1
|
Tóth DM, Szeri F, Ashaber M, Muazu M, Székvölgyi L, Arányi T. Tissue-specific roles of de novo DNA methyltransferases. Epigenetics Chromatin 2025; 18:5. [PMID: 39819598 PMCID: PMC11740433 DOI: 10.1186/s13072-024-00566-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/23/2024] [Indexed: 01/19/2025] Open
Abstract
DNA methylation, catalyzed by DNA methyltransferases (DNMT), plays pivotal role in regulating embryonic development, gene expression, adaption to environmental stress, and maintaining genome integrity. DNMT family consists of DNMT1, DNMT3A, DNMT3B, and the enzymatically inactive DNMT3L. DNMT3A and DNMT3B establish novel methylation patterns maintained by DNMT1 during replication. Genetic variants of DNMT3A and DNMT3B cause rare diseases such as Tatton-Brown-Rahman and ICF syndromes. Additionally, somatic mutations cause common conditions such as osteoarthritis, osteoporosis, clonal hematopoiesis of indeterminate potential (CHIP), hematologic malignancies, and cancer. While DNMTs have been extensively studied in vitro, in early development and in disease, their detailed physiologic roles remain less understood as in vivo investigations are hindered by the embryonic or perinatal lethality of the knockout mice. To circumvent this problem, tissue-specific Dnmt3a and Dnmt3b knockouts were engineered. This review explores their diverse molecular roles across various organs and cell types and characterizes the phenotype of the knockout mice. We provide a comprehensive collection of over forty tissue-specific knockout models generated by cre recombinase. We highlight the distinct functions of DNMT3A and DNMT3B in germ cells, early development, uterus, hematopoietic differentiation, musculoskeletal development, visceral organs, and nervous system. Our findings indicate that DNMT3A primarily regulates hematopoietic differentiation, while DNMT3B is crucial for cartilage homeostasis and ossification. We emphasize the context-dependent roles of DNMT3A and DNMT3B and demonstrate that they also complement DNMT1 maintenance methyltransferase activity. Overall, the expression patterns of DNMTs across tissues provide insights into potential therapeutic applications for treating neurologic diseases, cancer, and osteoporosis.
Collapse
Affiliation(s)
- Dániel Márton Tóth
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary.
| | - Flóra Szeri
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
| | - Mária Ashaber
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Muhyiddeen Muazu
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Lóránt Székvölgyi
- Department of Molecular and Nanopharmaceutics, Genome Architecture and Recombination Research Group, Faculty of Pharmacy, MTA-DE Momentum, University of Debrecen, Debrecen, Hungary.
| | - Tamás Arányi
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary.
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
| |
Collapse
|
2
|
Saga T, Kanagawa M, Harada T, Lang L, Yamawaki F, Ishihara T. Prognostic Value of Pretreatment Fetal Hemoglobin Levels in Patients with Myelodysplastic Syndromes and Acute Myeloid Leukemia Treated with Azacitidine: A Single-center Retrospective Study. Intern Med 2024; 63:781-790. [PMID: 37495538 PMCID: PMC11008988 DOI: 10.2169/internalmedicine.1216-22] [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: 11/02/2022] [Accepted: 06/05/2023] [Indexed: 07/28/2023] Open
Abstract
Objective Azacitidine (AZA) has been the standard of care for elderly patients with high-risk myelodysplastic syndromes (MDS). However, reliable clinical predictors of outcome have yet to be identified. The prognostic value of fetal hemoglobin (HbF) levels has been reported for decitabine therapy. We evaluated pretreatment HbF levels in AZA monotherapy as a prognostic marker in MDS/acute myeloid leukemia (AML). Methods This study included chemotherapy-naïve patients who had received seven-day treatment schedules of AZA and whose HbF levels were measured at the onset of treatment between March 2011 and July 2020. Patients were grouped into HbF-normal (<1.0%) or HbF-elevated (≥1.0%) groups. Responses were classified according to the International Working Group 2006 criteria. Patients Twenty-nine patients were included and classified as having either MDS (n=21), chronic myelomonocytic leukemia (n=5), myelodysplastic/myeloproliferative neoplasm unclassifiable (n=1), or AML with <30% marrow blasts (n=2) based on the World Health Organization 2016 diagnostic criteria. According to the revised International Prognostic Scoring System classification, 20/29 patients were at intermediate, high, or very high risk. Pretreatment HbF levels were elevated in 13/29 patients. Results The median follow-up duration was 13.0 (range 1.5-93.5) months. The HbF-elevated group was associated with a significantly higher hematologic improvement rate (76.9% vs. 25%, p=0.009) and better overall survival (median, 21.0 vs. 13.0 months, p=0.048) than the HbF-normal group. Conclusion These results suggest that elevated pretreatment HbF levels can predict better outcomes in patients with MDS/AML treated with AZA.
Collapse
Affiliation(s)
- Tomoyuki Saga
- Department of Hematology, Kin-ikyo Chuo Hospital, Japan
| | | | - Tomoya Harada
- Department of Hematology, Kin-ikyo Chuo Hospital, Japan
| | - Lang Lang
- Department of Hematology, Kin-ikyo Chuo Hospital, Japan
| | | | | |
Collapse
|
3
|
Caballero JC, Dávila J, López-Pavía M, Such E, Bernal T, Ramos F, Calabuig M, Hernández Sánchez JM, Pomares H, Sánchez Barba M, Abáigar M, González B, Merchán B, Sancho-Tello R, Callejas M, Muñoz-Novas C, Cerveró C, Sanz G, Hernández Rivas JM, Díez Campelo M. Outcomes and effect of somatic mutations after erythropoiesis stimulating agents in patients with lower-risk myelodysplastic syndromes. Ther Adv Hematol 2024; 15:20406207231218157. [PMID: 38186638 PMCID: PMC10768603 DOI: 10.1177/20406207231218157] [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: 03/23/2023] [Accepted: 11/15/2023] [Indexed: 01/09/2024] Open
Abstract
Background Erythropoiesis stimulating agents (ESAs) are the first-line therapy in patients with lower-risk myelodysplastic syndromes (LR-MDS). Some predictive factors for ESAs response have been identified. Type and number of somatic mutations have been associated with prognosis and response to therapies in MDS patients. Objectives The objective was to evaluate the outcomes after ESAs in patients with LR-MDS and to address the potential predictive value of somatic mutations in ESAs-treated patients. Design Multi-center retrospective study of a cohort of 722 patients with LR-MDS included in the SPRESAS (Spanish Registry of Erythropoietic Stimulating Agents Study) study. Retrospective analysis of 65 patients with next generation sequencing (NGS) data from diagnosis. Methods ESAs' efficacy and safety were evaluated in patients receiving ESAs and best supportive care (BSC). To assess the potential prognostic value of somatic mutations in erythroid response (ER) rate and outcome, NGS was performed in responders and non-responders. Results ER rate for ESAs-treated patients was 65%. Serum erythropoietin (EPO) level <200 U/l was the only variable significantly associated with a higher ER rate (odds ratio, 2.45; p = 0.036). Median overall survival (OS) in patients treated with ESAs was 6.7 versus 3.1 years in patients receiving BSC (p < 0.001). From 65 patients with NGS data, 57 (87.7%) have at least one mutation. We observed a trend to a higher frequency of ER among patients with a lower number of mutated genes (40.4% in <3 mutated genes versus 22.2% in ⩾3; p = 0.170). The presence of ⩾3 mutated genes was also significantly associated with worse OS (hazard ratio, 2.8; p = 0.015), even in responders. A higher cumulative incidence of acute myeloid leukemia progression at 5 years was also observed in patients with ⩾3 mutated genes versus <3 (33.3% and 10.7%, respectively; p < 0.001). Conclusion This large study confirms the beneficial effect of ESAs and the adverse effect of somatic mutations in patients with LR-MDS.
Collapse
Affiliation(s)
- Juan Carlos Caballero
- Hematology Department, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Julio Dávila
- Hematology Department, Hospital Nuestra Señora de Sonsoles, Ávila, Spain
| | - María López-Pavía
- Hematology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Universidad de Valencia, Valencia, Spain
| | - Esperanza Such
- Hematology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Universidad de Valencia, Valencia, Spain
| | - Teresa Bernal
- Hematology Department, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Fernando Ramos
- Hematology Department, Hospital Universitario de León, León, Spain
| | - Marisa Calabuig
- Hematology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Jesús María Hernández Sánchez
- Instituto de Biología Molecular y Celular del Cáncer, Instituto Biosanitario de Salamanca, Centro de Investigación del Cáncer, USAL-CSIC, Salamanca, Spain
| | - Helena Pomares
- Hematology Department, Hospital Universitario de Bellvitge-Hospital Duran I Reynals, Instituto Catalán de Oncología, L’Hospitalet del Llobregat, Spain
| | | | - María Abáigar
- Instituto de Biología Molecular y Celular del Cáncer, Instituto Biosanitario de Salamanca, Centro de Investigación del Cáncer, USAL-CSIC, Salamanca, Spain
| | - Bernardo González
- Hematology Department, Hospital Universitario de Canarias, La Laguna, Spain
| | - Brayan Merchán
- Hematology Department, Hospital Universitario Vall d’Hebron, Barcelona, Spain
| | | | - Marta Callejas
- Hematology Department, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Spain
| | | | - Carlos Cerveró
- Hematology Department, Hospital Virgen de la Luz, Cuenca, Spain
| | - Guillermo Sanz
- Hematology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
- Universidad de Valencia, Valencia, Spain
| | - Jesús María Hernández Rivas
- Instituto de Biología Molecular y Celular del Cáncer, Instituto Biosanitario de Salamanca, Centro de Investigación del Cáncer, USAL-CSIC, Salamanca, Spain Hematology Department, Hospital Universitario de Salamanca, Salamanca, Spain Instituto Biosanitario de Salamanca, Salamanca, Spain
- Hematology Department, Hospital Universitario de Salamanca, Salamanca, Spain Instituto Biosanitario de Salamanca, Salamanca, Spain
- Instituto Biosanitario de Salamanca, Salamanca, Spain
| | - María Díez Campelo
- Hematology Department, Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, Salamanca 37007, Spain
- Instituto Biosanitario de Salamanca, Salamanca, Spain
| |
Collapse
|
4
|
Gao J, Hu Y, Gao L, Xiao P, Lu J, Hu S. The effect of decitabine-combined minimally myelosuppressive regimen bridged allo-HSCT on the outcomes of pediatric MDS from 10 years' experience of a single center. BMC Pediatr 2022; 22:312. [PMID: 35624441 PMCID: PMC9137053 DOI: 10.1186/s12887-022-03376-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 05/19/2022] [Indexed: 11/21/2022] Open
Abstract
Background Myelodysplastic syndrome (MDS) is a rare disease in children and the treatment option before the allogeneic hematopoietic stem cell transplantation (allo-HSCT) is rarely reported. Our main objective was to report our single-center experience with the DNA-hypomethylating agent, decitabine-combined minimally myelosuppressive regimen (DAC + MMR) bridged allo-HSCT in children with MDS. Methods Twenty-eight children with de novo MDS who underwent allo-HSCT between 2011 and 2020 were enrolled. Patients were divided into subgroups (refractory cytopenia of childhood [RCC] and advanced MDS [aMDS]) and treated by HSCT alone or pre-transplant combination treatment based on risk stratification. The patients’ clinical characteristics, treatment strategies and outcomes were retrospectively evaluated. Results Twenty patients with aMDS had received pre-transplant treatment (three were treated with decitabine alone, thirteen with DAC + MMR, and four with acute myeloid leukemia type [AML-type] induction therapy). DAC + MMR was well tolerated and the most common adverse events were myelosuppression and gastrointestinal reaction. DAC + MMR had shown an improved marrow complete remission (mCR) compared with AML-type chemotherapy (13/13, 100% versus 2/4, 50%, P = 0.044). The median follow-up for total cohort was 53.0 months (range, 2.3-127.0 months) and the 4-year overall survival (OS) was 71.4 ± 8.5%. In the subgroup of aMDS, pretreatment of DAC + MMR resulted in a much better survival rate than AML-type chemotherapy (84.6 ± 10.0% versus 0.0 ± 0.0%, P < 0.001). Conclusions The DAC + MMR bridged allo-HSCT may be recommended as a novel and effective approach. Supplementary Information The online version contains supplementary material available at 10.1186/s12887-022-03376-1.
Collapse
Affiliation(s)
- Junyan Gao
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Pediatrics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Yixin Hu
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Li Gao
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Peifang Xiao
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun Lu
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shaoyan Hu
- Department of Hematology & Oncology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
5
|
Hu C, Wang X. Predictive and prognostic value of gene mutations in myelodysplastic syndrome treated with hypomethylating agents: a meta-analysis. Leuk Lymphoma 2022; 63:2336-2351. [PMID: 35543621 DOI: 10.1080/10428194.2022.2070913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although the effect of gene mutations on overall response rate (ORR) and overall survival (OS) in myelodysplastic syndrome (MDS) treated with hypomethylating agents (HMAs) has been explored, the effect is still controversial. We performed this meta-analysis to investigate the effect. The pooled odds ratio (OR) and 95% confidence interval (CI) for ORR and the pooled hazard ratio (HR) and 95%CI for OS were chosen to estimate the effect. The pooled OR of TET2 was 0.73 (95%CI: 0.59-0.91, p = 0.005) and the pooled OR of ASXL1 was 1.38 (95%CI: 1.12-1.71, p = 0.003). As for prognosis, the pooled HR of RUNX1 was 1.45 (95%CI: 1.15-1.85, p = 0.002). The pooled HR of TP53 was 2.30 (95%CI: 1.83-2.90, p < 0.001) and the pooled HR of U2AF1 was 1.41 (95%CI: 1.15-1.74, p = 0.001). There was no statistical difference shown in other genes. Therefore, TET2 mutation and ASXL1 wild-type were the predictor of better response to HMAs. Mutations of TP53, RUNX1, and U2AF1 were associated with poor prognosis in MDS.
Collapse
Affiliation(s)
- Chaolu Hu
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoqin Wang
- Department of Hematology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Brunner AM, Fell G, Steensma DP. Historical expectations with DNA methyltransferase inhibitor monotherapy in MDS: when is combination therapy truly "promising"? Blood Adv 2022; 6:2854-2866. [PMID: 35143613 PMCID: PMC9092413 DOI: 10.1182/bloodadvances.2021006357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 02/09/2022] [Indexed: 11/20/2022] Open
Abstract
DNA methyltransferase inhibitors (DNMTIs) for patients with higher risk myelodysplastic syndromes (HR-MDS) have low complete remission rates and are not curative. Early DNMTI combination clinical trials in HR-MDS are often termed "promising," but many randomized trials subsequently failed to show benefit. Clearer understanding of when a combination is likely to improve upon DNMTI monotherapy would inform randomized studies. We reviewed MDS azacitidine or decitabine monotherapy studies. We collected baseline demographics including International Prognostic Scoring System (IPSS) risk, DNMTI, disease characteristics; and response variables including survival and marrow and hematologic responses. Aggregate estimates across studies were calculated using meta-analyses techniques. Using a binomial design, we estimated the necessary operating characteristics to design a phase 2 study showing improved efficacy of a combination over monotherapy. Among 1908 patients, the overall response rate (ORR) was 24% (n = 464; 95% confidence interval [CI], 0.22-0.26): 267 complete response (CR, 14%), 68 partial response (4%), and 129 marrow complete remission (7%). Among 1604 patients for whom a hematologic response was reported, 476 (30%; 95% CI, 0.27-0.32) reported hematologic improvement (HI). More patients treated with azacitidine achieved HI (38%; 95% CI, 0.35-0.41) compared with decitabine (15%; 95% CI, 0.13-0.19), whereas the marrow ORR rate was higher with decitabine (29%; 95% CI, 0.26-0.33) compared with azacitidine (21%; 95% CI, 0.19-0.23). CR rates were similar between DNMTIs: 13% with azacitidine and 16% with decitabine. Variables that influence MDS response include the specific DNMTI backbone and the distribution of IPSS risk of patients enrolled on a trial. Considering these factors can help identify which early combination approaches are worth assessing in larger randomized trials.
Collapse
Affiliation(s)
- Andrew M Brunner
- Department of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Geoffrey Fell
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA; and
| | | |
Collapse
|
7
|
Can the New and Old Drugs Exert an Immunomodulatory Effect in Acute Myeloid Leukemia? Cancers (Basel) 2021; 13:cancers13164121. [PMID: 34439275 PMCID: PMC8393879 DOI: 10.3390/cancers13164121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/11/2021] [Accepted: 08/14/2021] [Indexed: 12/30/2022] Open
Abstract
Simple Summary The advent of novel immunotherapeutic strategies has revealed the importance of immune dysregulation and of a tolerogenic microenvironment for acute myeloid leukemia (AML) fitness. We reviewed the “off-target” effects on the immune system of different drugs used in the treatment of AML to explore the advantages of this unexpected interaction. Abstract Acute myeloid leukemia (AML) is considered an immune-suppressive neoplasm capable of evading immune surveillance through cellular and environmental players. Increasing knowledge of the immune system (IS) status at diagnosis seems to suggest ever more attention of the crosstalk between the leukemic clone and its immunologic counterpart. During the last years, the advent of novel immunotherapeutic strategies has revealed the importance of immune dysregulation and suppression for leukemia fitness. Considering all these premises, we reviewed the “off-target” effects on the IS of different drugs used in the treatment of AML, focusing on the main advantages of this interaction. The data reported support the idea that a successful therapeutic strategy should consider tailored approaches for performing leukemia eradication by both direct blasts killing and the engagement of the IS.
Collapse
|
8
|
Bouchla A, Thomopoulos TP, Papageorgiou SG, Apostolopoulou C, Loucari C, Mpazani E, Pappa V. Predicting outcome in higher-risk myelodysplastic syndrome patients treated with azacitidine. Epigenomics 2021; 13:1129-1143. [PMID: 34291653 DOI: 10.2217/epi-2021-0124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
5-Azacitidine (5-AZA) is widely used for the treatment of higher-risk myelodysplastic syndromes. However, response and survival rates vary considerably, while indicated treatment duration remains undefined. For these reasons, factors determining response and survival are of major importance. Clinical, morphological, flow cytometry, cytogenetic and molecular factors are discussed in this review. Biomarkers predictive of response and prognosis, as well as their link to the mode of action of 5-AZA are also addressed, shifting the focus from clinical practice to investigational research. Their use could further improve prognostic classification of 5-AZA treated higher-risk myelodysplastic syndromes in the near future.
Collapse
Affiliation(s)
- Anthi Bouchla
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| | - Thomas P Thomopoulos
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| | - Sotirios G Papageorgiou
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| | - Christina Apostolopoulou
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| | - Constantinos Loucari
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| | - Efthimia Mpazani
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| | - Vasiliki Pappa
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| |
Collapse
|
9
|
Watad A, Kacar M, Bragazzi NL, Zhou Q, Jassam M, Taylor J, Roman E, Smith A, Jones RA, Amital H, Cargo C, McGonagle D, Savic S. Somatic Mutations and the Risk of Undifferentiated Autoinflammatory Disease in MDS: An Under-Recognized but Prognostically Important Complication. Front Immunol 2021; 12:610019. [PMID: 33679746 PMCID: PMC7933213 DOI: 10.3389/fimmu.2021.610019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/25/2021] [Indexed: 01/13/2023] Open
Abstract
Objectives: We theorized that myelodysplastic syndrome (MDS) with somatic mutations and karyotype abnormalities are associated with autoinflammation, and that the presence of autoinflammatory disease affected prognosis in MDS. Methods: One hundred thirty-four MDS patients were assessed for the prevalence of autoinflammatory complications and its link with karyotypes and somatic mutation status. Autoinflammatory complications were described either as well-defined autoinflammatory diseases (AD) or undifferentiated "autoinflammatory disease" (UAD) (defined as CRP over 10.0 mg/L on five consecutive occasions, taken at separate times and not explained by infection). Several patient characteristics including demographic, clinical, laboratory, cytogenetics charts, and outcomes, were compared between different groups. Results: Sixty-two (46.3%) patients had an autoinflammatory complication manifesting as arthralgia (43.5% vs. 23.6%, p = 0.0146), arthritis (30.6% vs. 15.3%, p = 0.0340), skin rash (27.4% vs. 12.5%, p = 0.0301), pleuritis (14.5% vs. 4.2%, p = 0.0371) and unexplained fever (27.4% vs. 0%, p < 0.0001). AD were found in 7.4% of MDS patients (with polymyalgia rheumatic being the most frequently one). Classical autoimmune diseases were found only in 4 MDS patients (3.0%). Transcription factor pathway mutations (RUNX1, BCOR, WTI, TP53) (OR 2.20 [95%CI 1.02-4.75], p = 0.0451) and abnormal karyotypes (OR 2.76 [95%CI 1.22-6.26], p = 0.0153) were associated with autoinflammatory complications. Acute leukaemic transformation was more frequent in MDS patients with autoinflammatory features than those without (27.4% vs. 9.7%, p = 0.0080). Conclusions: Autoinflammatory complications are common in MDS. Somatic mutations of transcription factor pathways and abnormal karyotypes are associated with greater risk of autoinflammatory complications, which are themselves linked to malignant transformation and a worse prognosis.
Collapse
Affiliation(s)
- Abdulla Watad
- National Institute for Health Research—Leeds Biomedical Research Centre and Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, St James's University Hospital, Leeds, United Kingdom
- Department of Medicine B and Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Mark Kacar
- National Institute for Health Research—Leeds Biomedical Research Centre and Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, St James's University Hospital, Leeds, United Kingdom
- Department of Clinical Immunology and Allergy, St James's University Hospital, Leeds, United Kingdom
| | - Nicola Luigi Bragazzi
- Laboratory for Industrial and Applied Mathematics (LIAM), Department of Mathematics and Statistics, York University, Toronto, ON, Canada
| | - Qiao Zhou
- National Institute for Health Research—Leeds Biomedical Research Centre and Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, St James's University Hospital, Leeds, United Kingdom
- Department of Rheumatology & Immunology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Miriam Jassam
- Department of Clinical Immunology and Allergy, St James's University Hospital, Leeds, United Kingdom
| | - Jan Taylor
- Department of Haematology, St James's University Hospital, Leeds, United Kingdom
| | - Eve Roman
- Epidemiology & Cancer Statistics Group, Department of Health Sciences, University of York, York, United Kingdom
| | - Alexandra Smith
- Epidemiology & Cancer Statistics Group, Department of Health Sciences, University of York, York, United Kingdom
| | - Richard A. Jones
- HMDS Department, Leeds Teaching Hospitals, Leeds, United Kingdom
| | - Howard Amital
- Department of Medicine B and Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Catherine Cargo
- HMDS Department, Leeds Teaching Hospitals, Leeds, United Kingdom
| | - Dennis McGonagle
- National Institute for Health Research—Leeds Biomedical Research Centre and Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, St James's University Hospital, Leeds, United Kingdom
| | - Sinisa Savic
- National Institute for Health Research—Leeds Biomedical Research Centre and Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, St James's University Hospital, Leeds, United Kingdom
- Department of Clinical Immunology and Allergy, St James's University Hospital, Leeds, United Kingdom
| |
Collapse
|
10
|
Unravelling the Epigenome of Myelodysplastic Syndrome: Diagnosis, Prognosis, and Response to Therapy. Cancers (Basel) 2020; 12:cancers12113128. [PMID: 33114584 PMCID: PMC7692163 DOI: 10.3390/cancers12113128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/19/2020] [Accepted: 10/24/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Myelodysplastic syndrome (MDS) is a type of blood cancer that mostly affects older individuals. Invasive tests to obtain bone samples are used to diagnose MDS and many patients do not respond to therapy or stop responding to therapy in the short-term. Less invasive tests to help diagnose, prognosticate, and predict response of patients is a felt need. Factors that influence gene expression without changing the DNA sequence (epigenetic modifiers) such as DNA methylation, micro-RNAs and long-coding RNAs play an important role in MDS, are potential biomarkers and may also serve as targets for therapy. Abstract Myelodysplastic syndrome (MDS) is a malignancy that disrupts normal blood cell production and commonly affects our ageing population. MDS patients are diagnosed using an invasive bone marrow biopsy and high-risk MDS patients are treated with hypomethylating agents (HMAs) such as decitabine and azacytidine. However, these therapies are only effective in 50% of patients, and many develop resistance to therapy, often resulting in bone marrow failure or leukemic transformation. Therefore, there is a strong need for less invasive, diagnostic tests for MDS, novel markers that can predict response to therapy and/or patient prognosis to aid treatment stratification, as well as new and effective therapeutics to enhance patient quality of life and survival. Epigenetic modifiers such as DNA methylation, long non-coding RNAs (lncRNAs) and micro-RNAs (miRNAs) are perturbed in MDS blasts and the bone marrow micro-environment, influencing disease progression and response to therapy. This review focusses on the potential utility of epigenetic modifiers in aiding diagnosis, prognosis, and predicting treatment response in MDS, and touches on the need for extensive and collaborative research using single-cell technologies and multi-omics to test the clinical utility of epigenetic markers for MDS patients in the future.
Collapse
|
11
|
Dahn ML, Cruickshank BM, Jackson AJ, Dean C, Holloway RW, Hall SR, Coyle KM, Maillet H, Waisman DM, Goralski KB, Giacomantonio CA, Weaver ICG, Marcato P. Decitabine Response in Breast Cancer Requires Efficient Drug Processing and Is Not Limited by Multidrug Resistance. Mol Cancer Ther 2020; 19:1110-1122. [PMID: 32156786 DOI: 10.1158/1535-7163.mct-19-0745] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/30/2020] [Accepted: 03/05/2020] [Indexed: 11/16/2022]
Abstract
Dysregulation of DNA methylation is an established feature of breast cancers. DNA demethylating therapies like decitabine are proposed for the treatment of triple-negative breast cancers (TNBC) and indicators of response need to be identified. For this purpose, we characterized the effects of decitabine in a panel of 10 breast cancer cell lines and observed a range of sensitivity to decitabine that was not subtype specific. Knockdown of potential key effectors demonstrated the requirement of deoxycytidine kinase (DCK) for decitabine response in breast cancer cells. In treatment-naïve breast tumors, DCK was higher in TNBCs, and DCK levels were sustained or increased post chemotherapy treatment. This suggests that limited DCK levels will not be a barrier to response in patients with TNBC treated with decitabine as a second-line treatment or in a clinical trial. Methylome analysis revealed that genome-wide, region-specific, tumor suppressor gene-specific methylation, and decitabine-induced demethylation did not predict response to decitabine. Gene set enrichment analysis of transcriptome data demonstrated that decitabine induced genes within apoptosis, cell cycle, stress, and immune pathways. Induced genes included those characterized by the viral mimicry response; however, knockdown of key effectors of the pathway did not affect decitabine sensitivity suggesting that breast cancer growth suppression by decitabine is independent of viral mimicry. Finally, taxol-resistant breast cancer cells expressing high levels of multidrug resistance transporter ABCB1 remained sensitive to decitabine, suggesting that the drug could be used as second-line treatment for chemoresistant patients.
Collapse
Affiliation(s)
- Margaret L Dahn
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Ainsleigh J Jackson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Cheryl Dean
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ryan W Holloway
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Steven R Hall
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Krysta M Coyle
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hillary Maillet
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - David M Waisman
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Kerry B Goralski
- Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada.,College of Pharmacy, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carman A Giacomantonio
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Surgery, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ian C G Weaver
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada.,Brain Repair Centre, Halifax, Nova Scotia, Canada
| | - Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada. .,Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
12
|
Liang S, Zhou X, Pan H, Yang Y, Shi L, Wang L. Prognostic value of DNMT3A mutations in myelodysplastic syndromes: a meta-analysis. ACTA ACUST UNITED AC 2020; 24:613-622. [PMID: 31482762 DOI: 10.1080/16078454.2019.1657613] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objectives: Although DNA (cytosine-5)-methyltransferase 3 alpha (DNMT3A) gene mutations have been widely reported in myelodysplastic syndromes (MDS), the prognostic significance of DNMT3A mutations is still controversial. In this study, we conducted a meta-analysis to determine the prognostic effect of DNMT3A mutations in patients with MDS. Methods: Eligible studies from PubMed, Embase, Web of Science, Clinical Trials and the Cochrane Library were searched. Hazard ratios (HRs) and their 95% confidence intervals (CIs) for overall survival (OS) and leukemia-free survival (LFS) were pooled to assess the effect of DNMT3A mutations on the prognosis in MDS patients. Results: A total of 12 studies with 2236 patients were included in this meta-analysis. The pooled HRs for OS and LFS revealed that MDS patients with DNMT3A mutations had a significantly poor prognosis as compared with those without mutations (OS: HR = 1.654, 95% CI = 1.387-1.973, p < 0.001; LFS: HR = 4.624, 95% CI = 3.121-6.851, p < 0.001). Discussion and Conclusion: This meta-analysis showed an adverse prognostic effect of DNMT3A mutations in patients with MDS, which will contribute to risk stratification and prognostic assessment in the disease.
Collapse
Affiliation(s)
- Simin Liang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Xiaojia Zhou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Hui Pan
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Yichun Yang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Lin Shi
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University , Chongqing , People's Republic of China
| |
Collapse
|