1
|
Milosevic J, Fransson S, Svensson J, Otte J, Olsen TK, Sveinbjornsson B, Hertwig F, Bartenhagen C, Abel F, Reinsbach SE, Djos A, Javanmardi N, Shi Y, Hehir-Kwa JY, Mensenkamp A, Tytgat GA, Holmberg J, Molenaar JJ, Jongmans M, Fischer M, Baryawno N, Gisselsson D, Martinsson T, Kogner P, Johnsen JI. Gain of chromosome 17 is an early genetic abnormality in neuroblastoma with PPM1D emerging as a strong candidate oncogene driving tumor progression. Cancer Lett 2025:217769. [PMID: 40320038 DOI: 10.1016/j.canlet.2025.217769] [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: 02/25/2025] [Revised: 04/15/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Segmental gain of chromosome 17q is the most common genetic aberration in high-risk neuroblastoma, but its role in disease progression is poorly understood. This study aims to address the contribution of 17q gain to neuroblastoma malignancy. We analyzed the genetic and transcriptional landscape of 417 neuroblastoma patients across various risk groups and clinical stages using multi-omic approaches. Single-cell RNA/DNA sequencing and SNP arrays were combined to characterize genomic aberrations, while evolutionary trajectories were mapped to explore the accumulation of genetic changes in patients with neuroblastoma. Additionally, DNA and RNA sequencing were used to assess mutational burden and gene expression patterns. Our findings suggest that chromosome 17 gain is an early genetic event acquired during neuroblastoma development, correlating with the accumulation of additional chromosomal aberrations and poor prognosis. Increased segmental gains of chromosome 17q were observed during clonal evolution, relapse disease and metastasis. We identified PPM1D, a p53-inducible Ser/Thr phosphatase located on chr17q22.3, as a key player activated by segmental 17q-gain, gene-fusion, or gain-of-function somatic and germline mutations, further promoting neuroblastoma development/progression. Gain of chromosome 17 is an early driver of genetic instability in neuroblastoma, with PPM1D emerging as a potential candidate gene implicated in high-risk disease progression.
Collapse
Affiliation(s)
- Jelena Milosevic
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, United States.
| | - Susanne Fransson
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Johanna Svensson
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Jörg Otte
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Thale K Olsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Department of Immunology, Genetics, and Pathology, Uppsala university, Uppsala, Sweden
| | - Baldur Sveinbjornsson
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Falk Hertwig
- Department of Experimental Pediatric Oncology, University Children's Hospital, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
| | - Christoph Bartenhagen
- Department of Experimental Pediatric Oncology, University Children's Hospital, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
| | - Frida Abel
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Susanne E Reinsbach
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Anna Djos
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Niloufar Javanmardi
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Yao Shi
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jane Y Hehir-Kwa
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Arjen Mensenkamp
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Johan Holmberg
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden; Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Jan J Molenaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | | | - Matthias Fischer
- Department of Experimental Pediatric Oncology, University Children's Hospital, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
| | - Ninib Baryawno
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - David Gisselsson
- Division of Clinical Genetics, Faculty of Medicine, Lund University, Lund, Sweden; Section for Pathology, Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office of Medical Services, Region Skåne, Sweden
| | - Tommy Martinsson
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - John Inge Johnsen
- Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
2
|
Kim H, Lee JK, Hong YJ, Kang HJ, Byun BH, Lee SS. PPM1D Mutation as a Distinct Feature of Myeloid Neoplasms in B-Cell Non-Hodgkin Lymphoma Patients: A Retrospective Analysis. Cancers (Basel) 2025; 17:1185. [PMID: 40227763 PMCID: PMC11988103 DOI: 10.3390/cancers17071185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Myeloid neoplasms are the most common secondary blood cancer in B-cell non-Hodgkin lymphoma (BNHL) patients treated with cytotoxic therapies. We aimed to characterize the genetic and clinicopathologic features of myeloid neoplasms arising after B-cell non-Hodgkin lymphoma (MN-BNHL) by comparing their features with myeloid neoplasms developing after solid cancer (MN-SC). METHODS We retrospectively analyzed the clinicopathologic and genetic data of myeloid neoplasm patients diagnosed between 2008 and 2023, categorized as MN-BNHL or MN-SC. Further NGS analysis was performed on available bone marrow samples with missing genetic data. The genetic profiles of myeloid neoplasms between BNHL and solid cancer groups were compared. RESULTS Sixteen patients developed MN-BNHL. Among the 11 MN-BNHL patients undergoing NGS, all harbored tier 1 mutations. PPM1D mutations (PPM1Dms) were most frequent (73%), followed by DNMT3A (46%) and TP53 (36%). PPM1Dms were significantly more prevalent than in MN-SC (n = 21), where TP53 mutations were most common (64%) (p < 0.001). PPM1Dms often co-occurred with DNMT3A. They were associated with prior radioimmunotherapy (relative risk (RR): 3.3 and RR 3.57). MN-BNHL patients with PPM1Dms exhibited improved survival compared to those without (p = 0.0376), but this benefit was negated by the presence of TP53 mutations (p = 0.0049). CONCLUSIONS PPM1Dms are a prominent genetic feature in MN-BNHL, suggesting a distinct role in its development compared to MN-SC. Further investigation is needed to elucidate the precise contribution of PPM1D and its interaction with other mutations in BNHL-related myeloid neoplasm development and prognosis.
Collapse
Affiliation(s)
- Heyjin Kim
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.K.); (Y.J.H.)
- Medical Science Demonstration Center, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Jin Kyung Lee
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.K.); (Y.J.H.)
| | - Young Jun Hong
- Department of Laboratory Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea; (H.K.); (Y.J.H.)
| | - Hye Jin Kang
- Division of Hematology-Oncology, Department of Internal Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea;
| | - Byung Hyun Byun
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea;
| | - Seung-Sook Lee
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea;
| |
Collapse
|
3
|
Shaw S, Jiang W, Rush J, Dumont N, Kim J, Singh R, Skepner A, Khodier C, Raffier C, Murphy Z, Yan N, Schluter C, Yu X, Szuchnicki M, Sathappa M, Kahn J, Sperling AS, Wagner F, McKinney DC, Gould AE, Garvie CW, Miller PG. Identification of small molecule inhibitors of PPM1D using an integrated drug discovery platform. iScience 2025; 28:112069. [PMID: 40124519 PMCID: PMC11930361 DOI: 10.1016/j.isci.2025.112069] [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: 06/25/2024] [Revised: 10/23/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
PPM1D is a serine/threonine phosphatase recurrently activated in cancer, regulates the DNA damage response (DDR), and suppresses p53. Though PPM1D inhibition impairs tumor growth in cancer models and is the subject of multiple drug discovery efforts, no PPM1D inhibitors with clinical potential have been identified. We screened 600,000 compounds in a displacement assay and generated a hit series with nanomolar activity. We optimized our leads using internally developed assays to interrogate PPM1D, p53, and the DDR and defined important structure-activity relationships. Using an in vivo bioluminescent readout of p53 activation, we compared different DDR and p53 modulators and showed that despite having a distinct chemical structure, our lead compound had comparable in vivo activity to established PPM1D inhibitors. Our approach yielded multiple allosteric inhibitors of PPM1D, deepened our understanding of PPM1D as a drug target, and is highly amenable to studying other modulators of the DDR and p53.
Collapse
Affiliation(s)
- Subrata Shaw
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Wei Jiang
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Jason Rush
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Nancy Dumont
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - John Kim
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Ritu Singh
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Adam Skepner
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Carol Khodier
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Cerise Raffier
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Zachary Murphy
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Ni Yan
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Krantz Family Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Cameron Schluter
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Krantz Family Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Xiao Yu
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Mateusz Szuchnicki
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Murugappan Sathappa
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Josephine Kahn
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Adam S. Sperling
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Florence Wagner
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - David C. McKinney
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Alexandra E. Gould
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Colin W. Garvie
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Peter G. Miller
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Krantz Family Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
4
|
Kucheryavenko AS, Muzyko EA, Perfilova VN, Kaplanov KD, Frolov MY. The role of the PPM1D gene in tumor pathogenesis. BIOMEDITSINSKAIA KHIMIIA 2025; 71:19-28. [PMID: 40045720 DOI: 10.18097/pbmcr1495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
The PPM1D gene and its protein product (serine-threonine protein phosphatase, PPM1D or Wip1) are involved in regulation of cell's DNA damage response, cell cycle control, and repair. Amplification, overexpression, or mutations of the PPM1D gene have a significant impact on cell responses to stress factors and genetic instability as well as impairments of processes of double-strand break repair, nucleotide excision repair, base excision repair, cell cycle, and apoptosis. PPM1D dephosphorylates and thus inactivates p53, proteins that respond to DNA strand integrity damage, cell cycle checkpoint proteins, and apoptotic proteins. This contributes to tumor development, growth, and maintenance of the tumor phenotype. In this review we consider data on the role of the PPM1D gene in the formation and maintenance of various oncological processes, including tumors of the mammary glands, ovaries, prostate gland, esophagus, stomach, intestines, liver and pancreas, hemoblastoses, and others.
Collapse
Affiliation(s)
| | - E A Muzyko
- Volgograd State Medical University, Volgograd, Russia
| | - V N Perfilova
- Volgograd State Medical University, Volgograd, Russia; Volgograd Medical Research Center, Volgograd, Russia
| | | | - M Yu Frolov
- Volgograd State Medical University, Volgograd, Russia
| |
Collapse
|
5
|
Dou X, Dan C, Zhang D, Zhou H, He R, Zhou G, Zhu Y, Fu N, Niu B, Xu S, Liao Y, Luo Z, Yang L, Zhang H, Xu Y, Zhan Q, Chen W, Yang Z, Tang X, Zhang H, Xiao Q, Chen J, Liu L, Wang Y, Pei L, Wang L. Genomic mutation patterns and prognostic value in de novo and secondary acute myeloid leukemia: A multicenter study from China. Int J Cancer 2024; 155:2253-2264. [PMID: 39109820 DOI: 10.1002/ijc.35125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 10/25/2024]
Abstract
Acute myeloid leukemia (AML) can manifest as de novo AML (dn-AML) or secondary AML (s-AML), with s-AML being associated with inferior survival and distinct genomic characteristics. The underlying reasons for this disparity remain to be elucidated. In this multicenter study, next-generation sequencing (NGS) was employed to investigate the mutational landscape of AML in 721 patients from June 2020 to May 2023.Genetic mutations were observed in 93.34% of the individuals, with complex variations (more than three gene mutations) present in 63.10% of them. TET2, ASXL1, DNMT3A, TP53 and SRSF2 mutations showed a higher prevalence among older individuals, whereas WT1 and KIT mutations were more commonly observed in younger patients. BCOR, BCORL1, ZRSR2, ASXL1 and SRSF2 exhibited higher mutation frequencies in males. Additionally, ASXL1, NRAS, PPMID, SRSF2, TP53 and U2AF1 mutations were more common in patients with s-AML, which PPM1D was more frequently associated with therapy-related AML (t-AML). Advanced age and hyperleukocytosis independently served as adverse prognostic factors for both types of AML; however, s-AML patients demonstrated a greater number of monogenic adverse prognostic factors compared to dn-AML cases (ASXL1, PPM1D, TP53 and U2AF1 in s-AML vs. FLT3, TP53 and U2AF1 in dn-AML). Age and sex-related gene mutations suggest epigenetic changes may be key in AML pathogenesis. The worse prognosis of s-AML compared to dn-AML could be due to the older age of s-AML patients and more poor-prognosis gene mutations. These findings could improve AML diagnosis and treatment by identifying potential therapeutic targets and risk stratification biomarkers.
Collapse
Affiliation(s)
- Xi Dou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunli Dan
- Department of Hematology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Duanzhong Zhang
- Department of Hematology, Dazhou Central Hospital, Sichuan, China
| | - Hongjing Zhou
- Department of Hematology, Jining No. 1 People's Hospital, Shandong, China
| | - Renke He
- Department of Hematology, Chongqing General Hospital, Chongqing, China
| | - Guangyu Zhou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Zhu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Nan Fu
- Department of Hematology, Shaanxi Provincial People's Hospital, Shaanxi, China
| | - Ben Niu
- Department of Hematology, Shaanxi Provincial People's Hospital, Shaanxi, China
| | - Shuangnian Xu
- Department of Hematology, Third Military Medical University Southwest Hospital, Chongqing, China
| | - Yi Liao
- Department of Oncology and Hematology, Chongqing University Central Hospital, Chongqing, China
| | - Zhangqin Luo
- Department of Hematology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Lihua Yang
- Department of Hematology, Dazhou Central Hospital, Sichuan, China
| | - Haiguo Zhang
- Department of Hematology, Jining No. 1 People's Hospital, Shandong, China
| | - Yizhi Xu
- Department of Hematology, Chongqing General Hospital, Chongqing, China
| | - Qian Zhan
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Chen
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zesong Yang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqiong Tang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongbin Zhang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Xiao
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianbin Chen
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Liu
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Wang
- Department of Hematology, Shaanxi Provincial People's Hospital, Shaanxi, China
| | - Li Pei
- Department of Hematology, Third Military Medical University Southwest Hospital, Chongqing, China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
6
|
Kudo R, Safonov A, Jones C, Moiso E, Dry JR, Shao H, Nag S, da Silva EM, Yildirim SY, Li Q, O'Connell E, Patel P, Will M, Fushimi A, Benitez M, Bradic M, Fan L, Nakshatri H, Sudhan DR, Denz CR, Huerga Sanchez I, Reis-Filho JS, Goel S, Koff A, Weigelt B, Khan QJ, Razavi P, Chandarlapaty S. Long-term breast cancer response to CDK4/6 inhibition defined by TP53-mediated geroconversion. Cancer Cell 2024; 42:1919-1935.e9. [PMID: 39393354 DOI: 10.1016/j.ccell.2024.09.009] [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: 11/09/2023] [Revised: 07/02/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024]
Abstract
Inhibition of CDK4/6 kinases has led to improved outcomes in breast cancer. Nevertheless, only a minority of patients experience long-term disease control. Using a large, clinically annotated cohort of patients with metastatic hormone receptor-positive (HR+) breast cancer, we identify TP53 loss (27.6%) and MDM2 amplification (6.4%) to be associated with lack of long-term disease control. Human breast cancer models reveal that p53 loss does not alter CDK4/6 activity or G1 blockade but instead promotes drug-insensitive p130 phosphorylation by CDK2. The persistence of phospho-p130 prevents DREAM complex assembly, enabling cell-cycle re-entry and tumor progression. Inhibitors of CDK2 can overcome p53 loss, leading to geroconversion and manifestation of senescence phenotypes. Complete inhibition of both CDK4/6 and CDK2 kinases appears to be necessary to facilitate long-term response across genomically diverse HR+ breast cancers.
Collapse
Affiliation(s)
- Rei Kudo
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA; Department of Surgery, The Jikei University School of Medicine, Tokyo 1058461, Japan
| | - Anton Safonov
- Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - Catherine Jones
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Enrico Moiso
- Department of Medicine, MSK, New York, NY 10065, USA; Department of Epidemiology and Biostatistics, MSK, New York, NY 10065, USA
| | | | - Hong Shao
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Sharanya Nag
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Edaise M da Silva
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Selma Yeni Yildirim
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Qing Li
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Elizabeth O'Connell
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Payal Patel
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA
| | - Marie Will
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA; Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Clinical Genetics Service, Department of Medicine, MSK, New York, NY 10065, USA
| | - Atsushi Fushimi
- Department of Surgery, The Jikei University School of Medicine, Tokyo 1058461, Japan
| | - Marimar Benitez
- Program in Molecular Biology, Sloan Kettering Institute, MSK, New York, NY 10065, USA
| | - Martina Bradic
- Program in Molecular Biology, Sloan Kettering Institute, MSK, New York, NY 10065, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | - Jorge S Reis-Filho
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Shom Goel
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Andrew Koff
- Program in Molecular Biology, Sloan Kettering Institute, MSK, New York, NY 10065, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, MSK, New York, NY 10065, USA
| | - Qamar J Khan
- Division of Medical Oncology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Pedram Razavi
- Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA.
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center (MSK), New York, NY 10065, USA; Breast Medicine Service, Department of Medicine, MSK, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
7
|
Pilevneli H, Döger F, Karagenç L, Kozacı D, Kilic Eren M. PPM1D/Wip1 is amplified, overexpressed, and mutated in human non-Hodgkin's lymphomas. Mol Biol Rep 2024; 51:1115. [PMID: 39489796 DOI: 10.1007/s11033-024-10029-2] [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: 08/19/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Wip1, is a p53-dependent Ser/Thr phosphatase involved in the timely termination of DDR. The PPM1D gene encoding Wip1 is deregulated and thus gained an oncogene character in common human solid tumors and cell lines. This study assessed the oncogenic potential of the PPM1D gene in human non- Hodgkin's lymphomas (NHL), the most common hematological malignancy worldwide. METHODS AND RESULTS FFPE human lymphoid hyperplasia (LH) (n = 17) and NHL tumor lymph node samples (n = 65) and human NHL cell lines were used to assess the oncogenic potential of the PPM1D gene in the present study. Copy number gain and mRNA expression analysis of the PPM1D/Wip1 gene were assessed by qRT-PCR analysis. Mutational analysis of Exon 6 of the PPM1D gene was performed by PCR amplification and Sanger sequencing. Expressions of Wip1 and p53 proteins were assessed by immunohistochemistry and Western blot analysis. CONCLUSIONS We found that PPM1D gained gene copy number in NHL tumors by 0.7-8 times compared to the control (p < 0.01). Increased PPM1D/Wip1 gene copy number was associated with higher mRNA and protein expression in human NHL samples (p < 0.01). Overexpression of Wip1 in NHL tumors and NHL cell lines was associated with amplification level and was unaffected by p53 status. Furthermore, a heterozygous type insertion mutation was detected in exon 6 (c.1553_1554insA) of the PPM1D gene particularly in DLBCL samples. Wip1 may have oncogenic potential, perhaps playing a role in the onset and progression of human NHL. The possible significance of Wip1 overexpression to chemotherapy response in NHL remains an intriguing question that requires more exploration.
Collapse
Affiliation(s)
- Hatice Pilevneli
- Department of Medical Biology, Faculty of Medicine, Aydın Adnan Menderes University, Aytepe Campus, Aydin, Turkey
| | - Firuzan Döger
- Department of Pathology, Faculty of Medicine, Aydın Adnan Menderes University, Aytepe Campus, Aydin, Turkey
| | - Levent Karagenç
- Department of Histology and Embryology, Faculty of Veterinary Sciences, Aydın Adnan Menderes University, Aydin, Turkey
| | - Didem Kozacı
- Department of Medical Biochemistry, Faculty of Medicine, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Mehtap Kilic Eren
- Department of Medical Biology, Faculty of Medicine, Aydın Adnan Menderes University, Aytepe Campus, Aydin, Turkey.
| |
Collapse
|
8
|
Takahashi K, Nakada D, Goodell M. Distinct landscape and clinical implications of therapy-related clonal hematopoiesis. J Clin Invest 2024; 134:e180069. [PMID: 39352380 PMCID: PMC11444158 DOI: 10.1172/jci180069] [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] [Indexed: 10/03/2024] Open
Abstract
Therapy-related clonal hematopoiesis (t-CH) is defined as clonal hematopoiesis detected in individuals previously treated with chemotherapy and/or radiation therapy. With the increased use of genetic analysis in oncological care, the detection of t-CH among cancer patients is becoming increasingly common. t-CH arises through the selective bottleneck imposed by chemotherapies and potentially through direct mutagenesis from chemotherapies, resulting in a distinct mutational landscape enriched with mutations in DNA damage-response pathway genes such as TP53, PPM1D, and CHEK2. Emerging evidence sheds light on the mechanisms of t-CH development and potential strategies to mitigate its emergence. Due to its unique characteristics that predominantly affect cancer patients, t-CH has clinical implications distinct from those of CH in the general population. This Review discusses the potential mechanisms of t-CH development, its mutational landscape, mutant-drug relationships, and its clinical significance. We highlight the distinct nature of t-CH and call for intensified research in this field.
Collapse
Affiliation(s)
- Koichi Takahashi
- Departments of Leukemia and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Margaret Goodell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
9
|
Jiang W, Shaw S, Rush J, Dumont N, Kim J, Singh R, Skepner A, Khodier C, Raffier C, Yan N, Schluter C, Yu X, Szuchnicki M, Sathappa M, Kahn J, Sperling AS, McKinney DC, Gould AE, Garvie CW, Miller PG. Identification of Small Molecule Inhibitors of PPM1D Using a Novel Drug Discovery Platform. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.595001. [PMID: 38826457 PMCID: PMC11142126 DOI: 10.1101/2024.05.20.595001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Protein phosphatase, Mg2+/Mn2+ dependent 1D (PPM1D), is a serine/threonine phosphatase that is recurrently activated in cancer, regulates the DNA damage response (DDR), and suppresses the activation of p53. Consistent with its oncogenic properties, genetic loss or pharmacologic inhibition of PPM1D impairs tumor growth and sensitizes cancer cells to cytotoxic therapies in a wide range of preclinical models. Given the therapeutic potential of targeting PPM1D specifically and the DDR and p53 pathway more generally, we sought to deepen our biological understanding of PPM1D as a drug target and determine how PPM1D inhibition differs from other therapeutic approaches to activate the DDR. We performed a high throughput screen to identify new allosteric inhibitors of PPM1D, then generated and optimized a suite of enzymatic, cell-based, and in vivo pharmacokinetic and pharmacodynamic assays to drive medicinal chemistry efforts and to further interrogate the biology of PPM1D. Importantly, this drug discovery platform can be readily adapted to broadly study the DDR and p53. We identified compounds distinct from previously reported allosteric inhibitors and showed in vivo on-target activity. Our data suggest that the biological effects of inhibiting PPM1D are distinct from inhibitors of the MDM2-p53 interaction and standard cytotoxic chemotherapies. These differences also highlight the potential therapeutic contexts in which targeting PPM1D would be most valuable. Therefore, our studies have identified a series of new PPM1D inhibitors, generated a suite of in vitro and in vivo assays that can be broadly used to interrogate the DDR, and provided important new insights into PPM1D as a drug target.
Collapse
Affiliation(s)
- Wei Jiang
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Subrata Shaw
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Jason Rush
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Nancy Dumont
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - John Kim
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Ritu Singh
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Adam Skepner
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Carol Khodier
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Cerise Raffier
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Ni Yan
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Cameron Schluter
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Xiao Yu
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Mateusz Szuchnicki
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Murugappan Sathappa
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Josephine Kahn
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Adam S. Sperling
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David C. McKinney
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Alexandra E. Gould
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Colin W. Garvie
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for the Development of Therapeutics, Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Peter G. Miller
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
10
|
Martinikova AS, Stoyanov M, Oravetzova A, Kok YP, Yu S, Dobrovolna J, Janscak P, van Vugt M, Macurek L. PPM1D activity promotes the replication stress caused by cyclin E1 overexpression. Mol Oncol 2024; 18:6-20. [PMID: 37067201 PMCID: PMC10766204 DOI: 10.1002/1878-0261.13433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/09/2023] [Accepted: 04/13/2023] [Indexed: 04/18/2023] Open
Abstract
Oncogene-induced replication stress has been recognized as a major cause of genome instability in cancer cells. Increased expression of cyclin E1 caused by amplification of the CCNE1 gene is a common cause of replication stress in various cancers. Protein phosphatase magnesium-dependent 1 delta (PPM1D) is a negative regulator of p53 and has been implicated in termination of the cell cycle checkpoint. Amplification of the PPM1D gene or frameshift mutations in its final exon promote tumorigenesis. Here, we show that PPM1D activity further increases the replication stress caused by overexpression of cyclin E1. In particular, we demonstrate that cells expressing a truncated mutant of PPM1D progress faster from G1 to S phase and fail to complete licensing of the replication origins. In addition, we show that transcription-replication collisions and replication fork slowing caused by CCNE1 overexpression are exaggerated in cells expressing the truncated PPM1D. Finally, replication speed and accumulation of focal DNA copy number alterations caused by induction of CCNE1 expression was rescued by pharmacological inhibition of PPM1D. We propose that increased activity of PPM1D suppresses the checkpoint function of p53 and thus promotes genome instability in cells expressing the CCNE1 oncogene.
Collapse
Affiliation(s)
- Andra S. Martinikova
- Laboratory of Cancer Cell Biology, Institute of Molecular GeneticsCzech Academy of SciencesPragueCzech Republic
| | - Miroslav Stoyanov
- Laboratory of Cancer Cell Biology, Institute of Molecular GeneticsCzech Academy of SciencesPragueCzech Republic
| | - Anna Oravetzova
- Laboratory of Cancer Cell Biology, Institute of Molecular GeneticsCzech Academy of SciencesPragueCzech Republic
| | - Yannick P. Kok
- Department of Medical Oncology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Shibo Yu
- Department of Pathology and Medical Biology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Jana Dobrovolna
- Laboratory of Cancer Cell Biology, Institute of Molecular GeneticsCzech Academy of SciencesPragueCzech Republic
| | - Pavel Janscak
- Laboratory of Cancer Cell Biology, Institute of Molecular GeneticsCzech Academy of SciencesPragueCzech Republic
- Institute of Molecular Cancer ResearchUniversity of ZurichSwitzerland
| | - Marcel van Vugt
- Department of Medical Oncology, University Medical Center GroningenUniversity of GroningenThe Netherlands
| | - Libor Macurek
- Laboratory of Cancer Cell Biology, Institute of Molecular GeneticsCzech Academy of SciencesPragueCzech Republic
| |
Collapse
|
11
|
Burocziova M, Danek P, Oravetzova A, Chalupova Z, Alberich-Jorda M, Macurek L. Ppm1d truncating mutations promote the development of genotoxic stress-induced AML. Leukemia 2023; 37:2209-2220. [PMID: 37709843 PMCID: PMC10624630 DOI: 10.1038/s41375-023-02030-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023]
Abstract
Hematopoietic stem cells (HSCs) ensure blood cell production during the life-time of an organism, and to do so they need to balance self-renewal, proliferation, differentiation, and migration in a steady state as well as in response to stress or injury. Importantly, aberrant proliferation of HSCs leads to hematological malignancies, and thus, tight regulation by various tumor suppressor pathways, including p53, is essential. Protein phosphatase magnesium-dependent 1 delta (PPM1D) is a negative regulator of p53 and promotes cell survival upon induction of genotoxic stress. Truncating mutations in the last exon of PPM1D lead to the production of a stable, enzymatically active protein and are commonly associated with clonal hematopoiesis. Using a transgenic mouse model, we demonstrate that truncated PPM1D reduces self-renewal of HSCs in basal conditions but promotes the development of aggressive AML after exposure to ionizing radiation. Inhibition of PPM1D suppressed the colony growth of leukemic stem and progenitor cells carrying the truncated PPM1D, and remarkably, it provided protection against irradiation-induced cell growth. Altogether, we demonstrate that truncated PPM1D affects HSC maintenance, disrupts normal hematopoiesis, and that its inhibition could be beneficial in the context of therapy-induced AML.
Collapse
Affiliation(s)
- Monika Burocziova
- Department Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic
- Department of Hemato-oncology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic
| | - Petr Danek
- Department of Hemato-oncology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic
| | - Anna Oravetzova
- Department Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic
| | - Zuzana Chalupova
- Department Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic
| | - Meritxell Alberich-Jorda
- Department of Hemato-oncology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic.
- Childhood Leukaemia Investigation Prague, Department of Pediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague, University Hospital Motol, V Uvalu 84, Praha, 150 06, Czech Republic.
| | - Libor Macurek
- Department Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Prague, Czech Republic.
| |
Collapse
|
12
|
Franch-Expósito S, Mehine M, Ptashkin RN, Bolton KL, Bandlamudi C, Srinivasan P, Zhang L, Goodell MA, Gedvilaite E, Menghrajani K, Sánchez-Vela P, Mandelker D, Comen E, Norton L, Benayed R, Gao T, Papaemmanuil E, Taylor B, Levine R, Offit K, Stadler Z, Berger MF, Zehir A. Associations Between Cancer Predisposition Mutations and Clonal Hematopoiesis in Patients With Solid Tumors. JCO Precis Oncol 2023; 7:e2300070. [PMID: 37561983 PMCID: PMC10581611 DOI: 10.1200/po.23.00070] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/31/2023] [Accepted: 07/05/2023] [Indexed: 08/12/2023] Open
Abstract
PURPOSE Clonal hematopoiesis (CH), the expansion of clones in the hematopoietic system, has been linked to different internal and external features such as aging, genetic ancestry, smoking, and oncologic treatment. However, the interplay between mutations in known cancer predisposition genes and CH has not been thoroughly examined in patients with solid tumors. METHODS We used prospective tumor-blood paired sequencing data from 46,906 patients who underwent Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT) testing to interrogate the associations between CH and rare pathogenic or likely pathogenic (P/LP) germline variants. RESULTS We observed an enrichment of CH-positive patients among those carrying P/LP germline mutations and identified a significant association between P/LP germline variants in ATM and CH. Germline and CH comutation patterns in ATM, TP53, and CHEK2 suggested biallelic inactivation as a potential mediator of clonal expansion. Moreover, we observed that CH-PPM1D mutations, similar to somatic tumor-associated PPM1D mutations, were depleted in patients with P/LP germline mutations in the DNA damage response (DDR) genes ATM, CHEK2, and TP53. Patients with solid tumors and harboring P/LP germline mutations, CH mutations, and mosaicism chromosomal alterations might be at an increased risk of developing secondary leukemia while germline variants in TP53 were identified as an independent risk factor (hazard ratio, 36; P < .001) for secondary leukemias. CONCLUSION Our results suggest a close relationship between inherited variants and CH mutations within the DDR genes in patients with solid tumors. Associations identified in this study might translate into enhanced clinical surveillance for CH and associated comorbidities in patients with cancer harboring these germline mutations.
Collapse
Affiliation(s)
- Sebastià Franch-Expósito
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Miika Mehine
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ryan N. Ptashkin
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- C2i Genomics, New York, NY
| | - Kelly L. Bolton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Chaitanya Bandlamudi
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Preethi Srinivasan
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Natera Inc, San Carlos, CA
| | - Linda Zhang
- Department of Molecular and Cellular Biology, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX
| | - Margaret A. Goodell
- Department of Molecular and Cellular Biology, Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX
| | - Erika Gedvilaite
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kamal Menghrajani
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Pablo Sánchez-Vela
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Diana Mandelker
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Elizabeth Comen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Larry Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ryma Benayed
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Precision Medicine and Biosamples, AstraZeneca, New York, NY
| | - Teng Gao
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA
| | - Elli Papaemmanuil
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Barry Taylor
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ross Levine
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kenneth Offit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Zsofia Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael F. Berger
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ahmet Zehir
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Precision Medicine and Biosamples, AstraZeneca, New York, NY
| |
Collapse
|