1
|
Pan X, Huang C, Bai X, Li F. Causal relationship between breast cancer and acute myeloid leukemia based on two-sample bidirectional Mendelian randomization and transcriptome overlap analysis. Discov Oncol 2025; 16:492. [PMID: 40198525 PMCID: PMC11979033 DOI: 10.1007/s12672-025-02288-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 04/01/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Breast cancer is the most prevalent malignancy and the leading cause of cancer-related deaths among women worldwide. Several case reports have shown that some breast cancer patients subsequently develop acute myeloid leukemia (AML) within a short period. However, the causal relationship and pathogenic mechanisms between breast cancer and AML remain incompletely understood. METHODS Mendelian randomization (MR) analyses were conducted to explore the bidirectional causal relationships between breast cancer and AML. Additionally, we applied the Bayesian Weighted Mendelian Randomization (BWMR) approach to validate the results of the MR analysis. Subsequently, we utilized RNA-seq data from various sources to explore the potential molecular signaling pathways between breast cancer and AML. RESULTS Both IVW method and BWMR approach demonstrated that data from three distinct sources consistently indicated breast cancer as a risk factor for AML, with all sources showing statistically significant results (all P < 0.05, Odds Ratios [ORs] > 1). Bioinformatic analyses suggested that extracellular vesicle functions and p53 signaling pathway may mediate molecular links between breast cancer and AML. Using machine learning, we identified 8 genes with high diagnostic efficacy for predicting the occurrence of AML in breast cancer patients. CONCLUSIONS MR analyses indicated a causal relationship between breast cancer and AML. Additionally, transcriptome analysis offered a theoretical basis for understanding the potential mechanisms and therapeutic targets of AML in breast cancer patients.
Collapse
Affiliation(s)
- Xin'an Pan
- Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yongwai Zheng Street, East Lake District, Nanchang City, 330006, Jiangxi Province, China
| | - Cuihan Huang
- The First Clinical Medical College of Nanchang University, Xuefu Road, Nanchang, 330006, Jiangxi, China
| | - Xinyi Bai
- School of Public, Health of Nanchang University, Xuefu Road, Nanchang, 330006, Jiangxi, China
| | - Fei Li
- Department of Hematology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yongwai Zheng Street, East Lake District, Nanchang City, 330006, Jiangxi Province, China.
- Jiangxi Clinical Research Center for Hematologic Disease, Nanchang, China.
| |
Collapse
|
2
|
Wang Y, Cai Y. Risk of secondary myeloid neoplasms following treatment in patients with grade I-II follicular lymphoma: a retrospective cohort study. Discov Oncol 2025; 16:397. [PMID: 40138078 PMCID: PMC11947400 DOI: 10.1007/s12672-025-02149-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND To assess the association between initial treatment modalities and the risk of developing subtypes of myeloid neoplasms (MNs) in survivors with grade I-II follicular lymphoma (FL) and to evaluate their impact on survival outcomes. METHODS Patients diagnosed with grade I-II FL as their first malignancy were identified from the Surveillance, Epidemiology, and End Results (SEER) database. Fine-Gray competing risk regression and Poisson regression were used to evaluate the treatment-associated risk (RR) for MNs and the Kaplan-Meier method was applied to assess the survival outcomes. RESULTS Among 19,326 FL patients, 9539 patients (49.36%) received chemotherapy, and 2890 patients (14.95%) received radiotherapy as part of their initial treatment. With a median follow-up time of 103 months, 90, 82, and 23 patients developed myelodysplastic syndromes (MDS), acute myeloid leukemia (AML), and myeloproliferative neoplasms (MPN), respectively. In both multivariable competing risk regression analysis and Poisson regression analysis, chemotherapy was found to be associated with a higher risk of developing MDS (adjusted hazard ratio (HR), 1.85; 95% confidence interval (CI), 1.13-3.02; adjusted RR, 1.88; 95% CI, 1.18-3.04), total AML (adjusted HR, 2.22; 95% CI, 1.33-3.71; adjusted RR, 2.24; 95% CI, 1.37-3.78), and AML with myelodysplasia-related changes (AML-MRC) (adjusted HR, 3.42; 95% CI, 1.24-9.44; adjusted RR, 3.48; 95% CI, 1.52-9.07). Additionally, radiotherapy also increased the risk of AML-MRC (adjusted HR, 2.74; 95% CI, 1.12-6.72; adjusted RR, 2.73; 95% CI, 1.10-6.08). The development of AML or MDS was associated with worse overall survival (OS) and disease-specific survival (DSS) in grade I-II FL survivors. CONCLUSION Initial chemotherapy and radiotherapy in patients with grade I-II FL were associated with increased risk of certain subtypes of MNs, such as MDS and AML. The importance of balancing risks and benefits should be emphasized in initial FL treatment.
Collapse
Affiliation(s)
- Yuebo Wang
- Research and Foreign Affairs Department, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Yanan Cai
- Department of Hematology, Henan Key Laboratory for Hematology, Henan Provincial People's Hospital; Zhengzhou University People's Hospital, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
3
|
Singhal D, Kutyna MM, Hahn CN, Shah MV, Hiwase DK. Therapy-Related Myeloid Neoplasms: Complex Interactions among Cytotoxic Therapies, Genetic Factors, and Aberrant Microenvironment. Blood Cancer Discov 2024; 5:400-416. [PMID: 39422544 PMCID: PMC11528189 DOI: 10.1158/2643-3230.bcd-24-0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/21/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
Therapy-related myeloid neoplasm (t-MN), characterized by its association with prior exposure to cytotoxic therapy, remains poorly understood and is a major impediment to long-term survival even in the era of novel targeted therapies due to its aggressive nature and treatment resistance. Previously, cytotoxic therapy-induced genomic changes in hematopoietic stem cells were considered sine qua non in pathogenesis; however, recent research demonstrates a complex interaction between acquired and hereditary genetic predispositions, along with a profoundly senescent bone marrow (BM) microenvironment. We review emerging data on t-MN risk factors and explore the intricate interplay among clonal hematopoiesis, genetic predisposition, and the abnormal BM microenvironment. Significance: t-MN represents a poorly understood blood cancer with extremely poor survival and no effective therapies. We provide a comprehensive review of recent preclinical research highlighting complex interaction among emerging therapies, hereditary and acquired genetic factors, and BM microenvironment. Understanding the risk factors associated with t-MN is crucial for clinicians, molecular pathologists, and cancer biologists to anticipate and potentially reduce its incidence in the future. Moreover, better understanding of the molecular pathogenesis of t-MN may enable preemptive screening and even intervention in high-risk patients.
Collapse
Affiliation(s)
- Deepak Singhal
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Monika M. Kutyna
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Christopher N. Hahn
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
- Genetics and Molecular Pathology, SA Pathology, Adelaide, Australia
| | | | - Devendra K. Hiwase
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| |
Collapse
|
4
|
Pourhassan H, Zhang J, Tinajero J, Pullarkat V, Agrawal V, Koller P, Al Malki M, Aribi A, Salhotra A, Sandhu K, Ali H, Stein A, Marcucci G, Forman S, Aldoss I. Therapy-related acute lymphoblastic leukaemia in women with antecedent breast cancer. Br J Haematol 2024; 205:158-165. [PMID: 38556836 DOI: 10.1111/bjh.19432] [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: 02/14/2024] [Revised: 03/14/2024] [Accepted: 03/16/2024] [Indexed: 04/02/2024]
Abstract
Therapy-related acute lymphoblastic leukaemia (tr-ALL) is a disease entity attributed to previous exposure to chemotherapy and/or radiation for antecedent malignancy. There is observed female predominance for tr-ALL, likely due to high prevalence and excellent curable rate for non-metastatic breast cancer as well as the frequent use of carcinogenic agents as part of adjuvant therapy. Here, we reviewed 37 women with diagnosis of ALL following breast cancer treatment with focus on cytogenetic categorization. Philadelphia chromosome positivity (Ph+), KMT2A alterations and other cytogenetic change groups were observed in 32%, 22% and 46% of patients respectively. Median overall survival (OS) and relapse-free survival (RFS) were 19.4 and 12.9 months, overall while both OS and RFS were superior in tr-ALL with Ph+ disease compared to KMT2Ar and other cytogenetics respectively. Seventeen (45.9%) patients underwent consolidative allogeneic haematopoietic cell transplantation (alloHCT) in CR1 out of which 4 (24%) relapsed following transplant. Both OS and RFS were superior in the KMT2Ar cytogenetics group following alloHCT. Ph chromosome represents the largest genetic entity of tr-ALL following breast cancer therapy, and it may be associated with superior survival outcomes while KMT2Ar may be associated with poorer outcomes that can perhaps be mitigated by alloHSCT.
Collapse
Affiliation(s)
- Hoda Pourhassan
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Jianying Zhang
- Division of Biostatistics, Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Duarte, California, USA
| | - Jose Tinajero
- Pharmacy, City of Hope National Medical Center, Duarte, California, USA
| | - Vinod Pullarkat
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Vaibhav Agrawal
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Paul Koller
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Monzr Al Malki
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Ahmed Aribi
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Amandeep Salhotra
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Karamjeet Sandhu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Haris Ali
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Anthony Stein
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Guido Marcucci
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Stephen Forman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| | - Ibrahim Aldoss
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
5
|
Bataller A, Gener-Ricos G, Almanza-Huante E, Chien KS, Urrutia S, Bazinet A, Rodriguez-Sevilla JJ, Hammond D, Sasaki K, Takahashi K, DiNardo CD, Ravandi F, Borthakur G, Kadia TM, Kanagal-Shamanna R, Kantarjian HM, Garcia-Manero G, Montalban-Bravo G. Therapy-related chronic myelomonocytic leukemia does not have the high-risk features of a therapy-related neoplasm. Blood Adv 2024; 8:2695-2706. [PMID: 38513082 PMCID: PMC11170163 DOI: 10.1182/bloodadvances.2024012565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/08/2024] [Accepted: 02/15/2024] [Indexed: 03/23/2024] Open
Abstract
ABSTRACT Therapy-related myeloid neoplasms (t-MNs) arise after exposure to cytotoxic therapies and are associated with high-risk genetic features and poor outcomes. We analyzed a cohort of patients with therapy-related chronic myelomonocytic leukemia (tCMML; n = 71) and compared its features to that of de novo CMML (dnCMML; n = 461). Median time from cytotoxic therapy to tCMML diagnosis was 6.5 years. Compared with dnCMML, chromosome-7 abnormalities (4% vs 13%; P = .005) but not complex karyotype (3% vs 7%; P = .15), were more frequent in tCMML. tCMML was characterized by higher TP53 mutation frequency (4% vs 12%; P = .04) and lower NRAS (6% vs 22%, P = .007) and CBL (4% vs 12%, P = .04) mutation frequency. Prior therapy with antimetabolites (odd ratio [OR], 1.22; 95% confidence interval [CI], 1.05-1.42; P = .01) and mitotic inhibitors (OR, 1.24; 95% CI, 1.06-1.44; P = .009) was associated with NF1 and SETBP1 mutations whereas prior mitotic inhibitor therapy was associated with lower TET2 mutation frequency (OR, 0.71; 95% CI, 0.55-0.92; P = .01). Although no differences in median overall survival (OS) were observed among tCMML and dnCMML (34.7 months vs 35.9 months, P = .26), multivariate analysis for OS revealed that prior chemotherapy was associated with increased risk of death (hazard ratio, 1.76; 95% CI, 1.07-2.89; P = .026). Compared with a cohort of therapy-related myelodysplastic syndrome, tCMML had lower TP53 mutation frequency (12% vs 44.4%, P < .001) and less unfavorable outcomes. In summary, tCMML does not exhibit the high-risk features and poor outcomes of t-MNs.
Collapse
Affiliation(s)
- Alex Bataller
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Georgina Gener-Ricos
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Kelly S. Chien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Samuel Urrutia
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Alexandre Bazinet
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Danielle Hammond
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Courtney D. DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tapan M. Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hagop M. Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | |
Collapse
|
6
|
Cowell IG, Austin CA. Myeloperoxidase inhibition protects bone marrow mononuclear cells from DNA damage induced by the TOP2 poison anti-cancer drug etoposide. FEBS Open Bio 2024; 14:1001-1010. [PMID: 38531625 PMCID: PMC11148113 DOI: 10.1002/2211-5463.13799] [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: 01/15/2024] [Revised: 02/13/2024] [Accepted: 03/20/2024] [Indexed: 03/28/2024] Open
Abstract
Myeloperoxidase (MPO) is found almost exclusively in granulocytes and immature myeloid cells. It plays a key role in the innate immune system, catalysing the formation of reactive oxygen species that are important in anti-microbial action, but MPO also oxidatively transforms the topoisomerase II (TOP2) poison etoposide to chemical forms that have elevated DNA damaging properties. TOP2 poisons such as etoposide are widely used anti-cancer drugs, but they are linked to cases of secondary acute myeloid leukaemias through a mechanism that involves DNA damage and presumably erroneous repair leading to leukaemogenic chromosome translocations. This leads to the possibility that myeloperoxidase inhibitors could reduce the rate of therapy-related leukaemia by protecting haematopoietic cells from TOP2 poison-mediated genotoxic damage while preserving the anti-cancer efficacy of the treatment. We show here that myeloperoxidase inhibition reduces etoposide-induced TOP2B-DNA covalent complexes and resulting DNA double-strand break formation in primary ex vivo expanded CD34+ progenitor cells and unfractionated bone marrow mononuclear cells. Since MPO inhibitors are currently being developed as anti-inflammatory agents this raises the possibility that repurposing of these potential new drugs could provide a means of suppressing secondary acute myeloid leukaemias associated with therapies containing TOP2 poisons.
Collapse
|
7
|
Travaglini S, Marinoni M, Visconte V, Guarnera L. Therapy-Related Myeloid Neoplasm: Biology and Mechanistic Aspects of Malignant Progression. Biomedicines 2024; 12:1054. [PMID: 38791019 PMCID: PMC11118122 DOI: 10.3390/biomedicines12051054] [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: 04/01/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Therapy-related myeloid neoplasms (t-MN) arise after a documented history of chemo/radiotherapy as treatment for an unrelated condition and account for 10-20% of myelodysplastic syndromes and acute myeloid leukemia. T-MN are characterized by a specific genetic signature, aggressive features and dismal prognosis. The nomenclature and the subsets of these conditions have changed frequently over time, and despite the fact that, in the last classification, they lost their autonomous entity status and became disease qualifiers, the recognition of this feature remains of major importance. Furthermore, in recent years, extensive studies focusing on clonal hematopoiesis and germline variants shed light on the mechanisms of positive pressure underpinning the rise of driver gene mutations in t-MN. In this manuscript, we aim to review the evolution of defining criteria and characteristics of t-MN from a clinical and biological perspective, the advances in mechanistic aspects of malignant progression and the challenges in prevention and management.
Collapse
Affiliation(s)
- Serena Travaglini
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Massimiliano Marinoni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Valeria Visconte
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Luca Guarnera
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
8
|
Murt A, Bayram B, Yılmaz U, Seyahi N, Eşkazan AE. Chronic Myeloid Leukemia in Renal Transplantation Patients in the Era of Tyrosine Kinase Inhibitors: A Case Report and Review of the Literature. Nephron Clin Pract 2024; 148:563-568. [PMID: 38574488 DOI: 10.1159/000538532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 03/16/2024] [Indexed: 04/06/2024] Open
Abstract
Lifelong immunosuppression, cytotoxic effects of some immunosuppressive drugs, and opportunistic oncogenic viruses increase malignancy risks in solid organ recipients. The risk of myeloid neoplasms including chronic myeloid leukemia (CML) is also increased in this patient population. Tyrosine kinase inhibitors (TKIs), the key element of CML therapy, should be used cautiously in transplantation patients as they may interact with calcineurin inhibitors. With this report, a 63-year-old female kidney transplant recipient who developed CML 9 years after kidney transplantation is presented. CML in this patient was treated with a slightly reduced dose of imatinib (300 mg) due to concerns of adverse events including its interaction with tacrolimus. Deep molecular response (DMR) was achieved at 12 months under imatinib treatment. The patient is still in DMR after 30 months of follow-up, and she did not experience any adverse events or acute rejection episodes. CML and the use of TKIs in kidney transplant patients have been discussed with an extensive literature review. In this patient population, TKIs are generally well tolerated with achievement of treatment responses and good prognosis. Graft functions are also well maintained as long as drug interactions are monitored.
Collapse
Affiliation(s)
- Ahmet Murt
- Division of Nephrology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Batuhan Bayram
- Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Umut Yılmaz
- Division of Hematology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Nurhan Seyahi
- Division of Nephrology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Ahmet Emre Eşkazan
- Division of Hematology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
9
|
Fabiani E, Cristiano A, Hajrullaj H, Falconi G, Leone G, Voso M. Therapy-Related Myeloid Neoplasms: Predisposition and Clonal Evolution. Mediterr J Hematol Infect Dis 2023; 15:e2023064. [PMID: 38028397 PMCID: PMC10631709 DOI: 10.4084/mjhid.2023.064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Therapy-related Myeloid Neoplasm (t-MN) represents one of the worst long-term consequences of cytotoxic therapy for primary tumors and autoimmune disease. Poor survival and refractoriness to current treatment strategies characterize affected patients from a clinical point of view. In our aging societies, where newer therapies and ameliorated cancer management protocols are improving the life expectancy of cancer patients, therapy-related Myeloid Neoplasms are an emerging problem. Although several research groups have contributed to characterizing the main risk factors in t-MN development, the multiplicity of primary tumors, in association with the different therapeutic strategies available and the new drugs in development, make interpreting the current data still complex. The main risk factors involved in t-MN pathogenesis can be subgrouped into patient-specific, inherited, and acquired predispositions. Although t-MN can occur at any age, the risk tends to increase with advancing age, and older patients, characterized by a higher number of comorbidities, are more likely to develop the disease. Thanks to the availability of deep sequencing techniques, germline variants have been reported in 15-20% of t-MN patients, highlighting their role in cancer predisposition. It is becoming increasingly evident that t-MN with driver gene mutations may arise in the background of Clonal Hematopoiesis of Indeterminate Potential (CHIP) under the positive selective pressure of chemo and/or radiation therapies. Although CHIP is generally considered benign, it has been associated with an increased risk of t-MN. In this context, the phenomenon of clonal evolution may be described as a dynamic process of expansion of preexisting clones, with or without acquisition of additional genetic alterations, that, by favoring the proliferation of more aggressive and/or resistant clones, may play a crucial role in the progression from preleukemic states to t-MN.
Collapse
Affiliation(s)
- Emiliano Fabiani
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
- UniCamillus-Saint Camillus International University of Health Sciences, Rome, Italy
| | - A. Cristiano
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - H. Hajrullaj
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - G. Falconi
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| | - G. Leone
- Università Cattolica del Sacro Cuore, Roma, Italy
| | - M.T. Voso
- Department of Biomedicine and Prevention, University of Tor Vergata, Rome, Italy
| |
Collapse
|
10
|
Lee JW, Oh H, You JY, Lee ES, Lee JH, Song SE, Lee NK, Jung SP, An JS, Cho KR, Kim CY, Park KH. Therapy-related myeloid neoplasm in early breast cancer patients treated with adjuvant chemotherapy. Eur J Cancer 2023; 191:112952. [PMID: 37473463 DOI: 10.1016/j.ejca.2023.112952] [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: 01/24/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Long-term complications are becoming more important as the survival rate of breast cancer improves. Treatment-related myeloid neoplasm is an important long-term complication in breast cancer survivors as it has a poor prognosis. OBJECTIVE We evaluated the incidence and risk factors for the development of treatment-related acute myeloid leukaemia (AML)/myelodysplastic syndrome (MDS) in patients treated with early breast cancer. METHODS We accessed the national Korean database to identify 153,565 patients diagnosed with breast cancer between January 2007 and October 2016 who underwent surgery for breast cancer. We estimated the cumulative incidence of AML/MDS and analysed the risk factors for developing AML/MDS. RESULTS Of 153,575 patients, 79,321 received anthracycline-based adjuvant therapy, 14,317 received adjuvant therapy without anthracyclines and 46,657 did not receive adjuvant chemotherapy. Overall, 120 developed AML (105 in the anthracycline group, 9 in the non-anthracycline group and 6 in the control group), and 128 developed MDS (96, 9 and 23 in each group). The 10-year cumulative incidence of AML/MDS was the highest in the anthracycline group (0.221% and 0.199%), followed by the non-anthracycline group (0.122% and 0.163%) and the control group (0.024% and 0.089%). The risk of developing AML/MDS was significantly higher in patients treated with anthracyclines (hazard ratio [HR] 9.531; p < 0.0001 for AML and HR 2.559; p < 0.0001 for MDS) compared to patients in the control group. CONCLUSION This study found that anthracycline-based adjuvant therapy significantly increased the risk of AML/MDS in Korean breast cancer patients, with the risk persisting for at least 10 years. While the cumulative incidence was low, the long-term risks of AML/MDS should be taken into account considering the poor outcomes associated with these neoplasms.
Collapse
MESH Headings
- Humans
- Female
- Breast Neoplasms/complications
- Leukemia, Myeloid, Acute/chemically induced
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/epidemiology
- Myelodysplastic Syndromes/chemically induced
- Myelodysplastic Syndromes/epidemiology
- Chemotherapy, Adjuvant/adverse effects
- Combined Modality Therapy
- Anthracyclines
- Neoplasms, Second Primary/chemically induced
- Neoplasms, Second Primary/epidemiology
- Neoplasms, Second Primary/drug therapy
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
Collapse
Affiliation(s)
- Ji Won Lee
- Division of Medical Oncology, Department of Internal Medicine, Korea University Anam Hospital, Seoul, South Korea
| | - Hoonji Oh
- Department of Biostatistics, Korea University College of Medicine, Seoul, South Korea
| | - Ji Young You
- Division of Breast and Endocrine, Department of Surgery, Departments of Breast Surgery, Korea University Anam Hospital, Seoul, South Korea
| | - Eun-Shin Lee
- Division of Breast and Endocrine, Department of Surgery, Departments of Breast Surgery, Korea University Anam Hospital, Seoul, South Korea
| | - Jung Hyun Lee
- Department of Pathology, Korea University Anam Hospital, Seoul, South Korea
| | - Sung Eun Song
- Department of Radiology, Korea University Anam Hospital, Seoul, South Korea
| | - Nam Kwon Lee
- Department of Radiation Oncology, Korea University Anam Hospital, Seoul, South Korea
| | - Seung Pil Jung
- Division of Breast and Endocrine, Department of Surgery, Departments of Breast Surgery, Korea University Anam Hospital, Seoul, South Korea
| | - Jung Seok An
- Department of Pathology, Korea University Anam Hospital, Seoul, South Korea
| | - Kyu Ran Cho
- Department of Radiology, Korea University Anam Hospital, Seoul, South Korea
| | - Cheol Yong Kim
- Department of Radiation Oncology, Korea University Anam Hospital, Seoul, South Korea
| | - Kyong Hwa Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University Anam Hospital, Seoul, South Korea.
| |
Collapse
|
11
|
Testa U, Castelli G, Pelosi E. TP53-Mutated Myelodysplasia and Acute Myeloid Leukemia. Mediterr J Hematol Infect Dis 2023; 15:e2023038. [PMID: 37435040 PMCID: PMC10332352 DOI: 10.4084/mjhid.2023.038] [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: 05/08/2023] [Accepted: 06/01/2023] [Indexed: 07/13/2023] Open
Abstract
TP53-mutated myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) form a distinct and heterogeneous group of myeloid malignancies associated with poor outcomes. Studies carried out in the last years have in part elucidated the complex role played by TP53 mutations in the pathogenesis of these myeloid disorders and in the mechanisms of drug resistance. A consistent number of studies has shown that some molecular parameters, such as the presence of a single or multiple TP53 mutations, the presence of concomitant TP53 deletions, the association with co-occurring mutations, the clonal size of TP53 mutations, the involvement of a single (monoallelic) or of both TP53 alleles (biallelic) and the cytogenetic architecture of concomitant chromosome abnormalities are major determinants of outcomes of patients. The limited response of these patients to standard treatments, including induction chemotherapy, hypomethylating agents and venetoclax-based therapies and the discovery of an immune dysregulation have induced a shift to new emerging therapies, some of which being associated with promising efficacy. The main aim of these novel immune and nonimmune strategies consists in improving survival and in increasing the number of TP53-mutated MDS/AML patients in remission amenable to allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Rome Italy
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Rome Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Rome Italy
| |
Collapse
|
12
|
Morton LM, Curtis RE, Linet MS, Schonfeld SJ, Advani PG, Dalal NH, Sasse EC, Dores GM. Trends in risk for therapy-related myelodysplastic syndrome/acute myeloid leukemia after initial chemo/immunotherapy for common and rare lymphoid neoplasms, 2000-2018. EClinicalMedicine 2023; 61:102060. [PMID: 37457112 PMCID: PMC10344829 DOI: 10.1016/j.eclinm.2023.102060] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Background Historically, survivors of common lymphoid neoplasms (LNs) had increased risks for therapy-related myelodysplastic syndrome/acute myeloid leukemia (tMDS/AML). Despite major treatment advances in the treatment of LNs over the last two decades, a comprehensive evaluation of tMDS/AML trends following both common and rare LNs treated in this contemporary period is lacking. Methods In US cancer registries during 2000-2018, we identified 1496 tMDS/AML cases among 186,503 adults who were treated with initial chemo/immunotherapy for first primary LN and survived ≥1 year. We quantified tMDS/AML standardized incidence ratios (SIRs), excess absolute risks (EARs, per 10,000 person-years), and cumulative incidence. Findings The highest tMDS/AML risks occurred after precursor leukemia/lymphoma (SIR = 39, EAR = 30), Burkitt leukemia/lymphoma (SIR = 20, EAR = 24), peripheral T-cell lymphoma (SIR = 12, EAR = 23), chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL; SIR = 9.0, EAR = 27), and mantle cell lymphoma (SIR = 8.5, EAR = 25). Elevated risks (SIRs = 4.2-6.9, EARs = 4.9-15) also were observed after all other LN subtypes except hairy cell leukemia and mycosis fungoides/Sézary syndrome. Among patients treated more recently, tMDS/AML risks were significantly higher after CLL/SLL (SIR2000-2005 = 4.8, SIR2012-2017 = 10, Ptrend = 0.0043), significantly lower after Hodgkin (SIR2000-2005 = 15, SIR2012-2017 = 6.3, Ptrend = 0.024) and marginal zone (SIR2000-2005 = 7.5, SIR2012-2017 = 2.3, Ptrend = 0.015) lymphomas, and non-significantly lower after mantle cell lymphoma (SIR2000-2005 = 10, SIR2012-2017 = 3.2, Ptrend = 0.054), lymphoplasmacytic lymphoma/Waldenström macroglobulinemia (SIR2000-2005 = 6.9, SIR2012-2017 = 1.0, Ptrend = 0.067), and plasma cell neoplasms (SIR2000-2005 = 5.4, SIR2012-2017 = 3.1, Ptrend = 0.051). EAR and cumulative incidence trends generally were similar to SIR trends. Median survival after tMDS/AML was 8.0 months (interquartile range, 3.0-22.0). Interpretation Although tMDS/AML risks are significantly elevated after initial chemo/immunotherapy for most LNs, patients treated more recently have lower tMDS/AML risks, except after CLL/SLL. Though rare, the poor prognosis following tMDS/AML emphasizes the importance of continued efforts to reduce treatment-associated toxicity. Funding This research was supported in part by the Intramural Research Program of the National Cancer Institute, National Institutes of Health. LMM, GMD, REC, and CBS verified the data, and all authors had access to the data and made the decision to submit for publication.
Collapse
Affiliation(s)
- Lindsay M. Morton
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Rochelle E. Curtis
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Martha S. Linet
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Sara J. Schonfeld
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Pragati G. Advani
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Nicole H. Dalal
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
- Department of Internal Medicine, University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Elizabeth C. Sasse
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Graça M. Dores
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| |
Collapse
|
13
|
Jahn J, Diamond B, Hsu J, Montoya S, Totiger TM, Landgren O, Maura F, Taylor J. Therapy-selected clonal hematopoiesis and its role in myeloid neoplasms. Leuk Res 2023; 126:107020. [PMID: 36696829 PMCID: PMC11305114 DOI: 10.1016/j.leukres.2023.107020] [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: 01/06/2023] [Accepted: 01/19/2023] [Indexed: 01/21/2023]
Abstract
Therapy-related myeloid neoplasms (t-MN) account for approximately 10-15% of all myeloid neoplasms and are associated with poor prognosis. Genomic characterization of t-MN to date has been limited in comparison to the considerable sequencing efforts performed for de novo myeloid neoplasms. Until recently, targeted deep sequencing (TDS) or whole exome sequencing (WES) have been the primary technologies utilized and thus limited the ability to explore the landscape of structural variants and mutational signatures. In the past decade, population-level studies have identified clonal hematopoiesis as a risk factor for the development of myeloid neoplasms. However, emerging research on clonal hematopoiesis as a risk factor for developing t-MN is evolving, and much is unknown about the progression of CH to t-MN. In this work, we will review the current knowledge of the genomic landscape of t-MN, discuss background knowledge of clonal hematopoiesis gained from studies of de novo myeloid neoplasms, and examine the recent literature studying the role of therapeutic selection of CH and its evolution under the effects of antineoplastic therapy. Finally, we will discuss the potential implications on current clinical practice and the areas of focus needed for future research into therapy-selected clonal hematopoiesis in myeloid neoplasms.
Collapse
Affiliation(s)
- Jacob Jahn
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States
| | - Benjamin Diamond
- Myeloma Division, Department of Medicine, University of Miami Miller School of Medicine, United States
| | - Jeffrey Hsu
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States
| | - Skye Montoya
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States
| | - Tulasigeri M Totiger
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States
| | - Ola Landgren
- Myeloma Division, Department of Medicine, University of Miami Miller School of Medicine, United States
| | - Francesco Maura
- Myeloma Division, Department of Medicine, University of Miami Miller School of Medicine, United States
| | - Justin Taylor
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States; Leukemia Program, Department of Medicine, University of Miami Miller School of Medicine, United States.
| |
Collapse
|
14
|
Shih AJ, Jun T, Skol AD, Bao R, Huang L, Vora S, McNerney ME, Hungate EA, Le Beau MM, Larson RA, Elliott A, Lu HM, Huether R, Hernandez F, Stölzel F, Allan JM, Onel K. Inherited cancer predisposing mutations in patients with therapy-related myeloid neoplasms. Br J Haematol 2023; 200:489-493. [PMID: 36349721 PMCID: PMC11246702 DOI: 10.1111/bjh.18543] [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: 09/16/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022]
Abstract
Some patients with therapy-related myeloid neoplasms (t-MN) may have unsuspected inherited cancer predisposition syndrome (CPS). We propose a set of clinical criteria to identify t-MN patients with high risk of CPS (HR-CPS). Among 225 t-MN patients with an antecedent non-myeloid malignancy, our clinical criteria identified 52 (23%) HR-CPS patients. Germline whole-exome sequencing identified pathogenic or likely pathogenic variants in 10 of 27 HR-CPS patients compared to 0 of 9 low-risk CPS patients (37% vs. 0%, p = 0.04). These simple clinical criteria identify t-MN patients most likely to benefit from genetic testing for inherited CPS.
Collapse
Affiliation(s)
- Andrew J Shih
- The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030
| | | | - Andrew D Skol
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637
| | - Riyue Bao
- Department of Medicine, The University of Pittsburgh
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15232
| | - Lei Huang
- Center for Research Informatics, The University of Chicago, Chicago, IL 60637
| | - Sapana Vora
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637
| | - Megan E McNerney
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637
- Department of Pathology, The University of Chicago, Chicago, IL 60637
| | - Eric A Hungate
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637
| | | | - Richard A Larson
- Department of Medicine, The University of Chicago, Chicago, IL 60637
| | | | | | | | | | - Friedrich Stölzel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus Dresden, Dresden University of Technology, Dresden, Germany 01307
| | - James M Allan
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4AD, UK
| | - Kenan Onel
- Sema4, Stamford, CT 06902
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
15
|
Tian H, Yang L, Hou W, Wu Y, Dai Y, Yu J, Liu D. Case report: Identification of acute promyelocytic leukemia during osimertinib resistance followed by granulocyte colony-stimulating factor and pembrolizumab. Front Oncol 2023; 12:1032225. [PMID: 36713543 PMCID: PMC9880289 DOI: 10.3389/fonc.2022.1032225] [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: 08/30/2022] [Accepted: 12/21/2022] [Indexed: 01/15/2023] Open
Abstract
Background The occurrence of acute promyelocytic leukemia (APL) during the management of lung cancer is rare and life-threatening. It was mainly reported to be secondary to chemoradiotherapy. A few studies reported an increased incidence of therapy-related acute promyelocytic leukemia (t-APL) after gefitinib became available. Case presentation We reported a patient who developed thrombocytopenia after receiving oral osimertinib in combination with intensity-modulated radiotherapy (IMRT). For half a year, she had an unrecoverable low platelet count, which progressed to concomitant leukopenia and the transient appearance of orthochromatic normoblasts in the peripheral blood test, indicating a dormant myeloid disorder. Due to simultaneous resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI), pembrolizumab and granulocyte colony-stimulating factor (G-CSF) were administered, revealing prominent signs of hematological malignancy in a peripheral blood test that was later identified as t-APL. Conclusion In general, patients undergoing EGFR-TKI combined with local radiotherapy should be concerned about their hematological assessment. If patients exhibit unrecoverable abnormalities in routine blood tests, a secondary nonsolid malignancy other than myelosuppression should be considered, and further lung cancer treatment should be discontinued.
Collapse
Affiliation(s)
- Huohuan Tian
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Linhui Yang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wang Hou
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu Wu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Dai
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiang Yu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dan Liu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Dan Liu,
| |
Collapse
|
16
|
Testa U, Castelli G, Pelosi E. Clonal Hematopoiesis: Role in Hematologic and Non-Hematologic Malignancies. Mediterr J Hematol Infect Dis 2022; 14:e2022069. [PMID: 36119457 PMCID: PMC9448266 DOI: 10.4084/mjhid.2022.069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/18/2022] [Indexed: 02/08/2023] Open
Abstract
Hematopoietic stem cells (HSCs) ensure the coordinated and balanced production of all hematopoietic cell types throughout life. Aging is associated with a gradual decline of the self-renewal and regenerative potential of HSCs and with the development of clonal hematopoiesis. Clonal hematopoiesis of indeterminate potential (CHIP) defines the clonal expansion of genetically variant hematopoietic cells bearing one or more gene mutations and/or structural variants (such as copy number alterations). CHIP increases exponentially with age and is associated with cancers, including hematologic neoplasia, cardiovascular and other diseases. The presence of CHIP consistently increases the risk of hematologic malignancy, particularly in individuals who have CHIP in association with peripheral blood cytopenia.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
17
|
Pommier Y, Nussenzweig A, Takeda S, Austin C. Human topoisomerases and their roles in genome stability and organization. Nat Rev Mol Cell Biol 2022; 23:407-427. [PMID: 35228717 PMCID: PMC8883456 DOI: 10.1038/s41580-022-00452-3] [Citation(s) in RCA: 230] [Impact Index Per Article: 76.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 12/15/2022]
Abstract
Human topoisomerases comprise a family of six enzymes: two type IB (TOP1 and mitochondrial TOP1 (TOP1MT), two type IIA (TOP2A and TOP2B) and two type IA (TOP3A and TOP3B) topoisomerases. In this Review, we discuss their biochemistry and their roles in transcription, DNA replication and chromatin remodelling, and highlight the recent progress made in understanding TOP3A and TOP3B. Because of recent advances in elucidating the high-order organization of the genome through chromatin loops and topologically associating domains (TADs), we integrate the functions of topoisomerases with genome organization. We also discuss the physiological and pathological formation of irreversible topoisomerase cleavage complexes (TOPccs) as they generate topoisomerase DNA–protein crosslinks (TOP-DPCs) coupled with DNA breaks. We discuss the expanding number of redundant pathways that repair TOP-DPCs, and the defects in those pathways, which are increasingly recognized as source of genomic damage leading to neurological diseases and cancer. Topoisomerases have essential roles in transcription, DNA replication, chromatin remodelling and, as recently revealed, 3D genome organization. However, topoisomerases also generate DNA–protein crosslinks coupled with DNA breaks, which are increasingly recognized as a source of disease-causing genomic damage.
Collapse
|
18
|
Diagnosis and Treatment of Therapy-related Acute Myeloid Leukemia. Crit Rev Oncol Hematol 2022; 171:103607. [DOI: 10.1016/j.critrevonc.2022.103607] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 12/01/2021] [Accepted: 01/26/2022] [Indexed: 11/21/2022] Open
|
19
|
Li G, Holly T, Kelly DR, Reddy V, Mikhail FM, Carroll AJ, Kutny MA. Therapy-related Myeloid Neoplasms in Children: A Single-institute Study. J Pediatr Hematol Oncol 2022; 44:e109-e113. [PMID: 33625084 DOI: 10.1097/mph.0000000000002097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/10/2021] [Indexed: 11/26/2022]
Abstract
Therapy-related myeloid neoplasm (t-MN) in the pediatric population is not well characterized. We studied 12 pediatric patients diagnosed with t-MN in our institution since 2006. The median age at the t-MN diagnoses was 14.8 years (range, 9 to 20 y). The primary malignancies included 9 solid tumors and 3 hematopoietic malignancies. Rhabdomyosarcoma (n=4) was the most common primary malignancy. Five of the 9 patients with solid tumors and all 3 patients with hematopoietic malignancies had primary neoplasms involving bone marrow. The median latency period was 5.2 years (range, 1.8 to 13.8 y). Thrombocytopenia was present in all patients at the t-MN diagnoses. Complete or partial monosomy of chromosome 5 or 7 were the 2 most common cytogenetic abnormalities. A quarter of patients demonstrated a genetic predisposition to t-MN: 1 with Li-Fraumeni syndrome with a germline TP53 R248Q mutation, 1 with Noonan syndrome with a somatic mutation (PTPN11 S502T), and 1 with a constitutive chromosomal translocation [t(X;9)(p22;q34)] and a germline TP53 L130V mutation. Outcomes remain poor. Two patients survived 3 and 5.1 years after hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Geling Li
- Department of Pathology and Laboratory Medicine, Children's of Alabama
- Departments of Pathology
| | - Taylor Holly
- Department of Pediatrics, Division of Hematology and Oncology
| | - David R Kelly
- Department of Pathology and Laboratory Medicine, Children's of Alabama
- Departments of Pathology
| | | | - Fady M Mikhail
- Genetics, University of Alabama at Birmingham, Birmingham, AL
| | | | - Matthew A Kutny
- Department of Pediatrics, Division of Hematology and Oncology
| |
Collapse
|
20
|
Emerging trends of therapy related myeloid neoplasms following modern cancer therapeutics in the United States. Sci Rep 2021; 11:23284. [PMID: 34857802 PMCID: PMC8639740 DOI: 10.1038/s41598-021-02497-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022] Open
Abstract
Clonal hematopoiesis (CH) is a risk factor for the development of therapy-related myelodysplastic syndromes (tMDS) and acute myeloid leukemia (tAML). Adoption of targeted-immunotherapeutics since 2011, may alter the risk of CH progression to tMDS/AML. To study this, we evaluated risk of tMDS and tAML in 667 588 ≥ 1-year survivors of non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC), melanoma and multiple-myeloma (MM) diagnosed during: 2000–2005, 2006–2010 and 2011–2016. The risk of tMDS increased significantly after NSCLC across all time periods (Ptrend = 0.002) while tAML risk decreased from 2006–2010 to 2011–2016, coinciding with increasing use of non-chemotherapeutic agents. tAML risk after RCC decreased (Ptrend = 0.007) whereas tMDS risk did not significantly change over time. After melanoma, tMDS and tAML risks were similar to the general population. tMDS and tAML risk after MM increased from the first to second time-period, however, only risk of tMDS decreased during last period. We report diverging trends in the risk of tAML and tMDS after adoption of modern cancer therapies for specific cancers. It is imperative to further explore impact of contemporary treatment strategies on clonal evolution. Modern treatments via their discrete mechanism of actions on pre-existing CH may alter the risk of subsequent tMDS and tAML.
Collapse
|
21
|
Hawkins CJ, Miles MA. Mutagenic Consequences of Sublethal Cell Death Signaling. Int J Mol Sci 2021; 22:ijms22116144. [PMID: 34200309 PMCID: PMC8201051 DOI: 10.3390/ijms22116144] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/03/2021] [Accepted: 06/05/2021] [Indexed: 02/06/2023] Open
Abstract
Many human cancers exhibit defects in key DNA damage response elements that can render tumors insensitive to the cell death-promoting properties of DNA-damaging therapies. Using agents that directly induce apoptosis by targeting apoptotic components, rather than relying on DNA damage to indirectly stimulate apoptosis of cancer cells, may overcome classical blocks exploited by cancer cells to evade apoptotic cell death. However, there is increasing evidence that cells surviving sublethal exposure to classical apoptotic signaling may recover with newly acquired genomic changes which may have oncogenic potential, and so could theoretically spur the development of subsequent cancers in cured patients. Encouragingly, cells surviving sublethal necroptotic signaling did not acquire mutations, suggesting that necroptosis-inducing anti-cancer drugs may be less likely to trigger therapy-related cancers. We are yet to develop effective direct inducers of other cell death pathways, and as such, data regarding the consequences of cells surviving sublethal stimulation of those pathways are still emerging. This review details the currently known mutagenic consequences of cells surviving different cell death signaling pathways, with implications for potential oncogenic transformation. Understanding the mechanisms of mutagenesis associated (or not) with various cell death pathways will guide us in the development of future therapeutics to minimize therapy-related side effects associated with DNA damage.
Collapse
Affiliation(s)
- Christine J. Hawkins
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia;
| | - Mark A. Miles
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia;
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
- Correspondence:
| |
Collapse
|
22
|
Wang H, Yin Y, Wang R, Huang J, Xue H, Cheng Y, Zhang L, Chen C. Clinicopathological features, risk and survival in lung cancer survivors with therapy-related acute myeloid leukaemia. BMC Cancer 2020; 20:1081. [PMID: 33172389 PMCID: PMC7654570 DOI: 10.1186/s12885-020-07603-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/30/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A secondary malignancy is the most serious complication in lung cancer (LC) survivors. This study aimed to evaluate the clinicopathological features, predictable risk factors and survival of patients with LC who developed therapy-related acute myeloid leukaemia (t-AML). METHODS Patients from the Surveillance, Epidemiology, and End Results (SEER) database diagnosed with t-AML after LC between 1975 and 2015 were included. Standardized incidence ratios (SIRs) were used to perform multiple primary analyses. The risk of t-AML development among LC patients was assessed using a logistic regression model. Kaplan-Meier analysis was used to construct overall survival (OS) curves. Cox regression was used to assess the influence of various prognostic factors. RESULTS A total of 104 patients with t-AML after LC-targeting chemotherapy were included. The median latency period to the development of t-AML was 35.5 months. The calculated SIR of t-AML was 4.00. Chemoradiotherapy, small cell lung cancer (SCLC), or localized/regional-stage LC was a risk factor for the development of t-AML. The median OS was only 1 month, and those younger than 65 years were predicted to have a better OS time. CONCLUSIONS t-AML is a rare but serious late complication in LC patients and is associated with a poor prognosis. It is necessary to carry out long-term follow-up and screen for t-AML in LC patients, especially among those undergoing both radiotherapy and chemotherapy, with SCLC or with localized/regional-stage LC.
Collapse
Affiliation(s)
- Huabin Wang
- Pediatric Blood Center, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Guangming, Shenzhen, Guangdong, 518107, P.R. China
| | - Yin Yin
- Department of Thoracic Surgery, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Ru Wang
- Department of Radiation Oncology, Shandong Provincial ENT Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Junbin Huang
- Pediatric Blood Center, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Guangming, Shenzhen, Guangdong, 518107, P.R. China
| | - Hongman Xue
- Pediatric Blood Center, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Guangming, Shenzhen, Guangdong, 518107, P.R. China
| | - Yucai Cheng
- Pediatric Blood Center, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Guangming, Shenzhen, Guangdong, 518107, P.R. China
| | - Lidan Zhang
- Pediatric Blood Center, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Guangming, Shenzhen, Guangdong, 518107, P.R. China
| | - Chun Chen
- Pediatric Blood Center, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Guangming, Shenzhen, Guangdong, 518107, P.R. China.
| |
Collapse
|
23
|
Tiruneh T, Enawgaw B, Shiferaw E. Genetic Pathway in the Pathogenesis of Therapy-Related Myeloid Neoplasms: A Literature Review. Oncol Ther 2020; 8:45-57. [PMID: 32700075 PMCID: PMC7360004 DOI: 10.1007/s40487-020-00111-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
Therapy-related myeloid neoplasms are a life-threatening and often fatal complication, associated with poor prognosis outcomes and with high-risk unfavorable cytogenetic abnormalities including complex karyotype. They occur after the treatment of primary malignancies using chemotherapy and/or radiation therapy. Such therapy is not specific to cancer cells, and also damages the deoxyribonucleic acid (DNA) of normal cells, resulting in unbalanced and balanced translocations. There are eight genetic pathways, whose details are summarized in this review, depending on the cytogenetic abnormalities induced. This abnormality is the major contributor to the development of therapy-related myeloid neoplasms. The etiology of these neoplasms depends on the complex interaction between the nature and dose of the cytotoxic agent, the environment, and the presence of subsequent inherited mutations. This review aims to elaborate upon recent knowledge regarding the etiology, pathogenesis, and genetic pathways of therapy-related myeloid neoplasms. A deeper understanding of their etiology would aid physicians in more careful monitoring of patients during or after cytotoxic therapy for hematological malignancy. Ultimately, this knowledge could influence initial treatment strategies, with the aim of reducing both the incidence and serious complications of neoplasms. Therefore, early detection of DNA lesions is vital. The authors recommend that primary malignancy be treated with targeted therapy.
Collapse
Affiliation(s)
- Tegenaw Tiruneh
- Department Hematology and Immunohematology, College of Medicine and Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia. .,School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia.
| | - Bamlaku Enawgaw
- School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Elias Shiferaw
- School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
24
|
Linet MS, Gilbert ES, Vermeulen R, Dores GM, Yin SN, Portengen L, Hayes RB, Ji BT, Lan Q, Li GL, Rothman N. Benzene Exposure Response and Risk of Myeloid Neoplasms in Chinese Workers: A Multicenter Case-Cohort Study. J Natl Cancer Inst 2020; 111:465-474. [PMID: 30520970 DOI: 10.1093/jnci/djy143] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/21/2018] [Accepted: 07/17/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND There is international consensus that benzene exposure is causally related to acute myeloid leukemia (AML), and more recent evidence of association with myelodysplastic syndromes (MDS). However, there are uncertainties about the exposure response, particularly risks by time since exposure and age at exposure. METHODS In a case-cohort study in 110 631 Chinese workers followed up during 1972-1999 we evaluated combined MDS/AML (n = 44) and chronic myeloid leukemia (n = 18). We estimated benzene exposures using hierarchical modeling of occupational factors calibrated with historical routine measurements, and evaluated exposure response for cumulative exposure and average intensity using Cox regression; P values were two-sided. RESULTS Increased MDS/AML risk with increasing cumulative exposure in our a priori defined time window (2 to <10 years) before the time at risk was suggested (Ptrend = 08). For first exposure (within the 2 to <10-year window) before age 30 years, the exposure response was stronger (P = .004) with rate ratios of 1.12 (95% confidence interval [CI] = 0.27 to 4.29), 5.58 (95% CI = 1.65 to 19.68), and 4.50 (95% CI = 1.22 to 16.68) for cumulative exposures of more than 0 to less than 40, 40 to less than 100, and at least 100 ppm-years, respectively, compared with no exposure. There was little evidence of exposure response after at least 10 years (Ptrend = .94), regardless of age at first exposure. Average intensity results were generally similar. The risk for chronic myeloid leukemia was increased in exposed vs unexposed workers, but appeared to increase and then decrease with increasing exposure. CONCLUSION For myeloid neoplasms, the strongest effects were apparent for MDS/AML arising within 10 years of benzene exposure and for first exposure in the 2 to less than 10-year window before age 30 years.
Collapse
Affiliation(s)
- Martha S Linet
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD
| | - Ethel S Gilbert
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD
| | - Roel Vermeulen
- Division of Environmental Epidemiology, Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Graça M Dores
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD
| | - Song-Nian Yin
- National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, Peoples Republic of China
| | - Lutzen Portengen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands
| | - Richard B Hayes
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, NY
| | - Bu-Tian Ji
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD
| | - Qing Lan
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD
| | - Gui-Lan Li
- National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, Peoples Republic of China
| | - Nathaniel Rothman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD
| | | |
Collapse
|
25
|
Inv(11)(q21q23); KMT2A-MAML2, a Recurrent Genetic Abnormality in T-Cell Therapy-related Acute Lymphoblastic Leukemia. J Pediatr Hematol Oncol 2020; 42:e258-e261. [PMID: 31343482 DOI: 10.1097/mph.0000000000001572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
T-cell therapy-related acute lymphoblastic leukemia (T-t-ALL) is a rare condition associated with previous cytotoxic therapy for another disease. Here we report T-t-ALL with inv(11)(q21q23), which involves KMT2A and MAML2, a transcriptional coactivator of NOTCH proteins, that occurred after chemotherapy for Philadelphia chromosome-positive B-cell acute lymphoblastic leukemia. This case describes the youngest patient with T-t-ALL harboring inv(11)(q21q23) and is the first independent report following an initial series also occurring in children. Our results lend further support to the observation that the KMT2A-MAML2 fusion gene resulting from inv(11)(q21q23) is likely a recurrent cytogenetic abnormality in T-t-ALL and appears to be associated with pediatric cases.
Collapse
|
26
|
Seo B, Kim J, Kim S, Lee E. Bibliometric analysis of studies about acute myeloid leukemia conducted globally from 1999 to 2018. Blood Res 2020; 55:1-9. [PMID: 32269969 PMCID: PMC7106114 DOI: 10.5045/br.2020.55.1.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/20/2020] [Accepted: 02/03/2020] [Indexed: 01/12/2023] Open
Abstract
A bibliometric study is performed to analyze publication patterns in a specific research area and to establish a landscape model that can be used to quantitatively weigh publications. This study aimed to investigate AML research networks and to conduct a trend-related keyword analysis. We analyzed 48,202 studies about AML published from 1999 to 2019 in the Web of Science Core Collection. The network analysis was conducted using the R&R studio software. The journal Blood had the highest number of published articles with an h-index of 410. The USA had the highest number of total publications (18,719, 38.3%) and research funded by the government, institutions, and pharmaceutical companies (5,436, 10.8%). The institute with the largest number of publications was the MD Anderson Cancer Center. Kantarjian H, Garcia-Manero G, and Ravandi F were the leading authors of publications about AML. Keyword analysis revealed that FLT 3, micro-RNA, and NK cell topics were the hotspots in the cell and gene area in all publications. The overall AML research landscape is popular in the field of translational research as it can identify molecular, cell, and gene studies conducted by different funding agencies, countries, institutions, and author networks. With active funding and support from the Chinese government, the productivity of scientific research is increasing not only in the AML field but also in the medical/health-related science field.
Collapse
Affiliation(s)
- Beomjun Seo
- Department of Public Health, Graduate School, Korea University, Seoul, Korea
| | - Jeeyoon Kim
- Department of Clinical Pharmacy, Graduate School, Cha University, Seoul, Korea
| | - Seungwook Kim
- Graduate School of Interdisciplinary Management, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Eunil Lee
- Department of Public Health, Graduate School, Korea University, Seoul, Korea
| |
Collapse
|
27
|
Li Y, Xue Z, Dong X, Liu Q, Liu Z, Li H, Xing H, Xu Y, Tang K, Tian Z, Wang M, Rao Q, Wang J. Mitochondrial dysfunction and oxidative stress in bone marrow stromal cells induced by daunorubicin leads to DNA damage in hematopoietic cells. Free Radic Biol Med 2020; 146:211-221. [PMID: 31706989 DOI: 10.1016/j.freeradbiomed.2019.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 11/04/2019] [Accepted: 11/04/2019] [Indexed: 12/26/2022]
Abstract
Cytotoxic chemotherapies could cause the dysregulation of hematopoiesis and even put patients at increased risk of hematopoietic malignancy. Therapy-related leukemia is mainly caused by cytotoxic chemotherapy-induced genetic mutations in hematopoietic stem/progenitor cells (HSPCs). In addition to the intrinsic mechanism, some extrinsic events occurring in the bone marrow (BM) microenvironment are also possible mechanisms involved in genetic alteration. In the present study, we investigated the damage to BM stromal cells induced by a chemotherapy drug, daunorubicin (DNR) and further identified the DNA damage in hematopoietic cells caused by drug-treated stromal cells. It was found that treatment with DNR in mice caused a temporary reduction in cell number in each BM stromal cell subpopulation and the impairment of clonal growth potential in BM stromal cells. DNR treatment led to a tendency of senescence, generation of intracellular reactive oxygen species, production of cytokines and chemokines, and dysfunction of mitochondrial in stromal cells. Transcriptome microarray data and gene ontology (GO) or gene set enrichment analysis (GSEA) showed that differentially expressed genes that were down-regulated in response to DNR treatment were significantly enriched in mitochondrion function, and negative regulators of reactive oxygen species. Surprisingly, it was found that DNR-treated stromal cells secreted high levels of H2O2 into the culture supernatant. Furthermore, coculture of hematopoietic cells with DNR-treated stromal cells led to the accumulation of DNA damage as determined by the levels of histone H2AX phosphorylation and 8-oxo-2'-deoxyguanosine in hematopoietic cells. Overall, our results suggest that DNR-induced BM stromal cell damage can lead to genomic instability in hematopoietic cells.
Collapse
Affiliation(s)
- Yihui Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China
| | - Zhenya Xue
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China
| | - Xuanjia Dong
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China
| | - Qian Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China
| | - Zhe Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China
| | - Huan Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China
| | - Haiyan Xing
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China
| | - Yingxi Xu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China
| | - Kejing Tang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China
| | - Zheng Tian
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China
| | - Min Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China
| | - Qing Rao
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China.
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China; National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, PR China.
| |
Collapse
|
28
|
Thanarajasingam G, Minasian LM, Baron F, Cavalli F, De Claro RA, Dueck AC, El-Galaly TC, Everest N, Geissler J, Gisselbrecht C, Gribben J, Horowitz M, Ivy SP, Jacobson CA, Keating A, Kluetz PG, Krauss A, Kwong YL, Little RF, Mahon FX, Matasar MJ, Mateos MV, McCullough K, Miller RS, Mohty M, Moreau P, Morton LM, Nagai S, Rule S, Sloan J, Sonneveld P, Thompson CA, Tzogani K, van Leeuwen FE, Velikova G, Villa D, Wingard JR, Wintrich S, Seymour JF, Habermann TM. Beyond maximum grade: modernising the assessment and reporting of adverse events in haematological malignancies. Lancet Haematol 2018; 5:e563-e598. [PMID: 29907552 PMCID: PMC6261436 DOI: 10.1016/s2352-3026(18)30051-6] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/28/2018] [Accepted: 03/29/2018] [Indexed: 02/06/2023]
Abstract
Tremendous progress in treatment and outcomes has been achieved across the whole range of haematological malignancies in the past two decades. Although cure rates for aggressive malignancies have increased, nowhere has progress been more impactful than in the management of typically incurable forms of haematological cancer. Population-based data have shown that 5-year survival for patients with chronic myelogenous and chronic lymphocytic leukaemia, indolent B-cell lymphomas, and multiple myeloma has improved markedly. This improvement is a result of substantial changes in disease management strategies in these malignancies. Several haematological malignancies are now chronic diseases that are treated with continuously administered therapies that have unique side-effects over time. In this Commission, an international panel of clinicians, clinical investigators, methodologists, regulators, and patient advocates representing a broad range of academic and clinical cancer expertise examine adverse events in haematological malignancies. The issues pertaining to assessment of adverse events examined here are relevant to a range of malignancies and have been, to date, underexplored in the context of haematology. The aim of this Commission is to improve toxicity assessment in clinical trials in haematological malignancies by critically examining the current process of adverse event assessment, highlighting the need to incorporate patient-reported outcomes, addressing issues unique to stem-cell transplantation and survivorship, appraising challenges in regulatory approval, and evaluating toxicity in real-world patients. We have identified a range of priority issues in these areas and defined potential solutions to challenges associated with adverse event assessment in the current treatment landscape of haematological malignancies.
Collapse
Affiliation(s)
| | - Lori M Minasian
- National Cancer Institute, National Institutes of Health, Department of Health & Human Services, Bethesda, MD, USA
| | - Frederic Baron
- Division of Haematology, University of Liege, Liege, Belgium
| | - Franco Cavalli
- Oncology Institute of Southern Switzerland, Bellinzona, Switzlerand
| | - R Angelo De Claro
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Amylou C Dueck
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ, USA
| | - Tarec C El-Galaly
- Department of Haematology, Aalborg University Hospital, Aalborg Denmark
| | - Neil Everest
- Haematology Clinical Evaluation Unit, Therapeutic Goods Administration, Department of Health, Symondston, ACT, Australia
| | - Jan Geissler
- Leukaemia Patient Advocates Foundation, Bern, Switzerland
| | - Christian Gisselbrecht
- Haemato-Oncology Department, Hopital Saint-Louis, Paris Diderot University VII, Paris, France
| | - John Gribben
- Centre for Haemato-Oncology, Barts Cancer Institute, London, UK
| | - Mary Horowitz
- Division of Haematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - S Percy Ivy
- National Cancer Institute, National Institutes of Health, Department of Health & Human Services, Bethesda, MD, USA
| | - Caron A Jacobson
- Division of Haematologic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Armand Keating
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Paul G Kluetz
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Aviva Krauss
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Yok Lam Kwong
- Department of Haematology and Haematologic Oncology, University of Hong Kong, Hong Kong, China
| | - Richard F Little
- National Cancer Institute, National Institutes of Health, Department of Health & Human Services, Bethesda, MD, USA
| | | | - Matthew J Matasar
- Lymphoma and Adult BMT Services, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Robert S Miller
- CancerLinQ, American Society of Clinical Oncology, Alexandria, VA, USA
| | - Mohamad Mohty
- Haematology and Cellular Therapy Department, Saint-Antoine Hospital, University Pierre & Marie Curie, Paris, France
| | | | - Lindsay M Morton
- National Cancer Institute, National Institutes of Health, Department of Health & Human Services, Bethesda, MD, USA
| | - Sumimasa Nagai
- University of Tokyo, Tokyo, Japan; Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Simon Rule
- Plymouth University Medical School, Plymouth, UK
| | - Jeff Sloan
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Pieter Sonneveld
- Department of Haematology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | | | | | | | - Galina Velikova
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Diego Villa
- Division of Medical Oncology, British Columbia Cancer Agency, University of British Columbia, Vancouver, BC, Canada
| | - John R Wingard
- Division of Haematology & Oncology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Sophie Wintrich
- Myelodysplastic Syndrome (MDS) Alliance and MDS UK Patient Support Group, London, UK
| | - John F Seymour
- Department of Haematology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Royal Melbourne Hospital, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | | |
Collapse
|
29
|
Demoor-Goldschmidt C, de Vathaire F. Review of risk factors of secondary cancers among cancer survivors. Br J Radiol 2018; 92:20180390. [PMID: 30102558 DOI: 10.1259/bjr.20180390] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Improvements in cancer survival have made the long-term risks from treatments more important, in particular among the children, adolescents and young adults who are more at risk particularly due to a longer life expectancy and a higher sensitivity to treatments. Subsequent malignancies in cancer survivors now constitute 15 to 20% of all cancer diagnoses in the cancer registries. Lots of studies are published to determine risk factors, with some controversial findings. Just data from large cohorts with detailed information on individual treatments and verification of what is called "secondary cancers" can add some knowledge, because their main difficulty is that the number of events for most second cancer sites are low, which impact the statistical results. In this review of the literature, we distinguish second and secondary cancers and discuss the factors contributing to this increased risk of secondary cancers. The article concludes with a summary of current surveillance and screening recommendations.
Collapse
Affiliation(s)
- Charlotte Demoor-Goldschmidt
- CESP University, Paris-Sud, UVSQ, INSERM, Université Paris-Saclay, Villejuif, France.,Cancer and Radiation Team, Gustave Roussy, Villejuif, France.,Pediatric Oncology, Hematology, Immunology, CHU d'Angers, Angers, France
| | - Florent de Vathaire
- CESP University, Paris-Sud, UVSQ, INSERM, Université Paris-Saclay, Villejuif, France.,Cancer and Radiation Team, Gustave Roussy, Villejuif, France
| |
Collapse
|
30
|
Yang LH, Su P, Luedke C, Lu CM, Louissaint A, McCall CM, Rapisardo S, Vallangeon B, Wang E. Chronic Myeloid Leukemia Following Treatment for Primary Neoplasms or Other Medical Conditions. Am J Clin Pathol 2018; 150:246-258. [PMID: 29992292 DOI: 10.1093/ajcp/aqy050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Therapy-related chronic myeloid leukemia (CML) has been reported, but its clinical presentation and pathologic features have not yet been well characterized. METHODS Twenty-one cases of CML following treatment for primary diseases were collected and retrospectively analyzed. RESULTS The clinical presentation, pathologic features, and cytogenetic profile were similar to de novo CML. In particular, those with an isolated Philadelphia chromosome constituted 88.9% of our cases, and additional aberrations characteristic of therapy-related acute myeloid leukemia/myelodysplastic syndrome (AML/MDS) were not identified in this study. The patients responded to imatinib/derivatives and survived with limited follow-up. CONCLUSIONS Therapy-related CML has a clinical presentation, pathologic features, and cytogenetic profile akin to de novo CML. Absence of additional significant aberrations seems to suggest a pathogenesis different from therapy-related AML/MDS. Therapy-related CML exhibits a robust therapeutic response to imatinib/derivatives and favorable clinical outcomes similar to de novo CML.
Collapse
Affiliation(s)
- Lian-He Yang
- Department of Pathology, First Affiliated Hospital and College of Basic Sciences of China Medical University, Shenyang, China
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Pu Su
- Department of Pathology, Duke University Medical Center, Durham, NC
- Department of Medicine, East Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Catherine Luedke
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Chuanyi Mark Lu
- Department of Laboratory Medicine, University of California San Francisco
| | - Abner Louissaint
- Department of Pathology and Laboratory Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Chad M McCall
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Sarah Rapisardo
- Department of Pathology, Duke University Medical Center, Durham, NC
| | - Bethany Vallangeon
- Department of Pathology, East Carolina University Medical Center, Greenville, NC
| | - Endi Wang
- Department of Pathology, Duke University Medical Center, Durham, NC
| |
Collapse
|
31
|
Teepen JC, Curtis RE, Dores GM, Berrington de Gonzalez A, van den Heuvel-Eibrink MM, Kremer LCM, Gilbert ES, van Leeuwen FE, Ronckers CM, Morton LM. Risk of subsequent myeloid neoplasms after radiotherapy treatment for a solid cancer among adults in the United States, 2000-2014. Leukemia 2018; 32:2580-2589. [PMID: 29795414 DOI: 10.1038/s41375-018-0149-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/24/2018] [Accepted: 04/11/2018] [Indexed: 12/31/2022]
Abstract
Although increased risk of acute myeloid leukemia (AML) has been observed after chemotherapy and radiotherapy, less is known about radiotherapy-related risks of specific AML subtypes and other specific myeloid neoplasms. We used the US population-based cancer registry data to evaluate risk of myeloid neoplasms among three cohorts of cancer survivors initially treated with radiotherapy only. We included 1-year survivors of first primary thyroid (radioiodine only, stages I-IV; N = 49 879), prostate (excluding stage IV; N = 237 439), or uterine corpus cancers (stage I-II; N = 16 208) diagnosed during 2000-2013. We calculated standardized incidence ratios (SIRs) and excess absolute risks (EARs). Thyroid cancer survivors had significantly elevated risks of total AML (SIR = 2.77, 95% CI: 1.99-3.76), AML with cytogenetic abnormalities (SIR = 3.90, 95% CI: 1.57-8.04), AML with myelodysplasia-related changes (SIR = 2.87, 95% CI: 1.05-6.25), and BCR-ABL1-positive chronic myelogenous leukemia (CML) (SIR = 5.38, 95% CI: 2.58-9.89). Irradiated prostate and uterine corpus cancer survivors were at elevated risk for total AML (SIR = 1.14, 95% CI: 1.03-1.27 and SIR = 1.77, 95% CI: 1.01-2.87, respectively), AML with cytogenetic abnormalities (SIR = 2.52, 95% CI: 1.84-3.37 and SIR = 7.21, 95% CI: 2.34-16.83, respectively), and acute promyelocytic leukemia (SIR = 3.20, 95% CI: 2.20-4.49 and SIR = 8.88, 95% CI: 2.42-22.73, respectively). In addition, prostate cancer survivors were at increased risk of BCR-ABL1-positive CML (SIR = 2.11, 95% CI: 1.52-2.85). Our findings support the importance of diagnostic precision in myeloid neoplasm classification since susceptibility following radiotherapy may vary by myeloid neoplasm subtype, thereby informing risk/benefit discussions in first primary cancer treatment.
Collapse
Affiliation(s)
- Jop C Teepen
- Department of Pediatric Oncology, Emma Children's Hospital/Academic Medical Center, Amsterdam, The Netherlands. .,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| | - Rochelle E Curtis
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Graça M Dores
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Amy Berrington de Gonzalez
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | | | - Leontien C M Kremer
- Department of Pediatric Oncology, Emma Children's Hospital/Academic Medical Center, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Ethel S Gilbert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Flora E van Leeuwen
- Department of Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Cécile M Ronckers
- Department of Pediatric Oncology, Emma Children's Hospital/Academic Medical Center, Amsterdam, The Netherlands
| | - Lindsay M Morton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| |
Collapse
|
32
|
Allogeneic Hematopoietic Stem Cell Transplantation In Therapy-Related Myeloid Neoplasms (t-MN) of the Adult: Monocentric Observational Study and Review of the Literature. Mediterr J Hematol Infect Dis 2018; 10:e2018005. [PMID: 29326802 PMCID: PMC5760063 DOI: 10.4084/mjhid.2018.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/01/2017] [Indexed: 02/07/2023] Open
Abstract
Background Therapy related myeloid neoplasms (t-MN) occur due to direct mutational events of chemotherapeutic agents and radiotherapy. Disease latency, mutational events and prognosis vary with drugs categories. Methods We describe a cohort of 30 patients, 18 females and 12 males, with median age of 52.5 years (range, 20 to 64), submitted to allogeneic stem cell transplantation (HSCT) in our department between September 1999 and March 2017. Patients had a history of solid tumour in 14 cases, haematological disease in 15 cases and both of them in one case. After a median of 36.5 months (range, 4 to 190) from first neoplasm, patients developed t-AML in 19 cases and t-MDS in 11 cases. Molecular abnormalities were detected in 5 patients, while karyotype aberrations were found in 17 patients. Patients received conventional chemotherapy in 14 cases, azacitidine in 10 cases and both of them in one case. Five patients were submitted to HSCT without previous treatment except for supportive therapy. Results Seventeen patients obtained sustained CR after SCT, while 8 patients showed resistant or relapsed disease. The remaining five patients died early after SCT. At follow up time (May 2017) 13 patients were alive with a median OS of 48 months (range 3–195), while 17 patients died after a median of 4 months (range 1–27) by relapse mortality in 6 cases and non-relapse mortality in the other 11 patients. Conclusions Global OS was 43%. After SCT, 72.2% of patients with t-MN maintained a sustained CR.
Collapse
|
33
|
Tikku G, Jain M, Shukla P. Chronic Myeloid Leukemia with a Complex Variant 'Ph' Translocation That Develops in Breast Carcinoma, Postchemotherapy: A Rare but Treatable Entity. J Breast Cancer 2017; 20:208-211. [PMID: 28690659 PMCID: PMC5500406 DOI: 10.4048/jbc.2017.20.2.208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 01/14/2017] [Indexed: 12/04/2022] Open
Abstract
We report a case of chronic myeloid leukemia (CML) that developed after postoperative chemotherapy with cyclophosphamide, doxorubicin and 5-fluorouracil (CAF) for breast cancer. A 55-year-old woman was diagnosed with invasive ductal carcinoma which was treated with a modified radical mastectomy followed by six cycles of CAF chemotherapy. Nine years later, she developed CML and locoregional recurrence. Her breast recurrence showed strong estrogen receptor, weak progesterone receptor and strong human epidermal growth factor 2 (score 3+) expression. Her secondary CML in the chronic phase showed a complex variant translocation (CVT) involving chromosomes 9, 22, and 17. Considering that the HER2/neu gene is also located on chromosome 17, this secondary CML in chronic phase with CVT is indeed a rare occurrence. We discuss the associated genetic factors and the possible role of breast cancer chemo/radiotherapy in the development of such CML as well as its treatment and prognosis compared with de novo CML.
Collapse
Affiliation(s)
- Gargi Tikku
- Department of Oncopathology, Delhi State Cancer Institute, Delhi, India
| | - Monica Jain
- Department of Oncopathology, Delhi State Cancer Institute, Delhi, India
| | - Pragya Shukla
- Department of Clinical Oncology, Delhi State Cancer Institute, Delhi, India
| |
Collapse
|
34
|
Lu QS, Xu N, Zhou X, Cai GX, Li L, Li YL, Lu ZY, Huang JX, Liu QF, Liu XL. [Clinical characteristics and prognosis of 35 patients with therapy-related hematological neoplasms]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2017; 37:221-6. [PMID: 27033760 PMCID: PMC7342953 DOI: 10.3760/cma.j.issn.0253-2727.2016.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
目的 探讨治疗相关血液肿瘤的临床特征及预后。 方法 采用细胞形态学、流式细胞术、间期荧光原位杂交技术(I-FISH)、染色体核型分析对35例治疗相关血液肿瘤患者进行诊断和分型并回顾性分析其临床特征及预后。 结果 35例患者中,治疗相关急性髓系白血病(t-AML)20例,治疗相关急性淋巴细胞白血病(t-ALL)4例,治疗相关急性混合细胞白血病1例,治疗相关非霍奇金淋巴瘤(t-NHL) 8例,治疗相关骨髓增生异常综合征(t-MDS)2例。第一肿瘤至治疗相关恶性血液肿瘤的中位发病间隔期为29(16~90)个月,中位生存时间14(1~60)个月,3年累积生存率为17.1%。在25例治疗相关性急性白血病患者中,40.0%(10/25)合并复杂核型,36.0%(9/25)合并MLL断裂基因重排,12.0%(3/25)合并AML-ETO融合基因阳性,1例合并NPM1点突变,1例合并P16基因缺失。 结论 治疗相关血液肿瘤患者的预后差。
Collapse
Affiliation(s)
- Q S Lu
- Department of Hematology, Nanfang Hospital, Southern Medical University, 510515 Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
van de Ven M, van der Burg E, van der Gulden H, Klarenbeek S, Bouwman P, Jonkers J. Prophylactic window therapy with the clinical poly(ADP-ribose) polymerase inhibitor olaparib delays BRCA1-deficient mammary tumour formation in mice. J Pathol 2017; 241:511-521. [PMID: 27943283 DOI: 10.1002/path.4857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 11/07/2016] [Accepted: 11/17/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Marieke van de Ven
- Division of Molecular Pathology and Cancer Genomics Netherlands; The Netherlands Cancer Institute; Amsterdam The Netherlands
- MCCA Preclinical Intervention Unit; The Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Eline van der Burg
- Division of Molecular Pathology and Cancer Genomics Netherlands; The Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Hanneke van der Gulden
- Division of Molecular Pathology and Cancer Genomics Netherlands; The Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Sjoerd Klarenbeek
- Division of Molecular Pathology and Cancer Genomics Netherlands; The Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Peter Bouwman
- Division of Molecular Pathology and Cancer Genomics Netherlands; The Netherlands Cancer Institute; Amsterdam The Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology and Cancer Genomics Netherlands; The Netherlands Cancer Institute; Amsterdam The Netherlands
| |
Collapse
|
36
|
Atwal M, Lishman EL, Austin CA, Cowell IG. Myeloperoxidase Enhances Etoposide and Mitoxantrone-Mediated DNA Damage: A Target for Myeloprotection in Cancer Chemotherapy. Mol Pharmacol 2017; 91:49-57. [PMID: 27974636 PMCID: PMC5198516 DOI: 10.1124/mol.116.106054] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 11/08/2016] [Indexed: 01/17/2023] Open
Abstract
Myeloperoxidase is expressed exclusively in granulocytes and immature myeloid cells and transforms the topoisomerase II (TOP2) poisons etoposide and mitoxantrone to chemical forms that have altered DNA damaging properties. TOP2 poisons are valuable and widely used anticancer drugs, but they are associated with the occurrence of secondary acute myeloid leukemias. These factors have led to the hypothesis that myeloperoxidase inhibition could protect hematopoietic cells from TOP2 poison-mediated genotoxic damage and, therefore, reduce the rate of therapy-related leukemia. We show here that myeloperoxidase activity leads to elevated accumulation of etoposide- and mitoxantrone-induced TOP2A and TOP2B-DNA covalent complexes in cells, which are converted to DNA double-strand breaks. For both drugs, the effect of myeloperoxidase activity was greater for TOP2B than for TOP2A. This is a significant finding because TOP2B has been linked to genetic damage associated with leukemic transformation, including etoposide-induced chromosomal breaks at the MLL and RUNX1 loci. Glutathione depletion, mimicking in vivo conditions experienced during chemotherapy treatment, elicited further MPO-dependent increase in TOP2A and especially TOP2B-DNA complexes and DNA double-strand break formation. Together these results support targeting myeloperoxidase activity to reduce genetic damage leading to therapy-related leukemia, a possibility that is enhanced by the recent development of novel specific myeloperoxidase inhibitors for use in inflammatory diseases involving neutrophil infiltration.
Collapse
Affiliation(s)
- Mandeep Atwal
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne. United Kingdom
| | - Emma L Lishman
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne. United Kingdom
| | - Caroline A Austin
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne. United Kingdom
| | - Ian G Cowell
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne. United Kingdom
| |
Collapse
|
37
|
Cho HW, Choi YB, Yi ES, Lee JW, Sung KW, Koo HH, Yoo KH. Therapy-related myeloid neoplasms in children and adolescents. Blood Res 2016; 51:242-248. [PMID: 28090486 PMCID: PMC5234240 DOI: 10.5045/br.2016.51.4.242] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 08/18/2016] [Accepted: 09/06/2016] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND This retrospective study aimed to characterize and analyze the outcome of therapy-related myeloid neoplasms (t-MNs) in children and adolescents. METHODS The medical records of 16 patients under 21 years of age at the time of t-MN diagnosis were reviewed. RESULTS The median patient age was 11.5 years (range, 1.6-20.4 yr). Twelve patients had therapy-related acute myeloid leukemia, 3 patients had myelodysplastic syndrome, and 1 patient had chronic myelomonocytic leukemia. The median latency period was 29 months (range, 11-68 mo). Fourteen patients had cytogenetic aberrations, 8 of whom had an 11q23 abnormality. Of the 13 patients treated with curative intent, 12 patients received myeloid-type induction therapy that led to complete remission (CR) in 8 patients. Nine patients underwent allogeneic transplantation; 4 patients did not undergo transplantation due to chemotherapy-related toxic death (N=3) or parental refusal (N=1). The 5-year overall survival and event-free survival of the 13 patients treated with a curative intent were 46.2% and 30.8%, respectively. For the 9 patients who underwent allogeneic transplantation, the 5-year event-free survival was 66.7%. CONCLUSION A significant proportion of young patients with t-MNs can experience long-term survival, and allogeneic transplantation plays a key role for attaining cure in these patients.
Collapse
Affiliation(s)
- Hee Won Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Bae Choi
- Department of Pediatrics, Chung-Ang University Hospital, Seoul, Korea
| | - Eun Sang Yi
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Won Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.; Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
38
|
Takahashi K, Wang F, Kantarjian H, Doss D, Khanna K, Thompson E, Zhao L, Patel K, Neelapu S, Gumbs C, Bueso-Ramos C, DiNardo CD, Colla S, Ravandi F, Zhang J, Huang X, Wu X, Samaniego F, Garcia-Manero G, Futreal PA. Preleukaemic clonal haemopoiesis and risk of therapy-related myeloid neoplasms: a case-control study. Lancet Oncol 2016; 18:100-111. [PMID: 27923552 PMCID: PMC5405697 DOI: 10.1016/s1470-2045(16)30626-x] [Citation(s) in RCA: 288] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 10/04/2016] [Accepted: 10/13/2016] [Indexed: 01/01/2023]
Abstract
Background Therapy-related myeloid neoplasms (t-MNs) are often fatal secondary
malignancies. Risk factors for t-MNs are not well understood. Recent studies
suggested that individuals with clonal hematopoiesis have higher risk of
developing hematological malignancies. We hypothesized that cancer patients
with clonal hematopoiesis have increased risk of developing t-MNs. Methods We conducted a retrospective case-control study to compare the
prevalence of clonal hematopoiesis between patients who developed t-MNs
(cases) and who did not develop t-MNs (control). For cases, we studied14
patients with various types of cancers who developed t-MNs and whose paired
samples of t-MN bone marrow (BM) and peripheral blood (PB) that were
previously obtained at the time of primary cancer diagnosis were available.
Fifty four patients with lymphoma who received combination chemotherapy and
did not develop t-MNs after at least 5 years of follow up were studied as a
control. We performed molecular barcode sequencing of 32 genes on the
pre-treatment PB samples to detect clonal hematopoiesis. For the t-MN cases,
we also performed targeted gene sequencing on t-MN BM samples and
investigated clonal evolution from clonal hematopoiesis to t-MNs. To confirm
association between clonal hematopoiesis and t-MN development, we also
analyzed prevalence of clonal hematopoiesis in a separate cohort of 74
patients with lymphoma. All of these patients were treated under the
prospective randomized trial of frontline chemotherapy with
cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) with or
without melatonin and 5 (7%) of them had developed t-MNs. Findings In 14 patients with t-MNs, we detected pre-leukemic mutations in 10
of their prior PB samples (71%). In control, clonal hematopoiesis
was detected in 17 patients (31%), and the cumulative incidence of
t-MNs at 5 years was significantly higher in patients with clonal
hematopoiesis (30% [95% CI: 16% –
51%] vs. 7% [95% CI: 2%
– 21%], P = 0.016). In the separate cohort,
5 patients (7%) developed t-MNs and 4 (80%) of them had
clonal hematopoiesis. The cumulative incidence of t-MNs at 10 years was
significantly higher in patients with clonal hematopoiesis (29%
[95% CI: 8%–53%] vs.
0% [95% CI: 0%–0%],
P = 0.0009). Multivariate Fine and Gray model showed that having
clonal hematopoiesis significantly increased the risk of t-MN development
(HR = 13.7, P = 0.013). Interpretation Pre-leukemic clonal hematopoiesis is frequently detected in patients
with t-MNs at the time of their primary cancer diagnosis and before patients
were exposed to chemotherapy/radiation therapy. Detection of clonal
hematopoiesis significantly increased the risk of t-MN development in
patients with lymphoma. These data suggest potential approaches of screening
clonal hematopoiesis in cancer patients to identify patients at risk of
t-MNs and warrants a validation in prospective trial investigating a role of
clonal hematopoiesis as a predictive marker for t-MNs.
Collapse
Affiliation(s)
- Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Feng Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Denaha Doss
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kanhav Khanna
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Erika Thompson
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Zhao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sattva Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Curtis Gumbs
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carlos Bueso-Ramos
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Institute of Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Felipe Samaniego
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - P Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Therapy-related myeloid neoplasms (tMN) are increasingly recognized and studied diseases which have traditionally been defined clinically. With advances in methods used to study the genetics of aging and myeloid disease biology, novel insights are emerging which are expected to improve our understanding of the genetics and pathogenesis of tMN. RECENT FINDINGS Clinical outcomes in tMN and de novo MDS/AML appear to be largely determined by genetics, and data are emerging to show how DNA mutations may enhance tMN risk stratification. The discovery of skewed hematopoieses and mutations in healthy older adults suggests an alternate predisposition mechanism for the genesis of tMN. Patients with tMN do respond to standard therapy and can benefit from allogeneic transplant in a manner similar to their genetically matched de novo counterpart. SUMMARY De novo MDS/AML and tMN have shared genetic features, and tMN clinical outcomes may depend more on the genetics at presentation than the clinical history of an antecedent malignancy. Acquired somatic mutations in genes such as TP53 and myeloid skewing with associated mutations in cancer-free older adults may predispose such individuals to tMN under the influence of myelosuppressive therapy, and this may be a route to the development of a subset of tMN.
Collapse
|
40
|
Leung G, Papademetriou M, Chang S, Arena F, Katz S. Interactions Between Inflammatory Bowel Disease Drugs and Chemotherapy. ACTA ACUST UNITED AC 2016; 14:507-534. [DOI: 10.1007/s11938-016-0109-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
41
|
Megías-Vericat JE, Montesinos P, Herrero MJ, Bosó V, Martínez-Cuadrón D, Poveda JL, Sanz MÁ, Aliño SF. Pharmacogenomics and the treatment of acute myeloid leukemia. Pharmacogenomics 2016; 17:1245-1272. [DOI: 10.2217/pgs-2016-0055] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clinically and biologically heterogeneous malignancy that is primarily treated with combinations of cytarabine and anthracyclines. Although this scheme remains effective in most of the patients, variability of outcomes in patients has been partly related with their genetic variability. Several pharmacogenetic studies have analyzed the impact of polymorphisms in genes encoding transporters, metabolizers or molecular targets of chemotherapy agents. A systematic review on all eligible studies was carried out in order to estimate the effect of polymorphisms of anthracyclines and cytarabine pathways on efficacy and toxicity of AML treatment. Other emerging genes recently studied in AML, such as DNA repair genes, genes potentially related to chemotherapy response or AML prognosis, have also been included.
Collapse
Affiliation(s)
- Juan Eduardo Megías-Vericat
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
- Servicio de Farmacia, Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - Pau Montesinos
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - María José Herrero
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
- Departamento Farmacología, Facultad de Medicina, Universidad de Valencia, Avda, Blasco Ibáñez 15, 46010 – Valencia, Spain
| | - Virginia Bosó
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
- Servicio de Farmacia, Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - David Martínez-Cuadrón
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - José Luis Poveda
- Servicio de Farmacia, Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - Miguel Ángel Sanz
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - Salvador F Aliño
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
- Departamento Farmacología, Facultad de Medicina, Universidad de Valencia, Avda, Blasco Ibáñez 15, 46010 – Valencia, Spain
- Unidad de Farmacología Clínica, Área del Medicamento, Hospital Universitario y Politécnico La Fe. Avda. Fernando Abril Martorell 106, 46026 – Valencia, Spain
| |
Collapse
|
42
|
ESPíRITO Santo AE, Chacim S, Ferreira I, Leite L, Moreira C, Pereira D, Dantas Brito MD, Nunes M, Domingues N, Oliveira I, Moreira I, Martins A, Viterbo L, Mariz JM, Medeiros R. Effect of therapy-related acute myeloid leukemia on the outcome of patients with acute myeloid leukemia. Oncol Lett 2016; 12:262-268. [PMID: 27347135 DOI: 10.3892/ol.2016.4591] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 10/28/2015] [Indexed: 01/30/2023] Open
Abstract
Therapy-related acute myeloid leukemia (t-AML) is a rare and almost always fatal late side effect of antineoplastic treatment involving chemotherapy, radiotherapy or the two combined. The present retrospective study intended to characterize t-AML patients that were diagnosed and treated in a single referral to an oncological institution in North Portugal. Over the past 10 years, 231 cases of AML were diagnosed and treated at the Portuguese Institute of Oncology of Porto, of which 38 t-AML cases were identified. Data regarding the patient demographics, primary diagnosis and treatment, age at onset of therapy-related myeloid neoplasm, latency time of the neoplasm, cytogenetic characteristics, AML therapy and outcome were collected from medical records. A previous diagnosis with solid tumors was present in 28 patients, and 10 patients possessed a history of hematological conditions, all a lymphoproliferative disorder. Breast cancer was the most frequent solid tumor identified (39.5% of all solid tumors diagnosed). The mean latency time was 3 years. In the present study, t-AML patients were older (P<0.001) and more frequently carried cytogenetic abnormalities (P=0.009) compared with de novo AML patients. The overall survival time was observed to be significantly poorer among individuals with t-AML (P<0.001). However, in younger patients (age, <50 years) there was no difference between the overall survival time of patients with t-AML and those with de novo AML (P=0.983). Additionally, patients with promyelocytic leukemia possess a good prognosis, even when AML occurs as a secondary event (P=0.98). To the best of our knowledge, the present study is the first to evaluate t-AML in Portugal and the results are consistent with the data published previously in other populations. The present study concludes that although t-AML demonstrates a poor prognosis, this is not observed among younger patients or promyelocytic leukemia patients.
Collapse
Affiliation(s)
- Ana Espírito ESPíRITO Santo
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal; Faculty of Medicine, University of Porto, Porto, Portugal
| | - Sérgio Chacim
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Isabel Ferreira
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Luís Leite
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Claudia Moreira
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Dulcineia Pereira
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | | | - Marta Nunes
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Nelson Domingues
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Isabel Oliveira
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Ilídia Moreira
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Angelo Martins
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Luísa Viterbo
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - José Mário Mariz
- Department of Onco-Hematology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group Investigation Centre, Portuguese Institute of Oncology of Porto, Porto, Portugal; Abel Salazar Institute of Biomedical Sciences, University of Porot, Porto, Portugal; Department of Oncology, Portuguese Institute of Oncology, Porto, Portugal; Biomedical Research Centre, Faculty of Health Sciences of Fernando Pessoa University, Porto, Portugal; Department of Research, Portuguese League Against Cancer (Regional North Core), Porto, Portugal
| |
Collapse
|
43
|
van Leeuwen FE, Ronckers CM. Anthracyclines and Alkylating Agents: New Risk Factors for Breast Cancer in Childhood Cancer Survivors? J Clin Oncol 2016; 34:891-4. [DOI: 10.1200/jco.2015.65.0465] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
| | - Cécile M. Ronckers
- Emma Children’s Hospital/Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
44
|
Morton LM, Onel K, Curtis RE, Hungate EA, Armstrong GT. The rising incidence of second cancers: patterns of occurrence and identification of risk factors for children and adults. Am Soc Clin Oncol Educ Book 2015:e57-67. [PMID: 24857148 DOI: 10.14694/edbook_am.2014.34.e57] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As the population of cancer survivors has increased and continues to age, the occurrence of second cancers has risen dramatically-from 9% of all cancer diagnoses in 1975-1979 to 19% in 2005-2009. The Childhood Cancer Survivor Study, a cohort of more than 14,000 childhood cancer survivors with detailed exposure data and long-term follow-up, has substantially contributed to our understanding of the roles of radiotherapy and chemotherapy in second cancer occurrence. In particular, dose-related risks have been demonstrated for second cancers of the breast, thyroid, central nervous system, gastrointestinal tract, and sarcomas following radiation. Cytotoxic chemotherapy-which has long been known to be leukemogenic-also appears to contribute to risk for a range of other second cancer types. Individuals who develop a second cancer are at particularly high risk for developing additional second cancers. A genome-wide association study of survivors of Hodgkin lymphoma who received radiotherapy identified a locus on chromosome 6q21 as being associated with second cancer risk, demonstrating that recent advances in genomics are likely to prove invaluable for elucidating the contribution of genetic susceptibility to second cancer etiology. Among adults, risk of second cancers varies substantially by type of first and second cancer, patient age, and prevalence of second cancer risk factors, including primary cancer treatments, environmental and lifestyle exposures, and genetic susceptibility. Further research is needed to quantify second cancer risks associated with specific etiologic factors and to identify the patients at highest risk of developing a second cancer to target prevention and screening efforts.
Collapse
Affiliation(s)
- Lindsay M Morton
- From the Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, MD; Department of Pediatrics, The University of Chicago, Chicago, IL; and Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| | - Kenan Onel
- From the Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, MD; Department of Pediatrics, The University of Chicago, Chicago, IL; and Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| | - Rochelle E Curtis
- From the Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, MD; Department of Pediatrics, The University of Chicago, Chicago, IL; and Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| | - Eric A Hungate
- From the Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, MD; Department of Pediatrics, The University of Chicago, Chicago, IL; and Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| | - Gregory T Armstrong
- From the Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, MD; Department of Pediatrics, The University of Chicago, Chicago, IL; and Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
45
|
Adhikari J, Sharma P, Bhatt VR. Risk of secondary solid malignancies after allogeneic hematopoietic stem cell transplantation and preventive strategies. Future Oncol 2015; 11:3175-85. [PMID: 26551415 DOI: 10.2217/fon.15.252] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The risk of secondary solid malignancies is increased after allogeneic hematopoietic stem cell transplantation (HSCT). The risk starts at about 10 years after HSCT and continues even 20 years later. The most common secondary malignancies include squamous cell carcinoma of skin, genitourinary tract and oral cavity; lung and breast cancers. The use of total body irradiation or conditioning chemotherapy, chronic graft-versus-host disease and duration since HSCT can influence the risk of secondary solid malignancies. Secondary solid malignancies are common causes of nonrelapse mortality in long-term survivors and may account for up to 10% of late deaths. Avoiding smoking, alcohol use and excess sun exposure may reduce the risk. Cancer prevention guidelines are largely consensus-driven and follow the recommendations for general population.
Collapse
Affiliation(s)
- Janak Adhikari
- Department of Medicine, Kathmandu University School of Medical Sciences, Dhulikhel, Kavre, Nepal
| | - Priyadarshani Sharma
- Department of Medicine, Kathmandu University School of Medical Sciences, Dhulikhel, Kavre, Nepal
| | - Vijaya Raj Bhatt
- Division of Hematology-Oncology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
46
|
Palmitelli M, de Campos-Nebel M, González-Cid M. Progression of chromosomal damage induced by etoposide in G2 phase in a DNA-PKcs-deficient context. Chromosome Res 2015; 23:719-32. [PMID: 26152239 DOI: 10.1007/s10577-015-9478-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/11/2015] [Accepted: 06/16/2015] [Indexed: 11/29/2022]
Abstract
Etoposide (ETO), a drug used for the treatment of human tumors, is associated with the development of secondary malignancies. Recently, therapeutic strategies have incorporated chemosensitizing agents to improve the tumoral response to this drug. ETO creates DNA double-strand breaks (DSB) via inhibition of DNA topoisomerase II (Top2). To repair DSB, homologous recombination (HR) and non-homologous end-joining (NHEJ), involving D-NHEJ (dependent of the catalytic subunit of DNA-dependent protein kinase, DNA-PKcs) and B-NHEJ (backup repair pathway) are activated. We evaluated the progression of the DNA damage induced by the Top2 poison ETO in G2 phase of human HeLa cells after chemical inhibition of DNA-PKcs with NU7026. Compared to ETO treatment alone, this combined treatment resulted in a twofold higher rate of chromatid breaks and exchanges when analysis was performed in the following metaphase. Moreover, when analysis was performed in the second metaphase following treatment, increases in the percentage of micronuclei with H2AX (biomarker for DSB) foci in binucleated cells and dicentric chromosomes were seen. In post-mitotic G1 phase, a close association between unresolved DSB and meiotic recombination 11 homolog A (MRE11) signals was observed, demonstrating the contribution of MRE11 in the DSB repair by B-NHEJ. Hence, chemical inhibition of DNA-PKcs impaired both D-NHEJ and HR repair pathways, altering the maintenance of chromosomal integrity and cell proliferation. Our results suggest that the chemosensitizing effectiveness of the DNA-PKcs inhibitor and the survival rate of aberrant cells may contribute to the development of therapy-related tumors.
Collapse
Affiliation(s)
- Micaela Palmitelli
- Laboratorio de Mutagénesis, Instituto de Medicina Experimental, IMEX-CONICET, Academia Nacional de Medicina, J. A. Pacheco de Melo 3081, 1425, Buenos Aires, Argentina
| | - Marcelo de Campos-Nebel
- Laboratorio de Mutagénesis, Instituto de Medicina Experimental, IMEX-CONICET, Academia Nacional de Medicina, J. A. Pacheco de Melo 3081, 1425, Buenos Aires, Argentina
| | - Marcela González-Cid
- Laboratorio de Mutagénesis, Instituto de Medicina Experimental, IMEX-CONICET, Academia Nacional de Medicina, J. A. Pacheco de Melo 3081, 1425, Buenos Aires, Argentina.
| |
Collapse
|
47
|
Cho MS, Lee JK, Bae KS, Han EA, Jang SJ, Ha WH, Lee SS, Barquinero JF, Kim WT. Retrospective biodosimetry using translocation frequency in a stable cell of occupationally exposed to ionizing radiation. JOURNAL OF RADIATION RESEARCH 2015; 56:709-16. [PMID: 25922373 PMCID: PMC4497401 DOI: 10.1093/jrr/rrv028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 03/10/2015] [Accepted: 03/30/2015] [Indexed: 05/18/2023]
Abstract
Two cases of hematological malignancies were reported in an industrial radiography company over a year, which were reasonably suspected of being consequences of prolonged exposure to ionizing radiation because of the higher incidence than expected in the general population. We analyzed chromosomal aberrations in the peripheral blood lymphocytes from the other workers who had been working under similar circumstances as the patients in the company. Among the subjects tested, 10 workers who belonged to the highest band were followed up periodically for 1.5 years since the first analysis. The aim of this study was to clarify pertinence of translocation analysis to an industrial set-up where chronic exposure was commonly expected. To be a useful tool for a retrospective biodosimetry, the aberrations need to be persistent for a decade or longer. Therefore we calculated the decline rates and half-lives of frequency for both a reciprocal translocation and a dicentric chromosome and compared them. In this study, while the frequency of reciprocal translocations was maintained at the initial level, dicentric chromosomes were decreased to 46.9% (31.0-76.5) of the initial frequency over the follow-up period. Our results support the long-term stability of reciprocal translocation through the cell cycle and validate the usefulness of translocation analysis as a retrospective biodosimetry for cases of occupational exposure.
Collapse
Affiliation(s)
- Min Su Cho
- Department of Emergency Medical Preparedness, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-gil, Nowon-gu, Seoul, South Korea Department of Surgery, Yonsei University Wonju College of Medicine, 162 Ilsan-dong, Wonju 220-701, Gangwon-do, South Korea
| | - Jin Kyung Lee
- Department of Emergency Medical Preparedness, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-gil, Nowon-gu, Seoul, South Korea Department of Biological Dosimetry, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-gil, Nowon-gu, Seoul, South Korea
| | - Keum Seok Bae
- Department of Surgery, Yonsei University Wonju College of Medicine, 162 Ilsan-dong, Wonju 220-701, Gangwon-do, South Korea
| | - Eun-Ae Han
- Department of Biological Dosimetry, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-gil, Nowon-gu, Seoul, South Korea
| | - Seong Jae Jang
- Department of Biological Dosimetry, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-gil, Nowon-gu, Seoul, South Korea
| | - Wi-Ho Ha
- Department of Health Physics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-gil, Nowon-gu, Seoul, South Korea
| | - Seung-Sook Lee
- Department of Emergency Medical Preparedness, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-gil, Nowon-gu, Seoul, South Korea
| | - Joan Francesc Barquinero
- Department of Animal Biology, Plant Biology and Ecology, Autonomous University of Barcelona, 08193 Bellaterra, Catalonia, Spain
| | - Wan Tae Kim
- Division of Radiation Regulation, Korea Institute of Nuclear Safety, 62 Gwahak-ro, Yuseong-gu, Daejeon 305-338, South Korea
| |
Collapse
|
48
|
Inhibition of Bcl-2 or IAP proteins does not provoke mutations in surviving cells. Mutat Res 2015; 777:23-32. [PMID: 25916945 DOI: 10.1016/j.mrfmmm.2015.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/31/2015] [Accepted: 04/07/2015] [Indexed: 11/21/2022]
Abstract
Chemotherapy and radiotherapy can cause permanent damage to the genomes of surviving cells, provoking severe side effects such as second malignancies in some cancer survivors. Drugs that mimic the activity of death ligands, or antagonise pro-survival proteins of the Bcl-2 or IAP families have yielded encouraging results in animal experiments and early phase clinical trials. Because these agents directly engage apoptosis pathways, rather than damaging DNA to indirectly provoke tumour cell death, we reasoned that they may offer another important advantage over conventional therapies: minimisation or elimination of side effects such as second cancers that result from mutation of surviving normal cells. Disappointingly, however, we previously found that concentrations of death receptor agonists like TRAIL that would be present in vivo in clinical settings provoked DNA damage in surviving cells. In this study, we used cell line model systems to investigate the mutagenic capacity of drugs from two other classes of direct apoptosis-inducing agents: the BH3-mimetic ABT-737 and the IAP antagonists LCL161 and AT-406. Encouragingly, our data suggest that IAP antagonists possess negligible genotoxic activity. Doses of ABT-737 that were required to damage DNA stimulated Bax/Bak-independent signalling and exceeded concentrations detected in the plasma of animals treated with this drug. These findings provide hope that cancer patients treated by BH3-mimetics or IAP antagonists may avoid mutation-related illnesses that afflict some cancer survivors treated with conventional DNA-damaging anti-cancer therapies.
Collapse
|
49
|
Singh ZN, Jethava Y, Post GR, Alapat D, Sawyer J, Waheed S, Nair B, Usmani SZ, Bailey C, Petty N, Van Rhee F, Barlogie B. Fulminant onset of acute leukemia from normal hematopoiesis within 3 months of follow up for multiple myeloma treated with total therapy protocols. Clin Case Rep 2015; 3:183-92. [PMID: 25838910 PMCID: PMC4377252 DOI: 10.1002/ccr3.180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 10/02/2014] [Accepted: 10/25/2014] [Indexed: 01/12/2023] Open
Abstract
Assiduous surveillance for genetic aberrations is necessary in patients on cytotoxic therapies to detect therapy-related myeloid neoplasms (t-MN). Current modalities include metaphase cytogenetics and FISH. Since t-MN may develop abruptly in cytogenetically normal patients, a discussion exploring additional methods such as SNP-array and targeted-deep-sequencing to detect subchromosomal abnormalities is needed.
Collapse
Affiliation(s)
- Zeba N Singh
- Department of Pathology, University of Arkansas for Medical Sciences 4301 W. Markham Street Slot 502, Little Rock, Arkansas, 72205
| | - Yogesh Jethava
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Ginell R Post
- Department of Pathology, University of Arkansas for Medical Sciences 4301 W. Markham Street Slot 502, Little Rock, Arkansas, 72205
| | - Daisy Alapat
- Department of Pathology, University of Arkansas for Medical Sciences 4301 W. Markham Street Slot 502, Little Rock, Arkansas, 72205
| | - Jeffrey Sawyer
- Department of Pathology, University of Arkansas for Medical Sciences 4301 W. Markham Street Slot 502, Little Rock, Arkansas, 72205
| | - Sarah Waheed
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Bijay Nair
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Saad Z Usmani
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Clyde Bailey
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Nathan Petty
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Frits Van Rhee
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| | - Bart Barlogie
- Myeloma Institute for Research and Therapy, University of Arkansas for Medical Sciences 4301 West Markham Slot 816, Little Rock, Arkansas, 72205
| |
Collapse
|
50
|
Insights into the application of let-7 family as promising biomarker in cancer screening. Tumour Biol 2015; 36:5233-9. [PMID: 25801240 DOI: 10.1007/s13277-015-3180-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/27/2015] [Indexed: 10/24/2022] Open
Abstract
Cancer is a leading cause of death worldwide with its low 5-year survival rate. Studies on the accuracy of let-7 family for human cancers have inconsistent conclusions, leading us to conduct this meta-analysis. This meta-analysis comprised of 11 studies from eight articles involving 805 cancer patients and 483 controls. The pooled parameters were as follows: sensitivity, 77 % (95 % confidence interval (CI) 73-81 %); specificity, 80 % (95 % CI 68-88 %); positive likelihood ratio (PLR), 3.8; negative likelihood ratio (NLR), 0.29; and diagnostic odds ratio (DOR) 13.0. In addition, we plotted the SROC and calculated the area under the curve (AUC) of 0.81 (95 % CI 0.78-0.84), which indicated a relatively high descriptive accuracy. In summary, our data suggested that let-7 family might be applied in noninvasive screening tests for human cancers, which needed to be validated in further large-scale studies.
Collapse
|