: 04/04/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
Primary urethral adenocarcinoma is an extremely rare malignancy with an unclear pathogenesis. Previously, we reported 4 brachytherapy-associated (BA) urethral mucinous adenocarcinomas that developed following treatment for prostate cancer. In the present study, we report one additional BA and 3 radiation-independent (RI) urethral adenocarcinomas. The aim of this study is to explore the molecular alterations and to compare the clinicopathologic features. RNA sequencing was performed on 5 tumors, and a next-generation sequencing (NGS)-based fusion assay was used to identify gene fusions in 6 tumors. Additionally, NGS-based targeted genomic DNA sequencing was employed to analyze one metastatic BA tumor and one metastatic RI tumor. The 8 patients had a mean age of 67 (range: 37-87) years, with one being female in the RI cohort. Cystoscopy revealed the following urethral findings: a papillary lesion (4/7), mass causing obstruction (1/7) and irregular friable tissue (2/7). Seven patients underwent urethrectomy with cystectomy/prostatectomy/hysterectomy. The mean tumor size was 3.4 cm (range: 1.5-6.5). Adenocarcinoma in situ was noted in 5 tumors. All 5 BA tumors originated from the prostatic urethra, with 4 showing mucinous morphology and one enteric morphology, and showed moderate to poor differentiation and tumor stages of pT2 (2/4), pT3 (1/3) and pT4 (1/4). Two patients developed metastasis, one at 3.3 and one at 4.2 years after diagnosis, and all patients were alive at a median follow-up of 4.5 (range: 2-14) years. In contrast, 3 RI tumors arose from bulbar, prostatic, or female mid/distal urethra, presenting as enteric, mucinous, and not otherwise specified (NOS) subtypes, with well to moderate differentiation and a tumor stage of pT4 (2/2). Two died of the disease, while one was alive without disease at a median follow-up of 4 (range: 2.2-14.5) years. All tumors were diffusely positive for CK20, CDX2 (7/7), and AMACR (3/3), and lacked nuclear β-catenin expression (5/5). Most expressed CK7 (5/7). KRAS mutations (p.Gly12Val and p.Gly13Asp) were observed in one BA mucinous tumor and one RI NOS tumor with the p.Gly13Asp mutation also detected in the metastatic RI tumor. The EGFR p.Ser784Phe mutation was detected in one RI enteric tumor. TP53 p.Val172Phe, CDKN2A p.Leu32_Leu37del, and amplifications of EGFR and MDM2, were identified in a metastatic BA enteric tumor. No fusion transcripts were identified. In conclusion, urethral adenocarcinoma harbors recurrent KRAS and EGFR alterations, independent of prior radiotherapy. RI tumors appear to be associated with a worse prognosis compared to BA tumors.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Chin-Lee Wu
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
15
|
Rahmé R, Resnick-Silverman L, Anguiano V, Campbell MJ, Fenaux P, Manfredi JJ. Mutant p53 regulates a distinct gene set by a mode of genome occupancy that is shared with wild type. EMBO Rep 2025; 26:1315-1343. [PMID: 39875582 PMCID: PMC11893899 DOI: 10.1038/s44319-025-00375-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/05/2025] [Accepted: 01/14/2025] [Indexed: 01/30/2025] Open
Abstract
To directly examine the interplay between mutant p53 or Mdm2 and wild type p53 in gene occupancy and expression, an integrated RNA-seq and ChIP-seq analysis was performed in vivo using isogenically matched mouse strains. Response to radiation was used as an endpoint to place findings in a biologically relevant context. Unexpectedly, mutant p53 and Mdm2 only inhibit a subset of wild type p53-mediated gene expression. In contrast to a dominant-negative or inhibitory role, the presence of either mutant p53 or Mdm2 actually enhances the occupancy of wild type p53 on many canonical targets. The C-terminal 19 amino acids of wild type p53 suppress the p53 response allowing for survival at sublethal doses of radiation. Further, the p53 mutant 172H is shown to occupy genes and regulate their expression via non-canonical means that are shared with wild type p53. This results in the heterozygous 172H/+ genotype having an expanded transcriptome compared to wild type p53 + /+.
Collapse
Affiliation(s)
- Ramy Rahmé
- Department of Oncological Sciences and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Institut de Recherche Saint Louis (IRSL), INSERM U1131, Université de Paris, Paris, France
- Ecole Doctorale Hématologie-Oncogenèse-Biothérapies, Université de Paris, Paris, France
| | - Lois Resnick-Silverman
- Department of Oncological Sciences and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Vincent Anguiano
- Department of Oncological Sciences and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Pierre Fenaux
- Institut de Recherche Saint Louis (IRSL), INSERM U1131, Université de Paris, Paris, France
- Service Hématologie Seniors, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris, Paris, France
| | - James J Manfredi
- Department of Oncological Sciences and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
16
|
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
|
17
|
Louis J, Rolain M, Levacher C, Baudry K, Pujol P, Ruminy P, Baert Desurmont S, Bou J, Bouvignies E, Coutant S, Kasper E, Lienard G, Vasseur S, Vezain M, Houdayer C, Charbonnier F, Bougeard G. Li-Fraumeni syndrome: a germline TP53 splice variant reveals a novel physiological alternative transcript. J Med Genet 2025; 62:160-168. [PMID: 39788694 DOI: 10.1136/jmg-2024-110449] [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: 10/21/2024] [Accepted: 12/22/2024] [Indexed: 01/12/2025]
Abstract
BACKGROUND Li-Fraumeni syndrome (LFS) predisposes individuals to a wide range of cancers from childhood onwards, underscoring the crucial need for accurate interpretation of germline TP53 variants for optimal clinical management of patients and families. Several unclassified variants, particularly those potentially affecting splicing, require specialised testing. One such example is the NM_000546.6:c.1101-2A>C (rs587781664) variant, located at the splice acceptor site of the last intron of TP53, identified in a female patient with breast cancer diagnosed in her 20s. METHODS To interpret this variant, which has been classified as a variant of uncertain significance (VUS), we developed specific assays including a p53 functional assay, RT-QMPSF, Splice and Expression Analyses by exon Ligation and High-Throughput Sequencing and long RT-droplet digital PCR. RESULTS We demonstrated a loss of p53 transcriptional activity, and a half reduction in TP53 mRNA expression. Additionally, we detected the use of a novel alternative last exon downstream of exon 11, which we have named exon 12. This transcript, typically detectable at low levels in most individuals, was found to be more highly expressed in the c.1101-2A>C carrier, predominantly transcribed from the mutant allele due to the disruption of the splice acceptor site in intron 10. CONCLUSION By combining these approaches, we successfully reclassified this intronic VUS as 'pathogenic', enabling appropriate genetic counselling for the patient and her family. Additionally, we identified a novel TP53 alternative transcript that is expressed in both physiological and pathological contexts, with heightened expression in the patient with LFS. This discovery provides a basis for further investigation into the role of TP53 isoforms in LFS oncogenesis.
Collapse
Affiliation(s)
- Jeanne Louis
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| | - Marion Rolain
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| | - Corentin Levacher
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| | - Karen Baudry
- CHU Montpellier, Département d'oncogénétique, F-34000, Montpellier, France
| | - Pascal Pujol
- CHU Montpellier, Département d'oncogénétique, F-34000, Montpellier, France
- Univ Montpellier et CREEC, UMR IRD 224-CNRS 5290, F-34000, Montpellier, France
| | - Philippe Ruminy
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, Centre Henri Becquerel, F-76000, Rouen, France
| | - Stéphanie Baert Desurmont
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| | - Jacqueline Bou
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| | - Emilie Bouvignies
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| | - Sophie Coutant
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| | - Edwige Kasper
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| | - Gwendoline Lienard
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| | - Stéphanie Vasseur
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| | - Myriam Vezain
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| | - Claude Houdayer
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| | - Françoise Charbonnier
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| | - Gaëlle Bougeard
- Univ Rouen Normandie, Inserm U1245, Normandie Univ, CHU Rouen, Department of Genetics, F-76000, Rouen, France
| |
Collapse
|
18
|
Danishevich A, Fedorova D, Bodunova N, Makarova M, Byakhova M, Semenova A, Galkin V, Litvinova M, Nikolaev S, Efimova I, Osinin P, Lisitsa T, Khakhina A, Shipulin G, Nasedkina T, Shumilova S, Gusev O, Bilyalov A, Shagimardanova E, Shigapova L, Nemtsova M, Sagaydak O, Woroncow M, Gadzhieva S, Khatkov I. Assessing germline TP53 mutations in cancer patients: insights into Li-Fraumeni syndrome and genetic testing guidelines. Hered Cancer Clin Pract 2025; 23:5. [PMID: 39962599 PMCID: PMC11834258 DOI: 10.1186/s13053-025-00307-w] [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: 12/06/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Germline TP53 gene variants are intricately linked to Li-Fraumeni syndrome, a rare and aggressive hereditary cancer syndrome. This study investigated the frequency and spectrum of TP53 pathogenic variants associated with Li-Fraumeni syndrome in a large cohort of mainly breast cancer patients from Russia. METHODS The study analyzed 3,455 genomic DNA samples from cancer patients using next-generation sequencing panels and whole-genome sequencing. Clinically significant TP53 variants were identified and validated using Sanger sequencing. The clinical and family history characteristics of patients with TP53 variants were analyzed. RESULTS The analysis identified 13 (0.4%) individuals with clinically significant germline TP53 variants, all of whom were females with either unilateral breast cancer or breast cancer as part of multiple primary malignant neoplasms. The average age of breast cancer manifestation was 39.9 years, with a median of 36 years. Only 38.5% of the TP53 mutation carriers met the modified Chompret criteria for TP53 testing. CONCLUSIONS The findings underscore the necessity of thorough phenotype and family history analysis in genetic counseling to effectively diagnose LFS, and emphasize the importance of identifying TP53 variant carriers for developing treatment strategies, prognosis, and monitoring, as well as for identifying high-risk family members. The study also highlights that the current guidelines fail to identify over half of the TP53 mutation carriers, suggesting the need for a more comprehensive approach to genetic testing in suspected hereditary cancer cases.
Collapse
Affiliation(s)
- Anastasiia Danishevich
- SBHI Moscow Clinical Scientific Center Named After Loginov of Moscow Healthcare Department, Moscow, 111123, Russia.
| | - Daria Fedorova
- SBHI Moscow Clinical Scientific Center Named After Loginov of Moscow Healthcare Department, Moscow, 111123, Russia
| | - Natalia Bodunova
- SBHI Moscow Clinical Scientific Center Named After Loginov of Moscow Healthcare Department, Moscow, 111123, Russia
| | - Maria Makarova
- Evogen LLC, Moscow, 115191, Russia
- Russian Scientific Center of Roentgenoradiology of the Ministry of Health of the Russian Federation, Moscow, 117997, Russia
| | - Maria Byakhova
- City Clinical Oncological Hospital No. 1 of Moscow Healthcare Department, Moscow, 117152, Russia
| | - Anna Semenova
- City Clinical Oncological Hospital No. 1 of Moscow Healthcare Department, Moscow, 117152, Russia
| | - Vsevolod Galkin
- City Clinical Oncological Hospital No. 1 of Moscow Healthcare Department, Moscow, 117152, Russia
| | - Maria Litvinova
- SBHI Moscow Clinical Scientific Center Named After Loginov of Moscow Healthcare Department, Moscow, 111123, Russia
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russian Federation (Sechenov University), Moscow, 119991, Russia
| | - Sergey Nikolaev
- SBHI Moscow Clinical Scientific Center Named After Loginov of Moscow Healthcare Department, Moscow, 111123, Russia
| | - Irina Efimova
- Medical Genetic Research Center Named After Academician N.P. Bochkov, Moscow, 115522, Russia
| | - Pavel Osinin
- SBHI Moscow Clinical Scientific Center Named After Loginov of Moscow Healthcare Department, Moscow, 111123, Russia
| | - Tatyana Lisitsa
- FSBI "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical and Biological Agency, Moscow, 119435, Russia
- FSBI "National Medical Research Center of Oncology Named After N.N. Blokhin" of the Ministry of Health of the Russian Federation, Moscow, 115522, Russia
| | - Anastasiya Khakhina
- FSBI "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical and Biological Agency, Moscow, 119435, Russia
| | - German Shipulin
- FSBI "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical and Biological Agency, Moscow, 119435, Russia
| | - Tatiana Nasedkina
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, Moscow, 119991, Russia
| | - Syuykum Shumilova
- Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, Moscow, 119991, Russia
| | - Oleg Gusev
- Life Improvement By Future Technologies (LIFT) Center, Skolkovo, Moscow, 143025, Russia
| | - Airat Bilyalov
- SBHI Moscow Clinical Scientific Center Named After Loginov of Moscow Healthcare Department, Moscow, 111123, Russia
- Kazan Federal University, Kazan, 420008, Russia
| | - Elena Shagimardanova
- SBHI Moscow Clinical Scientific Center Named After Loginov of Moscow Healthcare Department, Moscow, 111123, Russia
- Life Improvement By Future Technologies (LIFT) Center, Skolkovo, Moscow, 143025, Russia
| | | | - Marina Nemtsova
- Evogen LLC, Moscow, 115191, Russia
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of Russian Federation (Sechenov University), Moscow, 119991, Russia
- Medical Genetic Research Center Named After Academician N.P. Bochkov, Moscow, 115522, Russia
| | | | - Mary Woroncow
- National Medical Research Center of Endocrinology, Moscow, 117292, Russia
- Lomonosov Moscow State University, Moscow, 119991, Russia
| | | | - Igor Khatkov
- SBHI Moscow Clinical Scientific Center Named After Loginov of Moscow Healthcare Department, Moscow, 111123, Russia.
| |
Collapse
|
19
|
Islam SM, Hasan M, Alam J, Dey A, Molineaux D. In Silico Screening, Molecular Dynamics Simulation and Binding Free Energy Identify Single-Point Mutations That Destabilize p53 and Reduce Binding to DNA. Proteins 2025; 93:498-514. [PMID: 39264222 PMCID: PMC11695177 DOI: 10.1002/prot.26747] [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: 06/18/2024] [Revised: 08/09/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Considering p53's pivotal role as a tumor suppressor protein, proactive identification and characterization of potentially harmful p53 mutations are crucial before they appear in the population. To address this, four computational prediction tools-SIFT, Polyphen-2, PhD-SNP, and MutPred2-utilizing sequence-based and machine-learning algorithms, were employed to identify potentially deleterious p53 nsSNPs (nonsynonymous single nucleotide polymorphisms) that may impact p53 structure, dynamics, and binding with DNA. These computational methods identified three variants, namely, C141Y, C238S, and L265P, as detrimental to p53 stability. Furthermore, molecular dynamics (MD) simulations revealed that all three variants exhibited heightened structural flexibility compared to the native protein, especially the C141Y and L265P mutations. Consequently, due to the altered structure of mutant p53, the DNA-binding affinity of all three variants decreased by approximately 1.8 to 9.7 times compared to wild-type p53 binding with DNA (14 μM). Notably, the L265P mutation exhibited an approximately ten-fold greater reduction in binding affinity. Consequently, the presence of the L265P mutation in p53 could pose a substantial risk to humans. Given that p53 regulates abnormal tumor growth, this research carries significant implications for surveillance efforts and the development of anticancer therapies.
Collapse
Affiliation(s)
- Shahidul M. Islam
- Department of Chemistry, Delaware State University, Dover, DE 19901, USA
| | - Mehedi Hasan
- Department of Chemistry, Delaware State University, Dover, DE 19901, USA
| | - Jahidul Alam
- Department of Molecular Biology and Biotechnology, Queen’s University Belfast, Northern Ireland, BT7 1NN, United Kingdom
| | - Anonya Dey
- Department of Genetic Engineering and Biotechnology, University of Chittagong, Chittagong, 4331, Bangladesh
| | - Dylan Molineaux
- Department of Chemistry, Delaware State University, Dover, DE 19901, USA
| |
Collapse
|
20
|
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
|
21
|
Opiso DMS, Gregorio GEV, Silao CLT. Association of TP53 Germline Variant and Choledochal Cyst among Clinically Diagnosed Filipino Pediatric Patients. ACTA MEDICA PHILIPPINA 2025; 59:7-14. [PMID: 39967707 PMCID: PMC11831089 DOI: 10.47895/amp.vi0.9091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Background and Objective Choledochal cysts (CC) are rare congenital, cystic dilations of the biliary tree occurring predominantly in Asian populations and in females. Patients are usually children presenting with any of the following: abdominal pain, palpable abdominal mass, and jaundice. Its congenital nature hints at a potential genetic cause. A possible causal gene is TP53, a tumor suppressor with a germline variant called rs201753350 (c.91G>A) that changed from a G allele to an A allele, decreasing the cell proliferation suppressing activity of its functional protein. Currently, there is no information on the TP53 rs201753350 germline variant available for the Filipino population. This study determined the prevalence of rs201753350 and the association between the functional G allele, the rs201753350 germline variant A allele, and the occurrence of CCs in Filipino pediatric patients in a tertiary government hospital. Methods Genomic DNA was extracted from blood samples of pediatric patients clinically diagnosed with CC. Controls were DNA samples collected from a previous study. The samples underwent PCR, electrophoresis, and sequencing. Results A total of 109 participants (22 cases and 87 controls) were included in the study. The A allele (22.94%) occurs at a lower frequency than the G allele (77.06%) among both cases and controls. More individuals have a homozygous G/G genotype (54.13%) than a heterozygous A/G genotype (45.87%) while the homozygous A/A genotype was not observed. The estimated risk of choledochal cyst occurrence is significantly lower in individuals with the A allele (PR: 0.08, 95% CI: 0.01 - 0.55) and the A/G genotype (PR: 0.06, 95% CI: 0.01 - 0.40). Conclusion There is no significant evidence to suggest an association between the TP53 rs201753350 germline variant and the occurrence of choledochal cysts in Filipinos. It is recommended that other mutations within and beyond the TP53 gene be investigated for possible associations with choledochal cyst occurrence.
Collapse
Affiliation(s)
| | - Germana Emerita V. Gregorio
- Department of Pediatrics, College of Medicine and Philippine General Hospital, University of the Philippines Manila
| | - Catherine Lynn T. Silao
- Department of Pediatrics, College of Medicine and Philippine General Hospital, University of the Philippines Manila
- Institute of Human Genetics, National Institutes of Health, University of the Philippines Manila
| |
Collapse
|
22
|
Tseng YH, Tran TTM, Tsai Chang J, Huang YT, Nguyen AT, Chang IYF, Chen YT, Hsieh HW, Juang YL, Chang PMH, Huang TY, Chang YC, Chen YM, Liu H, Huang CYF. Utilizing TP53 hotspot mutations as effective predictors of gemcitabine treatment outcome in non-small-cell lung cancer. Cell Death Discov 2025; 11:26. [PMID: 39870629 PMCID: PMC11772833 DOI: 10.1038/s41420-025-02300-7] [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: 03/27/2024] [Revised: 12/11/2024] [Accepted: 01/14/2025] [Indexed: 01/29/2025] Open
Abstract
TP53 mutations are recognized to correlate with a worse prognosis in individuals with non-small cell lung cancer (NSCLC). There exists an immediate necessity to pinpoint selective treatment for patients carrying TP53 mutations. Potential drugs were identified by comparing drug sensitivity differences, represented by the half-maximal inhibitory concentration (IC50), between TP53 mutant and wild-type NSCLC cell lines using database analysis. In addition, clinical data from NSCLC patients were collected to evaluate both their TP53 status and their response to gemcitabine, thereby facilitating further validation. Subsequently, NSCLC cell lines with different TP53 status (A549 and H1299) were subjected to gemcitabine treatment to investigate the association between TP53 mutations and gemcitabine response. According to the dataset, NSCLC cell lines carrying TP53 mutations displayed heightened sensitivity to gemcitabine. From a clinical standpoint, patients exhibiting TP53 hotspot mutations demonstrated prolonged overall survival upon gemcitabine treatment. In vitro, overexpressing various hotspot TP53 mutations significantly sensitized H1299 cells to gemcitabine. Moreover, the knockdown of TP53 in A549 cells notably augmented sensitivity to gemcitabine treatment, as evidenced by cell viability and reproductive cell death assays. Conversely, the overexpression of wild-type TP53 in H1299 cells led to an increased resistance against gemcitabine. Gemcitabine is a treatment option for patients with non-small cell lung cancer (NSCLC) who carry TP53 hotspot mutations. This potential effectiveness might arise from its ability to disrupt DNA damage repair processes, leading to G2/M phase cell cycle arrest or an augmentation of mitotic abnormalities, eventually cause cell death. As a result, when planning treatment strategies for NSCLC patients possessing TP53 hotspot mutations, gemcitabine should be considered to incorporate into the indication.
Collapse
Affiliation(s)
- Yen-Han Tseng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Trieu Thi My Tran
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jinghua Tsai Chang
- Institute of Medicine, Chung Shan Medical University, Taichung City, Taiwan
| | - Yu-Tang Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Biomedical Industry Ph.D. Program, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Anh Thuc Nguyen
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Program in Molecular Medicine, Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Ian Yi-Feng Chang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
- Department of Neurosurgery, Lin-Kou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yi-Tung Chen
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Hao-Wen Hsieh
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yue-Li Juang
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
| | - Peter Mu-Hsin Chang
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzu-Yi Huang
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Chih Chang
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Hsuan Liu
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan.
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Chong Hin Loon Memorial Cancer and Biotherapy Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Cao X, Luo Y, Shen S, Ren X. Primary mucinous cystadenocarcinoma of the breast: A case report and literature review. Oncol Lett 2025; 29:60. [PMID: 39611063 PMCID: PMC11602826 DOI: 10.3892/ol.2024.14806] [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: 08/03/2024] [Accepted: 10/30/2024] [Indexed: 11/30/2024] Open
Abstract
Mucinous cystadenocarcinoma (MCA) is a rare breast cancer. The present study reports a case of primary MCA of the breast with a comprehensive evaluation of this rare tumour. A 51-year-old woman sought medical attention for a mass in the left breast. A core needle biopsy revealed an infiltrating adenocarcinoma with mucus secretion and papillary formation. The macroscopic appearance was of a greyish-white, tough and well-circumscribed solid mass, without a notable cyst. Microscopically, the tumour consisted of ducts and cysts of varying sizes. Varying degrees of branching papillary structures were observed in the lumen and cyst cavities. The tumour cells were highly columnar in shape, with high-grade nuclei arranged in a single-layer. Immunohistochemistry revealed that the tumour was a basal-like triple-negative breast cancer with a high proliferation index and tumour protein p53 diffuse strong expression. Mutations in breast cancer 1-associated RING domain 1 (BARD1), kinase domain containing receptor (KDR), mucin-6 (MUC6), tumour protein 53 (TP53) and breast cancer 1-interacting protein C-terminal helicase 1 (BRIP1) were identified using DNA analysis. The patient was followed up for 26 months and showed no signs of recurrence or metastasis. In conclusion, the current study presents a case of MCA of breast accompanied by mutations in the BARD1, KDR, MUC6, TP53 and BRIP1 genes, with no recurrence after a 26-month follow-up. Combining this case with a review of the literature helps us to better understand the clinicopathological and genetic characteristics of MCA, and guide treatment.
Collapse
Affiliation(s)
- Xi Cao
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Yongchao Luo
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Songjie Shen
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| | - Xinyu Ren
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P.R. China
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Chai D, Wang X, Fan C, Wang J, Lim JM, Yu X, Young KH, Li Y. Vaccines targeting p53 mutants elicit anti-tumor immunity. Cancer Lett 2024; 611:217421. [PMID: 39740750 DOI: 10.1016/j.canlet.2024.217421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 01/02/2025]
Abstract
The p53 tumor suppressor is commonly mutated in cancer; however, there are no effective treatments targeting p53 mutants. A DNA vaccine gWIZ-S237G targeting the p53 S237G mutant, which is highly expressed in A20 murine tumor cells, was developed and administered intramuscularly via electroporation, either alone or in combination with PD-1 blockade. The anti-p53-S237G immunization elicited a robust protective response against subcutaneous A20 tumors and facilitated the infiltration of immune cells including CD8+ T cells, NK cells, and DCs. The vaccine enhanced the induction and maturation of CD11c+, CD103+CD11c+, and CD8+CD11c+ cells, which in turn promoted tumor-specific antibody production, as well as Th1 and CD8+ T cell-mediated immune responses. Several antigenic epitopes of p53-S237G effectively stimulated multifunctional CD8+ T cells to secrete IFN-γ and TNF-α. The vaccine showed long-term anti-tumor effects that were dependent on memory CD8+ T cells. Furthermore, the anti-p53-S237G vaccine exhibited significant protective efficacy in the A20 liver metastasis models. When combined with PD-1 inhibition, the vaccine showed superior inhibition of tumor growth and liver metastasis. Targeting p53 mutants by vaccination represents a potential precision medicine strategy against cancers harboring p53 mutations.
Collapse
Affiliation(s)
- Dafei Chai
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Xu Wang
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chunmei Fan
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Junhao Wang
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jing Ming Lim
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xinfang Yu
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ken H Young
- Department of Pathology, Division of Hematopathology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Yong Li
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
29
|
Goyal K, Afzal M, Altamimi ASA, Babu MA, Ballal S, Kaur I, Kumar S, Kumar MR, Chauhan AS, Ali H, Shahwan M, Gupta G. Chronic kidney disease and aging: dissecting the p53/p21 pathway as a therapeutic target. Biogerontology 2024; 26:32. [PMID: 39725742 DOI: 10.1007/s10522-024-10173-z] [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: 11/09/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024]
Abstract
Chronic kidney diseases (CKD) are a group of multi-factorial disorders that markedly impair kidney functions with progressive renal deterioration. Aging contributes to age-specific phenotypes in kidneys, which undergo several structural and functional alterations, such as a decline in regenerative capacity and increased fibrosis, inflammation, and tubular atrophy, all predisposing them to disease and increasing their susceptibility to injury while impeding their recovery. A central feature of these age-related processes is the activation of the p53/p21 pathway signaling. The pathway is a key player in cellular senescence, apoptosis, and cell cycle regulation, which are all key to maintaining the health of the kidney. P53 is a transcription factor and a tumor suppressor protein that responds to cell stress and damage. Persistent activation of cell p53 can lead to the expression of p21, an inhibitor of the cell cycle known as a cyclin-dependent kinase. This causes cells to cease dividing and leads to senescence, where cells can no longer increase. The accumulation of senescent cells in the aging kidney impairs kidney function by altering the microenvironment. As the number of senescent cells increases, the capacity of the kidney to recover from injury decreases, accelerating the progression of end-stage renal disease. This article review extensively explores the relationship between the p53/p21 pathway and cellular senescence within an aging kidney and the emerging therapeutic strategies that target it to overcome the impacts of cellular senescence on CKD.
Collapse
Affiliation(s)
- Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | | | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - M Ravi Kumar
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab, 140307, India
| | - Ashish Singh Chauhan
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Haider Ali
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Moyad Shahwan
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
30
|
Nakamoto-Matsubara R, Nardi V, Horick N, Fukushima T, Han RS, Shome R, Ochi K, Panaroni C, Fulzele K, Rexha F, Branagan AR, Cirstea D, Yee AJ, Scadden DT, Raje NS. Integration of clinical outcomes and molecular features in extramedullary disease in multiple myeloma. Blood Cancer J 2024; 14:224. [PMID: 39715752 DOI: 10.1038/s41408-024-01190-9] [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: 08/10/2024] [Revised: 10/31/2024] [Accepted: 11/13/2024] [Indexed: 12/25/2024] Open
Abstract
Multiple myeloma (MM) remains incurable despite novel therapeutics. A major contributor to the development of relapsed/refractory and resistant MM is extraosseous extramedullary disease (EMD), whose molecular biology is still not fully understood. We analyzed 528 MM patients who presented to our institution between 2014 and 2021 and who had undergone molecular testing. We defined EMD as organ plasmacytoma distinct from bones and evaluated patients for the development of EMD with the goal of defining their molecular characteristics. Here, we show that RAS/BRAF mutations are likely essential for the development of EMD. Our results also indicate that the underlying reason for the negative outcomes in patients with poor prognostic factors such as duplication 1q and deletion 17p is largely due to the development of EMD. However, the presence of TP53 mutation remains a poor prognostic factor regardless of EMD development. Furthermore, mutation sites of TP53 were different between EMD versus non-EMD patients, with gain-of-function mutations enriched in patients with EMD. Our data highlights distinct molecular abnormalities in patients with EMD and provides potential mechanistic insights for novel therapeutic targets for the future.
Collapse
Affiliation(s)
- Rie Nakamoto-Matsubara
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Valentina Nardi
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nora Horick
- Biostatistics Center, Massachusetts General Hospital, Boston, MA, USA
| | - Tsuyoshi Fukushima
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryan S Han
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Rajib Shome
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Kiyosumi Ochi
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Cristina Panaroni
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Keertik Fulzele
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Farah Rexha
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Andrew R Branagan
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Diana Cirstea
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Andrew J Yee
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - David T Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Noopur S Raje
- Center for Multiple Myeloma, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
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
|
32
|
Sun S, You E, Hong J, Hoyos D, Del Priore I, Tsanov KM, Mattagajasingh O, Di Gioacchino A, Marhon SA, Chacon-Barahona J, Li H, Jiang H, Hozeifi S, Rosas-Bringas O, Xu KH, Song Y, Lang ER, Rojas AS, Nieman LT, Patel BK, Murali R, Chanda P, Karacay A, Vabret N, De Carvalho DD, Zenklusen D, LaCava J, Lowe SW, Ting DT, Iacobuzio-Donahue CA, Solovyov A, Greenbaum BD. Cancer cells restrict immunogenicity of retrotransposon expression via distinct mechanisms. Immunity 2024; 57:2879-2894.e11. [PMID: 39577413 PMCID: PMC12022969 DOI: 10.1016/j.immuni.2024.10.015] [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: 11/16/2023] [Revised: 06/28/2024] [Accepted: 10/29/2024] [Indexed: 11/24/2024]
Abstract
To thrive, cancer cells must navigate acute inflammatory signaling accompanying oncogenic transformation, such as via overexpression of repeat elements. We examined the relationship between immunostimulatory repeat expression, tumor evolution, and the tumor-immune microenvironment. Integration of multimodal data from a cohort of pancreatic ductal adenocarcinoma (PDAC) patients revealed expression of specific Alu repeats predicted to form double-stranded RNAs (dsRNAs) and trigger retinoic-acid-inducible gene I (RIG-I)-like-receptor (RLR)-associated type-I interferon (IFN) signaling. Such Alu-derived dsRNAs also anti-correlated with pro-tumorigenic macrophage infiltration in late stage tumors. We defined two complementary pathways whereby PDAC may adapt to such anti-tumorigenic signaling. In mutant TP53 tumors, ORF1p from long interspersed nuclear element (LINE)-1 preferentially binds Alus and decreases their expression, whereas adenosine deaminases acting on RNA 1 (ADAR1) editing primarily reduces dsRNA formation in wild-type TP53 tumors. Depletion of either LINE-1 ORF1p or ADAR1 reduced tumor growth in vitro. The fact that tumors utilize multiple pathways to mitigate immunostimulatory repeats implies the stress from their expression is a fundamental phenomenon to which PDAC, and likely other tumors, adapt.
Collapse
Affiliation(s)
- Siyu Sun
- Halvorsen Center for Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Eunae You
- Massachusetts General Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Jungeui Hong
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David Hoyos
- Tri-Institutional Program in Computational Biology and Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Isabella Del Priore
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kaloyan M Tsanov
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Om Mattagajasingh
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, Canada
| | - Andrea Di Gioacchino
- Laboratoire de Physique de l'Ecole Normale Supérieure, Sorbonne Université, Université de Paris, Paris, France
| | - Sajid A Marhon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Jonathan Chacon-Barahona
- Tri-Institutional Program in Computational Biology and Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Hao Li
- Halvorsen Center for Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hua Jiang
- Laboratory of Cellular and Structural Biology, The Rockefeller University, New York, NY, USA
| | - Samira Hozeifi
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, Groningen, The Netherlands
| | - Omar Rosas-Bringas
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, Groningen, The Netherlands
| | - Katherine H Xu
- Massachusetts General Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Yuhui Song
- Massachusetts General Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Evan R Lang
- Massachusetts General Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Alexandra S Rojas
- Massachusetts General Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Linda T Nieman
- Massachusetts General Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Bidish K Patel
- Massachusetts General Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Rajmohan Murali
- Last Wish Program and Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pharto Chanda
- Last Wish Program and Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ali Karacay
- Last Wish Program and Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicolas Vabret
- Precision Immunology Institute at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel D De Carvalho
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Daniel Zenklusen
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, Canada
| | - John LaCava
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; European Research Institute for the Biology of Ageing, University Medical Center Groningen, Groningen, The Netherlands
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - David T Ting
- Massachusetts General Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Christine A Iacobuzio-Donahue
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Last Wish Program and Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander Solovyov
- Halvorsen Center for Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benjamin D Greenbaum
- Halvorsen Center for Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Physiology, Biophysics & Systems Biology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
33
|
Malhotra L, Singh A, Kaur P, Ethayathulla AS. Phenotypical mapping of TP53 unique missense mutations spectrum in human cancers. J Biomol Struct Dyn 2024:1-14. [PMID: 39639563 DOI: 10.1080/07391102.2024.2435060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/04/2024] [Indexed: 12/07/2024]
Abstract
The p53 tumor suppressor is one of the most mutated genes responsible for tumorigenesis in most human cancers. Out of 29,891 genomic mutations reported in the TP53 Database (https://tp53.isb-cgc.org/), 1,297 are identified as unique missense somatic mutations excluding frameshift, intronic, deletion, nonsense, silent, splice, and other unknown mutations. We have comprehensively analyzed all these 1,297 unique missense mutations and created a phenotypical map based on the distribution of mutations in each domain, the functional state of the protein, and their occurrence in different types of tissues and organs. Our mutation map shows that almost 118 unique missense mutations are reported in the transactivation and proline-rich domains, 1,065 in the central DNA-binding domains, and 113 in the oligomerization and regulatory domains. Based on the phenotype, these mutations are subdivided into 46 super trans, 491 functional, 315 partially functional, and 415 non-functional mutations. The prevalence of these mutations was checked in 71 different types of tissues and found that the mutant R248Q is reported in 51 types of tissues followed by R175H and R273H in 46 types. We correlated the potential impact of mutation in target gene transcription and regulation with nucleosomal DNA and RNA-Pol II complexes. We have discussed the impact of mutation at post-translational modification sites in the structure and function of p53 highlighting the potential therapeutic drug targets with tremendous clinical applications.
Collapse
Affiliation(s)
- Lakshay Malhotra
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, New Delhi, India
| | - Alankrita Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Abdul S Ethayathulla
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
34
|
LeBlanc C, Stefani J, Soriano M, Lam A, Zintel MA, Kotha SR, Chase E, Pimentel-Solorio G, Vunnum A, Flug K, Fultineer A, Hummel N, Staller MV. Conservation of function without conservation of amino acid sequence in intrinsically disordered transcriptional activation domains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626510. [PMID: 39677729 PMCID: PMC11642888 DOI: 10.1101/2024.12.03.626510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Protein function is canonically believed to be more conserved than amino acid sequence, but this idea is only well supported in folded domains, where highly diverged sequences can fold into equivalent 3D structures. In contrast, intrinsically disordered protein regions (IDRs) do not fold into a stable 3D structure, thus it remains unknown when and how function is conserved for IDRs that experience rapid amino acid sequence divergence. As a model system for studying the evolution of IDRs, we examined transcriptional activation domains, the regions of transcription factors that bind to coactivator complexes. We systematically identified activation domains on 502 orthologs of the transcriptional activator Gcn4 spanning 600 MY of fungal evolution. We find that the central activation domain shows strong conservation of function without conservation of sequence. This conservation of function without conservation of sequence is facilitated by evolutionary turnover (gain and loss) of key acidic and aromatic residues, the positions most important for function. This high sequence flexibility of functional orthologs mirrors the physical flexibility of the activation domain coactivator interaction interface, suggesting that physical flexibility enables evolutionary plasticity. We propose that turnover of short functional elements, sometimes individual amino acids, is a general mechanism for conservation of function without conservation of sequence during IDR evolution.
Collapse
Affiliation(s)
- Claire LeBlanc
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Jordan Stefani
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Melvin Soriano
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Angelica Lam
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Marissa A. Zintel
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
| | - Sanjana R. Kotha
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Emily Chase
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Giovani Pimentel-Solorio
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
| | - Aditya Vunnum
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
| | - Katherine Flug
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
| | - Aaron Fultineer
- Department of Physics, University of California Berkeley, Berkeley, 94720
| | - Niklas Hummel
- Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Max V. Staller
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, 94720
- Center for Computational Biology, University of California Berkeley, Berkeley, 94720
- Chan Zuckerberg Biohub–San Francisco, San Francisco, CA 94158
| |
Collapse
|
35
|
Li Q, Zhang Y, Luo S, Zhang Z, Oberg AL, Kozono DE, Lu H, Sarkaria JN, Ma L, Wang L. Identify Non-mutational p53 Functional Deficiency in Human Cancers. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae064. [PMID: 39325855 PMCID: PMC11702981 DOI: 10.1093/gpbjnl/qzae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 07/23/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024]
Abstract
An accurate assessment of p53's functional statuses is critical for cancer genomic medicine. However, there is a significant challenge in identifying tumors with non-mutational p53 inactivation which is not detectable through DNA sequencing. These undetected cases are often misclassified as p53-normal, leading to inaccurate prognosis and downstream association analyses. To address this issue, we built the support vector machine (SVM) models to systematically reassess p53's functional statuses in TP53 wild-type (TP53WT) tumors from multiple The Cancer Genome Atlas (TCGA) cohorts. Cross-validation demonstrated the good performance of the SVM models with a mean area under the receiver operating characteristic curve (AUROC) of 0.9822, precision of 0.9747, and recall of 0.9784. Our study revealed that a significant proportion (87%-99%) of TP53WT tumors actually had compromised p53 function. Additional analyses uncovered that these genetically intact but functionally impaired (termed as predictively reduced function of p53 or TP53WT-pRF) tumors exhibited genomic and pathophysiologic features akin to TP53-mutant tumors: heightened genomic instability and elevated levels of hypoxia. Clinically, patients with TP53WT-pRF tumors experienced significantly shortened overall survival or progression-free survival compared to those with predictively normal function of p53 (TP53WT-pN) tumors, and these patients also displayed increased sensitivity to platinum-based chemotherapy and radiation therapy.
Collapse
Affiliation(s)
- Qianpeng Li
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Zhang
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sicheng Luo
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhang Zhang
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ann L Oberg
- Division of Computational Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - David E Kozono
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Hua Lu
- Department of Biochemistry & Molecular Biology and Tulane Cancer Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Lina Ma
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liguo Wang
- Division of Computational Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Bioinformatics and Computational Biology Graduate Program, University of Minnesota Rochester, Rochester, MN 55904, USA
| |
Collapse
|
36
|
Gungordu S, Aptullahoglu E. Targeting MDM2-mediated suppression of p53 with idasanutlin: a promising therapeutic approach for acute lymphoblastic leukemia. Invest New Drugs 2024; 42:603-611. [PMID: 39305365 DOI: 10.1007/s10637-024-01473-9] [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: 07/17/2024] [Accepted: 09/17/2024] [Indexed: 12/08/2024]
Abstract
Despite available treatments for acute lymphoblastic leukemia (ALL), the disease's high clinical variability necessitates new therapeutic strategies, particularly for patients with high-risk features. The tumor suppressor protein p53, encoded by the TP53 gene and known as the guardian of the genome, plays a crucial role in preventing tumor development. Over 90% of ALL cases initially harbor wild-type TP53. Reactivation of p53, which is encoded from the wild type TP53 but lost its function for several reasons, is an attractive therapeutic approach in cancer treatment. p53 can be activated in a non-genotoxic manner by targeting its primary repressor, the MDM2 protein. Clinical trials involving MDM2 inhibitors are currently being conducted in a growing body of investigation, reflecting of the interest in incorporating these treatments into cancer treatment strategies. Early-phase clinical trials have demonstrated the promise of idasanutlin (RG7388), one of the developed compounds. It is a second-generation MDM2-p53 binding antagonist with enhanced potency, selectivity, and bioavailability. The aim of this study is to evaluate the efficacy of RG7388 as a therapeutic strategy for ALL and to investigate its potential impact on improving treatment outcomes for high-risk patients. RG7388 potently decreased the viability in five out of six ALL cell lines with diverse TP53 mutation profiles, whereas only one cell line exhibited high resistance. RG7388 induced a pro-apoptotic gene expression signature with upregulation of p53-target genes involved in the intrinsic and extrinsic pathways of apoptosis. Consequently, RG7388 led to a concentration-dependent increase in caspase-3/7 activity and cleaved poly (ADP-ribose) polymerase. In this research, RG7388 was investigated with pre-clinical methods in ALL cells as a novel treatment strategy. This study suggests further functional research and in-vivo evaluation, and it highlights the prospect of treating p53-functional ALL with MDM2 inhibitors.
Collapse
Affiliation(s)
- Seyda Gungordu
- Biotechnology Application and Research Centre, Bilecik Şeyh Edebali University, 11100, Bilecik, Turkey
| | - Erhan Aptullahoglu
- Biotechnology Application and Research Centre, Bilecik Şeyh Edebali University, 11100, Bilecik, Turkey.
- Department of Molecular Biology and Genetics, Faculty of Science, Bilecik Şeyh Edebali University, 11100, Bilecik, Turkey.
| |
Collapse
|
37
|
Gibbs E, Miao Q, Ferrolino M, Bajpai R, Hassan A, Phillips AH, Pitre A, Kümmerle R, Miller S, Nagy G, Leite W, Heller W, Stanley C, Perrone B, Kriwacki R. p14 ARF forms meso-scale assemblies upon phase separation with NPM1. Nat Commun 2024; 15:9531. [PMID: 39528457 PMCID: PMC11555371 DOI: 10.1038/s41467-024-53904-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
NPM1 is an abundant nucleolar chaperone that, in addition to facilitating ribosome biogenesis, contributes to nucleolar stress responses and tumor suppression through its regulation of the p14 Alternative Reading Frame tumor suppressor protein (p14ARF). Oncogenic stress induces p14ARF to inhibit MDM2, stabilize p53 and arrest the cell cycle. Under non-stress conditions, NPM1 stabilizes p14ARF in nucleoli, preventing its degradation and blocking p53 activation. However, the mechanisms underlying the regulation of p14ARF by NPM1 are unclear because the structural features of the p14ARF-NPM1 complex were elusive. Here we show that p14ARF assembles into a gel-like meso-scale network upon phase separation with NPM1. This assembly is mediated by intermolecular contacts formed by hydrophobic residues in an α-helix and β-strands within a partially folded N-terminal portion of p14ARF. These hydrophobic interactions promote phase separation with NPM1, enhance p14ARF nucleolar partitioning, restrict NPM1 diffusion within condensates and nucleoli, and reduce cellular proliferation. Our structural analysis provides insights into the multifaceted chaperone function of NPM1 in nucleoli by mechanistically linking the nucleolar localization of p14ARF to its partial folding and meso-scale assembly upon phase separation with NPM1.
Collapse
Affiliation(s)
- Eric Gibbs
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Qi Miao
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Mylene Ferrolino
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Richa Bajpai
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Aila Hassan
- Bruker Switzerland AG, Fällanden, Switzerland
| | - Aaron H Phillips
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Aaron Pitre
- Cell and Tissue Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Shondra Miller
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Gergely Nagy
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Wellington Leite
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - William Heller
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Chris Stanley
- Computational Sciences and Engineering Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | | | - Richard Kriwacki
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Sciences Center, Memphis, Tennessee, USA.
| |
Collapse
|
38
|
Stieg DC, Casey K, Karisetty BC, Leu JIJ, Larkin F, Vogel P, Madzo J, Murphy ME. The Ashkenazi-Centric G334R Variant of TP53 is Severely Impaired for Transactivation but Retains Tumor Suppressor Function in a Mouse Model. Mol Cell Biol 2024; 44:607-621. [PMID: 39520074 PMCID: PMC11583612 DOI: 10.1080/10985549.2024.2421885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Mutations in the TP53 tumor suppressor gene are the most abundant genetic occurrences in cancer. Some of these mutations lead to loss of function of p53 protein, some are gain of function, and some variants are hypomorphic (partially functional). Currently, there is no clinical distinction between different p53 mutations and cancer therapy or prognosis. Mutations in the oligomerization domain of p53 appear to be quite distinct in function, compared to mutations in the DNA binding domain. Here we show that, like other p53 oligomerization domain mutants, the Ashkenazi-specific G334R mutant accumulates to very high levels in cells and is significantly impaired for the transactivation of canonical p53 target genes. Surprisingly, we find that this mutant retains the ability to bind to consensus p53 target sites. A mouse model reveals that mice containing the G334R variant show increased predisposition to cancer, but only a fraction of these mice develop late-onset cancer. We show that the G334R variant retains the ability to interact with the SP1 transcription factor and contributes to the transactivation of joint SP1-p53 target genes. The combined evidence indicates that G334R is a unique oligomerization domain mutant that retains some tumor suppressor function.
Collapse
Affiliation(s)
- David C. Stieg
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Kaitlyn Casey
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania, USA
- Cancer Biology Program, St Joseph’s University, Philadelphia, Pennsylvania, USA
| | | | - Julia I-Ju Leu
- Perelman School of Medicine, Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fiona Larkin
- Hunterdon County Academies, Annandale, New Jersey, USA
| | - Peter Vogel
- Comparative Pathology Core, St Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jozef Madzo
- Bioinformatics Facility, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Maureen E. Murphy
- Program in Molecular and Cellular Oncogenesis, The Wistar Institute, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
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
|
40
|
Thangavelu L, Altamimi ASA, Ghaboura N, Babu MA, Roopashree R, Sharma P, Pal P, Choudhary C, Prasad GVS, Sinha A, Balaraman AK, Rawat S. Targeting the p53-p21 axis in liver cancer: Linking cellular senescence to tumor suppression and progression. Pathol Res Pract 2024; 263:155652. [PMID: 39437639 DOI: 10.1016/j.prp.2024.155652] [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: 09/06/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Liver cancer is a major health epidemic worldwide, mainly due to its high mortality rates and limited treatment options. The association of cellular senescence to tumorigenesis and the cancer hallmarks remains a subject of interest in cancer biology. The p53-p21 signalling axis is an important regulator in restoring the cell's balance by supporting tumor suppression and tumorigenesis in liver cancer. We review the novel molecular mechanisms that p53 and its downstream effector, p21, employ to induce cellular senescence, making it last longer, and halt the proliferation of damaged hepatocytes to become tumorous cells. We also examine how dysregulation of this pathway contributes to HCC pathogenesis, proliferation, survival, acquired resistance to apoptosis, and increased invasiveness. Furthermore, we comprehensively describe the molecular cross-talk between the p53-p21 signalling axis and major cell cycle signalling pathways, including Wnt/β-catenin, PI3K/Akt, and TGF-β in liver cancer and provide an overview of promising candidates for chemoprevention and future therapeutic strategies. This review article explores the roles of the p53-p21 pathway in liver cancer, examining its function in promoting cellular senescence under normal conditions and its potential role in cancer progression. It also highlights novel therapeutic drugs and drug targets within the pathway and discusses the implications for treatment strategies and prognosis in liver cancer.
Collapse
Affiliation(s)
- Lakshmi Thangavelu
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Nehmat Ghaboura
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India.
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Pusparghya Pal
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Chhavi Choudhary
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - Sushama Rawat
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| |
Collapse
|
41
|
Byrjalsen A, Stoltze UK, Lautrup C, Christensen LL, Mikkelsen T, Hjalgrim L, Brok JS, Dahl C, Schmiegelow K, Borgwardt L, Diness BR, Hansen TVO, Wadt KAW. Novel germline TP53 variant (p.(Phe109Ile)) confers high risk of cancer. J Med Genet 2024; 61:1023-1025. [PMID: 39317423 DOI: 10.1136/jmg-2024-110255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/14/2024] [Indexed: 09/26/2024]
Affiliation(s)
- Anna Byrjalsen
- Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ulrik Kristoffer Stoltze
- Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Charlotte Lautrup
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | | | - Torben Mikkelsen
- Department of Pediatric and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Lisa Hjalgrim
- Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jesper Sune Brok
- Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Christine Dahl
- Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Kjeld Schmiegelow
- Department of Pediatric and Adolescent Medicine, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lotte Borgwardt
- Department of Diagnostic Radiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Birgitte Rode Diness
- Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Van Overeem Hansen
- Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karin A W Wadt
- Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
42
|
Bakhtiar H, Sharifi MN, Helzer KT, Shi Y, Bootsma ML, Shang TA, Chrostek MR, Berg TJ, Carson Callahan S, Carreno V, Blitzer GC, West MT, O'Regan RM, Wisinski KB, Sjöström M, Zhao SG. A phenocopy signature of TP53 loss predicts response to chemotherapy. NPJ Precis Oncol 2024; 8:220. [PMID: 39358429 PMCID: PMC11447220 DOI: 10.1038/s41698-024-00722-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024] Open
Abstract
In preclinical studies, p53 loss of function impacts chemotherapy response, but this has not been consistently validated clinically. We trained a TP53-loss phenocopy gene expression signature from pan-cancer clinical samples in the TCGA. In vitro, the TP53-loss phenocopy signature predicted chemotherapy response across cancer types. In a clinical dataset of 3003 breast cancer samples treated with neoadjuvant chemotherapy, the TP53-loss phenocopy samples were 56% more likely to have a pathologic complete response (pCR), with a significant association between TP53-loss phenocopy and pCR in both ER positive and ER negative tumors. In an independent clinical validation in the I-SPY2 trial (N = 987), we confirmed the association with neoadjuvant chemotherapy pCR and found higher rates of chemoimmunotherapy response in TP53-loss phenocopy tumors compared to non-TP53-loss phenocopy tumors (64% vs. 28%). The TP53-loss phenocopy signature predicts chemotherapy response across cancer types in vitro, and in a proof-of-concept clinical validation is associated with neoadjuvant chemotherapy response across multiple clinical breast cancer cohorts.
Collapse
Affiliation(s)
- Hamza Bakhtiar
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Marina N Sharifi
- Department of Medicine, Division of Hematology, Oncology, and Palliative Care, University of Wisconsin, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Kyle T Helzer
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Yue Shi
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Matthew L Bootsma
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Tianfu A Shang
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | | | - Tracy J Berg
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - S Carson Callahan
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Viridiana Carreno
- Department of Medicine, Division of Hematology, Oncology, and Palliative Care, University of Wisconsin, Madison, WI, USA
| | - Grace C Blitzer
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA
| | - Malinda T West
- Department of Medicine, Division of Hematology, Oncology, and Palliative Care, University of Wisconsin, Madison, WI, USA
| | - Ruth M O'Regan
- Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Kari B Wisinski
- Department of Medicine, Division of Hematology, Oncology, and Palliative Care, University of Wisconsin, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Martin Sjöström
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Shuang G Zhao
- Department of Human Oncology, University of Wisconsin, Madison, WI, USA.
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
43
|
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
|
44
|
Jiang B, Elkashif A, Coulter JA, Dunne NJ, McCarthy HO. Immunotherapy for HPV negative head and neck squamous cell carcinoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189138. [PMID: 38889878 DOI: 10.1016/j.bbcan.2024.189138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
Head and neck cancer (HNSCC) is the 8th most common cancer in the UK, with incidence increasing due to lifestyle factors such as tobacco and alcohol abuse. HNSCC is an immune-suppressive disease characterised by impaired cytokine secretion and dysregulation of immune infiltrate. As such, immunotherapy is a potential treatment option, with therapeutic cancer vaccination demonstrating the greatest potential. The success of cancer vaccination is dependent on informed antigen selection: an ideal antigen must be either tumour-specific or tumour-associated, as well as highly immunogenic. Stratification of the patient population for antigen expression and validated biomarkers are also vital. This review focuses on the latest developments in immunotherapy, specifically the development of therapeutic vaccines, and highlights successes, potential drawbacks and areas for future development. Immunotherapy approaches considered for HNSCC include monoclonal antibodies (mAb), Oncolytic viral (OV) therapies, Immune Checkpoint Inhibitors (ICIs) and cancer vaccines.
Collapse
Affiliation(s)
- Binyumeng Jiang
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Ahmed Elkashif
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Jonathan A Coulter
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Nicholas J Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| |
Collapse
|
45
|
Wong D, Tageldein M, Luo P, Ensminger E, Bruce J, Oldfield L, Gong H, Fischer NW, Laverty B, Subasri V, Davidson S, Khan R, Villani A, Shlien A, Kim RH, Malkin D, Pugh TJ. Cell-free DNA from germline TP53 mutation carriers reflect cancer-like fragmentation patterns. Nat Commun 2024; 15:7386. [PMID: 39191772 DOI: 10.1038/s41467-024-51529-w] [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: 09/11/2023] [Accepted: 08/07/2024] [Indexed: 08/29/2024] Open
Abstract
Germline pathogenic TP53 variants predispose individuals to a high lifetime risk of developing multiple cancers and are the hallmark feature of Li-Fraumeni syndrome (LFS). Our group has previously shown that LFS patients harbor shorter plasma cell-free DNA fragmentation; independent of cancer status. To understand the functional underpinning of cfDNA fragmentation in LFS, we conducted a fragmentomic analysis of 199 cfDNA samples from 82 TP53 mutation carriers and 30 healthy TP53-wildtype controls. We find that LFS individuals exhibit an increased prevalence of A/T nucleotides at fragment ends, dysregulated nucleosome positioning at p53 binding sites, and loci-specific changes in chromatin accessibility at development-associated transcription factor binding sites and at cancer-associated open chromatin regions. Machine learning classification resulted in robust differentiation between TP53 mutant versus wildtype cfDNA samples (AUC-ROC = 0.710-1.000) and intra-patient longitudinal analysis of ctDNA fragmentation signal enabled early cancer detection. These results suggest that cfDNA fragmentation may be a useful diagnostic tool in LFS patients and provides an important baseline for cancer early detection.
Collapse
Affiliation(s)
- Derek Wong
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Maha Tageldein
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Ping Luo
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Erik Ensminger
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey Bruce
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Leslie Oldfield
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
| | - Haifan Gong
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | | - Brianne Laverty
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Vallijah Subasri
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
| | - Scott Davidson
- Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Torotno, Ontario, Canada
| | - Reem Khan
- Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Torotno, Ontario, Canada
| | - Anita Villani
- Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Division of Hematology/Oncology, The Hospital for Sick Children, Toroton, Ontario, Canada
| | - Adam Shlien
- Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Torotno, Ontario, Canada
| | - Raymond H Kim
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada.
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada.
- Ontario Institute of Cancer Research, Toronto, Ontario, Canada.
| | - David Malkin
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
- Department of Pediatrics, University of Toronto, Torotno, Ontario, Canada.
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada.
| | - Trevor J Pugh
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
- Ontario Institute of Cancer Research, Toronto, Ontario, Canada.
| |
Collapse
|
46
|
Fallatah MMJ, Demir Ö, Law F, Lauinger L, Baronio R, Hall L, Bournique E, Srivastava A, Metzen LT, Norman Z, Buisson R, Amaro RE, Kaiser P. Pyrimidine Triones as Potential Activators of p53 Mutants. Biomolecules 2024; 14:967. [PMID: 39199355 PMCID: PMC11352488 DOI: 10.3390/biom14080967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
p53 is a crucial tumor suppressor in vertebrates that is frequently mutated in human cancers. Most mutations are missense mutations that render p53 inactive in suppressing tumor initiation and progression. Developing small-molecule drugs to convert mutant p53 into an active, wild-type-like conformation is a significant focus for personalized cancer therapy. Prior research indicates that reactivating p53 suppresses cancer cell proliferation and tumor growth in animal models. Early clinical evidence with a compound selectively targeting p53 mutants with substitutions of tyrosine 220 suggests potential therapeutic benefits of reactivating p53 in patients. This study identifies and examines the UCI-1001 compound series as a potential corrector for several p53 mutations. The findings indicate that UCI-1001 treatment in p53 mutant cancer cell lines inhibits growth and reinstates wild-type p53 activities, including DNA binding, target gene activation, and induction of cell death. Cellular thermal shift assays, conformation-specific immunofluorescence staining, and differential scanning fluorometry suggest that UCI-1001 interacts with and alters the conformation of mutant p53 in cancer cells. These initial results identify pyrimidine trione derivatives of the UCI-1001 series as candidates for p53 corrector drug development.
Collapse
Affiliation(s)
| | - Özlem Demir
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Fiona Law
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Linda Lauinger
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Roberta Baronio
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Linda Hall
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Elodie Bournique
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Ambuj Srivastava
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Landon Tyler Metzen
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Zane Norman
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Rémi Buisson
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| | - Rommie E. Amaro
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Peter Kaiser
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
47
|
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
|
48
|
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
|
49
|
Abreu RBV, Pereira AS, Rosa MN, Ashton-Prolla P, Silva VAO, Melendez ME, Palmero EI. Functional evaluation of germline TP53 variants identified in Brazilian families at-risk for Li-Fraumeni syndrome. Sci Rep 2024; 14:17187. [PMID: 39060302 PMCID: PMC11282216 DOI: 10.1038/s41598-024-67810-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Germline TP53 pathogenic variants can lead to a cancer susceptibility syndrome known as Li-Fraumeni (LFS). Variants affecting its activity can drive tumorigenesis altering p53 pathways and their identification is crucial for assessing individual risk. This study explored the functional impact of TP53 missense variants on its transcription factor activity. We selected seven TP53 missense variants (c.129G > C, c.320A > G, c.417G > T, c.460G > A, c,522G > T, c.589G > A and c.997C > T) identified in Brazilian families at-risk for LFS. Variants were created through site-directed mutagenesis and transfected into SK-OV-3 cells to assess their transcription activation capabilities. Variants K139N and V197M displayed significantly reduced transactivation activity in a TP53-dependent luciferase reporter assay. Additionally, K139N negatively impacted CDKN1A and MDM2 expression and had a limited effect on GADD45A and PMAIP1 upon irradiation-induced DNA damage. Variant V197M demonstrated functional impact in all target genes evaluated and loss of Ser15 phosphorylation. K139N and V197M variants presented a reduction of p21 levels after irradiation. Our data show that K139N and V197M negatively impact p53 functions, supporting their classification as pathogenic variants. This underscores the significance of conducting functional studies on germline TP53 missense variants classified as variants of uncertain significance to ensure proper management of LFS-related cancer risks.
Collapse
Affiliation(s)
- Renata B V Abreu
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba, Brazil
| | - Ariane S Pereira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Marcela N Rosa
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Patricia Ashton-Prolla
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Department of Genetics, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Viviane A O Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
- Department of Pathology, School of Medicine, Federal University of Bahia, Salvador, Bahia, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, Brazil
| | - Matias E Melendez
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
- Molecular Carcinogenesis Program, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Edenir I Palmero
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil.
- Department of Genetics, Brazilian National Cancer Institute, Rio de Janeiro, Brazil.
| |
Collapse
|
50
|
Cutrona MB, Wu J, Yang K, Peng J, Chen T. Pancreatic cancer organoid-screening captures personalized sensitivity and chemoresistance suppression upon cytochrome P450 3A5-targeted inhibition. iScience 2024; 27:110289. [PMID: 39055940 PMCID: PMC11269815 DOI: 10.1016/j.isci.2024.110289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 04/12/2024] [Accepted: 06/13/2024] [Indexed: 07/28/2024] Open
Abstract
Cytochrome P450 3A5 (CYP3A5) has been proposed as a predictor of therapy response in subtypes of pancreatic ductal adenocarcinoma cancer (PDAC). To validate CYP3A5 as a therapeutic target, we developed a high-content image organoid-based screen to quantify the phenotypic responses to the selective inhibition of CYP3A5 enzymatic activity by clobetasol propionate (CBZ), using a cohort of PDAC-derived organoids (PDACOs). The chemoresistance of PDACOs to a panel of standard-of-care drugs, alone or in combination with CBZ, was investigated. PDACO pharmaco-profiling revealed CBZ to have anti-cancer activity that was dependent on the CYP3A5 level. In addition, CBZ restored chemo-vulnerability to cisplatin in a subset of PDACOs. A correlative proteomic analysis established that CBZ caused the suppression of multiple cancer pathways sustained by or associated with a mutant form of p53. Limiting the active pool of CYP3A5 enables targeted and personalized therapy to suppress pro-oncogenic mechanisms that fuel chemoresistance in some PDAC tumors.
Collapse
Affiliation(s)
- Meritxell B. Cutrona
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA
| | - Jing Wu
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA
| | - Ka Yang
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN 38105-3678, USA
| |
Collapse
|