1
|
Chen Y, Zhu X, Lin C, Xu R, Xu P, Xin L, Li L, Zhang L. circKCNQ5 promotes the proliferation of DNA-methyltransferase 3A R882 mutated acute myeloid leukemia cells by elevating high-mobility group box 1 expression. Ann Med 2025; 57:2478309. [PMID: 40130394 PMCID: PMC11938305 DOI: 10.1080/07853890.2025.2478309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 09/18/2024] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Patients with acute myeloid leukemia (AML) harboring the DNA-methyltransferase 3 A (DNMT3A) R882 mutation (DR882MUT) usually have a high recurrence rate and poor prognosis. circKCNQ5 levels were aberrantly elevated in patients with AML according to the microarray platform. Therefore, the purpose of this study is to investigate the effect and mechanism of circKCNQ5 on DR882MUT AML cell proliferation. METHODS A DR882MUT cell line model was established. circKCNQ5 expression in AML cells expressing wild-type DNMT3A (DNMT3A-WT) or DR882MUT was analyzed using RT-qPCR. The proliferation of DNMT3A-WT and DR882MUT AML cells after transfection was measured using a CCK-8 assay. High-mobility group box 1 (HMGB1) protein expression was assessed by western blotting. The regulatory mechanism of circKCNQ5 on HMGB1 expression was studied using RNA pull-down and co-immunoprecipitation assays. RESULTS circKCNQ5 expression increased gradually in HS-5, DNMT3A-WT, and DR882MUT AML cells. circKCNQ5 overexpression facilitated the proliferation of DNMT3A-WT KG-1a and HL-60 cells, whereas circKCNQ5 silencing blocked DR882MUT KG-1a and HL-60 cell proliferation. CircKCNQ5 interacts with HMGB1 and enhanced HMGB1 protein levels by inhibiting HMGB1 ubiquitination. HMGB1 protein levels increased gradually in HS-5, DNMT3A-WT, and DR882MUT AML cells. Furthermore, circKCNQ5 overexpression elevated HMGB1 protein levels in DNMT3A-WT KG-1a and HL-60 cells, whereas circKCNQ5 silencing reduced HMGB1 protein levels in DR882MUT KG-1a and HL-60 cells. HMGB1 overexpression remarkably increased the proliferative ability of DR882MUT KG-1a and HL-60 cells and circKCNQ5 silencing. CONCLUSIONS These findings verified that circKCNQ5 promotes the proliferation of DR882MUT AML cells by increasing HMGB1 expression.
Collapse
Affiliation(s)
- Yijian Chen
- Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, P. R. China
| | - Xiaodan Zhu
- Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, P. R. China
| | - Chuanming Lin
- Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, P. R. China
| | - Rong Xu
- Department of Endocrinology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, P. R. China
| | - Pengxiang Xu
- Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, P. R. China
| | - Liuyan Xin
- Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, P. R. China
| | - Lin Li
- Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, P. R. China
| | - Liqun Zhang
- Quality Control Department, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, P. R. China
| |
Collapse
|
2
|
Gozdecka M, Dudek M, Wen S, Gu M, Stopforth RJ, Rak J, Damaskou A, Grice GL, McLoughlin MA, Bond L, Wilson R, Giotopoulos G, Shanmugiah VM, Bakar RB, Yankova E, Cooper JL, Narayan N, Horton SJ, Asby R, Pask DC, Mupo A, Duddy G, Marando L, Georgomanolis T, Carter P, Ramesh AP, Dunn WG, Barcena C, Gallipoli P, Yusa K, Petrovski S, Wright P, Quiros PM, Frezza C, Nathan JA, Kaser A, Kar S, Tzelepis K, Mitchell J, Fabre MA, Huntly BJP, Vassiliou GS. Mitochondrial metabolism sustains DNMT3A-R882-mutant clonal haematopoiesis. Nature 2025; 642:431-441. [PMID: 40239706 PMCID: PMC12158785 DOI: 10.1038/s41586-025-08980-6] [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: 03/19/2024] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
Somatic DNMT3A-R882 codon mutations drive the most common form of clonal haematopoiesis (CH) and are associated with increased acute myeloid leukaemia (AML) risk1,2. Preventing expansion of DNMT3A-R882-mutant haematopoietic stem/progenitor cells (HSPCs) may therefore avert progression to AML. To identify DNMT3A-R882-mutant-specific vulnerabilities, we conducted a genome-wide CRISPR screen on primary mouse Dnmt3aR882H/+ HSPCs. Among the 640 vulnerability genes identified, many were involved in mitochondrial metabolism, and metabolic flux analysis confirmed enhanced oxidative phosphorylation use in Dnmt3aR882H/+ versus Dnmt3a+/+ (WT) HSPCs. We selected citrate/malate transporter Slc25a1 and complex I component Ndufb11, for which pharmacological inhibitors are available, for downstream studies. In vivo administration of SLC25A1 inhibitor CTPI2 and complex I inhibitors IACS-010759 and metformin suppressed post-transplantation clonal expansion of Dnmt3aR882H/+, but not WT, long-term haematopoietic stem cells. The effect of metformin was recapitulated using a primary human DNMT3A-R882 CH sample. Notably, analysis of 412,234 UK Biobank participants showed that individuals taking metformin had a markedly lower prevalence of DNMT3A-R882-mutant CH, after controlling for potential confounders including glycated haemoglobin, diabetes and body mass index. Collectively, our data propose modulation of mitochondrial metabolism as a therapeutic strategy for prevention of DNMT3A-R882-mutant AML.
Collapse
MESH Headings
- Animals
- Mice
- DNA (Cytosine-5-)-Methyltransferases/genetics
- DNA Methyltransferase 3A
- Humans
- Mitochondria/metabolism
- Mitochondria/drug effects
- Mitochondria/genetics
- Clonal Hematopoiesis/genetics
- Clonal Hematopoiesis/drug effects
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/prevention & control
- Leukemia, Myeloid, Acute/drug therapy
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/cytology
- Mutation/genetics
- Female
- Male
- Metformin/pharmacology
- Oxidative Phosphorylation/drug effects
- Clone Cells/metabolism
- Clone Cells/drug effects
- Clone Cells/cytology
- Electron Transport Complex I/antagonists & inhibitors
- Electron Transport Complex I/metabolism
Collapse
Affiliation(s)
- Malgorzata Gozdecka
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
| | - Monika Dudek
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Sean Wen
- Department of Haematology, University of Cambridge, Cambridge, UK
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals Research and Development, AstraZeneca, Cambridge, UK
| | - Muxin Gu
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Richard J Stopforth
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Justyna Rak
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Aristi Damaskou
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Guinevere L Grice
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Matthew A McLoughlin
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Laura Bond
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Rachael Wilson
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - George Giotopoulos
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Vijaya Mahalingam Shanmugiah
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Rula Bany Bakar
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Eliza Yankova
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Jonathan L Cooper
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Nisha Narayan
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Sarah J Horton
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Ryan Asby
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Dean C Pask
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | | | | | - Ludovica Marando
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Theodoros Georgomanolis
- Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University Hospital Cologne, Cologne, Germany
| | - Paul Carter
- Section of Cardiovascular Medicine, The Victor Phillip Dahdalleh Heart and Lung Research Institute, The University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, UK
| | - Amirtha Priya Ramesh
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - William G Dunn
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Clea Barcena
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Paolo Gallipoli
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Kosuke Yusa
- Stem Cell Genetics, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals Research and Development, AstraZeneca, Cambridge, UK
| | - Penny Wright
- Department of Anatomic Pathology, Canterbury Health Laboratories, Christchurch, New Zealand
| | - Pedro M Quiros
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Christian Frezza
- Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University Hospital Cologne, Cologne, Germany
- Institute of Genetics, Faculty of Mathematics and Natural Sciences, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - James A Nathan
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Arthur Kaser
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Siddhartha Kar
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Konstantinos Tzelepis
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- Milner Therapeutics Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Jonathan Mitchell
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals Research and Development, AstraZeneca, Cambridge, UK
| | - Margarete A Fabre
- Department of Haematology, University of Cambridge, Cambridge, UK
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals Research and Development, AstraZeneca, Cambridge, UK
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Brian J P Huntly
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - George S Vassiliou
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
| |
Collapse
|
3
|
Lawrence EM, Cooray A, Kueh AJ, Pal M, Tai L, Garnham AL, Li-Wai-Suen CSN, Vanyai H, Gouil Q, Lancaster J, Callegari S, Whelan L, Lieschke E, Thomas A, Strasser A, Liao Y, Shi W, Wei AH, Herold MJ. Transcriptomic changes including p53 dysregulation prime DNMT3A mutant cells for transformation. EMBO Rep 2025:10.1038/s44319-025-00450-4. [PMID: 40307617 DOI: 10.1038/s44319-025-00450-4] [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: 11/22/2024] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
DNMT3A mutations are prevalent in haematologic malignancies. In our mouse model the murine homologue (R878H) of the human 'hotspot' R882H mutation is introduced into the mouse Dnmt3a locus. This results in globally reduced DNA methylation in all tissues. Mice with heterozygous R878H DNMT3A mutations develop γ-radiation induced thymic lymphoma more rapidly than control mice, suggesting a vulnerability to stress stimuli in Dnmt3aR878H/+ cells. In competitive transplantations, Dnmt3aR878H/+ Lin-Sca-1+Kit+ (LSK) haematopoietic stem/progenitor cells (HSPCs) have a competitive advantage over WT HSPCs, indicating a self-renewal phenotype at the expense of differentiation. RNA sequencing of Dnmt3aR878H/+ LSKs exposed to low dose γ-radiation shows downregulation of the p53 pathway compared to γ-irradiated WT LSKs. Accordingly, reduced PUMA expression is observed by flow cytometry in the bone marrow of γ-irradiated Dnmt3aR878H/+ mice due to impaired p53 signalling. These findings provide new insights into how DNMT3A mutations cause subtle changes in the transcriptome of LSK cells which contribute to their increased self-renewal and propensity for malignant transformation.
Collapse
Affiliation(s)
- Erin M Lawrence
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Amali Cooray
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Andrew J Kueh
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Olivia Newton John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Martin Pal
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- School of Dentistry and Medical Sciences, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Lin Tai
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Olivia Newton John Cancer Research Institute, Heidelberg, Australia
| | - Alexandra L Garnham
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Connie S N Li-Wai-Suen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Hannah Vanyai
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Quentin Gouil
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Olivia Newton John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - James Lancaster
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Sylvie Callegari
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Lauren Whelan
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth Lieschke
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Oncogene Biology Laboratory, Francis Crick Institute, London, UK
| | - Annabella Thomas
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Andreas Strasser
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Yang Liao
- Olivia Newton John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Wei Shi
- Olivia Newton John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Andrew H Wei
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Marco J Herold
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
- Olivia Newton John Cancer Research Institute, Heidelberg, Australia.
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia.
| |
Collapse
|
4
|
Corvigno S, Yao J, Asare A, Zhao L, Celestino J, Hajek RA, Goette EAA, Rogers RT, Montoya RN, Song P, Zhang QC, Song X, Mohammad MM, Shaw KR, Zhang J, Lu KH, Jazaeri AA, Westin SN, Sood AK, Lee S. Longitudinal genomic profiling of chemotherapy-related CHIP variants in patients with ovarian cancer. Front Oncol 2025; 15:1538446. [PMID: 40365343 PMCID: PMC12069037 DOI: 10.3389/fonc.2025.1538446] [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: 12/02/2024] [Accepted: 03/14/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Clonal hematopoiesis (CH) is characterized by the presence of hematopoietic stem cells (HSCs) with the potential of clonally expanding and giving rise to hematological malignancies. Clonal hematopoiesis of indeterminate potential (CHIP) is the outgrowth of a single HSC clone with an acquired somatic mutation in the absence of hematological abnormalities. CHIP variants occur with a variant allele frequency (VAF) of at least 2% in peripheral blood. This definition does not account for less frequent mutations that give rise to hematopoietic clones. Previous studies indicate an association between CH and secondary hematologic malignancies in cancer patients who receive chemotherapy. Methods To discover novel candidate CHIP mutations, including those with extremely low VAFs, we performed an in-depth characterization of low-frequency CHIP variants in a highly selected group of patients with high-grade serous ovarian cancer (HGSC) before and after neoadjuvant chemotherapy (NACT). We performed comprehensive ultra-high-depth whole-exome sequencing of circulating free DNA (cfDNA) and matched white blood cell (WBC) DNA from pre- (n=9) and post-NACT (n=9) samples from HGSC patients who had excellent response (ER; n=4) or poor response (PR; n=5) to NACT. Results Variants present in both the WBC DNA and cfDNA from a patient were considered candidate CHIP variants. We identified 93,088 candidate CHIP variants in 13,780 genes. Compared with pre-NACT samples, post-NACT samples tended to have fewer CHIP mutations with VAFs of less than 5%, which may reflect the negative selective pressure of chemotherapy on rare hematopoietic clones. Finally, we identified CHIP variants in tumor samples matched to the liquid biopsies. Discussion Our innovative sequencing approach enabled the discovery of a large number of novel low-frequency candidate CHIP mutations, whose frequency and composition are affected by chemotherapy, in the cfDNA of patients with HGSC. The CHIP variants that were enriched after chemotherapy, if validated, might become essential predictive markers for therapy-related myeloid neoplasia.
Collapse
Affiliation(s)
- Sara Corvigno
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jun Yao
- Department of Molecular & Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Amma Asare
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Li Zhao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joseph Celestino
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Richard A. Hajek
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ency A. Arboleda Goette
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ridge T. Rogers
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Raymond N. Montoya
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ping Song
- Institute for Personalized Cancer Therapy (IPCT) Genomic Laboratory (IPCT Lab), Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Qingxiu C. Zhang
- Institute for Personalized Cancer Therapy (IPCT) Genomic Laboratory (IPCT Lab), Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xingzhi Song
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mohammad M. Mohammad
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kenna R. Shaw
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Karen H. Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Amir A. Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shannon N. Westin
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anil K. Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sanghoon Lee
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
5
|
Hosseini M, Voisin V, Chegini A, Varesi A, Cathelin S, Ayyathan DM, Liu ACH, Yang Y, Wang V, Maher A, Grignano E, Reisz JA, D'Alessandro A, Young K, Wu Y, Fiumara M, Ferrari S, Naldini L, Gaiti F, Pai S, Egan G, Schimmer AD, Bader GD, Dick JE, Xie SZ, Trowbridge JJ, Chan SM. Metformin reduces the competitive advantage of Dnmt3a R878H HSPCs. Nature 2025:10.1038/s41586-025-08871-w. [PMID: 40240595 DOI: 10.1038/s41586-025-08871-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/05/2025] [Indexed: 04/18/2025]
Abstract
Clonal haematopoiesis arises when a haematopoietic stem cell (HSC) acquires a mutation that confers a competitive advantage over wild-type HSCs, resulting in its clonal expansion. Individuals with clonal haematopoiesis are at increased risk of developing haematologic neoplasms and other age-related inflammatory illnesses1-4. Suppressing the expansion of mutant HSCs may prevent these outcomes; however, such interventions have not yet been identified. The most common clonal haematopoiesis driver mutations are in the DNMT3A gene, with arginine 882 (R882) being a mutation hotspot1-3,5-7. Here we show that mouse haematopoietic stem and progenitor cells (HSPCs) carrying the Dnmt3aR878H/+ mutation, equivalent to human DNMT3AR882H/+, have increased mitochondrial respiration compared with wild-type cells and are dependent on this metabolic reprogramming for their competitive advantage. Treatment with metformin, an anti-diabetic drug that inhibits mitochondrial respiration8, reduced the competitive advantage of Dnmt3aR878H/+ HSCs. Through a multi-omics approach, we found that metformin acts by enhancing methylation potential in Dnmt3aR878H/+ HSPCs and reversing the aberrant DNA CpG methylation and histone H3 K27 trimethylation profiles in these cells. Metformin also reduced the competitive advantage of human DNMT3AR882H HSPCs generated by prime editing. Our findings provide preclinical rationale for investigating metformin as a preventive intervention against DNMT3A R882 mutation-driven clonal haematopoiesis in humans.
Collapse
Affiliation(s)
| | - Veronique Voisin
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
| | - Ali Chegini
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Angelica Varesi
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Alex C H Liu
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Yitong Yang
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Vivian Wang
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Abdula Maher
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Eric Grignano
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kira Young
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - Yiyan Wu
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Martina Fiumara
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Samuele Ferrari
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Federico Gaiti
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Shraddha Pai
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Grace Egan
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Gary D Bader
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Steven M Chan
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
6
|
Mechahougui H, Gutmans J, Gouasmi R, Smekens L, Friedlaender A. BRAF Targeting Across Solid Tumors: Molecular Aspects and Clinical Applications. Int J Mol Sci 2025; 26:3757. [PMID: 40332392 PMCID: PMC12027668 DOI: 10.3390/ijms26083757] [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: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
BRAF mutations are critical drivers in cancers such as melanoma, colorectal cancer, and non-small-cell lung cancer. The most common mutation, BRAF V600E, is a key therapeutic target. Targeted treatments with BRAF and MEK inhibitors have significantly improved progression-free and overall survival in melanoma patients. However, in cancers like metastatic colorectal cancer, BRAF mutations are associated with poor outcomes due to aggressive disease behavior and resistance to conventional chemotherapy. Despite progress, resistance to BRAF/MEK inhibitors remains a major challenge, often driven by secondary mutations in the mitogen-activated protein kinase (MAPK) pathway, activation of alternative pathways such as phosphoinositide 3-kinases (PI3Ks)/protein kinase B (AKT), or changes in the tumor microenvironment. These challenges have motivated ongoing research into combining BRAF inhibitors with immunotherapies to enhance and prolong treatment effectiveness. Future research must also account for the role of the cancer's tissue of origin, as the biological context significantly influences response to targeted therapies, highlighting the need for a deeper understanding of tumor biology, micro-environment, and genetics.
Collapse
Affiliation(s)
- Hiba Mechahougui
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (J.G.); (L.S.)
| | - James Gutmans
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (J.G.); (L.S.)
| | - Roumaïssa Gouasmi
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, 69100 Lyon, France;
| | - Laure Smekens
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (J.G.); (L.S.)
| | | |
Collapse
|
7
|
Yan X, Chen HY, Wang L, Tian YL, Gu Y, Liu N, Ge Z. [Dynamic changes in genetic mutations in myelodysplastic neoplasms with progressive disease and leukemic transformation]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2025; 46:252-260. [PMID: 40355355 PMCID: PMC12038471 DOI: 10.3760/cma.j.cn121090-20240708-00254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Indexed: 05/14/2025]
Abstract
Objective: To investigate the key genetic mutations during the progressive disease (PD) /leukemic transformation (LT) course in MDS by analyzing the dynamic changes of genetic mutations in patients with myelodysplastic neoplasms (MDS) with or without PD/LT. Methods: This study enrolled 84 patients with sequential MDS from May 2019 to August 2023 at ZhongDa Hospital Southeast University and used the next generation sequencing to detect gene mutations. The dynamic changes of genetic mutations in patients with MDS with or without PD/LT were retrospectively analyzed. Results: ①This study analyzed data from 84 patients diagnosed with MDS with a median age of 63 (range: 31-95) years and consisting of 51 males and 33 females. Participants were distributed to the PD cohort (n=20), LT cohort (n=13), and non-PD/LT cohort (n=51). Patients from the PD/LT cohorts demonstrated a higher proportion of bone marrow blasts than the non-PD/LT cohort at the first sequencing (1.6% vs. 0.4%, P=0.013). ②The most frequently mutated genes that were detected at first sequencing were ASXL1 (n=21, 25.0%), TP53 (n=17, 20.2%), TET2 (n=12, 14.3%), DNMT3A (n=11, 13.1%), and U2AF1 (n=11, 13.1%). Further, patients from the PD/LT cohorts exhibited a higher median number of mutated genes than the non-PD/LT cohort (2 vs.1, P=0.014) at first sequencing. TET2 (27.3% vs. 5.9%, P=0.010), SETBP1 (15.2% vs.2.0%, P=0.033), and RUNX1 (18.2% vs. 2.0%, P=0.013) mutations were enriched in the PD/LT cohorts than in the non-PD/LT cohort. ③The most frequently detected acquired mutations (Ⅰ mutations) and clonally expanded mutations (Ⅱ mutations) were TP53 (n=9, 10.7%), TET2 (n=7, 8.3%), ASXL1 (n=7, 8.3%), and RAS pathway (n=7, 8.3%). Furthermore, patients from the PD/LT cohorts showed a higher median number of Ⅰ/Ⅱ genes than the non-PD/LT cohort (2 vs. 0, P<0.001), and Ⅰ/Ⅱ RAS pathway (21.2% vs. 0, P=0.001), TP53 (27.3% vs. 0, P<0.001), and TET2 (18.2% vs. 2.0%, P=0.013) mutations were enriched in PD/LT cohorts than in the non-PD/LT cohorts. ④Most of the TP53 mutations (9/12, 75.0%) in PD/LT cohorts were Ⅰ/Ⅱ mutations, whereas all of the TP53 mutations in non-PD/LT cohort were clone-decrease mutations (Ⅲ mutations) (5/8, 62.5%) or clone-stable mutations (Ⅳ mutations) (3/8, 37.5%). Most of the RAS pathway mutations (7/8,87.5%) in the PD/LT cohorts were Ⅰ/Ⅱ mutations, whereas only one patient in the non-PD/LT cohort demonstrated RAS pathway mutations, which belonged to Ⅳ mutations. Conclusion: Patients from the PD/LT cohorts demonstrated a higher proportion of bone marrow blasts and a higher median number of mutations than the non-PD/LT cohort at first sequencing; TET2, SETBP1, and RUNX1 mutations were enriched in the PD/LT cohorts than in the non-PD/LT cohort at first sequencing. Patients from the PD/LT cohorts exhibited a higher number of Ⅰ/Ⅱ mutations than the non-PD/LT cohort. Further, Ⅰ/Ⅱ TP53, RAS pathway, and TET2 mutations were enriched in the PD/LT cohorts, and Ⅰ/Ⅱ TP53 and RAS pathway mutations may contribute to the PD/LT.
Collapse
Affiliation(s)
- X Yan
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing 210009, China
| | - H Y Chen
- Xuyi People's Hospital, Xuyi 211700, China
| | - L Wang
- Xuyi People's Hospital, Xuyi 211700, China
| | - Y L Tian
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing 210009, China
| | - Y Gu
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing 210009, China
| | - N Liu
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing 210009, China
| | - Z Ge
- Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, Nanjing 210009, China
| |
Collapse
|
8
|
Fathima S, Alsugair A, He R, Mangaonkar AA, Begna KH, Pardanani A, Zepeda Mendoza CJ, Reichard KK, Gangat N, Tefferi A. Myeloid neoplasms with PHF6 mutations: context-dependent genomic and prognostic characterization in 176 informative cases. Blood Cancer J 2025; 15:28. [PMID: 40025027 PMCID: PMC11873042 DOI: 10.1038/s41408-025-01231-x] [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: 11/18/2024] [Revised: 01/10/2025] [Accepted: 02/11/2025] [Indexed: 03/04/2025] Open
Abstract
Recent reports suggest a favorable prognosis for PHF6 mutation (PHF6MUT) in chronic myelomonocytic leukemia (CMML) and unfavorable in acute myeloid leukemia (AML). We accessed 176 consecutive patients with a spectrum of myeloid neoplasms with PHF6MUT, including AML (N = 67), CMML (N = 49), myelodysplastic syndromes (MDS; N = 36), myeloproliferative neoplasms (MPN; N = 16), and MDS/MPN (N = 8). PHF6 mutations were classified as nonsense (43%) or frameshift (30%) with the PHD2 domain being the most frequently (64%) affected region. Median follow-up was 25 months with 110 (63%) deaths and 44 allogenic transplants. Our top-line observations include (a) a distinctly superior overall survival (OS; 81 vs. 18 months; p < 0.01) and blast transformation-free survival (BTFS; "not reached" vs. 44 months; p < 0.01) in patients with CMML vs. those with other myeloid neoplasms, (ii) a higher than expected frequency of isolated loss of Y chromosome, in the setting of CMML (16% vs. expected 6%) and MDS (8% vs expected 2.5%), (iii) a significant association, in MDS, between PHF6MUT variant allele fraction (VAF) > 20% and inferior OS (HR 3.0, 95% CI 1.1-8.1, multivariate p = 0.02) as well as female gender and inferior BTFS (HR 26.8, 95% CI 1.9-368.3, multivariate p = 0.01), (iv) a relatively favorable median post-transplant survival of 46 months. Multivariable analysis also identified high-risk karyotype (HR 5.1, 95% CI 1.2-20.9, p = 0.02), and hemoglobin <10 g/dL (HR 2.7, 95% CI 1.0-7.2, p = 0.04), as independent predictors of inferior OS in patients with MDS. The current study provides disease-specific information on genotype and prognosis of PHF6-mutated myeloid neoplasms.
Collapse
Affiliation(s)
| | - Ali Alsugair
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Rong He
- Department of Laboratory Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | - Ayalew Tefferi
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
9
|
Huang G, Cai X, Li D. Significance of targeting DNMT3A mutations in AML. Ann Hematol 2025; 104:1399-1414. [PMID: 39078434 PMCID: PMC12031811 DOI: 10.1007/s00277-024-05885-8] [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: 04/18/2024] [Accepted: 07/05/2024] [Indexed: 07/31/2024]
Abstract
Acute myeloid leukemia (AML) is the most prevalent form of leukemia among adults, characterized by aggressive behavior and significant genetic diversity. Despite decades of reliance on conventional chemotherapy as the mainstay treatment, patients often struggle with achieving remission, experience rapid relapses, and have limited survival prospects. While intensified induction chemotherapy and allogeneic stem cell transplantation have enhanced patient outcomes, these benefits are largely confined to younger AML patients capable of tolerating intensive treatments. DNMT3A, a crucial enzyme responsible for establishing de novo DNA methylation, plays a pivotal role in maintaining the delicate balance between hematopoietic stem cell differentiation and self-renewal, thereby influencing gene expression programs through epigenetic regulation. DNMT3A mutations are the most frequently observed genetic abnormalities in AML, predominantly in older patients, occurring in approximately 20-30% of adult AML cases and over 30% of AML with a normal karyotype. Consequently, the molecular underpinnings and potential therapeutic targets of DNMT3A mutations in AML are currently being thoroughly investigated. This article provides a comprehensive summary and the latest insights into the structure and function of DNMT3A, examines the impact of DNMT3A mutations on the progression and prognosis of AML, and explores potential therapeutic approaches for AML patients harboring DNMT3A mutations.
Collapse
MESH Headings
- Humans
- DNA Methyltransferase 3A
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/drug therapy
- DNA (Cytosine-5-)-Methyltransferases/genetics
- DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors
- DNA (Cytosine-5-)-Methyltransferases/metabolism
- Mutation
- DNA Methylation
- Epigenesis, Genetic
- Molecular Targeted Therapy
- Gene Expression Regulation, Leukemic
- Prognosis
Collapse
Affiliation(s)
- Guiqin Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoya Cai
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dengju Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
10
|
Shen Y, Fu S, Liu X, Liu J, Fu Y, Zhao Y, Wang X, Jiang X, Zhang J. Gene Mutation Characteristics and Prognostic Significance in Acute Myeloid Leukemia Patients From Northeast China. Hum Mutat 2025; 2025:7730186. [PMID: 40226311 PMCID: PMC11918257 DOI: 10.1155/humu/7730186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 12/18/2024] [Accepted: 01/17/2025] [Indexed: 04/15/2025]
Abstract
A great part of studies on the correlation between gene mutations and prognosis in acute myeloid leukemia (AML) patients are based on Western populations. To profile the genomic landscape of AML patients in Northeast China, we retrospectively analyzed the clinical data of 377 newly diagnosed AML patients in Shengjing Hospital of China Medical University from 2016 to 2022 and compared them with data from other populations with different genetic backgrounds. The mutation status of NPM1, FLT3-ITD, FLT3-TKD, CEBPA (CCAT enhancer binding protein alpha), ASXL1, TET2, KIT, DNMT3A (DNA methyltransferase 3A), IDH1, IDH2, EZH2 (enhancer of zeste 2), RUNX1, TP53, NRAS, and GATA2 was acquired by next-generation sequencing (NGS) technology; meanwhile, the clinical data of the patients were collected. The Cox regression model was used to analyze factors affecting patient survival and the impact of CEBPA and DNMT3A mutation on prognosis, and the results were different from those in other populations. Seventy-seven of 377 patients (20.4%) were detected with CEBPA mutations, which was higher than the 2%-6% in the Caucasian population. In the CEBPAdm patients who did not receive bone marrow transplantation, the prognosis of male patients (n = 18) was significantly better than that of female patients (n = 21) (p = 0.0242). Sixty-three of 377 patients (16.7%) carried the DNMT3A mutation, which was lower than the mutation frequency of 20.9% in the German-Austrian population, and the prognosis of these patients was significantly poorer (p = 0.0052). In addition, the prognostic evaluation value of the DNMT3A mutation in AML patients was not affected regardless of the presence of the NPM1 and FLT3-ITD comutation (p > 0.05), nor the mutation site of DNMT3A. In conclusion, for the Northeastern Chinese population, the prognosis of male patients with CEBPAdm was more favorable than that of female patients, and the DNMT3A mutation serves as an independent predictor of poor prognosis in AML. These results highlighted the central role of genetic background in precision medicine strategies and further emphasized the importance of the clinical characteristics of AML gene mutations in the Chinese population.
Collapse
Affiliation(s)
- Yiyang Shen
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuang Fu
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuan Liu
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianing Liu
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Fu
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yue Zhao
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xinxin Wang
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xujian Jiang
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jihong Zhang
- Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Pattalachinti VK, Haque E, Yousef M, Yousef A, Chowdhury S, Overman M, Parseghian CM, Morris VK, Kee B, Huey RW, Raghav K, Court CM, Shen JP. BRAF mutant appendiceal adenocarcinoma differs from colorectal cancer but responds to BRAF-targeted therapy. NPJ Precis Oncol 2025; 9:38. [PMID: 39910160 PMCID: PMC11799341 DOI: 10.1038/s41698-025-00821-z] [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: 07/17/2024] [Accepted: 01/21/2025] [Indexed: 02/07/2025] Open
Abstract
Appendiceal Adenocarcinoma (AA) is a rare gastrointestinal cancer with no FDA-approved targeted therapies. Here, we retrospectively compare BRAF-mutant AA and colorectal cancer (CRC). BRAF mutation is rare in AA (3%). Unlike CRC, BRAFV600E AA is not associated with poor prognosis, female sex, microsatellite instability, mucinous histology, or poor differentiation. In both cancers, BRAFV600E but not atypical BRAF mutations are mutually exclusive with other Ras-activating mutations. BRAFV600E + EGFR inhibition shows efficacy in BRAFV600E AA (disease control rate = 80%, median progression-free survival = 7.1 months).
Collapse
Affiliation(s)
- Vinay K Pattalachinti
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Joe R. and Teresa Lozano Long School of Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Emaan Haque
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Abdelrahman Yousef
- Internal Medicine Department, University of New Mexico Hospital, Albuquerque, NM, USA
| | - Saikat Chowdhury
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christine M Parseghian
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Van K Morris
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bryan Kee
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ryan W Huey
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kanwal Raghav
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Colin M Court
- Department of Surgical Oncology and Endocrine Surgery, Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, USA
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
12
|
Rasool M, Haque A, Alharthi M, Sibiany A, Alamri MS, Alqarni SMH, Rather IA, Chaudhary AG, Karim S, Pushparaj PN. The mutational spectrum of NRAS gene discovers a novel frameshift mutation (E49R) in Saudi colorectal cancer patients. Cancer Cell Int 2025; 25:21. [PMID: 39844204 PMCID: PMC11755827 DOI: 10.1186/s12935-025-03652-x] [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: 04/18/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
Colorectal cancer (CRC) is a major health problem the world face currently and one of the leading causes of death worldwide. CRC is genetically heterogeneous and multiple genetic aberrations may appear on course of the disease throughout patient's lifetime. Genetic biomarkers such as BRAF, KRAS, and NRAS may provide early precision treatment options that are crucial for patient survival and well-being. The aim of this study was to identify pathogenic mutations in the NRAS gene causing colorectal cancer in the Saudi population. We enrolled 80 CRC tumor tissue samples and performed molecular analyses to establish the mutation spectrum status in the western region of Saudi Arabia. We identified 5 different mutations in 10 patients, 4 of whom were reported previously (G10R, E37K, Q61K, and Q61*) in the literature while we discovered one novel lethal insertion mutation (E49R). A novel identified insertion mutation was present in the third codon of the NRAS gene [c.145 insA (p.Glu49ArgTer85)], causing a frameshift in the amino acid sequence of the protein, and leading to an aberrant and truncated protein of 85 amino acids. Subsequent bioinformatics analysis showed that the mutation was highly deleterious and affected protein function to a greater extent. This identification may further improve the prognosis of CRC and benefit subsequent treatment choices.
Collapse
Affiliation(s)
- Mahmood Rasool
- Institute of Genomic Medicine Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Absarul Haque
- King Fahd Medical Research Center, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Alharthi
- Faculty of Medicine, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulrahman Sibiany
- Faculty of Medicine, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Saad Alamri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Irfan A Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adeel G Chaudhary
- Institute of Genomic Medicine Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sajjad Karim
- Institute of Genomic Medicine Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | | |
Collapse
|
13
|
Garbe C, Amaral T, Peris K, Hauschild A, Arenberger P, Basset-Seguin N, Bastholt L, Bataille V, Brochez L, Del Marmol V, Dréno B, Eggermont AMM, Fargnoli MC, Forsea AM, Höller C, Kaufmann R, Kelleners-Smeets N, Lallas A, Lebbé C, Leiter U, Longo C, Malvehy J, Moreno-Ramirez D, Nathan P, Pellacani G, Saiag P, Stockfleth E, Stratigos AJ, Van Akkooi ACJ, Vieira R, Zalaudek I, Lorigan P, Mandala M. European consensus-based interdisciplinary guideline for melanoma. Part 1: Diagnostics - Update 2024. Eur J Cancer 2025; 215:115152. [PMID: 39700658 DOI: 10.1016/j.ejca.2024.115152] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024]
Abstract
This guideline was developed in close collaboration with multidisciplinary experts from the European Association of Dermato-Oncology (EADO), the European Dermatology Forum (EDF) and the European Organization for Research and Treatment of Cancer (EORTC). Recommendations for the diagnosis and treatment of melanoma were developed on the basis of systematic literature research and consensus conferences. Cutaneous melanoma (CM) is the most dangerous form of skin tumor and accounts for 90 % of skin cancer mortality. The diagnosis of melanoma can be made clinically and must always be confirmed by dermoscopy. If melanoma is suspected, a histopathological examination is always required. Sequential digital dermoscopy and whole-body photography can be used in high-risk patients to improve the detection of early-stage melanoma. If available, confocal reflectance microscopy can also improve the clinical diagnosis in special cases. Melanoma is classified according to the 8th version of the American Joint Committee on Cancer classification. For thin melanomas up to a tumor thickness of 0.8 mm, no further diagnostic imaging is required. From stage IB, lymph node sonography is recommended, but no further imaging examinations. From stage IIB/C, whole-body examinations with computed tomography or positron emission tomography CT in combination with magnetic resonance imaging of the brain are recommended. From stage IIB/C and higher, a mutation test is recommended, especially for the BRAF V600 mutation. It is important to perform a structured follow-up to detect relapses and secondary primary melanomas as early as possible. A stage-based follow-up regimen is proposed, which in the experience of the guideline group covers the optimal requirements, although further studies may be considered. This guideline is valid until the end of 2026.
Collapse
Affiliation(s)
- Claus Garbe
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany.
| | - Teresa Amaral
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | - Ketty Peris
- Institute of Dermatology, Università Cattolica, Rome, and Fondazione Policlinico Universitario A. Gemelli - IRCCS, Rome, Italy
| | - Axel Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Kiel, Germany
| | - Petr Arenberger
- Department of Dermatovenereology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Nicole Basset-Seguin
- Université Paris Cite, AP-HP department of Dermatology INSERM U 976 Hôpital Saint Louis Paris France
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Denmark
| | - Veronique Bataille
- Twin Research and Genetic Epidemiology Unit, School of Basic & Medical Biosciences, King's College London, London SE1 7EH, UK
| | - Lieve Brochez
- Department of Dermatology, Ghent University Hospital, Ghent, Belgium
| | - Veronique Del Marmol
- Department of Dermatology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Brigitte Dréno
- Nantes Université, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302/EMR6001, F-44000 Nantes, France
| | - Alexander M M Eggermont
- University Medical Center Utrecht & Princess Maxima Center, Utrecht, the Netherlands; Comprehensive Cancer Center Munich of the Technical University Munich and the Ludwig Maximilians University, Munich, Germany
| | | | - Ana-Maria Forsea
- Dermatology Department, Elias University Hospital, Carol Davila University of Medicine and Pharmacy Bucharest, Romania
| | - Christoph Höller
- Department of Dermatology, Medical University of Vienna, Austria
| | - Roland Kaufmann
- Department of Dermatology, Venereology and Allergology, Frankfurt University Hospital, Frankfurt, Germany
| | - Nicole Kelleners-Smeets
- Department of Dermatology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Aimilios Lallas
- First Department of Dermatology, Aristotle University, Thessaloniki, Greece
| | - Celeste Lebbé
- Université Paris Cite, AP-HP department of Dermatology INSERM U 976 Hôpital Saint Louis Paris France
| | - Ulrike Leiter
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | - Caterina Longo
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, and Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Skin Cancer Centre, Reggio Emilia, Italy
| | - Josep Malvehy
- Melanoma Unit, Department of Dermatology, Hospital Clinic, IDIBAPS, Barcelona, Spain; University of Barcelona, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - David Moreno-Ramirez
- Medical-&-Surgical Dermatology Service. Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Paul Nathan
- Mount Vernon Cancer Centre, Northwood United Kingdom
| | | | - Philippe Saiag
- University Department of Dermatology, Université de Versailles-Saint Quentin en Yvelines, APHP, Boulogne, France
| | - Eggert Stockfleth
- Skin Cancer Center, Department of Dermatology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Alexander J Stratigos
- 1st Department of Dermatology, National and Kapodistrian University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece
| | - Alexander C J Van Akkooi
- Melanoma Institute Australia, The University of Sydney, and Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Ricardo Vieira
- Department of Dermatology and Venereology, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Iris Zalaudek
- Dermatology Clinic, Maggiore Hospital, University of Trieste, Trieste, Italy
| | - Paul Lorigan
- The University of Manchester, Oxford Rd, Manchester M13 9PL, UK
| | - Mario Mandala
- University of Perugia, Unit of Medical Oncology, Santa Maria della Misericordia Hospital, Perugia, Italy
| |
Collapse
|
14
|
Melosky B, Juergens RA, Banerji S, Sacher A, Wheatley-Price P, Snow S, Tsao MS, Leighl NB, Martins I, Cheema P, Liu G, Chu QSC. The continually evolving landscape of novel therapies in oncogene-driven advanced non-small-cell lung cancer. Ther Adv Med Oncol 2025; 17:17588359241308784. [PMID: 39776537 PMCID: PMC11705342 DOI: 10.1177/17588359241308784] [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: 10/09/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Non-small-cell lung cancer (NSCLC) is a highly heterogeneous disease that is frequently associated with a host of known oncogenic alterations. Advances in molecular diagnostics and drug development have facilitated the targeting of novel alterations such that the majority of NSCLC patients have driver mutations that are now clinically actionable. The goal of this review is to gain insights into clinical research and development principles by summary, analysis, and discussion of data on agents targeting known alterations in oncogene-driven, advanced NSCLC beyond those in the epidermal growth factor receptor (EGFR) and the anaplastic lymphoma kinase (ALK). A search of published and presented literature was conducted to identify prospective trials and integrated analyses reporting outcomes for agents targeting driver gene alterations (except those in EGFR and ALK) in molecularly selected, advanced NSCLC. Clinical efficacy data were extracted from eligible reports and summarized in text and tables. Findings show that research into alteration-directed therapies in oncogene-driven, advanced NSCLC is an extremely active research field. Ongoing research focuses on the expansion of new agents targeting both previously identified targets (particularly hepatocyte growth factor receptor (MET), human epidermal growth factor receptor 2 (HER2), and Kirsten rat sarcoma viral oncogene homolog (KRAS)) as well as novel, potentially actionable targets (such as neuregulin-1 (NRG1) and phosphatidylinositol 3-kinase (PI3K)). The refinement of biomarker selection criteria and the development of more selective and potent agents are allowing for increasingly specific and effective therapies and the expansion of clinically actionable alterations. Clinical advances in this field have resulted in a large number of regulatory approvals over the last 3 years. Future developments should focus on the continued application of alteration therapy matching principles and the exploration of novel ways to target oncogene-driven NSCLC.
Collapse
Affiliation(s)
- Barbara Melosky
- Medical Oncology, BC Cancer Agency—Vancouver, University of British Columbia, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
| | | | - Shantanu Banerji
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Adrian Sacher
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Paul Wheatley-Price
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Stephanie Snow
- QEII Health Sciences Centre, Dalhousie University, Halifax, NS, Canada
| | - Ming-Sound Tsao
- University Health Network and Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Natasha B. Leighl
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | | | - Parneet Cheema
- William Osler Health System, University of Toronto, Brampton, ON, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Quincy S. C. Chu
- Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
15
|
Afsar S, Syed RU, Khojali WMA, Masood N, Osman ME, Jyothi JS, Hadi MA, Khalifa AAS, Aboshouk NAM, Alsaikhan HA, Alafnan AS, Alrashidi BA. Non-coding RNAs in BRAF-mutant melanoma: targets, indicators, and therapeutic potential. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:297-317. [PMID: 39167168 DOI: 10.1007/s00210-024-03366-3] [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: 06/12/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Melanoma, a highly aggressive skin cancer, is often driven by BRAF mutations, such as the V600E mutation, which promotes cancer growth through the MAPK pathway and contributes to treatment resistance. Understanding the role of non-coding RNAs (ncRNAs) in these processes is crucial for developing new therapeutic strategies. This review aims to elucidate the relationship between ncRNAs and BRAF mutations in melanoma, focusing on their regulatory roles and impact on treatment resistance. We comprehensively reviewed current literature to synthesize evidence on ncRNA-mediated regulation of BRAF-mutant melanoma and their influence on therapeutic responses. Key ncRNAs, including microRNAs and long ncRNAs, were identified as significant regulators of melanoma development and therapy resistance. MicroRNAs such as miR-15/16 and miR-200 families modulate critical pathways like Wnt signaling and melanogenesis. Long ncRNAs like ANRIL and SAMMSON play roles in cell growth, invasion, and drug susceptibility. Specific ncRNAs, such as BANCR and RMEL3, intersect with the MAPK pathway, highlighting their potential as therapeutic targets or biomarkers in BRAF-mutant melanoma. Additionally, ncRNAs involved in drug resistance, such as miR-579-3p and miR-1246, target processes like autophagy and immune checkpoint regulation. This review highlights the pivotal roles of ncRNAs in regulating BRAF-mutant melanoma and their contribution to drug resistance. These findings underscore the potential of ncRNAs as biomarkers and therapeutic targets, paving the way for innovative treatments to improve outcomes for melanoma patients.
Collapse
Affiliation(s)
- S Afsar
- Department of Virology, Sri Venkateswara University, Tirupathi, Andhra Pradesh, 517502, India.
| | - Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, 81442, Hail, Saudi Arabia.
| | - Weam M A Khojali
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hail, 81442, Hail, Saudi Arabia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Omdurman Islamic University, Omdurman, 14415, Sudan
| | - Najat Masood
- Chemistry Department, Faculty of Science, University of Ha'il, P.O. Box 2440, 81451, Ha'il,, Saudi Arabia
| | - Mhdia Elhadi Osman
- Department of Clinical Pharmacy, Faculty of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - J Siva Jyothi
- Department of Pharmaceutics, Hindu College of Pharmacy, Andhra Pradesh, India
| | - Mohd Abdul Hadi
- Department of Pharmaceutics, Bhaskar Pharmacy College, Moinabad, R.R.District, Hyderabad, 500075, Telangana, India
| | - Amna Abakar Suleiman Khalifa
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, 81442, Hail, Saudi Arabia
| | - Nayla Ahmed Mohammed Aboshouk
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, 81442, Hail, Saudi Arabia
| | | | | | | |
Collapse
|
16
|
Yang X, Li X, Zhang YL, Zhang SN, Zhang M, Sun C, Yang L, Liu HM, Yuan S. Targeting undruggable protein KRAS for cancer therapy: novel opportunities and challenges. Future Med Chem 2025; 17:143-147. [PMID: 39713985 PMCID: PMC11749466 DOI: 10.1080/17568919.2024.2444865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024] Open
Affiliation(s)
- Xin Yang
- Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Xiang Li
- Department of Obstetrics and Gynecology, Zhengzhou Key Laboratory of Endometrial Disease Prevention and Treatment Zhengzhou China, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yue-Lin Zhang
- Department of Obstetrics and Gynecology, Zhengzhou Key Laboratory of Endometrial Disease Prevention and Treatment Zhengzhou China, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sheng-Nan Zhang
- Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Miao Zhang
- Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Chong Sun
- Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Li Yang
- Department of Obstetrics and Gynecology, Zhengzhou Key Laboratory of Endometrial Disease Prevention and Treatment Zhengzhou China, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong-Min Liu
- Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou, China
| | - Shuo Yuan
- Henan Children’s Hospital, Zhengzhou Children’s Hospital, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou, China
- School of Pharmaceutical Sciences & Key Laboratory of Advanced Drug Preparation Technologies, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Stein RA, Gomaa FE, Raparla P, Riber L. Now and then in eukaryotic DNA methylation. Physiol Genomics 2024; 56:741-763. [PMID: 39250426 DOI: 10.1152/physiolgenomics.00091.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024] Open
Abstract
Since the mid-1970s, increasingly innovative methods to detect DNA methylation provided detailed information about its distribution, functions, and dynamics. As a result, new concepts were formulated and older ones were revised, transforming our understanding of the associated biology and catalyzing unprecedented advances in biomedical research, drug development, anthropology, and evolutionary biology. In this review, we discuss a few of the most notable advances, which are intimately intertwined with the study of DNA methylation, with a particular emphasis on the past three decades. Examples of these strides include elucidating the intricacies of 5-methylcytosine (5-mC) oxidation, which are at the core of the reversibility of this epigenetic modification; the three-dimensional structural characterization of eukaryotic DNA methyltransferases, which offered insights into the mechanisms that explain several disease-associated mutations; a more in-depth understanding of DNA methylation in development and disease; the possibility to learn about the biology of extinct species; the development of epigenetic clocks and their use to interrogate aging and disease; and the emergence of epigenetic biomarkers and therapies.
Collapse
Affiliation(s)
- Richard A Stein
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, New York, United States
| | - Faris E Gomaa
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, New York, United States
| | - Pranaya Raparla
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, New York, United States
| | - Leise Riber
- Department of Plant and Environmental Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
18
|
Wang H, Tang R, Jiang L, Jia Y. The role of PIK3CA gene mutations in colorectal cancer and the selection of treatment strategies. Front Pharmacol 2024; 15:1494802. [PMID: 39555098 PMCID: PMC11565213 DOI: 10.3389/fphar.2024.1494802] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
PIK3CA gene encodes the p110α catalytic subunit of PI3K, which regulates the PI3K/AKT/mTOR signaling pathway. PIK3CA gene mutation is one of the most common mutations in colorectal cancer (CRC), affecting about 15%-20% of CRC patients. PIK3CA gene mutation leads to the persistent activation of the PI3K/AKT/mTOR signaling pathway, which promotes the proliferation, invasion, metastasis, and drug resistance of CRC. This article provides a summary of the key detection methods for PIK3CA gene mutation, and provides an introduction to the existing colorectal cancer treatments and their practical applications in the clinic. Besides, this article summarizes the role and mechanism of PIK3CA gene mutation in the occurrence and development of CRC. It also explores the relationship between PIK3CA gene mutation and the clinical features and prognosis of CRC. This article focuses on the influence and mechanism of PIK3CA gene mutation on the targeted therapy and immunotherapy of CRC, and discusses the potential value and future direction of PIK3CA gene mutation in the personalized therapy of CRC. We aim to provide new perspectives and ideas for the precise diagnosis and treatment of CRC.
Collapse
Affiliation(s)
- Haitao Wang
- The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Rui Tang
- Chengdu Anorectal Hospital, Chengdu, China
| | - Ling Jiang
- Chengdu Anorectal Hospital, Chengdu, China
| | - Yingtian Jia
- Department of Anorectal, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
19
|
Wang M, Chen P, Li D, Zhao M. Prognostic impact of DTA mutation and co-occurring mutations in patients with myelodysplastic syndrome. Mol Biol Rep 2024; 51:985. [PMID: 39278886 DOI: 10.1007/s11033-024-09922-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/06/2024] [Indexed: 09/18/2024]
Abstract
OBJECTIVE To evaluate the frequency and prognostic significance of DTA (DNMT3A、TET2、ASXL1) gene mutation and co-occurring mutations in patients with myelodysplastic syndrome (MDS). METHODS The clinical data of 102 newly diagnosed MDS patients who accepted Next Generation Sequencing (NGS) was retrospectively analyzed. According to whether the patients had DTA gene mutation, the patients were divided into DTA mutated (DTA-mut) group and wild type (DTA-wt) group, and the relationship between gene mutation and clinical characteristics and prognosis was analyzed. RESULTS Among the 102 MDS patients, 96% (98/102) presented with mutation, while the mean number of mutations was 3.04 mutations/patient. DTA-mut was detected in 56.9% (58/102) patients. The most frequent co-mutated genes in DTA-mut group were SF3B1 (25.8%), RUNX1 (24.1%), U2AF1 (18.9%), SRSF2, EZH2, SETBP1 (17.2%), STAG2 (15.5%), IDH2 (12.1%) and BCOR, CBL (10.3%). The two groups showed no significant differences in ages, blood parameters, bone marrow blasts, WHO 2022 classification, IPSS-R risk category and rate of conversion to leukemia. Compared with the DTA-wt group, the mutation frequency of RUNX1 was higher (P = 0.02), while mutation frequency of TP53 was lower (P = 0.001) and the mutation frequency of ≥ 3 co-mutated genes was higher in DTA-mut group (P = 0.00). Survival analysis showed that the overall survivals (OS) of DTA-mut patients was significantly inferior to that of DTA-wt patients (P = 0.0332). According to IPSS-R classification, a statistically significant difference in OS was only observed in higher risk (IPSS-R > 3.5) group (P = 0.0058). In the context of DTA mutation, the OS of patients with RUNX1 mutation was shorter than that of patients without RUNX1 mutation significantly (P = 0.0074). The OS of patients with SF3B1 mutation was longer than that of patients without SF3B1 mutation, but there was no statistical difference (P = 0.0827). DTA mutations were not independent prognostic factors when DTA and co-mutated genes with frequency > 10% were considered in Cox regression model (P = 0.329). However, multivariate analysis confirmed an independently adverse prognosis of RUNX1 co-mutation (P = 0.042, HR = 2.426, 95% CI:1.031-5.711) in DTA-mut cohort. Moreover, our multivariable analysis suggests that SRSF2-mut was an independent poor prognostic factor for all MDS patients (P = 0.047), but lost significance (P = 0.103) for DTA-mut patients. CONCLUSIONS DTA mutations are frequently observed in patients with MDS, often accompanied by genes involved in RNA splicing and transcription factors like SF3B1 and RUNX1. DTA and concomitant mutations affect prognosis in MDS patients and RUNX1 was identified as an independent poor prognostic factor in patients with DTA mutations.
Collapse
Affiliation(s)
- Min Wang
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
- Department of Hematology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ping Chen
- Department of Hematology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Daqi Li
- Department of Hematology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China.
| |
Collapse
|
20
|
Hartwich TMP, Mansolf M, Demirkiran C, Greenman M, Bellone S, McNamara B, Nandi SP, Alexandrov LB, Yang‐Hartwich Y, Coma S, Pachter J, Santin AD. Preclinical evaluation of avutometinib and defactinib in high-grade endometrioid endometrial cancer. Cancer Med 2024; 13:e70210. [PMID: 39240189 PMCID: PMC11378359 DOI: 10.1002/cam4.70210] [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: 04/05/2024] [Revised: 08/02/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND High-grade endometrial cancers (EAC) are aggressive tumors with a high risk of progression after treatment. As EAC may harbor mutations in the RAS/MAPK pathways, we evaluated the preclinical in vitro and in vivo efficacy of avutometinib, a RAF/MEK clamp, in combination with the focal adhesion kinase (FAK) inhibitors defactinib or VS-4718, against multiple primary EAC cell lines and xenografts. METHODS Whole-exome sequencing (WES) was used to evaluate the genetic landscape of five primary EAC cell lines. The in vitro activity of avutometinib and defactinib as single agents and in combination was evaluated using cell viability, cell cycle, and cytotoxicity assays. Mechanistic studies were performed using Western blot assays while in vivo experiments were completed in UTE10 engrafted mice treated with either vehicle, avutometinib, VS-4718, or their combination through oral gavage. RESULTS WES results demonstrated multiple EAC cell lines to harbor genetic derangements in the RAS/MAPK pathway including KRAS/PTEN/PIK3CA/BRAF/ARID1A, potentially sensitizing to FAK and RAF/MEK inhibition. Five out of five of the EAC cell lines demonstrated in vitro sensitivity to FAK and/or RAF/MEK inhibition. By Western blot assays, exposure of EAC cell lines to defactinib, avutometinib, and their combination demonstrated decreased phosphorylated FAK (p-FAK) as well as decreased p-MEK and p-ERK. In vivo the combination of avutometinib/VS-4718 demonstrated superior tumor growth inhibition compared to single-agent treatment and controls starting at Day 9 (p < 0.02 and p < 0.04) in UTE10 xenografts. CONCLUSIONS Avutometinib, defactinib, and to a larger extent their combinations, demonstrated promising in vitro and in vivo activity against EAC cell lines and xenografts. These preclinical data support the potential clinical evaluation of this combination in high-grade EAC patients.
Collapse
Affiliation(s)
| | - Miranda Mansolf
- Department of Obstetrics, Gynecology, and Reproductive SciencesYale UniversityNew HavenConnecticutUSA
| | - Cem Demirkiran
- Department of Obstetrics, Gynecology, and Reproductive SciencesYale UniversityNew HavenConnecticutUSA
| | - Michelle Greenman
- Department of Obstetrics, Gynecology, and Reproductive SciencesYale UniversityNew HavenConnecticutUSA
| | - Stefania Bellone
- Department of Obstetrics, Gynecology, and Reproductive SciencesYale UniversityNew HavenConnecticutUSA
| | - Blair McNamara
- Department of Obstetrics, Gynecology, and Reproductive SciencesYale UniversityNew HavenConnecticutUSA
| | - Shuvro P. Nandi
- Department of Cellular and Molecular MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Ludmil B. Alexandrov
- Department of Cellular and Molecular MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Yang Yang‐Hartwich
- Department of Obstetrics, Gynecology, and Reproductive SciencesYale UniversityNew HavenConnecticutUSA
| | | | | | - Alessandro D. Santin
- Department of Obstetrics, Gynecology, and Reproductive SciencesYale UniversityNew HavenConnecticutUSA
| |
Collapse
|
21
|
Tomuleasa C, Tigu AB, Munteanu R, Moldovan CS, Kegyes D, Onaciu A, Gulei D, Ghiaur G, Einsele H, Croce CM. Therapeutic advances of targeting receptor tyrosine kinases in cancer. Signal Transduct Target Ther 2024; 9:201. [PMID: 39138146 PMCID: PMC11323831 DOI: 10.1038/s41392-024-01899-w] [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: 01/19/2024] [Revised: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 08/15/2024] Open
Abstract
Receptor tyrosine kinases (RTKs), a category of transmembrane receptors, have gained significant clinical attention in oncology due to their central role in cancer pathogenesis. Genetic alterations, including mutations, amplifications, and overexpression of certain RTKs, are critical in creating environments conducive to tumor development. Following their discovery, extensive research has revealed how RTK dysregulation contributes to oncogenesis, with many cancer subtypes showing dependency on aberrant RTK signaling for their proliferation, survival and progression. These findings paved the way for targeted therapies that aim to inhibit crucial biological pathways in cancer. As a result, RTKs have emerged as primary targets in anticancer therapeutic development. Over the past two decades, this has led to the synthesis and clinical validation of numerous small molecule tyrosine kinase inhibitors (TKIs), now effectively utilized in treating various cancer types. In this manuscript we aim to provide a comprehensive understanding of the RTKs in the context of cancer. We explored the various alterations and overexpression of specific receptors across different malignancies, with special attention dedicated to the examination of current RTK inhibitors, highlighting their role as potential targeted therapies. By integrating the latest research findings and clinical evidence, we seek to elucidate the pivotal role of RTKs in cancer biology and the therapeutic efficacy of RTK inhibition with promising treatment outcomes.
Collapse
Affiliation(s)
- Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania.
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania.
| | - Adrian-Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Cristian-Silviu Moldovan
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - David Kegyes
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Anca Onaciu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gabriel Ghiaur
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Department of Leukemia, Sidney Kimmel Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hermann Einsele
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Universitätsklinikum Würzburg, Medizinische Klinik II, Würzburg, Germany
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
22
|
Haisma B, Schols SEM, van Oerle RGM, Verbeek-Knobbe K, Hellenbrand D, Verwoerd EJ, Heubel-Moenen FCJI, Stroobants AK, Meijer D, Rijpma SR, Henskens YMC. Comparative analysis of thrombin generation platforms for patients with coagulation factor deficiencies: A comprehensive assessment. Thromb Res 2024; 240:109045. [PMID: 38834002 DOI: 10.1016/j.thromres.2024.109045] [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: 05/13/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
INTRODUCTION Thrombin generation assays (TGAs) assess the overall functionality of the hemostatic system and thereby provide a reflection of the hemostatic capacity of patients with disorders in this system. Currently, four (semi-)automated TGA platforms are available: the Calibrated Automated Thrombogram, Nijmegen Hemostasis Assay, ST Genesia and Ceveron s100. In this study, we compared their performance for detecting patients with congenital single coagulation factor deficiencies. MATERIALS AND METHODS Pooled patient samples, healthy control samples and normal pooled plasma were tested on all four platforms, using the available reagents that vary in tissue factor and phospholipid concentrations. The TGA parameters selected for analysis were peak height and thrombin potential. Results were normalized by using the calculated mean of healthy controls and a correction for between-run variation. Outcomes were presented as relative values, with the mean of healthy controls standardized to 100 %. RESULTS Across all platforms and reagents used, thrombin potentials and peak heights of samples with coagulation factor deficiencies were lower than those of healthy controls. Reagents designed for bleeding tendencies yielded the lowest values on all platforms (relative median peak height 19-32 %, relative median thrombin potential 19-45 %). Samples representing more severe coagulation factor deficiencies generally exhibited lower relative peak heights and thrombin potentials. CONCLUSIONS Thrombin generation assays prove effective in differentiating single coagulation factor deficient samples from healthy controls, with modest discrepancies observed between the platforms. Reagents designed for assessing bleeding tendencies, featuring the lowest tissue factor and phospholipid concentrations, emerged as the most suitable option for detecting coagulation factor deficiencies.
Collapse
Affiliation(s)
- Bauke Haisma
- Department of Hematology, Radboud university medical center, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands; Hemophilia Treatment Center Nijmegen-Eindhoven-Maastricht, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands.
| | - Saskia E M Schols
- Department of Hematology, Radboud university medical center, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands; Hemophilia Treatment Center Nijmegen-Eindhoven-Maastricht, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands.
| | - René G M van Oerle
- Central Diagnostic Laboratory, Maastricht University Medical Center+, P. Debyelaan 25, 6229 HX Maastricht, the Netherlands; Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.
| | - Kitty Verbeek-Knobbe
- Hemophilia Treatment Center Nijmegen-Eindhoven-Maastricht, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands; Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands.
| | - Dave Hellenbrand
- Hemophilia Treatment Center Nijmegen-Eindhoven-Maastricht, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands; Central Diagnostic Laboratory, Maastricht University Medical Center+, P. Debyelaan 25, 6229 HX Maastricht, the Netherlands.
| | - Evelien J Verwoerd
- Hemophilia Treatment Center Nijmegen-Eindhoven-Maastricht, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands; Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands.
| | - Floor C J I Heubel-Moenen
- Hemophilia Treatment Center Nijmegen-Eindhoven-Maastricht, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands; Department of Hematology, Internal Medicine, Maastricht University Medical Center+, P. Debyelaan 25, 6229 HX Maastricht, the Netherlands.
| | - An K Stroobants
- Department of Laboratory Medicine, Radboudumc Laboratory of Diagnostics, Radboud university medical center, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands.
| | - Danielle Meijer
- Hemophilia Treatment Center Nijmegen-Eindhoven-Maastricht, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands; Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands.
| | - Sanna R Rijpma
- Department of Hematology, Radboud university medical center, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands; Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands.
| | - Yvonne M C Henskens
- Hemophilia Treatment Center Nijmegen-Eindhoven-Maastricht, Geert Grooteplein Zuid 10, 5425 GA Nijmegen, the Netherlands; Central Diagnostic Laboratory, Maastricht University Medical Center+, P. Debyelaan 25, 6229 HX Maastricht, the Netherlands; Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.
| |
Collapse
|
23
|
Liu Y, Li M, Guo Y, Zhang Z, Du L, Zhang X, Wang Y, Zhang D, Xue L, Lei B, Su J, Zhang R, Chen J, Zhang X, Jia Q, Tian C. A patient with BRAF N581S mutation-positive lung adenocarcinoma demonstrates durable response to combined anlotinib and tislelizumab: A case report and literature review. Pathol Res Pract 2024; 259:155371. [PMID: 38820929 DOI: 10.1016/j.prp.2024.155371] [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: 12/25/2023] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND Targeted therapy with combined dabrafenib and trametinib has been proven to provide clinical benefits in patients with BRAF V600E mutation-positive NSCLC. Nevertheless, the treatment strategy for NSCLC patients with BRAF non-V600E mutations remains limited. CASE PRESENTATION Here, we present a NSCLC patient with a BRAF N581S mutation, which is a class III BRAF mutation, and this patient had a durable response to targeted therapy with combined anlotinib and tislelizumab. CONCLUSION We hope to bring more attention to rare non-V600 BRAF mutations by presenting this case of NSCLC.
Collapse
Affiliation(s)
- Ying Liu
- Department of Precision Medicine Center, Sanmenxia Central Hospital, Sanmenxia, China
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yu Guo
- Department of Precision Medicine Center, Sanmenxia Central Hospital, Sanmenxia, China
| | - Zhiyong Zhang
- Department of Traditional Chinese Medicine, Sanmenxia Central Hospital, Sanmenxia, China
| | - Liuyang Du
- Department of Statistics Branch, Sanmenxia Central Hospital, Sanmenxia, China
| | - Xiaotong Zhang
- Department of Oncology, Sanmenxia Central Hospital, Sanmenxia, China
| | - Yingping Wang
- Department of Oncology, Sanmenxia Central Hospital, Sanmenxia, China
| | - Dong Zhang
- Department of Oncology, Sanmenxia Central Hospital, Sanmenxia, China
| | - Lingfei Xue
- Department of Oncology, Sanmenxia Central Hospital, Sanmenxia, China
| | - Binhua Lei
- Department of Oncology, Sanmenxia Central Hospital, Sanmenxia, China
| | - Jing Su
- Department of Oncology, Sanmenxia Central Hospital, Sanmenxia, China
| | - Ruiwen Zhang
- Department of Oncology, Sanmenxia Central Hospital, Sanmenxia, China
| | - Jiaohong Chen
- Department of Oncology, Sanmenxia Central Hospital, Sanmenxia, China
| | - Xiangqian Zhang
- Department of Oncology, Sanmenxia Central Hospital, Sanmenxia, China
| | - Qingge Jia
- Department of Reproductive Medicine, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China.
| | - Chuntao Tian
- Department of Oncology, Sanmenxia Central Hospital, Sanmenxia, China.
| |
Collapse
|
24
|
Imyanitov EN, Mitiushkina NV, Kuligina ES, Tiurin VI, Venina AR. Pathways and targeting avenues of BRAF in non-small cell lung cancer. Expert Opin Ther Targets 2024; 28:613-622. [PMID: 38941191 DOI: 10.1080/14728222.2024.2374742] [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: 03/10/2024] [Accepted: 06/27/2024] [Indexed: 06/30/2024]
Abstract
INTRODUCTION BRAF is a serine-threonine kinase implicated in the regulation of MAPK signaling cascade. BRAF mutation-driven activation occurs in approximately 2-4% of treatment-naive non-small cell carcinomas (NSCLCs). BRAF upregulation is also often observed in tumors with acquired resistance to receptor tyrosine kinase inhibitors (TKIs). AREAS COVERED This review describes the spectrum of BRAF mutations and their functional roles, discusses treatment options available for BRAF p.V600 and non-V600 mutated NSCLCs, and identifies some gaps in the current knowledge. EXPERT OPINION Administration of combined BRAF/MEK inhibitors usually produces significant, although often a short-term, benefit to NSCLC patients with BRAF V600 (class 1) mutations. There are no established treatments for BRAF class 2 (L597, K601, G464, G469A/V/R/S, fusions, etc.) and class 3 (D594, G596, G466, etc.) mutants, which account for up to two-thirds of BRAF-driven NSCLCs. Many important issues related to the use of immune therapy for the management of BRAF-mutated NSCLC deserve further investigation. The rare occurrence of BRAF mutations in NSCLC is compensated by high overall incidence of lung cancer disease; therefore, clinical studies on BRAF-associated NSCLC are feasible.
Collapse
Affiliation(s)
- Evgeny N Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg, Russia
| | - Natalia V Mitiushkina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| | - Ekatherina Sh Kuligina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| | - Vladislav I Tiurin
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| | - Aigul R Venina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| |
Collapse
|
25
|
Schwede M, Jahn K, Kuipers J, Miles LA, Bowman RL, Robinson T, Furudate K, Uryu H, Tanaka T, Sasaki Y, Ediriwickrema A, Benard B, Gentles AJ, Levine R, Beerenwinkel N, Takahashi K, Majeti R. Mutation order in acute myeloid leukemia identifies uncommon patterns of evolution and illuminates phenotypic heterogeneity. Leukemia 2024; 38:1501-1510. [PMID: 38467769 PMCID: PMC11250774 DOI: 10.1038/s41375-024-02211-z] [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: 10/30/2023] [Revised: 02/23/2024] [Accepted: 02/28/2024] [Indexed: 03/13/2024]
Abstract
Acute myeloid leukemia (AML) has a poor prognosis and a heterogeneous mutation landscape. Although common mutations are well-studied, little research has characterized how the sequence of mutations relates to clinical features. Using published, single-cell DNA sequencing data from three institutions, we compared clonal evolution patterns in AML to patient characteristics, disease phenotype, and outcomes. Mutation trees, which represent the order of select mutations, were created for 207 patients from targeted panel sequencing data using 1 639 162 cells, 823 mutations, and 275 samples. In 224 distinct orderings of mutated genes, mutations related to DNA methylation typically preceded those related to cell signaling, but signaling-first cases did occur, and had higher peripheral cell counts, increased signaling mutation homozygosity, and younger patient age. Serial sample analysis suggested that NPM1 and DNA methylation mutations provide an advantage to signaling mutations in AML. Interestingly, WT1 mutation evolution shared features with signaling mutations, such as WT1-early being proliferative and occurring in younger individuals, trends that remained in multivariable regression. Some mutation orderings had a worse prognosis, but this was mediated by unfavorable mutations, not mutation order. These findings add a dimension to the mutation landscape of AML, identifying uncommon patterns of leukemogenesis and shedding light on heterogeneous phenotypes.
Collapse
Affiliation(s)
- Matthew Schwede
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, School of Medicine, Stanford, CA, USA
| | - Katharina Jahn
- Biomedical Data Science, Institute for Computer Science, Free University of Berlin, Berlin, Germany
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jack Kuipers
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Linde A Miles
- Division of Experimental Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| | - Robert L Bowman
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Troy Robinson
- Human Oncology and Pathogenesis Program, Molecular Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ken Furudate
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hidetaka Uryu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tomoyuki Tanaka
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuya Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Asiri Ediriwickrema
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
- Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Brooks Benard
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Andrew J Gentles
- Department of Biomedical Data Science, Stanford University, School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Medicine, Stanford Center for Biomedical Informatics Research, Stanford University, Stanford, CA, USA
| | - Ross Levine
- Human Oncology and Pathogenesis Program, Molecular Cancer Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA.
- Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
26
|
Salman G, Johannesmeyer HJ, Breen MJ. Key considerations in navigating ticagrelor's reported effect on heparin-induced thrombocytopenia functional assays in a landscape of limited data. Am J Health Syst Pharm 2024; 81:488-493. [PMID: 38365265 DOI: 10.1093/ajhp/zxae040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Indexed: 02/18/2024] Open
Abstract
PURPOSE This article discusses key considerations regarding ticagrelor's reported effect on heparin-induced thrombocytopenia functional assays, such as literature gaps and possible management strategies. SUMMARY Limited data indicate that ticagrelor may induce false-negative results in functional assays used in the diagnosis of heparin-induced thrombocytopenia. False-negative functional assays for heparin-induced thrombocytopenia could have catastrophic consequences. The manufacturer labeling of ticagrelor now includes a warning for this potential drug-laboratory interaction. This article suggests areas that would benefit from further research and strategies in navigating this possible interaction. CONCLUSION Clinicians should exercise caution when evaluating functional assays for heparin-induced thrombocytopenia in patients receiving ticagrelor. This article offers suggestions for future areas of research and potential management strategies.
Collapse
Affiliation(s)
- Genene Salman
- Department of Pharmacy Practice, College of Pharmacy, Marshall B. Ketchum University, Fullerton, CA, USA
| | - Herman J Johannesmeyer
- Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Martin J Breen
- Department of Pharmacy Services, St. Jude Medical Center, Fullerton, CA, USA
| |
Collapse
|
27
|
Douik H, Sahraoui G, Jemaà M, Doghri R, Charfi L, Mrad K. Concurrent NRAS-BRAF variants in metastatic colorectal cancer: a Tunisian case report. Anticancer Drugs 2024; 35:462-465. [PMID: 38451831 DOI: 10.1097/cad.0000000000001586] [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: 03/09/2024]
Abstract
Target therapy for metastatic colorectal cancer needs the determination of KRAS, NRAS, and BRAF mutation status to identify patients resistant to anti-EGFR treatment. RAS genes (KRAS/NRAS) are mutated in 40-60% of metastatic colorectal cancer and BRAF in 5-10%. The presence of a double mutation in RAS and BRAF is rare. Therefore, RAS and BRAF mutations were considered exclusive. Herein, we describe a novel concomitant NRAS/BRAF mutation identified in a series of 865 colorectal cancer patients.
Collapse
Affiliation(s)
- Hayet Douik
- Pathology Department, Salah Azaiz Institute
- Human Genetics Laboratory (LR99ES10), Faculty of Medicine of Tunis, Tunis El Manar University
| | - Ghada Sahraoui
- Pathology Department, Salah Azaiz Institute
- Precision Medicine and Oncology Investigation Laboratory (LR21SP01), Salah Azaiz Institute
| | - Mohamed Jemaà
- Human Genetics Laboratory (LR99ES10), Faculty of Medicine of Tunis, Tunis El Manar University
- Department of Biology, Faculty of Science of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Raoudha Doghri
- Pathology Department, Salah Azaiz Institute
- Precision Medicine and Oncology Investigation Laboratory (LR21SP01), Salah Azaiz Institute
| | - Lamia Charfi
- Pathology Department, Salah Azaiz Institute
- Precision Medicine and Oncology Investigation Laboratory (LR21SP01), Salah Azaiz Institute
| | - Karima Mrad
- Pathology Department, Salah Azaiz Institute
- Precision Medicine and Oncology Investigation Laboratory (LR21SP01), Salah Azaiz Institute
| |
Collapse
|
28
|
Alhamdan YR, Ayoub NM, Jaradat SK, Shatnawi A, Yaghan RJ. BRAF Expression and Copy Number Alterations Predict Unfavorable Tumor Features and Adverse Outcomes in Patients With Breast Cancer. Int J Breast Cancer 2024; 2024:6373900. [PMID: 38919805 PMCID: PMC11199069 DOI: 10.1155/2024/6373900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/15/2024] [Accepted: 05/07/2024] [Indexed: 06/27/2024] Open
Abstract
Background: The role of BRAF in breast cancer pathogenesis is still unclear. To address this knowledge gap, this study is aimed at evaluating the impact of BRAF gene expression and copy number alterations (CNAs) on clinicopathologic characteristics and survival in patients with breast cancer. Methods: The Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset was obtained from the cBioPortal public domain. Tumoral BRAF mRNA expression and CNAs along with demographic and tumor data for patients with breast cancer were retrieved. The association of BRAF expression and CNAs with breast cancer clinicopathologic characteristics was analyzed. The impact of BRAF mRNA expression on the overall survival of patients was assessed using Kaplan-Meier survival analysis. Results: BRAF gene mRNA log intensity expression was positively correlated with tumor size and the Nottingham Prognostic Index (NPI) (p < 0.001). Alternatively, BRAF gene expression was negatively correlated with the age at diagnosis (p = 0.003). The average BRAF mRNA expression was significantly higher in premenopausal patients, patients with high tumor grade, hormone receptor-negative status, and non-luminal tumors compared to postmenopausal patients, patients with low-grade, hormone receptor-positive, and luminal disease. BRAF gain and high-level amplification copy numbers were significantly associated with higher NPI scores and larger tumor sizes compared to neutral copy number status. Survival analysis revealed no discernible differences in overall survival for patients with low and high BRAF mRNA expression. Conclusion: High BRAF mRNA expression as well as the gain and high-level amplification copy numbers were associated with advanced tumor characteristics and unfavorable prognostic factors in breast cancer. BRAF could be an appealing target for the treatment of premenopausal patients with hormone receptor-negative breast cancer.
Collapse
Affiliation(s)
- Yazan R. Alhamdan
- Department of Clinical PharmacyFaculty of PharmacyJordan University of Science and Technology, PO Box 3030, Irbid 22110, Jordan
| | - Nehad M. Ayoub
- Department of Clinical PharmacyFaculty of PharmacyJordan University of Science and Technology, PO Box 3030, Irbid 22110, Jordan
| | - Sara K. Jaradat
- Department of Clinical PharmacyFaculty of PharmacyJordan University of Science and Technology, PO Box 3030, Irbid 22110, Jordan
| | - Aymen Shatnawi
- Department of Drug Discovery and Biomedical SciencesCollege of PharmacyMedical University of South Carolina, 70 President St., Charleston, South Carolina 29425, USA
| | - Rami J. Yaghan
- Department of SurgeryCollege of Medicine and Medical SciencesArabian Gulf University, Road 2904, Building 293, Manama, Bahrain
- Department of General Surgery and UrologyFaculty of MedicineJordan University of Science and Technology, PO Box 3030, Irbid 22110, Jordan
| |
Collapse
|
29
|
Tabatabaei T, Rezvany MR, Ghasemi B, Vafaei F, Zadeh MK, Zaker F, Salmaninejad A. Effect of DNMT3A R882H Hot Spot Mutations on DDX43 Promoter Methylation in Acute Myeloid Leukemia. BIOMED RESEARCH INTERNATIONAL 2024; 2024:9625043. [PMID: 38807916 PMCID: PMC11132831 DOI: 10.1155/2024/9625043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/21/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
Epigenetic alterations have been observed in many hematological malignancies, including acute myeloid leukemia (AML). Many of these alterations result from mutations in DNA methyl transferase (DNMT) enzymes, disabling them to methylate target genes in a proper way. In this case-control study, we investigated the association between R882H mutation in DNMT3A gene and DDX43 gene methylation in patients with AML. 47 AML patients and 6 controls were included in this study. After DNA extraction, amplification refractory mutation system (ARMS)-PCR was used to evaluate R882H mutations in DNMT3A gene. The high-resolution melting (HRM) method was used to determine the methylation changes of the DDX43 gene promoter. R882H mutation was only found in 10.6% (5 out of 47) of AML patients. The frequency of DDX43 gene methylation was significantly higher in patients without R882H mutations compared to patients with R882H mutations (P < 0.05). The DNMT3A R882H mutation is typically present in a minority of AML patients. Nevertheless, this mutation is associated with a reduced frequency of methylation in the DDX43 promoter region.
Collapse
Affiliation(s)
- Tahere Tabatabaei
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Rezvany
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Bahare Ghasemi
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Farzane Vafaei
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Isfahan, Iran
| | - Masoumeh Kiani Zadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Farhad Zaker
- Department of Hematology and Blood Transfusion, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Arash Salmaninejad
- Regenerative Medicine, Organ Procurement and Transplantation Multi-Disciplinary Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Abdulbaki R, Pullarkat ST. A Brief Overview of the Molecular Landscape of Myelodysplastic Neoplasms. Curr Oncol 2024; 31:2353-2363. [PMID: 38785456 PMCID: PMC11119831 DOI: 10.3390/curroncol31050175] [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: 03/13/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
Myelodysplastic neoplasm (MDS) is a heterogeneous group of clonal hematological disorders that originate from the hematopoietic and progenitor cells and present with cytopenias and morphologic dysplasia with a propensity to progress to bone marrow failure or acute myeloid leukemia (AML). Genetic evolution plays a critical role in the pathogenesis, progression, and clinical outcomes of MDS. This process involves the acquisition of genetic mutations in stem cells that confer a selective growth advantage, leading to clonal expansion and the eventual development of MDS. With the advent of next-generation sequencing (NGS) assays, an increasing number of molecular aberrations have been discovered in recent years. The knowledge of molecular events in MDS has led to an improved understanding of the disease process, including the evolution of the disease and prognosis, and has paved the way for targeted therapy. The 2022 World Health Organization (WHO) Classification and the International Consensus Classification (ICC) have incorporated the molecular signature into the classification system for MDS. In addition, specific germline mutations are associated with MDS development, especially in pediatrics and young adults. This article reviews the genetic abnormalities of MDS in adults with a brief review of germline predisposition syndromes.
Collapse
Affiliation(s)
- Rami Abdulbaki
- Department of Pathology, Laboratory Medicine, UCLA, David Geffen School of Medicine, Los Angeles, CA 90095, USA;
| | | |
Collapse
|
31
|
Patel A, Gulhati P. Molecular Landscape and Therapeutic Strategies against Colorectal Cancer. Cancers (Basel) 2024; 16:1551. [PMID: 38672633 PMCID: PMC11049251 DOI: 10.3390/cancers16081551] [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: 03/07/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer deaths worldwide. Although the overall incidence of CRC is decreasing, the incidence of young-onset CRC, characterized by a diagnosis of CRC before age 50, is increasing. Outcomes for CRC patients are improving, partly due to comprehensive molecular characterization of tumors and novel therapeutic strategies. Advances in genomic and transcriptomic analyses using blood- and tumor-tissue-based sequencing have facilitated identification of distinct tumor subtypes harboring unique biological characteristics and therapeutic vulnerabilities. These insights have led to the development and incorporation of targeted therapies and immunotherapy in CRC treatment. In this review, we discuss the molecular landscape and key oncogenes/tumor suppressors contributing to CRC tumorigenesis, metastasis, and therapeutic resistance. We also discuss personalized therapeutic strategies for subsets of CRC patients and provide an overview of evolving novel treatments being evaluated in clinical trials.
Collapse
Affiliation(s)
- Aakash Patel
- Division of Medical Oncology, Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| | - Pat Gulhati
- Division of Medical Oncology, Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ 08901, USA
| |
Collapse
|
32
|
Capezzone M, Rossi M, Macerola E, Cantara S, Pepe F, Morabito EM, Dalmazio G, Bardi S, Ognibene A, Alessandri M, Materazzi G, De Napoli L, Cirianni M, Torregrossa L. Identification of a Novel Non-V600E BRAF Mutation in Papillary Thyroid Cancer. Case Rep Endocrinol 2024; 2024:6621510. [PMID: 38532782 PMCID: PMC10965284 DOI: 10.1155/2024/6621510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/12/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Papillary thyroid cancer (PTC) is a common endocrine malignancy, and its incidence is reported to be constantly increasing. BRAF mutation is detected in approximately 44% of PTCs, and the most common BRAF mutation is thymine (T) to adenine (A) missense mutation in nucleotide 1796 (T1796A, V600E). Although BRAFV600E represents 95% of all BRAF mutations, uncommon BRAF mutations have been reported in thyroid carcinomas and represent an alternative mechanism of BRAF activation with unclear clinical significance. We report a novel non-V600E mutation (c.1799_1812delinsAT, p.V600_W604delinsD), identified preoperatively with next-generation sequencing (NGS) on the material obtained with fine-needle aspiration cytology (FNAC) performed on a thyroid nodule cytologically suspicious for malignancy in a 35-year-old male patient. The presence of this new variant of BRAF mutation was subsequently confirmed in the postoperative phase by direct Sanger sequencing. In conclusion, we report a new non-V600E variant previously undetected in papillary thyroid cancer. In addition, this case report shows that the NGS technique on cytological tissue allows to detect the presence of rare mutations, thus increasing the diagnostic specificity of molecular analysis.
Collapse
Affiliation(s)
- Marco Capezzone
- UOSD of Endocrinology, Misericordia Hospital, Grosseto 58100, Italy
| | - Maja Rossi
- UOS Molecular Pathology, Hospital Misericordia, Grosseto 58100, Italy
| | - Elisabetta Macerola
- Department of Surgical, Medical and Molecular Pathology, University Hospital of Pisa, Pisa 56124, Italy
| | - Silvia Cantara
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Siena 53100, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | | | - Gilda Dalmazio
- UOSD of Endocrinology, Misericordia Hospital, Grosseto 58100, Italy
| | - Sara Bardi
- UOS Molecular Pathology, Hospital Misericordia, Grosseto 58100, Italy
| | - Agostino Ognibene
- UOS Molecular Pathology, Hospital Misericordia, Grosseto 58100, Italy
| | | | - Gabriele Materazzi
- Division of Endocrine Surgery, Department of Surgical Pathology, University Hospital of Pisa, Pisa 56124, Italy
| | - Luigi De Napoli
- Division of Endocrine Surgery, Department of Surgical Pathology, University Hospital of Pisa, Pisa 56124, Italy
| | - Michele Cirianni
- UOS Molecular Pathology, Hospital Misericordia, Grosseto 58100, Italy
| | - Liborio Torregrossa
- Department of Surgical, Medical and Molecular Pathology, University Hospital of Pisa, Pisa 56124, Italy
| |
Collapse
|
33
|
Timofeev O, Giron P, Lawo S, Pichler M, Noeparast M. ERK pathway agonism for cancer therapy: evidence, insights, and a target discovery framework. NPJ Precis Oncol 2024; 8:70. [PMID: 38485987 PMCID: PMC10940698 DOI: 10.1038/s41698-024-00554-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/16/2024] [Indexed: 03/18/2024] Open
Abstract
At least 40% of human cancers are associated with aberrant ERK pathway activity (ERKp). Inhibitors targeting various effectors within the ERKp have been developed and explored for over two decades. Conversely, a substantial body of evidence suggests that both normal human cells and, notably to a greater extent, cancer cells exhibit susceptibility to hyperactivation of ERKp. However, this vulnerability of cancer cells remains relatively unexplored. In this review, we reexamine the evidence on the selective lethality of highly elevated ERKp activity in human cancer cells of varying backgrounds. We synthesize the insights proposed for harnessing this vulnerability of ERK-associated cancers for therapeutical approaches and contextualize these insights within established pharmacological cancer-targeting models. Moreover, we compile the intriguing preclinical findings of ERK pathway agonism in diverse cancer models. Lastly, we present a conceptual framework for target discovery regarding ERKp agonism, emphasizing the utilization of mutual exclusivity among oncogenes to develop novel targeted therapies for precision oncology.
Collapse
Affiliation(s)
- Oleg Timofeev
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University, 35043, Marburg, Germany
| | - Philippe Giron
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research group Genetics, Reproduction and Development, Centre for Medical Genetics, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Steffen Lawo
- CRISPR Screening Core Facility, Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Martin Pichler
- Translational Oncology, II. Med Clinics Hematology and Oncology, 86156, Augsburg, Germany
| | - Maxim Noeparast
- Translational Oncology, II. Med Clinics Hematology and Oncology, 86156, Augsburg, Germany.
| |
Collapse
|
34
|
Chen YK, Kanouni T, Arnold LD, Cox JM, Gardiner E, Grandinetti K, Jiang P, Kaldor SW, Lee C, Li C, Martin ES, Miller N, Murphy EA, Timple N, Tyhonas JS, Vassar A, Wang TS, Williams R, Yuan D, Kania RS. The Discovery of Exarafenib (KIN-2787): Overcoming the Challenges of Pan-RAF Kinase Inhibition. J Med Chem 2024; 67:1747-1757. [PMID: 38230963 DOI: 10.1021/acs.jmedchem.3c01830] [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: 01/18/2024]
Abstract
RAF, a core signaling component of the MAPK kinase cascade, is often mutated in various cancers, including melanoma, lung, and colorectal cancers. The approved inhibitors were focused on targeting the BRAFV600E mutation that results in constitutive activation of kinase signaling through the monomeric protein (Class I). However, these inhibitors also paradoxically activate kinase signaling of RAF dimers, resulting in increased MAPK signaling in normal tissues. Recently, significant attention has turned to targeting RAF alterations that activate dimeric signaling (class II and III BRAF and NRAS). However, the discovery of a potent and selective inhibitor with biopharmaceutical properties suitable to sustain robust target inhibition in the clinical setting has proven challenging. Herein, we report the discovery of exarafenib (15), a highly potent and selective inhibitor that intercepts the RAF protein in the dimer compatible αC-helix-IN conformation and demonstrates anti-tumor efficacy in preclinical models with BRAF class I, II, and III and NRAS alterations.
Collapse
Affiliation(s)
- Young K Chen
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Toufike Kanouni
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Lee D Arnold
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Jason M Cox
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Elisabeth Gardiner
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Kathryn Grandinetti
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Ping Jiang
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Stephen W Kaldor
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Catherine Lee
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Chun Li
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Eric S Martin
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Nichol Miller
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Eric A Murphy
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Noel Timple
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - John S Tyhonas
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Angie Vassar
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Tim S Wang
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Richard Williams
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Ding Yuan
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| | - Robert S Kania
- Kinnate Biopharma, 12830 El Camino Real, Suite 150, San Diego, California 92130, United States
| |
Collapse
|
35
|
Hosseini M, Voisin V, Chegini A, Varesi A, Cathelin S, Ayyathan DM, Liu AC, Yang Y, Wang V, Maher A, Grignano E, Reisz JA, D’Alessandro A, Young K, Wu Y, Fiumara M, Ferrari S, Naldini L, Gaiti F, Pai S, Schimmer AD, Bader GD, Dick JE, Xie SZ, Trowbridge JJ, Chan SM. Metformin reduces the clonal fitness of Dnmt3a R878H hematopoietic stem and progenitor cells by reversing their aberrant metabolic and epigenetic state. RESEARCH SQUARE 2024:rs.3.rs-3874821. [PMID: 38405837 PMCID: PMC10889081 DOI: 10.21203/rs.3.rs-3874821/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Clonal hematopoiesis (CH) arises when a hematopoietic stem cell (HSC) acquires a mutation that confers a competitive advantage over wild-type (WT) HSCs, resulting in its clonal expansion. Individuals with CH are at an increased risk of developing hematologic neoplasms and a range of age-related inflammatory illnesses1-3. Therapeutic interventions that suppress the expansion of mutant HSCs have the potential to prevent these CH-related illnesses; however, such interventions have not yet been identified. The most common CH driver mutations are in the DNA methyltransferase 3 alpha (DNMT3A) gene with arginine 882 (R882) being a mutation hotspot. Here we show that murine hematopoietic stem and progenitor cells (HSPCs) carrying the Dnmt3a R878H/+ mutation, which is equivalent to human DNMT3A R882H/+, have increased mitochondrial respiration compared with WT cells and are dependent on this metabolic reprogramming for their competitive advantage. Importantly, treatment with metformin, an oral anti-diabetic drug with inhibitory activity against complex I in the electron transport chain (ETC), reduced the fitness of Dnmt3a R878H/+ HSCs. Through a multi-omics approach, we discovered that metformin acts by enhancing the methylation potential in Dnmt3a R878H/+ HSPCs and reversing their aberrant DNA CpG methylation and histone H3K27 trimethylation (H3K27me3) profiles. Metformin also reduced the fitness of human DNMT3A R882H HSPCs generated by prime editing. Our findings provide preclinical rationale for investigating metformin as a preventive intervention against illnesses associated with DNMT3A R882 mutation-driven CH in humans.
Collapse
Affiliation(s)
| | - Veronique Voisin
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
| | - Ali Chegini
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Angelica Varesi
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Alex C.H. Liu
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Yitong Yang
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Vivian Wang
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Abdula Maher
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Eric Grignano
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kira Young
- The Jackson Laboratory, Bar Harbor, ME, USA
| | - Yiyan Wu
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Martina Fiumara
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
- Vita-Salute San Raffaele University, Milan, 20132, Italy
| | - Samuele Ferrari
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
- Vita-Salute San Raffaele University, Milan, 20132, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, 20132, Italy
- Vita-Salute San Raffaele University, Milan, 20132, Italy
| | - Federico Gaiti
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Shraddha Pai
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Aaron D. Schimmer
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Gary D. Bader
- Donnelly Centre for Cellular and Biomolecular Research, Toronto, Ontario, Canada
| | - John E. Dick
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Steven M. Chan
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
36
|
Kazandjian S, Rousselle E, Dankner M, Cescon DW, Spreafico A, Ma K, Kavan P, Batist G, Rose AAN. The Clinical, Genomic, and Transcriptomic Landscape of BRAF Mutant Cancers. Cancers (Basel) 2024; 16:445. [PMID: 38275886 PMCID: PMC10814895 DOI: 10.3390/cancers16020445] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/08/2024] [Accepted: 01/13/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND BRAF mutations are classified into four molecularly distinct groups, and Class 1 (V600) mutant tumors are treated with targeted therapies. Effective treatment has not been established for Class 2/3 or BRAF Fusions. We investigated whether BRAF mutation class differed according to clinical, genomic, and transcriptomic variables in cancer patients. METHODS Using the AACR GENIE (v.12) cancer database, the distribution of BRAF mutation class in adult cancer patients was analyzed according to sex, age, primary race, and tumor type. Genomic alteration data and transcriptomic analysis was performed using The Cancer Genome Atlas. RESULTS BRAF mutations were identified in 9515 (6.2%) samples among 153,834, with melanoma (31%), CRC (20.7%), and NSCLC (13.9%) being the most frequent cancer types. Class 1 harbored co-mutations outside of the MAPK pathway (TERT, RFN43) vs. Class 2/3 mutations (RAS, NF1). Across all tumor types, Class 2/3 were enriched for alterations in genes involved in UV response and WNT/β-catenin. Pathway analysis revealed enrichment of WNT/β-catenin and Hedgehog signaling in non-V600 mutated CRC. Males had a higher proportion of Class 3 mutations vs. females (17.4% vs. 12.3% q = 0.003). Non-V600 mutations were generally more common in older patients (aged 60+) vs. younger (38% vs. 15% p < 0.0001), except in CRC (15% vs. 30% q = 0.0001). Black race was associated with non-V600 BRAF alterations (OR: 1.58; p < 0.0001). CONCLUSIONS Class 2/3 BRAFs are more present in Black male patients with co-mutations outside of the MAPK pathway, likely requiring additional oncogenic input for tumorigenesis. Improving access to NGS and trial enrollment will help the development of targeted therapies for non-V600 BRAF mutations.
Collapse
Affiliation(s)
- Suzanne Kazandjian
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H4A 3T2, Canada; (S.K.); (K.M.); (P.K.); (G.B.)
- Segal Cancer Centre, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Emmanuelle Rousselle
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (E.R.); (M.D.)
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Matthew Dankner
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (E.R.); (M.D.)
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - David W. Cescon
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Center, Toronto, ON M5G 2M9, Canada; (D.W.C.); (A.S.)
| | - Anna Spreafico
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Center, Toronto, ON M5G 2M9, Canada; (D.W.C.); (A.S.)
| | - Kim Ma
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H4A 3T2, Canada; (S.K.); (K.M.); (P.K.); (G.B.)
- Segal Cancer Centre, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Petr Kavan
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H4A 3T2, Canada; (S.K.); (K.M.); (P.K.); (G.B.)
- Segal Cancer Centre, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Gerald Batist
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H4A 3T2, Canada; (S.K.); (K.M.); (P.K.); (G.B.)
- Segal Cancer Centre, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - April A. N. Rose
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H4A 3T2, Canada; (S.K.); (K.M.); (P.K.); (G.B.)
- Segal Cancer Centre, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (E.R.); (M.D.)
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada
| |
Collapse
|
37
|
Casalin I, De Stefano A, Ceneri E, Cappellini A, Finelli C, Curti A, Paolini S, Parisi S, Zannoni L, Boultwood J, McCubrey JA, Suh PG, Ramazzotti G, Fiume R, Ratti S, Manzoli L, Cocco L, Follo MY. Deciphering signaling pathways in hematopoietic stem cells: the molecular complexity of Myelodysplastic Syndromes (MDS) and leukemic progression. Adv Biol Regul 2024; 91:101014. [PMID: 38242820 DOI: 10.1016/j.jbior.2024.101014] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
Myelodysplastic Syndromes, a heterogeneous group of hematological disorders, are characterized by abnormalities in phosphoinositide-dependent signaling, epigenetic regulators, apoptosis, and cytokine interactions within the bone marrow microenvironment, contributing to disease pathogenesis and neoplastic growth. Comprehensive knowledge of these pathways is crucial for the development of innovative therapies that aim to restore normal apoptosis and improve patient outcomes.
Collapse
Affiliation(s)
- Irene Casalin
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy.
| | - Alessia De Stefano
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Eleonora Ceneri
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Alessandra Cappellini
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Carlo Finelli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Antonio Curti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Stefania Paolini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Sarah Parisi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Letizia Zannoni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna - Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Jacqueline Boultwood
- Bloodwise Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Pann-Ghill Suh
- Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Giulia Ramazzotti
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Roberta Fiume
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Lucia Manzoli
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| | - Matilde Y Follo
- Department of Biomedical and Neuromotor Science, Cellular Signaling Laboratory, University of Bologna, Bologna, Italy
| |
Collapse
|
38
|
Wu JB, Li XJ, Liu H, Liu YJ, Liu XP. Association of KRAS, NRAS, BRAF and PIK3CA gene mutations with clinicopathological features, prognosis and ring finger protein 215 expression in patients with colorectal cancer. Biomed Rep 2023; 19:104. [PMID: 38025833 PMCID: PMC10646763 DOI: 10.3892/br.2023.1686] [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: 06/28/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
The relationships of KRAS, NRAS, BRAF and PIK3CA gene mutations with the clinicopathological features and prognosis of colorectal cancer (CRC) in patient are lacking. Furthermore, the role of ring finger protein 215 (RNF215) in CRC patients with KRAS, NRAS, BRAF and PIK3CA mutations remains unclear. In the present study, 182 surgical resection specimens from patients with primary CRC for retrospective analysis, were collected. KRAS/NRAS/BRAF/PIK3CA gene mutations were confirmed by an amplification-refractory mutation system. Immunohistochemistry (IHC) was conducted to confirm KRAS, NRAS, BRAF and PIK3CA protein expression. RNF215 expression in patients with CRC was evaluated using TIMER 2.0 database and IHC. The individual mutation rates of KRAS, NRAS, BRAF and PIK3CA were 40.7% (74/182), 4.4% (8/182), 4.4% (8/182) and 3.3% (6/182), respectively. The KRAS exon 2 mutation rate was the highest (61.5%, 64/104), and these mutations mainly occurred at codons 12 and 13. KRAS/NRAS/BRAF/PIK3CA wild-type CRC patients had significantly longer overall survival and disease-free survival than mutated KRAS/NRAS/BRAF/PIK3CA CRC patients (P<0.05). Overall, 45.4% (5/11) of patients with PIK3CA mutations had concomitant KRAS mutations. The KRAS/NRAS/BRAF/PIK3CA gene mutation rate in patients with lymph node metastasis (76.1%, 35/46) was significantly higher than that in patients without lymph node metastasis (50.8%, 69/136) (P=0.0027). There were no significant differences in IHC expression between patients with and without KRAS, NRAS, BRAF and PIK3CA mutations (P>0.05). The TIMER 2.0 analysis showed that RNF215 expression was significantly higher in the mutated BRAF group than in the wild-type BRAF group in CRC (P<0.05). In conclusion, KRAS is the most commonly mutated gene, and KRAS mutations may be a poor prognostic factor for patients with CRC. KRAS wild-type patient resistance may be related to PIK3CA gene mutations, although this needs further verification in larger cohorts. BRAF mutations may be associated with RNF215 expression in patients with CRC.
Collapse
Affiliation(s)
- Jing-Bo Wu
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Xiao-Jing Li
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Hui Liu
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Yong-Juan Liu
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, P.R. China
| | - Xiu-Ping Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
39
|
Hutchings DA, Salimian KJ, Waters KM, Birkness-Gartman JE, Voltaggio L, Assarzadegan N, Huang J, Lin MT, Singhi AD, Montgomery EA. Aberrant p53 Expression in Gastric Biopsies and Resection Specimens Following Neoadjuvant Chemoradiation: A Diagnostic Pitfall. Int J Surg Pathol 2023; 31:1458-1465. [PMID: 36843539 PMCID: PMC10460459 DOI: 10.1177/10668969231157304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Abstract
Gastric mucosal biopsies and resections from patients treated with neoadjuvant radiation and/or chemotherapy are frequently encountered. These samples may show histologic features related to therapy including inflammation, ulceration, and epithelial atypia. In some cases, epithelial atypia may be marked, prompting the use of adjunct p53 immunohistochemistry. We examined p53 expression by immunohistochemistry in gastric mucosa following therapy. We evaluated the histology and p53 immunohistochemical expression in gastric mucosa from 57 resections and 3 mucosal biopsies, from 60 patients treated with radiation and/or chemotherapy for gastroesophageal carcinoma (n = 33) or pancreatic carcinoma (n = 27). We identified histomorphologic features of therapy-related epithelial changes in 50 of 60 cases (83%). Abnormal p53 expression was present at least focally in nearly half the cases (27 of 60 cases; 45%), all of which showed morphologic evidence of therapy-related epithelial changes. Neuroendocrine cell micronests were present in 37 of 60 cases (62%). Next-generation sequencing (NGS) of foci with therapy-related epithelial changes showing abnormal p53 expression and carcinoma from the same patient was attempted and yielded results in 1 patient. Interestingly, differing TP53 alterations in the patient's adenocarcinoma and in a histologically benign esophageal submucosal gland with therapy-related epithelial changes and abnormal p53 expression were identified. Our results demonstrate that abnormal p53 expression is relatively common in gastric mucosal samples following radiation and/or chemotherapy and suggest that p53 expression should be avoided when distinguishing therapy-related changes from dysplasia or carcinoma. Furthermore, our NGS results raise interesting biological questions, which may warrant further investigation.
Collapse
Affiliation(s)
- Danielle A. Hutchings
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kevan J. Salimian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kevin M. Waters
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Lysandra Voltaggio
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Jialing Huang
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ming-Tseh Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aatur D. Singhi
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | | |
Collapse
|
40
|
Sheng L, Liu Y, Zhu Y, Zhou J, Hua H. Analysis of the clinical characteristics and prognosis of adult de novo acute myeloid leukemia (none APL) with PTPN11 mutations. Open Med (Wars) 2023; 18:20230830. [PMID: 38025540 PMCID: PMC10655689 DOI: 10.1515/med-2023-0830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 09/13/2023] [Accepted: 09/29/2023] [Indexed: 12/01/2023] Open
Abstract
We discuss the clinical characteristics and prognostic significance of adult individuals with PTPN11 mutations who have developed acute myeloid leukemia (AML) (none acute promyelocytic leukemia). Next generation sequencing and Sanger sequencing were used to detect 51 gene mutations, and multiplex-PCR was used to detect 41 fusion genes from 232 de novo adult AML patients retrospectively. About 7.76% patients harbored PTPN11 mutations, 20 PTPN11 alterations were identified, all of which were missense mutations in the N-SH2 (n = 16) and PTP (n = 4) domains located in exon 3. Patients with PTPN11 mut had significantly higher platelet counts and hemoglobin levels (p < 0.001), which were mainly detected in M5 (n = 12, 66.67%, p < 0.001) subtype. Patients with MLL-AF6 positive showed a higher frequency of PTPN11 mut (p = 0.018) in the 118 AML cases. PTPN11 mut were accompanied by other mutations, which were NPM1 (44.44%), DNMT3A (38.89%), FLT3 (38.89%), and NRAS (17.2%). PTPN11 mut had a negative impact on the complete remission rate in M5 subtype patients (p < 0.001). However, no statistically significant effect on overall survival (OS) with PTPN11 mut patients in the whole cohort and age group (p > 0.05) was observed. Further analysis revealed no significant difference in OS among NPM1 mut/PTPN11 mut, NPM1 mut/PTPN11 wt, DNMT3A mut/PTPN11 mut, and DNMT3A mut/PTPN11 wt patients (p > 0.05). Multivariate analysis showed the proportion of bone marrow blasts ≥65.4% was a factor significantly affecting OS in PTPN11 mut patients (p = 0.043).
Collapse
Affiliation(s)
- Li Sheng
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yajiao Liu
- Nursing Department, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310000, China
| | - Yingying Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Jingfen Zhou
- Department of Hematology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Haiying Hua
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, China
- Department of Hematology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
41
|
Elshazly AM, Gewirtz DA. The Cytoprotective Role of Autophagy in Response to BRAF-Targeted Therapies. Int J Mol Sci 2023; 24:14774. [PMID: 37834222 PMCID: PMC10572960 DOI: 10.3390/ijms241914774] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
BRAF-targeted therapies are widely used for the treatment of melanoma patients with BRAF V600 mutations. Vemurafenib, dabrafenib as well as encorafenib have demonstrated substantial therapeutic activity; however, as is the case with other chemotherapeutic agents, the frequent development of resistance limits their efficacy. Autophagy is one tumor survival mechanism that could contribute to BRAF inhibitor resistance, and multiple studies support an association between vemurafenib-induced and dabrafenib-induced autophagy and tumor cell survival. Clinical trials have also demonstrated a potential benefit from the inclusion of autophagy inhibition as an adjuvant therapy. This review of the scientific literature relating to the role of autophagy that is induced in response to BRAF-inhibitors supports the premise that autophagy targeting or modulation could be an effective adjuvant therapy.
Collapse
Affiliation(s)
- Ahmed M. Elshazly
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA;
| |
Collapse
|
42
|
Humphries S, Bond DR, Germon ZP, Keely S, Enjeti AK, Dun MD, Lee HJ. Crosstalk between DNA methylation and hypoxia in acute myeloid leukaemia. Clin Epigenetics 2023; 15:150. [PMID: 37705055 PMCID: PMC10500762 DOI: 10.1186/s13148-023-01566-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Acute myeloid leukaemia (AML) is a deadly disease characterised by the uncontrolled proliferation of immature myeloid cells within the bone marrow. Altered regulation of DNA methylation is an important epigenetic driver of AML, where the hypoxic bone marrow microenvironment can help facilitate leukaemogenesis. Thus, interactions between epigenetic regulation and hypoxia signalling will have important implications for AML development and treatment. MAIN BODY This review summarises the importance of DNA methylation and the hypoxic bone marrow microenvironment in the development, progression, and treatment of AML. Here, we focus on the role hypoxia plays on signalling and the subsequent regulation of DNA methylation. Hypoxia is likely to influence DNA methylation through altered metabolic pathways, transcriptional control of epigenetic regulators, and direct effects on the enzymatic activity of epigenetic modifiers. DNA methylation may also prevent activation of hypoxia-responsive genes, demonstrating bidirectional crosstalk between epigenetic regulation and the hypoxic microenvironment. Finally, we consider the clinical implications of these interactions, suggesting that reduced cell cycling within the hypoxic bone marrow may decrease the efficacy of hypomethylating agents. CONCLUSION Hypoxia is likely to influence AML progression through complex interactions with DNA methylation, where the therapeutic efficacy of hypomethylating agents may be limited within the hypoxic bone marrow. To achieve optimal outcomes for AML patients, future studies should therefore consider co-treatments that can promote cycling of AML cells within the bone marrow or encourage their dissociation from the bone marrow.
Collapse
Affiliation(s)
- Sam Humphries
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Danielle R Bond
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Zacary P Germon
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Simon Keely
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Anoop K Enjeti
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
- Department of Haematology, Calvary Mater Hospital, Waratah, NSW, 2298, Australia
- New South Wales Health Pathology, John Hunter Hospital, New Lambton Heights, NSW, 2305, Australia
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2308, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia
| | - Heather J Lee
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, 2308, Australia.
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, 2305, Australia.
| |
Collapse
|
43
|
Li S, Qu J, Wang X, Zou Q, Li C. SHP2 is involved in the occurrence, development and prognosis of cancer. Oncol Lett 2023; 26:393. [PMID: 37600341 PMCID: PMC10433711 DOI: 10.3892/ol.2023.13979] [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: 03/11/2023] [Accepted: 07/14/2023] [Indexed: 08/22/2023] Open
Abstract
Src homology-2 domain-containing protein tyrosine phosphatase (SHP2), encoded by protein tyrosine phosphatase non-receptor type 11 (PTPN11), is widely expressed in several human tissue types, and plays an important role in a variety of diseases. The present study assessed the impact of SHP2 on the occurrence, development and prognosis of solid tumors. The transcriptome sequencing data of 33 cancer types were downloaded from The Cancer Genome Atlas database. Clinical information of the corresponding patients, tumor mutational burden and information pertinent to microsatellite instability were also downloaded. The log-rank test and univariate Cox's regression test were used to evaluate patient survival. The 'ESTIMATE' method was used to assess the tumor microenvironment, and the 'CIBERSORT' algorithm was used to evaluate tumor immune cell infiltration. Spearman's correlation analysis was used to evaluate the correlation between SHP2 expression and the targets identified. ELISA was used to assess the SHP2 expression levels in peripheral blood samples of patients with breast, ovarian, endometrial and cervical cancer. The data indicated that the expression levels of SHP2 were increased in a variety of tumor tissues, and were associated with tumor progression and prognosis. In peripheral blood, the positive rates of SHP2 expression in breast cancer (71.43%) and ovarian cancer (58.82%) were significantly higher than those in the corresponding control groups. However, the positive rates of SHP2 expression in patients with endometrial cancer (31.03%) and cervical cancer (41.30%) were significantly lower than those in the corresponding control groups. Increased SHP2 expression improved overall survival (OS) and disease free survival (DFS) time in patients with kidney renal clear cell carcinoma. However, increased SHP2 expression reduced OS and DFS in patients with urothelial carcinoma, and cervical and endocervical cancer types. Moreover, the elevated expression of SHP2 could also reduce the OS of patients with breast invasive carcinoma, mesothelioma and liver hepatocellular carcinoma. PTPN11 expression was associated with the tumor microenvironment of various tumor types. The tumor mutational burden of various tumor types was associated with microsatellite instability. PTPN11 inhibited T-cell activation and promoted M2 macrophage activation in several tumors. Therefore, SHP2 may be used in the evaluation of tumor progression and prognosis, and it may be an optimal potential biological target for cancer therapy.
Collapse
Affiliation(s)
- Shu Li
- Department of Clinical Laboratory, Women and Children's Hospital of Chongqing Medical University, Chongqing 401174, P.R. China
- Department of Clinical Laboratory, Chongqing Health Center for Women and Children, Chongqing 401174, P.R. China
| | - Jialing Qu
- Department of Clinical Laboratory, Women and Children's Hospital of Chongqing Medical University, Chongqing 401174, P.R. China
- Department of Clinical Laboratory, Chongqing Health Center for Women and Children, Chongqing 401174, P.R. China
| | - Xiaotong Wang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401174, P.R. China
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children, Chongqing 401174, P.R. China
| | - Qin Zou
- Department of Clinical Laboratory, Women and Children's Hospital of Chongqing Medical University, Chongqing 401174, P.R. China
- Department of Clinical Laboratory, Chongqing Health Center for Women and Children, Chongqing 401174, P.R. China
| | - Chunli Li
- Department of Clinical Laboratory, Women and Children's Hospital of Chongqing Medical University, Chongqing 401174, P.R. China
- Department of Clinical Laboratory, Chongqing Health Center for Women and Children, Chongqing 401174, P.R. China
| |
Collapse
|
44
|
Maitre E, Macro M, Troussard X. Hairy cell leukaemia with unusual BRAF mutations. J Cell Mol Med 2023; 27:2626-2630. [PMID: 37530550 PMCID: PMC10468650 DOI: 10.1111/jcmm.17890] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/06/2023] [Accepted: 07/22/2023] [Indexed: 08/03/2023] Open
Abstract
Hairy cell leukaemia (HCL) diagnosis is based on the morphologic detection of circulating abnormal hairy cells in the peripheral blood and/or bone marrow, an HCL immunological score of 3 or 4 based on the expression of the CD11c, CD25, CD103 and CD123 and also the presence of a BRAF V600E activating mutation in the B-raf proto-oncogene (BRAF gene) (7q34). When using new generation sequencing of 21 targeted genes in 124 HCL patients, we identified a cohort of 6/124 (2%) patients with unusual BRAF mutations: two patients presented non-V600 mutations (BRAF F595L, BRAF W604L respectively) and four other patients silent BRAF mutations. When using droplet digital PCR (ddPCR) three of the four patients with concomitant BRAF V600E and silent mutation were negative. The respective role of these mutations in the occurrence of HCL or its progression remains to be clarified, but BRAF sequencing is necessary in case of negative BRAF V600E by ddPCR.
Collapse
Affiliation(s)
- Elsa Maitre
- Laboratoire d'HématologieCHU Caen NormandieCaenFrance
| | - Margaret Macro
- Institut bas Normand d'HématologieCHU Caen NormandieCaenFrance
| | - Xavier Troussard
- Laboratoire d'HématologieCHU Caen NormandieCaenFrance
- Institut bas Normand d'HématologieCHU Caen NormandieCaenFrance
| |
Collapse
|
45
|
Li Y, Lou J, Hong S, Hou D, Lv Y, Guo Z, Wang K, Xu Y, Zhai Y, Liu H. The role of heavy metals in the development of colorectal cancer. BMC Cancer 2023; 23:616. [PMID: 37400750 PMCID: PMC10316626 DOI: 10.1186/s12885-023-11120-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/27/2023] [Indexed: 07/05/2023] Open
Abstract
OBJECTIVE To investigate the relationship among 18 heavy metals, microsatellite instability (MSI) status, ERCC1, XRCC1 (rs25487), BRAF V600E and 5 tumor markers and their role in the development of colorectal cancer (CRC). METHODS A total of 101 CRC patients and 60 healthy controls were recruited in the present study. The levels of 18 heavy metals were measured by ICP-MS. MSI status and the genetic polymorphism were determined by PCR (FP205-02, Tiangen Biochemical Technology Co., Ltd., Beijing, China) and Sanger sequencing. Spearman's rank correlation was used to analyze the relationship among various factors. RESULTS The level of selenium (Se) was lower in the CRC group compared with the control group (p < 0.01), while vanadium (V), arsenic (As), tin (Sn), barium (Ba) and lead (Pb) were higher (p < 0.05), chromium (Cr) and copper (Cu) were significantly higher (p < 0.0001) in the CRC group than those in the control group. Multivariate logistic regression analysis indicated that Cr, Cu, As and Ba were the risk factors for CRC. In addition, CRC was positively correlated with V, Cr, Cu, As, Sn, Ba and Pb, but negatively correlated with Se. MSI was positively correlated with BRAF V600E, but negatively correlated with ERCC1. BRAF V600E was positively correlated with antimony (Sb), thallium (Tl), CA19-9, NSE, AFP and CK19. XRCC1 (rs25487) was found to be positively correlated with Se but negatively correlated with Co. The levels of Sb and Tl were significantly higher in the BRAF V600E positive group compared to the negative group. The mRNA expression level of ERCC1 was significantly higher (P = 0.035) in MSS compared to MSI. And there was a significant correlation between XRCC1 (rs25487) polymorphism and MSI status (P<0.05). CONCLUSION The results showed that low level of Se and high levels of V, As, Sn, Ba, Pb, Cr, and Cu increased the risk of CRC. Sb and Tl may cause BRAF V600E mutations, leading to MSI. XRCC1 (rs25487) was positively correlated with Se but negatively correlated with Co. The expression of ERCC1 may be related to MSS, while the XRCC1 (rs25487) polymorphism is related to MSI.
Collapse
Affiliation(s)
- Yongsheng Li
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Jingwei Lou
- Shanghai Biotecan Pharmaceuticals Co., Ltd, Shanghai, 201204, China
| | - Shaozhong Hong
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Dengfeng Hou
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Yandong Lv
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Zhiqiang Guo
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Kai Wang
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Yue Xu
- Shanghai Biotecan Pharmaceuticals Co., Ltd, Shanghai, 201204, China
| | - Yufeng Zhai
- Shanghai Biotecan Pharmaceuticals Co., Ltd, Shanghai, 201204, China.
| | - Hongzhou Liu
- Department of Colorectal Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China.
| |
Collapse
|
46
|
Malapelle U, Angerilli V, Pepe F, Fontanini G, Lonardi S, Scartozzi M, Memeo L, Pruneri G, Marchetti A, Perrone G, Fassan M. The ideal reporting of RAS testing in colorectal adenocarcinoma: a pathologists' perspective. Pathologica 2023; 115:137-147. [PMID: 37314870 PMCID: PMC10462993 DOI: 10.32074/1591-951x-895] [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: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 06/16/2023] Open
Abstract
RAS gene mutational status represents an imperative predictive biomarker to be tested in the clinical management of metastatic colorectal adenocarcinoma. Even if it is one of the most studied biomarkers in the era of precision medicine, several pre-analytical and analytical factors may still impasse an adequate reporting of RAS status in clinical practice, with significant therapeutic consequences. Thus, pathologists should be aware on the main topics related to this molecular evaluation: (i) adopt diagnostic limit of detections adequate to avoid the interference of sub-clonal cancer cell populations; (ii) choose the most adequate diagnostic strategy according to the available sample and its qualification for molecular testing; (iii) provide all the information regarding the mutation detected, since many RAS mutation-specific targeted therapeutic approaches are in development and will enter into routine clinical practice. In this review, we give a comprehensive description of the current scenario about RAS gene mutational testing in the clinic focusing on the pathologist's role in patient selection for targeted therapies.
Collapse
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples (NA), Italy
| | | | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples (NA), Italy
| | - Gabriella Fontanini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa (PI), Italy
| | - Sara Lonardi
- Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Padua (PD), Italy
| | - Mario Scartozzi
- Medical Oncology, University Hospital and University of Cagliari, Cagliari (CA), Italy
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, Catania (CT), Italy
| | - Gianfranco Pruneri
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale Tumori and University of Milan, Milan (MI), Italy
| | - Antonio Marchetti
- Center for Advanced Studies and Technology (CAST), University Chieti-Pescara, Chieti (CH), Italy
- Diagnostic Molecular Pathology, Unit of Anatomic Pathology, SS Annunziata Hospital, Chieti (CH), Italy and Department of Medical, Oral, and Biotechnological Sciences University “G. D’Annunzio” of Chieti-Pescara, Chieti (CH), Italy
| | - Giuseppe Perrone
- Department of Medicine and Surgery, Research Unit of Anatomical Pathology, Università Campus Bio-Medico di Roma, Roma, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), University of Padua, Padua (PD), Italy
- Veneto Institute of Oncology (IOV-IRCCS), Padua (PD), Italy
| |
Collapse
|
47
|
Mirzapoor Abbasabadi Z, Hamedi Asl D, Rahmani B, Shahbadori R, Karami S, Peymani A, Taghizadeh S, Samiee Rad F. KRAS, NRAS, BRAF, and PIK3CA mutation rates, clinicopathological association, and their prognostic value in Iranian colorectal cancer patients. J Clin Lab Anal 2023; 37:e24868. [PMID: 36930789 PMCID: PMC10098058 DOI: 10.1002/jcla.24868] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/13/2023] [Accepted: 03/01/2023] [Indexed: 03/19/2023] Open
Abstract
AIM Mutations in KRAS, NRAS, BRAF, and PIK3CA genes are critical factors in clinical evaluation of colorectal cancer (CRC) development and progression. In Iran, however, the data regarding genetic profile of CRC patients is limited except for KRAS exon2 and BRAF V600F mutations. This study aimed to investigate the mutational spectrum and prognostic effects of these genes and explore the relationship between these mutations and clinicopathological features of CRC. METHOD To achieve these objectives, mutations in KRAS (exons 2, 3, and 4), NRAS (exons 2, 3, and 4), PIK3CA (exons 9 and 20), and BRAF (exon 15) was determined using PCR and pyrosequencing in a total of 151 patients with colorectal cancer. RESULTS KRAS, BRAF, NRAS, and PIK3CA mutations were identified in 41%, 5.96%, 3.97%, and 13.24% of the cases, respectively. There were some significant correlations between clinicopathological features and KRAS, PIK3CA, BRAF, and NRAS mutations. Mutations in KRAS and PIK3CA were shown to be independent risk factors for poor survival of the patients at stage I-IV (p < 0.0001 and p = 0.001, respectively). No significant impact on prognosis was observed in patients with BRAF mutations. CONCLUSION Our study revealed the prevalence of CRC biomarkers mutations in Iranian patients and emphasized the role of KRAS and PIK3CA on shorter overall survival rates in this population.
Collapse
Affiliation(s)
- Zohreh Mirzapoor Abbasabadi
- Department of Molecular Medicine, Faculty of Medical Sciences, Qazvin University of Medical Sciences, Qazvin, Iran.,Department of Pathology and Molecular Medicine, Behsotun Lab, Alborz University of Medical Sciences, Karaj, Iran
| | - Dariush Hamedi Asl
- Department of Pathology and Molecular Medicine, Mehr Lab, Alborz University of Medical Sciences, Hashtgerd, Iran
| | - Babak Rahmani
- Department of Molecular Medicine, Faculty of Medical Sciences, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Rozhin Shahbadori
- Department of Pathology and Molecular Medicine, Mehr Lab, Alborz University of Medical Sciences, Hashtgerd, Iran
| | - Sara Karami
- Department of Pathology and Molecular Medicine, Behsotun Lab, Alborz University of Medical Sciences, Karaj, Iran
| | - Amir Peymani
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sara Taghizadeh
- Translational Ophthalmology Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Samiee Rad
- Medical Microbiology Research Center, Qazvin University of Medical Sciences, Qazvin, Iran.,Department of Pathology, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
48
|
Shi S, Gong Y, Li X, Ding Y, Song G, Liu H, Zhang Z. Mutations in Classical Signaling Pathways and Their Functional Impact in Microsatellite Instability High Colorectal Cancer. Genet Test Mol Biomarkers 2023; 27:90-99. [PMID: 36989521 DOI: 10.1089/gtmb.2022.0118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
Aims: Colorectal carcinomas with microsatellite instability high (MSI-H) are a distinctive group among colorectal cancers (CRCs). This study investigated the mutations of genes in the common signaling pathways and their potential clinical implications in MSI-H CRC. Materials and Methods: Twenty-five MSI-H tumors were selected from 384 primary CRCs, and the related clinical and pathological information were also collected from medical records. A commercial kit was used to detect the mutational status of crucial oncogenes within these tumors using next generation sequencing (NGS). Fluorescence in situ hybridization and immunohistochemistry were used to validate the NGS findings. Result: In the present study, MSI-H cases accounted for 6.51% of primary CRCs, with special clinicopathological features. NGS showed that the average number of mutations per tumor in the target genes evaluated was 3.36 and ranged from 1 to 9. In total, there were 17 cases (68%) with mutations in the RAS-RAF pathway and 18 cases (72%) with mutations in the PI3K pathway among the MSI-H CRCs. The remaining two cases included an EMAP Like 4-ALK Receptor Tyrosine Kinase (EML4-ALK) fusion and one with a Erb-B2 Receptor Tyrosine Kinase 2 (ERBB2) missense mutation. Conclusion: This study found multiple variants within different signaling pathways that were mutually present in MSI-H CRCs, suggesting that such a heterogeneous group of tumors requires complex treatment responses. Thus, additional clinical molecular testing is recommended for such patients, such as NGS, to inform the appropriate treatment strategies.
Collapse
Affiliation(s)
- Shanshan Shi
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuxi Gong
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao Li
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Ding
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guoxin Song
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haiyan Liu
- Department of Biology, College of Ecology, Lishui University, Lishui City, China
| | - Zhihong Zhang
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
49
|
Hoff FW, Madanat YF. Molecular Drivers of Myelodysplastic Neoplasms (MDS)-Classification and Prognostic Relevance. Cells 2023; 12:627. [PMID: 36831294 PMCID: PMC9954608 DOI: 10.3390/cells12040627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/12/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Myelodysplastic neoplasms (MDS) form a broad spectrum of clonal myeloid malignancies arising from hematopoietic stem cells that are characterized by progressive and refractory cytopenia and morphological dysplasia. Recent advances in unraveling the underlying pathogenesis of MDS have led to the identification of molecular drivers and secondary genetic events. With the overall goal of classifying patients into relevant disease entities that can aid to predict clinical outcomes and make therapeutic decisions, several MDS classification models (e.g., French-American-British, World Health Organization, and International Consensus Classification) as well as prognostication models (e.g., International Prognostic Scoring system (IPSS), the revised IPSS (IPSS-R), and the molecular IPSS (IPSS-M)), have been developed. The IPSS-M is the first model that incorporates molecular data for individual genes and facilitates better prediction of clinical outcome parameters compared to older versions of this model (i.e., overall survival, disease progression, and leukemia-free survival). Comprehensive classification and accurate risk prediction largely depend on the integration of genetic mutations that drive the disease, which is crucial to improve the diagnostic work-up, guide treatment decision making, and direct novel therapeutic options. In this review, we summarize the most common cytogenetic and genomic drivers of MDS and how they impact MDS prognosis and treatment decisions.
Collapse
Affiliation(s)
- Fieke W. Hoff
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390-8565, USA
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390-8565, USA
| | - Yazan F. Madanat
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390-8565, USA
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390-8565, USA
| |
Collapse
|
50
|
Measurable Residual Disease and Clonal Evolution in Acute Myeloid Leukemia from Diagnosis to Post-Transplant Follow-Up: The Role of Next-Generation Sequencing. Biomedicines 2023; 11:biomedicines11020359. [PMID: 36830896 PMCID: PMC9953407 DOI: 10.3390/biomedicines11020359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
It has now been ascertained that acute myeloid leukemias-as in most type of cancers-are mixtures of various subclones, evolving by acquiring additional somatic mutations over the course of the disease. The complexity of leukemia clone architecture and the phenotypic and/or genotypic drifts that can occur during treatment explain why more than 50% of patients-in hematological remission-could relapse. Moreover, the complexity and heterogeneity of clone architecture represent a hindrance for monitoring measurable residual disease, as not all minimal residual disease monitoring methods are able to detect genetic mutations arising during treatment. Unlike with chemotherapy, which imparts a relatively short duration of selective pressure on acute myeloid leukemia clonal architecture, the immunological effect related to allogeneic hematopoietic stem cell transplant is prolonged over time and must be overcome for relapse to occur. This means that not all molecular abnormalities detected after transplant always imply inevitable relapse. Therefore, transplant represents a critical setting where a measurable residual disease-based strategy, performed during post-transplant follow-up by highly sensitive methods such as next-generation sequencing, could optimize and improve treatment outcome. The purpose of our review is to provide an overview of the role of next-generation sequencing in monitoring both measurable residual disease and clonal evolution in acute myeloid leukemia patients during the entire course of the disease, with special focus on the transplant phase.
Collapse
|