1
|
Nelles R, Tallack MR, Tate C, Hunt S, Aung H, Henden A, Jones L, Seymour L. p53 immunohistochemistry staining is a rapid screening method for TP53 mutation in myeloid malignancies suitable for integration into routine diagnostic laboratory practice. Pathology 2025; 57:489-494. [PMID: 40059001 DOI: 10.1016/j.pathol.2024.11.011] [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: 06/25/2024] [Revised: 11/02/2024] [Accepted: 11/13/2024] [Indexed: 05/06/2025]
Abstract
TP53 mutations are a recognised poor prognostic marker across myeloid malignancies associated with inferior overall survival. Immunohistochemistry (IHC) for p53 represents a promising adjunctive test with rapid turn-around; however, controversy exists around its utility and optimal positive staining threshold. The aims of this study were to determine the diagnostic testing characteristics and optimal threshold of positive staining for p53 IHC in comparison to next-generation sequencing (NGS) results across myeloid malignancies and compare haematopathologist review to digital analysis. A total of 117 bone marrow samples, including TP53 wild-type (n=50) and TP53 mutant (n=67) based on NGS results, were independently assessed by two blinded haematopathologists and analysed using image analysis software with reliability assessment. A receiver operating characteristic curve was used to determine the optimal cut-off for predicting TP53 mutation. There was high reliability between reviewers [intraclass correlation (ICC) 0.84; confidence interval (CI) 0.783-0.891] and between average reviewer and analysis software (ICC 0.794; CI 0.715-0.853). The area under the curve was similar (p=0.818) for computer versus average reviewer. The optimal cut-off for reviewer assessment was 2% strong positive staining with adequate sensitivity (70%) and specificity (90%). p53 IHC has adequate test characteristics to be considered as a rapid screen to identify cases of TP53 mutation. Issues remain in identifying truncating and some splicing mutations.
Collapse
Affiliation(s)
- Ricky Nelles
- Pathology Queensland, Royal Brisbane and Women's Hospital, Qld, Australia; Department of Cancer Care Services, Royal Brisbane and Women's Hospital, Brisbane, Qld, Australia.
| | - Michael R Tallack
- Pathology Queensland, Royal Brisbane and Women's Hospital, Qld, Australia
| | - Courtney Tate
- Pathology Queensland, Princess Alexandra Hospital, Brisbane, Qld, Australia; Department of Haematology, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - Stewart Hunt
- Department of Cancer Care Services, Royal Brisbane and Women's Hospital, Brisbane, Qld, Australia
| | - Hnin Aung
- Pathology Queensland, Royal Brisbane and Women's Hospital, Qld, Australia
| | - Andrea Henden
- Department of Cancer Care Services, Royal Brisbane and Women's Hospital, Brisbane, Qld, Australia; QIMR Berghofer Medical Research Institute, Herston, Qld, Australia; Faculty of Medicine, University of Queensland, St Lucia, Qld, Australia
| | - Lee Jones
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Louise Seymour
- Pathology Queensland, Royal Brisbane and Women's Hospital, Qld, Australia
| |
Collapse
|
2
|
Skuli S, Matthews A, Carroll M, Lai C. A line in shifting sand: Can we define and target TP53 mutated MDS? Semin Hematol 2024; 61:449-456. [PMID: 39542753 PMCID: PMC11960488 DOI: 10.1053/j.seminhematol.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/30/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024]
Abstract
Mutations in the tumor suppressor protein, TP53, lead to dismal outcomes in myeloid malignancies, including myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Recent pathological reclassifications have integrated TP53 mutated MDS and AML under a unified category of TP53 mutated myeloid neoplasms, which allows for more flexibility in treatment approaches. Therapeutic strategies have predominantly mirrored those for AML, with allogeneic stem cell transplantation emerging as critical for long-term disease control. The question remains whether there are physiological distinctions within TP53 mutated myeloid neoplasms that will significantly impact prognosis and therapeutic considerations. This review explores the unique aspects of classically defined "TP53 mutated MDS", focusing on its distinct biological characteristics and outcomes. Our current understanding is that TP53 mutated MDS and AML are globally quite similar, but as a group have unique features compared to TP53 wildtype (WT) disease. Optimizing immunotherapy and targeting vulnerabilities due to co-mutations and/or chromosome abnormalities should be the focus of future research.
Collapse
Affiliation(s)
- Sarah Skuli
- Division of Hematology and Oncology, Department of Medicine, The University of Pennsylvania, Philadelphia, PA
| | - Andrew Matthews
- Division of Hematology and Oncology, Department of Medicine, The University of Pennsylvania, Philadelphia, PA
| | - Martin Carroll
- Division of Hematology and Oncology, Department of Medicine, The University of Pennsylvania, Philadelphia, PA
| | - Catherine Lai
- Division of Hematology and Oncology, Department of Medicine, The University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
3
|
Ahmadi SE, Rahimian E, Rahimi S, Zarandi B, Bahraini M, Soleymani M, Safdari SM, Shabannezhad A, Jaafari N, Safa M. From regulation to deregulation of p53 in hematologic malignancies: implications for diagnosis, prognosis and therapy. Biomark Res 2024; 12:137. [PMID: 39538363 PMCID: PMC11565275 DOI: 10.1186/s40364-024-00676-9] [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: 07/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The p53 protein, encoded by the TP53 gene, serves as a critical tumor suppressor, playing a vital role in maintaining genomic stability and regulating cellular responses to stress. Dysregulation of p53 is frequently observed in hematological malignancies, significantly impacting disease progression and patient outcomes. This review aims to examine the regulatory mechanisms of p53, the implications of TP53 mutations in various hematological cancers, and emerging therapeutic strategies targeting p53. We conducted a comprehensive literature review to synthesize recent findings related to p53's multifaceted role in hematologic cancers, focusing on its regulatory pathways and therapeutic potential. TP53 mutations in hematological malignancies often lead to treatment resistance and poor prognosis. Current therapeutic strategies, including p53 reactivation and gene therapy, show promise in improving treatment outcomes. Understanding the intricacies of p53 regulation and the consequences of its mutations is essential for developing effective diagnostic and therapeutic strategies in hematological malignancies, ultimately enhancing patient care and survival.
Collapse
Affiliation(s)
- Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Rahimian
- Department of Medical Translational Oncology, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany
| | - Samira Rahimi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahman Zarandi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehran Bahraini
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maral Soleymani
- Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Mehrab Safdari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ashkan Shabannezhad
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Jaafari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Zhang L, Abro B, Campbell A, Ding Y. TP53 mutations in myeloid neoplasms: implications for accurate laboratory detection, diagnosis, and treatment. Lab Med 2024; 55:686-699. [PMID: 39001691 PMCID: PMC11532620 DOI: 10.1093/labmed/lmae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024] Open
Abstract
Genetic alterations that affect the function of p53 tumor suppressor have been extensively investigated in myeloid neoplasms, revealing their significant impact on disease progression, treatment response, and patient outcomes. The identification and characterization of TP53 mutations play pivotal roles in subclassifying myeloid neoplasms and guiding treatment decisions. Starting with the presentation of a typical case, this review highlights the complicated nature of genetic alterations involving TP53 and provides a comprehensive analysis of TP53 mutations and other alterations in myeloid neoplasms. Currently available methods used in clinical laboratories to identify TP53 mutations are discussed, focusing on the importance of establishing a robust testing protocol within clinical laboratories to ensure the delivery of accurate and reliable results. The treatment implications of TP53 mutations in myeloid neoplasms and clinical trial options are reviewed. Ultimately, we hope that this review provides valuable insights into the patterns of TP53 alterations in myeloid neoplasms and offers guidance to establish practical laboratory testing protocols to support the best practices of precision oncology.
Collapse
Affiliation(s)
- Linsheng Zhang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, US
| | - Brooj Abro
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, US
| | - Andrew Campbell
- Department of Laboratory Medicine, Geisinger Medical Center, Danville, PA, US
| | - Yi Ding
- Department of Laboratory Medicine, Geisinger Medical Center, Danville, PA, US
| |
Collapse
|
5
|
Bedekovics J, Madarász K, Mokánszki A, Molnár S, Mester Á, Miltényi Z, Méhes G. Exploring p53 protein expression and its link to TP53 mutation in myelodysplasia-related malignancies-Interpretive challenges and potential field of applications. Histopathology 2024; 85:143-154. [PMID: 38571438 DOI: 10.1111/his.15185] [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: 01/18/2024] [Revised: 02/28/2024] [Accepted: 03/16/2024] [Indexed: 04/05/2024]
Abstract
AIMS TP53 alterations have a significant prognostic effect in myeloid neoplasms. Our objective was to investigate the TP53 gene mutation status, p53 protein expression and their relationship in dysplasia-related myeloid neoplasms with varying levels of myeloblast counts. METHODS AND RESULTS A total of 76 bone marrow biopsy samples with different blast counts were analysed. Total and strong (3+) p53 expression was determined. Dual immunohistochemical staining was performed to determine the cell population associated with p53 expression. NGS analysis was performed using the Accel-Amplicon Comprehensive TP53 panel. Both p53 expression and TP53 VAF showed a significant correlation with the myeloblast ratio (P < 0.0001); however, p53 expression was also present in other cell lineages. The VAF value exhibited a significant correlation with p53 expression. A high specificity (0.9800) was observed for TP53 mutation using the ≥ 10% strong (3+) p53 cut-off value, although the sensitivity (0.4231) was low. CONCLUSIONS Strong (3+) p53 expression using a ≥ 10% cut-off value accurately predicts TP53 mutation but does not reveal the allelic state. The p53 expression is significantly influenced by myeloblast count, and histological interpretation should consider the presence of intermixed non-neoplastic marrow cells with varying physiological p53 expression.
Collapse
Affiliation(s)
- Judit Bedekovics
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Kristóf Madarász
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Mokánszki
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Sarolta Molnár
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ágnes Mester
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsófia Miltényi
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
6
|
Matthews A, Fenu E, Skuli SJ, Harris JC, Bagg A, Lai C. Immunohistochemistry-based detection of TP53 mutations in acute myeloid leukemia: a promising high sensitivity diagnostic approach. Leuk Lymphoma 2024; 65:1012-1015. [PMID: 38535517 DOI: 10.1080/10428194.2024.2332505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/06/2024] [Accepted: 03/13/2024] [Indexed: 05/16/2024]
Affiliation(s)
- Andrew Matthews
- Hematology Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Elena Fenu
- Pathology, Virginia Commonwealth University Medical Center, Richmond, VA, USA
| | - Sarah J Skuli
- Hematology Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jaryse C Harris
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Bagg
- Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Catherine Lai
- Hematology Oncology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
7
|
Yang G, Anderson Williams S, He F, He Y, McIntyre K, Beckman AK, Nelson AC, Yohe SL. Immunohistochemistry screening for TP53 mutation in myeloid neoplasms in AZF-fixed bone marrow biopsies. Pathology 2024; 56:404-412. [PMID: 38341302 DOI: 10.1016/j.pathol.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/18/2023] [Accepted: 11/01/2023] [Indexed: 02/12/2024]
Abstract
TP53 mutational status in myeloid neoplasms is prognostic and in acute myeloid leukaemia (AML) may lead to alternative induction therapy; therefore, rapid assessment is necessary for precision treatment. Assessment of multiple prognostic genes by next generation sequencing in AML is standard of care, but the turn-around time often cannot support rapid clinical decision making. Studies in haematological neoplasms suggest p53 immunohistochemistry (IHC) correlates with TP53 mutational status, but they have used variable criteria to define TP53 overexpression. p53 IHC was performed and interpreted on AZF-fixed, acid decalcified bone marrow biopsies on 47 cases of clonal myeloid neoplasms with TP53 mutations between 2016 and 2019 and 16 control samples. Results were scored by manual and digital analysis. Most TP53-mutated cases (81%) overexpressed p53 by digital analysis and manual analysis gave similar results. Among the nine TP53-mutated IHC-negative cases, seven (78%) were truncating mutations and two (22%) were single-hit missense mutations. Using a digital cut-off of at least 3% ≥1+ positive nuclei, the sensitivity and specificity are 81% and 100%; cases with loss-of-function mutations were more likely to be negative. In this cohort, p53 immunopositivity correlated with TP53 mutational status, especially missense mutations, with excellent specificity. Truncating TP53 mutations explain most IHC-negative cases, impacting the sensitivity. We demonstrate that p53 IHC can screen for TP53 mutations allowing quicker treatment decisions for most patients. However, not all patients will be identified, so molecular studies are required. Furthermore, cut-offs for positivity vary in the literature, consequently laboratories should independently validate their processes before adopting p53 IHC for clinical use. p53 IHC performs well to screen for TP53 mutations in AZF-fixed bone marrow. Performance in our setting differs from the literature, which shows variability of pre-analytic factors and cut-offs used to screen for TP53 mutations. Each laboratory should validate p53 IHC to screen for TP53 mutations in their unique setting.
Collapse
Affiliation(s)
- Guang Yang
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Sarah Anderson Williams
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Fiona He
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, MN, USA
| | - Yuyu He
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Kelsey McIntyre
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Amy K Beckman
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Andrew C Nelson
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Sophia L Yohe
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
8
|
Tuval A, Strandgren C, Heldin A, Palomar-Siles M, Wiman KG. Pharmacological reactivation of p53 in the era of precision anticancer medicine. Nat Rev Clin Oncol 2024; 21:106-120. [PMID: 38102383 DOI: 10.1038/s41571-023-00842-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2023] [Indexed: 12/17/2023]
Abstract
p53, which is encoded by the most frequently mutated gene in cancer, TP53, is an attractive target for novel cancer therapies. Despite major challenges associated with this approach, several compounds that either augment the activity of wild-type p53 or restore all, or some, of the wild-type functions to p53 mutants are currently being explored. In wild-type TP53 cancer cells, p53 function is often abrogated by overexpression of the negative regulator MDM2, and agents that disrupt p53-MDM2 binding can trigger a robust p53 response, albeit potentially with induction of p53 activity in non-malignant cells. In TP53-mutant cancer cells, compounds that promote the refolding of missense mutant p53 or the translational readthrough of nonsense mutant TP53 might elicit potent cell death. Some of these compounds have been, or are being, tested in clinical trials involving patients with various types of cancer. Nonetheless, no p53-targeting drug has so far been approved for clinical use. Advances in our understanding of p53 biology provide some clues as to the underlying reasons for the variable clinical activity of p53-restoring therapies seen thus far. In this Review, we discuss the intricate interactions between p53 and its cellular and microenvironmental contexts and factors that can influence p53's activity. We also propose several strategies for improving the clinical efficacy of these agents through the complex perspective of p53 functionality.
Collapse
Affiliation(s)
- Amos Tuval
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm, Sweden
| | | | - Angelos Heldin
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm, Sweden
| | | | - Klas G Wiman
- Karolinska Institutet, Department of Oncology-Pathology, Stockholm, Sweden.
| |
Collapse
|
9
|
Nahmod KA, Sasaki K, Ok CY, Loghavi S. Utility of p53 immunohistochemistry as a surrogate for sequencing in myeloid neoplasms: A tale of caution. Am J Hematol 2024; 99:329-330. [PMID: 37243496 DOI: 10.1002/ajh.26970] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/29/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023]
Affiliation(s)
- Karen A Nahmod
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chi Young Ok
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
10
|
Vose JM, Fu K, Wang L, Mansoor A, Stewart D, Cheng H, Smith L, Yuan J, Qureishi HN, Link BK, Cessna MH, Barr PM, Kahl BS, Mckinney MS, Khan N, Advani RH, Martin P, Goy AH, Phillips TJ, Mehta A, Kamdar M, Crump M, Pro B, Flowers CR, Jacobson CA, Smith SM, Stephens DM, Bachanova V, Jin Z, Wu S, Hernandez-Ilizaliturri F, Torka P, Anampa-Guzmán A, Kashef F, Li X, Sharma S, Greiner TC, Armitage JO, Lunning M, Weisenburger DD, Bociek RG, Iqbal J, Yu G, Bi C. Integrative analysis of clinicopathological features defines novel prognostic models for mantle cell lymphoma in the immunochemotherapy era: a report from The North American Mantle Cell Lymphoma Consortium. J Hematol Oncol 2023; 16:122. [PMID: 38104096 PMCID: PMC10725579 DOI: 10.1186/s13045-023-01520-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Patients with mantle cell lymphoma (MCL) exhibit a wide variation in clinical presentation and outcome. However, the commonly used prognostic models are outdated and inadequate to address the needs of the current multidisciplinary management of this disease. This study aims to investigate the clinical and pathological features of MCL in the immunochemotherapy era and improve the prognostic models for a more accurate prediction of patient outcomes. METHODS The North American Mantle Cell Lymphoma Project is a multi-institutional collaboration of 23 institutions across North America to evaluate and refine prognosticators for front-line therapy. A total of 586 MCL cases diagnosed between 2000 and 2012 are included in this study. A comprehensive retrospective analysis was performed on the clinicopathological features, treatment approaches, and outcomes of these cases. The establishment of novel prognostic models was based on in-depth examination of baseline parameters, and subsequent validation in an independent cohort of MCL cases. RESULTS In front-line strategies, the use of hematopoietic stem cell transplantation was the most significant parameter affecting outcomes, for both overall survival (OS, p < 0.0001) and progression-free survival (PFS, p < 0.0001). P53 positive expression was the most significant pathological parameter correlating with inferior outcomes (p < 0.0001 for OS and p = 0.0021 for PFS). Based on the baseline risk factor profile, we developed a set of prognostic models incorporating clinical, laboratory, and pathological parameters that are specifically tailored for various applications. These models, when tested in the validation cohort, exhibited strong predictive power for survival and showed a stratification resembling the training cohort. CONCLUSIONS The outcome of patients with MCL has markedly improved over the past two decades, and further enhancement is anticipated with the evolution of clinical management. The innovative prognostic models developed in this study would serve as a valuable tool to guide the selection of more suitable treatment strategies for patients with MCL.
Collapse
Affiliation(s)
- Julie M Vose
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Nebraska Medicine Fred and Pamela Buffett Cancer Center, 505 S 45Th St, Omaha, NE, 68105, USA
| | - Kai Fu
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lu Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai, China
| | - Adnan Mansoor
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Canada
| | - Douglas Stewart
- Departments of Oncology and Medicine, University of Calgary, Calgary, Canada
| | - Hongxia Cheng
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Lynette Smith
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ji Yuan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Hina Naushad Qureishi
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brian K Link
- Department of Internal Medicine, University of Iowa Hospitals & Clinics, Iowa City, Iowa, USA
| | - Melissa H Cessna
- Department of Pathology, Intermountain Medical Center, Murray, UT, USA
| | - Paul M Barr
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Brad S Kahl
- Department of Medicine, Oncology Division, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Matthew S Mckinney
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Nadia Khan
- Department of Hematology/Oncology, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Ranjana H Advani
- Division of Oncology, Stanford Cancer Institute, Stanford, CA, USA
| | - Peter Martin
- Division of Hematology and Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Andre H Goy
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, USA
| | - Tycel J Phillips
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Amitkumar Mehta
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Manali Kamdar
- Division of Hematology, University of Colorado, Denver, CO, USA
| | - Michael Crump
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre - University Health Network, Toronto, ON, Canada
| | - Barbara Pro
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Christopher R Flowers
- Division of Cancer Medicine, Department of Lymphoma-Myeloma, MD Anderson Cancer Center, Houston, TX, USA
| | - Caron A Jacobson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Sonali M Smith
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Deborah M Stephens
- Huntsman Cancer Institute at University of Utah, Salt Lake City, UT, USA
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Zhaohui Jin
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Shishou Wu
- Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Qingdao University, No.20 Yuhuangding East Road, Yantai, 264000, China
| | | | - Pallawi Torka
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea Anampa-Guzmán
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Farshid Kashef
- Department of Pathology, University at Buffalo, Buffalo, NY, USA
| | - Xing Li
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sunandini Sharma
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Timothy C Greiner
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - James O Armitage
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Nebraska Medicine Fred and Pamela Buffett Cancer Center, 505 S 45Th St, Omaha, NE, 68105, USA
| | - Matthew Lunning
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Nebraska Medicine Fred and Pamela Buffett Cancer Center, 505 S 45Th St, Omaha, NE, 68105, USA
| | - Dennis D Weisenburger
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Robert G Bociek
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Nebraska Medicine Fred and Pamela Buffett Cancer Center, 505 S 45Th St, Omaha, NE, 68105, USA
| | - Javeed Iqbal
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Guohua Yu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.
- Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Qingdao University, No.20 Yuhuangding East Road, Yantai, 264000, China.
| | - Chengfeng Bi
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Nebraska Medicine Fred and Pamela Buffett Cancer Center, 505 S 45Th St, Omaha, NE, 68105, USA.
| |
Collapse
|
11
|
Lee TD, Aisner DL, David MP, Eno CC, Gagan J, Gocke CD, Guseva NV, Haley L, Jajosky AN, Jones D, Mansukhani MM, Mroz P, Murray SS, Newsom KJ, Paulson V, Roy S, Rushton C, Segal JP, Senaratne TN, Siddon AJ, Starostik P, Van Ziffle JAG, Wu D, Xian RR, Yohe S, Kim AS. Current clinical practices and challenges in molecular testing: a GOAL Consortium Hematopathology Working Group report. Blood Adv 2023; 7:4599-4607. [PMID: 37236162 PMCID: PMC10425685 DOI: 10.1182/bloodadvances.2023010149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/02/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
While molecular testing of hematologic malignancies is now standard of care, there is variability in practice and testing capabilities between different academic laboratories, with common questions arising on how to best meet clinical expectations. A survey was sent to hematopathology subgroup members of the Genomics Organization for Academic Laboratories consortium to assess current and future practice and potentially establish a reference for peer institutions. Responses were received from 18 academic tertiary-care laboratories regarding next-generation sequencing (NGS) panel design, sequencing protocols and metrics, assay characteristics, laboratory operations, case reimbursement, and development plans. Differences in NGS panel size, use, and gene content were reported. Gene content for myeloid processes was reported to be generally excellent, while genes for lymphoid processes were less well covered. The turnaround time (TAT) for acute cases, including acute myeloid leukemia, was reported to range from 2 to 7 calendar days to 15 to 21 calendar days, with different approaches to achieving rapid TAT described. To help guide NGS panel design and standardize gene content, consensus gene lists based on current and future NGS panels in development were generated. Most survey respondents expected molecular testing at academic laboratories to continue to be viable in the future, with rapid TAT for acute cases likely to remain an important factor. Molecular testing reimbursement was reported to be a major concern. The results of this survey and subsequent discussions improve the shared understanding of differences in testing practices for hematologic malignancies between institutions and will help provide a more consistent level of patient care.
Collapse
Affiliation(s)
- Thomas D. Lee
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA
| | - Dara L. Aisner
- Department of Pathology, University of Colorado, Aurora, CO
| | - Marjorie P. David
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Celeste C. Eno
- Department of Pathology and Lab Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Jeffrey Gagan
- Department of Pathology, University of Texas Southwestern, Dallas, TX
| | - Christopher D. Gocke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Lisa Haley
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Audrey N. Jajosky
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY
| | - Daniel Jones
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Mahesh M. Mansukhani
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY
| | - Pawel Mroz
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Sarah S. Murray
- Department of Pathology, University of California San Diego, La Jolla, CA
| | - Kimberly J. Newsom
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Vera Paulson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Somak Roy
- Department of Pathology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Chase Rushton
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA
| | | | - T. Niroshini Senaratne
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA
| | - Alexa J. Siddon
- Departments of Laboratory Medicine & Pathology, Yale School of Medicine, New Haven, CT
| | - Petr Starostik
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL
| | | | - David Wu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| | - Rena R. Xian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sophia Yohe
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Annette S. Kim
- Department of Pathology, Brigham and Women’s Hospital/Harvard Medical School, Boston, MA
| |
Collapse
|
12
|
Rezvani A, Monabati A, Kargar Z, Safaei A, Mahmoodzadeh M, Moosapour H, Hosseini M, Taheri A, Kheiri S, Taheri E. P53 IHC Result as a Prognostic Tool in MDS. IRANIAN JOURNAL OF PATHOLOGY 2023; 18:327-334. [PMID: 37942201 PMCID: PMC10628383 DOI: 10.30699/ijp.2023.1971023.2991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/13/2023] [Indexed: 11/10/2023]
Abstract
Background & Objective Some of the patients with myelodysplastic syndrome (MDS) are categorized as good prognosis based on the Revised International Prognostic Scoring System (IPSS-R). However, these patients may have poor clinical outcomes. It seems that the current diagnostic tools and IPSS-R cannot consider genetic factors for determining the prognosis of MDS patients. Methods This cross-sectional study included all adult MDS patients of both genders who were admitted from March 2015 to March 2020 to the Hematology wards of two educational tertiary hospitals in Iran (Namazi and Faghihi, affiliated with Shiraz University of medical sciences). Study data included relevant retrospective data from medical records and the results of immunohistochemical p53 staining on bone marrow biopsies. Results Of the 84 patients, 65 (77.4%) showed p53 expression in bone marrow. They had shorter median survival than those without p53 expression. Considering both variables of P53 IHC results and IPSS-R score, the patients who died with low-risk IPSS-R score presented high p53 expression. Conclusion This study shows that the investigation of p53 expression by IHC at the time of diagnosis is a valuable indicator of survival rate in MDS patients. These data suggest that the immunohistochemical analysis of p53 can be a prognostic tool for MDS and should be used as an adjunct test to make decisions on the best therapeutic choice.
Collapse
Affiliation(s)
- Alireza Rezvani
- Department of Hematology, Medical Oncology and Stem Cell Transplantation, Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Monabati
- Department of Pathology, Molecular Pathology and Cytogenetic Ward, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Kargar
- Molecular Pathology and Cytogenetic Ward, Pathology Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Akbar Safaei
- Department of Molecular Pathology & Cytogenetics, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahdi Mahmoodzadeh
- Department of Hematology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamideh Moosapour
- Evidence-Based Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Hosseini
- Molecular Pathology and Cytogenetic Ward, Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abdolmajid Taheri
- Department of Radiology, School of Medicine, Hajar Hospital, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Soleiman Kheiri
- Department of Epidemiology and Biostatistics, School of Health, Modeling in Health Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Elham Taheri
- Molecular Pathology and Cytogenetic Ward, Pathology Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
13
|
Marks JA, Wang X, Fenu EM, Bagg A, Lai C. TP53 in AML and MDS: The new (old) kid on the block. Blood Rev 2023; 60:101055. [PMID: 36841672 DOI: 10.1016/j.blre.2023.101055] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023]
Abstract
MDS and AML are clonal hematopoietic stem cell disorders of increasing incidence, having a variable prognosis based, among others, on co-occurring molecular abnormalities. TP53 mutations are frequently detected in these myeloid neoplasms and portend a poor prognosis with known therapeutic resistance. This article provides a timely review of the complexity of TP53 alterations, providing updates in diagnosis and prognosis based on new 2022 International Consensus Classification (ICC) and World Health Organization (WHO) guidelines. The article addresses optimal testing strategies and reviews current and arising therapeutic approaches. While the treatment landscape for this molecular subgroup is under active development, further exploration is needed to optimize the care of this group of patients with unmet needs.
Collapse
Affiliation(s)
- Jennifer A Marks
- Department of Medicine, Division of Hematology and Oncology, Georgetown University, 3800 Reservoir Road NW, Washington, D.C. 20007, USA.
| | - Xin Wang
- Department of Medicine, Division of Hematology and Oncology, Georgetown University, 3800 Reservoir Road NW, Washington, D.C. 20007, USA; Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, 12 South Pavilion, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | - Elena M Fenu
- Department of Pathology and Laboratory Medicine, Division of Hematopathology, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, Division of Hematopathology, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| | - Catherine Lai
- Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania, 12 South Pavilion, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| |
Collapse
|
14
|
Barone P, Patel S. Myelodysplastic syndrome: Approach to diagnosis in the era of personalized medicine. Semin Diagn Pathol 2023; 40:172-181. [PMID: 37121781 DOI: 10.1053/j.semdp.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 05/02/2023]
Affiliation(s)
- Paul Barone
- NewYork-Presbyterian Hospital, Weill Cornell Campus, United States of America.
| | - Sanjay Patel
- Weill Cornell Medicine, United States of America
| |
Collapse
|
15
|
Reilly CR, Shimamura A. Predisposition to myeloid malignancies in Shwachman-Diamond syndrome: biological insights and clinical advances. Blood 2023; 141:1513-1523. [PMID: 36542827 PMCID: PMC10082379 DOI: 10.1182/blood.2022017739] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Shwachman-Diamond syndrome (SDS) is an inherited multisystem ribosomopathy characterized by exocrine pancreatic deficiency, bone marrow failure, and predisposition to myeloid malignancies. The pathobiology of SDS results from impaired ribosomal maturation due to the deficiency of SBDS and the inability to evict the antiassociation factor eIF6 from the 60S ribosomal subunit. Clinical outcomes for patients with SDS who develop myeloid malignancies are extremely poor because of high treatment-related toxicities and a high rate of refractory disease/relapse even after allogeneic hematopoietic stem cell transplant (HSCT). Registry data indicate that outcomes are improved for patients with SDS who undergo routine bone marrow surveillance and receive an HSCT before developing an overt malignancy. However, the optimal approach to hematologic surveillance and the timing of HSCT for patients with SDS is not clearly established. Recent studies have elucidated distinct patterns of somatic blood mutations in patients with SDS that either alleviate the ribosome defect via somatic rescue (heterozygous EIF6 inactivation) or disrupt cellular checkpoints, resulting in increased leukemogenic potential (heterozygous TP53 inactivation). Genomic analysis revealed that most myeloid malignancies in patients with SDS have biallelic loss-of-function TP53 mutations. Single-cell DNA sequencing of SDS bone marrow samples can detect premalignant biallelic TP53-mutated clones before clinical diagnosis, suggesting that molecular surveillance may enhance the detection of incipient myeloid malignancies when HSCT may be most effective. Here, we review the clinical, genetic, and biologic features of SDS. In addition, we present evidence supporting the hematologic surveillance for patients with SDS that incorporates clinical, pathologic, and molecular data to risk stratify patients and prioritize transplant evaluation for patients with SDS with high-risk features.
Collapse
Affiliation(s)
- Christopher R. Reilly
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Akiko Shimamura
- Department of Pediatric Hematology/Oncology, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, MA
| |
Collapse
|
16
|
Nabil R, Elshazly SS, Hassan NM, Nooh HA. The expression level of ARF and p53 in AML patients, and their relation to patients' outcome. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2023. [DOI: 10.1186/s43042-023-00410-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
Abstract
Background
Acute myeloid leukemia (AML) is a cancer of hematopoietic progenitors characterized by gene mutations. The most popular deregulations are mutation and altered expression in the p53 gene, which is considered the guardian of the genome. Its activity is controlled by regulatory genes, e.g., alternate open reading frame (ARF), whose defects could affect p53 activity.
Aim
To study the effect of altered expression of p53 and ARF genes in de novo AML patients and correlate the results to the patients’ characteristics and outcomes.
Methods
Expression levels of p53 and ARF were assessed in 96 AML adult patients compared to 20 healthy controls using quantitative reverse-transcription PCR (RT-qPCR).
Results
There was significant up-regulation of p53 [77.6 (3.8–9528.3)] compared to controls [1.031 (0.210–9.051)], p < 0.001]. The expression level of ARF was significantly upregulated [6.2 (0.5–964.0)] compared to controls [0.854 (0.357–2.519), p < 0.001]. All of the low ARF expressers had low p53 overexpression, 61.1% of patients with high ARF expression had high p53 over-expression, and 38.9% with high ARF expression had low p53 over-expression (p < 0.001). ARF expression shows a trend of association with FLT3 mutation, as 89.3% with FLT3 mutation have high ARF expression (p = 0.080). Low p53 over-expression was seen in 77% of APL patients, while high p53 expression was associated with non-APL (p = 0.040). The median DFS of mutant NPM1 patients was higher than wild NPM1 (46.15 vs. 5.89 days, p = 0.045). Patients aged ≤ 50 years had better OS and DFS than those > 50 (p = 0.05, p = 0.035, respectively).There were no significant statistical associations between DFS and p53, ARF, and FLT3 mutations.
Conclusion
The p53 and ARF genes are overexpressed in de novo AML patients and they are interrelated. low p53 overexpression is associated with APL phenotype and t(15;17) and patients with t(15;17) had slightly better survival than patients with negative t(15;17) (p = 0.061). AML patients with mutated NPM1 had better DFS than wild NPM1 (p = 0.045). p53 pathway regulation can occur by many alternative ways rather than gene mutation.
Collapse
|
17
|
Shallis RM, Daver NG, Altman JK, Hasserjian RP, Kantarjian H, Platzbecker U, Santini V, Wei AH, Sallman DA, Zeidan AM. TP53-altered acute myeloid leukemia and myelodysplastic syndrome with excess blasts should be approached as a single entity. Cancer 2023; 129:175-180. [PMID: 36397669 PMCID: PMC11938810 DOI: 10.1002/cncr.34535] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/28/2022] [Accepted: 09/19/2022] [Indexed: 11/19/2022]
Abstract
TP53-altered myelodysplastic syndrome with excess blasts and TP53-altered acute myeloid leukemia should be considered under one unifying classification term for their study in clinical trials. Ultimately, such a unification would simplify the screening processes for clinical trials and allow a focus on treating the patient for a genetically defined disorder rather than one based on an arbitrary blast threshold.
Collapse
Affiliation(s)
- Rory M. Shallis
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, Yale University, New Haven, CT, USA
| | - Naval G. Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jessica K. Altman
- Division of Hematology and Oncology, Robert H. Lurie Comprehensive Cancer, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Robert P. Hasserjian
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Uwe Platzbecker
- Department of Hematology and Cellular Therapy, Medical Clinic and Policlinic I, Leipzig University Hospital, Leipzig, Germany
| | - Valeria Santini
- MDS Unit, AOU Careggi, University of Florence, Florence, Italy
| | - Andrew H. Wei
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital, Walter and Eliza Hall, Institute of Medical Research and the University of Melbourne, Melbourne, Australia
| | - David A. Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Amer M. Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
18
|
Rogers KJ, Abukhiran IM, Syrbu S, Tomasson M, Bates M, Dhakal P, Bhagavathi S. Utilizing digital pathology and immunohistochemistry of p53 as an adjunct to molecular testing in myeloid disorders. Acad Pathol 2023; 10:100064. [PMID: 36970330 PMCID: PMC10031312 DOI: 10.1016/j.acpath.2022.100064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/06/2022] [Accepted: 11/26/2022] [Indexed: 02/04/2023] Open
Abstract
TP53 mutation status guides early therapeutic decisions in the treatment of clonal myeloid disorders and serves as a simple means of monitoring response to treatment. We aim here to develop a standardized protocol for evaluating TP53 mutation status in myeloid disorders using immunohistochemistry assisted by digital image analysis and further compare this approach to manual interpretation alone. To accomplish this, we obtained 118 bone marrow biopsies from patients with hematologic malignancy and molecular testing for mutations associated with acute myeloid leukemia was performed. Clot or core biopsy slides were stained for p53 and digitally scanned. Overall mutation burden was assessed digitally using two different metrics to determine positivity, compared to the results of manual review, and correlated with molecular results. Using this approach, we found that digital analysis of immunohistochemistry stained slides performed worse than manual categorization alone in predicting TP53 mutation status in our cohort (PPV 91%, NPV 100% vs. PPV 100%, NPV 98%). While digital analysis reduced inter- and intraobserver variability when assessing mutation burden, there was poor correlation between the quantity and intensity of p53 staining and molecular analysis (R2 = 0.204). Therefore, digital image analysis of p53 immunohistochemistry accurately predicts TP53 mutation status as confirmed by molecular testing but does not offer a significant advantage over manual categorization alone. However, this approach offers a highly standardized methodology for monitoring disease status or response to treatment once a diagnosis has been made.
Collapse
Affiliation(s)
- Kai J. Rogers
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Ibrahim M. Abukhiran
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Sergei Syrbu
- Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Michael Tomasson
- Division of Hematology/Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Melissa Bates
- Division of Hematology/Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Prajwal Dhakal
- Division of Hematology/Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | | |
Collapse
|
19
|
Fitzpatrick MJ, Boiocchi L, Fathi AT, Brunner AM, Hasserjian RP, Nardi V. Correlation of p53 immunohistochemistry with
TP53
mutational status and overall survival in newly diagnosed acute myeloid leukemia. Histopathology 2022; 81:496-510. [DOI: 10.1111/his.14726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/28/2022] [Accepted: 07/10/2022] [Indexed: 11/26/2022]
Affiliation(s)
| | - Leonardo Boiocchi
- Department of Pathology and Laboratory Medicine Memorial Sloan Kettering Cancer Center New York NY USA
| | - Amir T. Fathi
- Department of Hematology/Oncology Massachusetts General Hospital Boston MA USA
| | - Andrew M. Brunner
- Department of Hematology/Oncology Massachusetts General Hospital Boston MA USA
| | | | - Valentina Nardi
- Department of Pathology Massachusetts General Hospital Boston MA USA
| |
Collapse
|
20
|
Tashakori M, Kadia T, Loghavi S, Daver N, Kanagal-Shamanna R, Pierce S, Sui D, Wei P, Khodakarami F, Tang Z, Routbort M, Bivins CA, Jabbour EJ, Medeiros LJ, Bhalla K, Kantarjian HM, Ravandi F, Khoury JD. TP53 copy number and protein expression inform mutation status across risk categories in acute myeloid leukemia. Blood 2022; 140:58-72. [PMID: 35390143 PMCID: PMC9346958 DOI: 10.1182/blood.2021013983] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 03/28/2022] [Indexed: 11/20/2022] Open
Abstract
Mutant TP53 is an adverse risk factor in acute myeloid leukemia (AML), but large-scale integrated genomic-proteomic analyses of TP53 alterations in patients with AML remain limited. We analyzed TP53 mutational status, copy number (CN), and protein expression data in AML (N = 528) and provide a compilation of mutation sites and types across disease subgroups among treated and untreated patients. Our analysis shows differential hotspots in subsets of AML and uncovers novel pathogenic variants involving TP53 splice sites. In addition, we identified TP53 CN loss in 70.2% of TP53-mutated AML cases, which have more deleterious TP53 mutations, as well as copy neutral loss of heterozygosity in 5/32 (15.6%) AML patients who had intact TP53 CN. Importantly, we demonstrate that mutant p53 protein expression patterns by immunohistochemistry evaluated using digital image-assisted analysis provide a robust readout that integrates TP53 mutation and allelic states in patients with AML. Expression of p53 by immunohistochemistry informed mutation status irrespective of TP53 CN status. Genomic analysis of comutations in TP53-mutant AML shows a muted landscape encompassing primarily mutations in genes involved in epigenetic regulation (DNMT3A and TET2), RAS/MAPK signaling (NF1, KRAS/NRAS, PTPN11), and RNA splicing (SRSF2). In summary, our data provide a rationale to refine risk stratification of patients with AML on the basis of integrated molecular and protein-level TP53 analyses.
Collapse
Affiliation(s)
- Mehrnoosh Tashakori
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | | | - Sanam Loghavi
- Department of Hematopathology, 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
| | | | - Dawen Sui
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX; and
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX; and
| | | | - Zhenya Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mark Routbort
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | - Joseph D Khoury
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
21
|
Santini V, Lübbert M, Wierzbowska A, Ossenkoppele GJ. The Clinical Value of Decitabine Monotherapy in Patients with Acute Myeloid Leukemia. Adv Ther 2022; 39:1474-1488. [PMID: 34786648 PMCID: PMC8989816 DOI: 10.1007/s12325-021-01948-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/06/2021] [Indexed: 12/17/2022]
Abstract
Decitabine (5-aza-2′-deoxycytidine) is a hypomethylating agent used in the treatment of acute myeloid leukemia (AML). Decitabine inhibits DNA methyltransferases, causing DNA hypomethylation, and leading amongst others to re-expression of silenced tumor suppressor genes. Decitabine is indicated for the treatment of adult patients with newly diagnosed de novo or secondary AML who are not eligible for standard induction chemotherapy. The initial authorization in 2012 was based on the results of the open-label, randomized, multicenter phase 3 DACO-016 trial, and supported by data from the supportive phase 2 open-label DACO-017 trial. Compared with standard care, decitabine significantly improved overall survival, event-free survival, progression-free survival, and response rate. Decitabine was generally well tolerated, offering a valuable treatment option in patients with AML irrespective of age, especially for patients achieving a complete response. Several observational “real-life” studies confirmed these results. In contrast to standard chemotherapy, the presence of adverse-risk karyotypes or TP53 mutations does not negatively impact sensitivity to hypomethylating therapy albeit with lower durability. Data suggest a potential positive effect of decitabine in patients with monosomal karyotype-positive AML. For the time being, decitabine is an appropriate option as monotherapy for patients with AML who are unfit to receive more intensive combination therapies, but emerging data suggest that decitabine-based doublet or triplet combinations may be future treatment options for patients with AML.
Collapse
Affiliation(s)
- Valeria Santini
- Department of Experimental and Clinical Medicine, MDS Unit, AOUC- University of Florence, Florence, Italy.
| | - Michael Lübbert
- Department of Internal Medicine I, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg Medical Center, Freiburg, Germany
| | | | - Gert J Ossenkoppele
- Department of Haematology, Location VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Ferraro F, Gruszczynska A, Ruzinova MB, Miller CA, Percival ME, Uy GL, Pusic I, Jacoby MA, Christopher MJ, Kim MY, Westervelt P, Cashen AF, Schroeder MA, DiPersio JF, Abboud CN, Wartman LD, Gao F, Link DC, Ley TJ, Welch JS. Decitabine salvage for TP53-mutated, relapsed/refractory acute myeloid leukemia after cytotoxic induction therapy. Haematologica 2022; 107:1709-1713. [PMID: 35236053 PMCID: PMC9244807 DOI: 10.3324/haematol.2021.280153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Indexed: 01/21/2023] Open
Affiliation(s)
- Francesca Ferraro
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Agata Gruszczynska
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Marianna B. Ruzinova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Christopher A. Miller
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Mary Elizabeth Percival
- Department of Internal Medicine, Division of Hematology, University of Washington School of Medicine, Seattle, WA,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Geoffrey L. Uy
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Iskra Pusic
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Meagan A. Jacoby
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Mathew J. Christopher
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Miriam Y. Kim
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Peter Westervelt
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Amanda F. Cashen
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Mark A. Schroeder
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - John F. DiPersio
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Camille N. Abboud
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Lukas D. Wartman
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Feng Gao
- Department of Surgery, Division of Public Health Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel C. Link
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Timothy J. Ley
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - John S. Welch
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, MO,John S. Welch -
| |
Collapse
|
23
|
Gao L, Harbaugh B, Parr K, Patel P, Golem S, Zhang D, Woodroof J, Cui W. MYC Expression Is Associated With p53 Expression and TP53 Aberration and Predicts Poor Overall Survival in Acute Lymphoblastic Leukemia/Lymphoma. Am J Clin Pathol 2022; 157:119-129. [PMID: 34528662 DOI: 10.1093/ajcp/aqab105] [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: 01/30/2021] [Accepted: 05/13/2021] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES We evaluated MYC and p53 expression, TP53 aberration, their relationship, and their impact on overall survival (OS) in acute lymphoblastic leukemia (ALL)/lymphoblastic lymphoma (LBL). METHODS We identified 173 patients with ALL and LBL, including 12 cases of mixed-phenotype acute leukemia, 8 cases of therapy-related B-cell ALL (B-ALL), 119 cases of B-ALL, and 34 cases of T-cell ALL/LBL diagnosed from 2003 to 2019. We retrospectively assessed p53 and MYC expression by immunohistochemistry of bone marrow and correlated MYC expression with p53 expression and TP53 aberration. RESULTS Expression of p53 and MYC was present in 11.5% and 27.7% of ALL/LBL cases (n = 20 and n = 48), respectively. MYC expression was significantly correlated with p53 expression and TP53 aberration (P = .002 and P = .03), and p53 expression and MYC expression had an adverse impact on OS in patients with ALL/LBL (P < .05). MYC and p53 dual expression as well as combined MYC expression and TP53 aberration had a negative impact on OS in patients with ALL/LBL. CONCLUSIONS MYC expression is correlated with p53 overexpression, TP53 aberration, and poor OS in patients with ALL/LBL.
Collapse
Affiliation(s)
- Linlin Gao
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - Brent Harbaugh
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - Kevin Parr
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - Payal Patel
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - Shivani Golem
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - Da Zhang
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - Janet Woodroof
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - Wei Cui
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| |
Collapse
|
24
|
What Are the Prospects for Treating TP53 Mutated Myelodysplastic Syndromes and Acute Myeloid Leukemia? Cancer J 2022; 28:51-61. [DOI: 10.1097/ppo.0000000000000569] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
25
|
Pescia C, Boggio F, Croci GA, Cassin R, Barella M, Pettine L, Reda G, Sabattini E, Finelli C, Gianelli U. Lymphocytic Infiltrate and p53 Protein Expression as Predictive Markers of Response and Outcome in Myelodysplastic Syndromes Treated with Azacitidine. J Clin Med 2021; 10:jcm10214809. [PMID: 34768330 PMCID: PMC8584458 DOI: 10.3390/jcm10214809] [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: 09/20/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 11/16/2022] Open
Abstract
High-risk Myelodysplastic syndromes (MDS) represent therapeutical challenges and are usually managed with hypomethylating agents such as azacitidine. Given the lack of data in the literature concerning azacitidine effects on bone marrow, we retrospectively analyzed 57 high-risk MDS cases in order to identify any changes induced by azacitidine therapy or relevant correlations between therapy response and pre- or post-treatment features. Azacitidine treatment had no significant impact on bone marrow cellularity or morphological dysplastic features. On the contrary, although not statistically significant, we observed a slight decrease in CD34+ and CD117+ blasts and p53+ precursors after treatment. Moreover, pre-treatment IPSS-R cytogenetic score (p = 0.004), lymphocytic infiltrate (p = 0.017) and p53+ elements (p = 0.001) correlated with AML progression; pre-treatment lymphocytic infiltrate was also linked to better response to therapy (p = 0.004), suggesting an anti-tumoral role of bone marrow microenvironment. Post-treatment blast count impacted negatively on overall survival (p = 0.035) and risk of leukemic progression (p = 0.04), while both post-treatment lymphocytic infiltrate and p53+ elements showed significant correlation with treatment response (p = 0.004 and p = 0.003 respectively). Higher post-treatment p53+ elements correlated also with risk of leukemic progression (p = 0.013). Our results suggest the possible role of lymphocytic infiltrate and p53+ elements as predictive markers in MDS treated with azacitidine, disclosing new chapters in the understanding of MDS evolution and treatment.
Collapse
Affiliation(s)
- Carlo Pescia
- Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (C.P.); (F.B.); (G.A.C.); (M.B.)
- Department of Pathophysiology and Transplantation, Università degli studi di Milano, 20122 Milan, Italy
| | - Francesca Boggio
- Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (C.P.); (F.B.); (G.A.C.); (M.B.)
| | - Giorgio Alberto Croci
- Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (C.P.); (F.B.); (G.A.C.); (M.B.)
- Department of Pathophysiology and Transplantation, Università degli studi di Milano, 20122 Milan, Italy
| | - Ramona Cassin
- Division of Hematology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.C.); (L.P.); (G.R.)
| | - Marco Barella
- Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (C.P.); (F.B.); (G.A.C.); (M.B.)
- Department of Pathophysiology and Transplantation, Università degli studi di Milano, 20122 Milan, Italy
| | - Loredana Pettine
- Division of Hematology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.C.); (L.P.); (G.R.)
| | - Gianluigi Reda
- Division of Hematology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (R.C.); (L.P.); (G.R.)
| | - Elena Sabattini
- Hematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Carlo Finelli
- Division of Hematology “Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Umberto Gianelli
- Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (C.P.); (F.B.); (G.A.C.); (M.B.)
- Department of Pathophysiology and Transplantation, Università degli studi di Milano, 20122 Milan, Italy
- Correspondence: ; Tel.: +39-02-5032-0562
| |
Collapse
|
26
|
Aladily TN, Obiedat S, Bustami N, Alhesa A, Altantawi AM, Khader M, Mansour AT. Combined utility of CD177, P53, CD105 and c-kit immunohistochemical stains improves the detection of myelodysplastic syndrome. Ann Diagn Pathol 2021; 55:151810. [PMID: 34482217 DOI: 10.1016/j.anndiagpath.2021.151810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/01/2021] [Accepted: 08/17/2021] [Indexed: 11/28/2022]
Abstract
The diagnosis of myelodysplastic syndrome (MDS) relies primarily on identifying peripheral blood cytopenia and morphologic dysplasia as well as detecting cytogenetic aberrations in a subset of patients. Accumulating data points to the importance of examining certain immunophenotypic changes characteristic of MDS, most of which are tested by flow cytometry. The role of immunohistochemistry in the diagnostic workup of MDS is less known. In this study, we used immunohistochemistry to survey the expression patterns of CD177, P53, CD105 and c- kit in a cohort of MDS bone marrow specimens (n = 57) and compared the results with a control group of patients who had cytopenia for other benign conditions (n = 49). MDS cases showed significant higher rates of: CD177-loss (13/57, 23% vs 1/49, 2%; P = .0016), P53 overexpression (8/57, 14% vs none; P = .005) and the presence of clusters of CD105-positive cells (6/57, 11% vs none; P = .021). Increased c-kit-positive cells was more common in MDS patients, but not statistically significant (17/57, 30% vs 8/49, 16%; P = .102). On multivariate analysis, only loss of CD177 expression was significantly higher in MDS group (P = .014). These findings suggest that a panel of immunohistochemical stains could serve as an adjunct tool in investigating unexplained cytopenias and warrant further comparative studies with flow cytometry.
Collapse
Affiliation(s)
- Tariq N Aladily
- Department of Hematopathology, The University of Jordan, Amman 11910, Jordan.
| | - Sara Obiedat
- Department of Hematopathology, The University of Jordan, Amman 11910, Jordan
| | - Nadwa Bustami
- Department of Hematopathology, The University of Jordan, Amman 11910, Jordan
| | - Ahmad Alhesa
- Department of Hematopathology, The University of Jordan, Amman 11910, Jordan
| | - Ahmad M Altantawi
- Department of Hematopathology, The University of Jordan, Amman 11910, Jordan
| | - Majd Khader
- Department of Hematopathology, The University of Jordan, Amman 11910, Jordan
| | - Ahmad T Mansour
- Department of Hematopathology, The University of Jordan, Amman 11910, Jordan; Department of Pathology and Laboratory Medicine, University of Cincinnati, OH 45220, USA.
| |
Collapse
|
27
|
Oliva EN, Latagliata R, Sabattini E, Mammì C, Cuzzola M, D’Errigo MG, Cannatà MC, Bova I, Capodanno I, Palumbo GA, Pane F, Reda G, Fianchi L, Riva M, Poloni A. Accuracy of bone marrow histochemical TP53 expression compared to the detection of TP53 somatic mutations in patients with myelodysplastic syndromes harbouring a del5q cytogenetic abnormality. AMERICAN JOURNAL OF BLOOD RESEARCH 2021; 11:417-426. [PMID: 34540351 PMCID: PMC8446828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/18/2021] [Indexed: 06/13/2023]
Abstract
TP53 gene mutations are common in Myelodysplastic Syndromes (MDS) with del5q and have a clinical and prognostic significance. Next Generation Sequencing (NGS) is an accurate, but expensive, technique, and not commonly available. Immunohistochemistry (IHC) for TP53 expression has been recently used as a surrogate to assess TP53 mutations. To compare the concordance between TP53 expression in IHC and TP53 mutations by NGS, 30 cases with MDS harbouring a del5q abnormality were evaluated. Overall, 10/30 patients (33.3%) had TP53 mutations by NGS, while 16/29 (55.1%) had TP53 overexpression in IHC. TP53 expression by IHC had a 70% sensitivity to identify patients with TP53 mutation by NGS, but its specificity was low (52.6%, kappa = 0.198; P = 0.24). In addition, ROC curve analyses showed that the overall diagnostic value (accuracy) of TP53 expression in IHC to identify patients with TP53 mutation by NGS was 68% in the whole study sample and 67% in patients with isolated del5q-. In both cases, the areas under the curves did not attain the statistical significance (P = 0.11 and P = 0.29, respectively). Based on the ROC curve, the cut-off of 2.3% TP53 expression in IHC was shown to be the best cut-off to identify TP53 mutations: using this cut-off, the agreement between TP53 expression and TP53 mutation by NGS reached statistical significance (kappa = 0.42; P = 0.023). In conclusion, the agreement between TP53 expression in IHC and TP53 mutation analysis by NGS is rather unsatisfactory in MDS patients with del5q at the standard cut-off. Thus, the IHC technique cannot be considered a valid alternative to NGS evaluation of TP53 mutational status in these patients.
Collapse
Affiliation(s)
- Esther N Oliva
- U.O. Ematologia-Grande Ospedale Metropolitano Bianchi Melacrino Morelli Reggio CalabriaItaly
| | | | - Elena Sabattini
- U.O. di Emolinfopatologia-Policlinico S. Orsola Malpighi BolognaItaly
| | - Corrado Mammì
- U.O.S.D. Genetica Medica-Grande Ospedale Metropolitano Bianchi Melacrino Morelli Reggio CalabriaItaly
| | - Maria Cuzzola
- U.O.C. di Microbiologia e Virologia-Grande Ospedale Metropolitano Bianchi Melacrino Morelli Reggio CalabriaItaly
| | - Maria Grazia D’Errigo
- U.O.S.D. Genetica Medica-Grande Ospedale Metropolitano Bianchi Melacrino Morelli Reggio CalabriaItaly
| | - Maria Concetta Cannatà
- U.O.S.D. Genetica Medica-Grande Ospedale Metropolitano Bianchi Melacrino Morelli Reggio CalabriaItaly
| | - Irene Bova
- U.O.S.D. Genetica Medica-Grande Ospedale Metropolitano Bianchi Melacrino Morelli Reggio CalabriaItaly
| | | | | | - Fabrizio Pane
- U.O. Ematologia Oncologica-A.O.U. Federico II di NapoliItaly
| | - Gianluigi Reda
- U.O. Ematologia-Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di MilanoItaly
| | - Luana Fianchi
- U.O.C. Ematologia-Policlinico Universitario Agostino Gemelli di RomaItaly
| | - Marta Riva
- U.O. Ematologia-ASST Grande Ospedale Metropolitano Niguarda MilanoItaly
| | - Antonella Poloni
- Clinica di Ematologia-Università Politecnica delle Marche e A.O.U. Ospedali Riuniti di AnconaItaly
| |
Collapse
|
28
|
Bouchla A, Thomopoulos TP, Papageorgiou SG, Apostolopoulou C, Loucari C, Mpazani E, Pappa V. Predicting outcome in higher-risk myelodysplastic syndrome patients treated with azacitidine. Epigenomics 2021; 13:1129-1143. [PMID: 34291653 DOI: 10.2217/epi-2021-0124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
5-Azacitidine (5-AZA) is widely used for the treatment of higher-risk myelodysplastic syndromes. However, response and survival rates vary considerably, while indicated treatment duration remains undefined. For these reasons, factors determining response and survival are of major importance. Clinical, morphological, flow cytometry, cytogenetic and molecular factors are discussed in this review. Biomarkers predictive of response and prognosis, as well as their link to the mode of action of 5-AZA are also addressed, shifting the focus from clinical practice to investigational research. Their use could further improve prognostic classification of 5-AZA treated higher-risk myelodysplastic syndromes in the near future.
Collapse
Affiliation(s)
- Anthi Bouchla
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| | - Thomas P Thomopoulos
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| | - Sotirios G Papageorgiou
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| | - Christina Apostolopoulou
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| | - Constantinos Loucari
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| | - Efthimia Mpazani
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| | - Vasiliki Pappa
- Second Department of Internal Medicine & Research Unit Hematology Unit, University General Hospital Attikon, Rimini, 12462 Chaidari, Athens, Greece
| |
Collapse
|
29
|
TP53 Abnormalities and MMR Preservation in 5 Cases of Proliferating Trichilemmal Tumours. Dermatopathology (Basel) 2021; 8:147-158. [PMID: 34070291 PMCID: PMC8161811 DOI: 10.3390/dermatopathology8020021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/19/2021] [Accepted: 05/22/2021] [Indexed: 01/02/2023] Open
Abstract
Proliferating trichilemmal tumours (PTT) are defined by a benign squamous cell proliferation inside a trichilemmal cystic (TC) cavity. A possible explanation of this proliferative phenomenon within the cyst may be molecular alterations in genes associated to cell proliferation, which can be induced by ultraviolet radiation. Among other genes, alterations on TP53 and DNA mismatch repair proteins (MMR) may be involved in the cellular proliferation observed in PTT. Based on this assumption, but also taking into account the close relationship between the sebaceous ducts and the external root sheath where TC develop, a MMR, a p53 expression assessment and a TP53 study were performed in a series of 5 PTT cases, including a giant one. We failed to demonstrate a MMR disorder on studied PTT, but we agree with previous results suggesting increased p53 expression in these tumours, particularly in proliferative areas. TP53 alteration was confirmed with FISH technique, demonstrating TP53 deletion in most cells.
Collapse
|
30
|
Owattanapanich W, Herzig J, Jahn N, Panina E, Ruchutrakool T, Kungwankiattichai S, Issaragrisil S, Döhner H, Döhner K. Genetic alterations in Thai adult patients with acute myeloid leukemia and myelodysplastic syndrome-excess blasts detected by next-generation sequencing technique. Ann Hematol 2021; 100:1983-1993. [PMID: 33839881 PMCID: PMC8285357 DOI: 10.1007/s00277-021-04513-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 04/03/2021] [Indexed: 10/27/2022]
Abstract
Several molecular aberrations affect the prognosis of patients with acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) with excess blasts (EB). This study aimed to determine the incidence and clinical impact of molecular genetic aberrations in Thai patients with AML and MDS-EB, detected by the next-generation sequencing (NGS) technique. This prospective, observational study was conducted between 2018 and 2020 on newly diagnosed Thai AML or MDS-EB patients aged above 15 years. NGS was performed using a custom amplicon-based targeted enrichment assay for 42 genes recurrently mutated in myeloid neoplasms. The molecular results were correlated with baseline patient and disease characteristics as well as outcomes. Forty-nine patients were enrolled in this study. The median age was 56 years (interquartile range [IQR], 44-64), with nearly equal proportions of males and females. The median number of mutations was 3 (IQR, 2-4). The most frequent alterations were FLT3 internal tandem duplications (ITD) (28.6%), DNMT3A (24.5%), and WT1 (22.4%) mutations. FLT3-ITD was more frequent in the de novo AML group than in the MDS/secondary AML group, whereas in the MDS/secondary AML group, ASXL1, ETV6, and SRSF2 mutations were more frequent. Patients aged greater than 65 years and patients with mutated TP53 were more likely to have inferior overall survival from multivariate analysis. FLT3-ITD was the most common mutation among newly diagnosed Thai AML patients. TP53 mutation and advanced age were independent adverse factors for survival outcome. The genetic landscapes of AML patients vary between national populations. Thai Clinical Trials Registry identifier: TCTR20190227003.
Collapse
Affiliation(s)
- Weerapat Owattanapanich
- Department of Internal Medicine III, University Hospital of Ulm, 89081, Ulm, Germany.,Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Julia Herzig
- Department of Internal Medicine III, University Hospital of Ulm, 89081, Ulm, Germany
| | - Nikolaus Jahn
- Department of Internal Medicine III, University Hospital of Ulm, 89081, Ulm, Germany
| | - Ekaterina Panina
- Department of Internal Medicine III, University Hospital of Ulm, 89081, Ulm, Germany
| | - Theera Ruchutrakool
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Smith Kungwankiattichai
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Surapol Issaragrisil
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Hartmut Döhner
- Department of Internal Medicine III, University Hospital of Ulm, 89081, Ulm, Germany.
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, 89081, Ulm, Germany.
| |
Collapse
|
31
|
Eskandari M, Shi Y, Liu J, Albanese J, Goel S, Verma A, Wang Y. The expression of MDM2, MDM4, p53 and p21 in myeloid neoplasms and the effect of MDM2/MDM4 dual inhibitor. Leuk Lymphoma 2020; 62:167-175. [PMID: 32924682 DOI: 10.1080/10428194.2020.1817441] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
p53 together with its downstream product p21 plays an important role in tumorigenesis development. MDM2 and MDM4 are two p53 regulators. We studied the expression of p53, p21, MDM2, and MDM4 in a total of 120 cases of myeloid neoplasms including MDS, AML or MDS/MPN, and control, using single and double immunohistochemical stains. We found TP53 mutations had a worse outcome in patients with AML/MDS, and p53 expression detected by immunohistochemistry had a similar prognostic value. p21 expression was strongly related to TP53 mutation status, with loss of expression in almost all TP53 mutated cases. MDM2 and MDM4 were highly expressed in hematopoietic cells in both benign and neoplastic cells. MDM2/p53 double positive cells exceeded MDM4/p53 double positive cells in neoplastic cases. Finally, we observed that p21 protein expression was up regulated upon the use of ALRN-6924 (Aileron) while no significant changes were seen in p53, MDM2 and MDM4 expression.
Collapse
Affiliation(s)
| | - Yang Shi
- Department of Pathology, Montefiore Medical Center, New York, NY, USA
| | - John Liu
- Rensselaer Polytechnic Institute, Troy, MI, USA
| | - Joseph Albanese
- Department of Pathology, Montefiore Medical Center, New York, NY, USA
| | - Swati Goel
- Department of Oncology, Montefiore Einstein Center for Cancer Care, New York, NY, USA
| | - Amit Verma
- Department of Oncology, Montefiore Einstein Center for Cancer Care, New York, NY, USA
| | - Yanhua Wang
- Department of Pathology, Montefiore Medical Center, New York, NY, USA
| |
Collapse
|
32
|
Gao L, Saeed A, Golem S, Zhang D, Woodroof J, McGuirk J, Ganguly S, Abhyankar S, Lin TL, Cui W. High-level MYC expression associates with poor survival in patients with acute myeloid leukemia and collaborates with overexpressed p53 in leukemic transformation in patients with myelodysplastic syndrome. Int J Lab Hematol 2020; 43:99-109. [PMID: 32812335 DOI: 10.1111/ijlh.13316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/02/2020] [Accepted: 07/29/2020] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Patients with mutated and overexpressed p53 have an aggressive course in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Studies on the impact of MYC expression in AML are limited. This is the first study to evaluate MYC expression and p53 status in AML and MDS. METHODS We identified 214 patients, 101 AML, 79 MDS, and 34 negative control patients. We retrospectively assessed p53 and MYC expression by immunohistochemistry and correlated MYC expression with p53 expression and aberrational status of TP53. RESULTS The level of both p53 and MYC expression was significantly higher in AML (mean: 9.7%; 12.1%) and MDS (mean: 5.2%; 5.5%) patients compared with control cases (mean: 0.18%; 2.3%; P = .001-0.02). p53 and MYC expression levels were even more elevated in AML when compared to MDS patients (P < .001). MYC expression was significantly associated with p53 expression and TP53 aberration in AML patients but not in MDS patients (P < .001). p53 expression and >20% MYC expression showed an adverse impact on overall survival (OS) (P < .05) in AML patients while p53 but not MYC expression showed an adverse impact on OS in MDS patients. MYC and p53 dual expression, as well as combined MYC expression and TP53 aberration, showed negative impact on OS in AML patients. MDS patients with leukemic transformation revealed an interval increase in expression of both p53 and MYC. CONCLUSION High-level MYC expression associates with p53 abnormality and poor survival in AML. MYC may provide proliferative advantage for leukemic progression in p53 dependent and independent manner.
Collapse
Affiliation(s)
- Linlin Gao
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Azhar Saeed
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Shivani Golem
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Da Zhang
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Janet Woodroof
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Joseph McGuirk
- Division of Hematologic Malignancies and Cellular Therapeutics, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Siddhartha Ganguly
- Division of Hematologic Malignancies and Cellular Therapeutics, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sunil Abhyankar
- Division of Hematologic Malignancies and Cellular Therapeutics, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Tara L Lin
- Division of Hematologic Malignancies and Cellular Therapeutics, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Wei Cui
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
33
|
Cumbo C, Tota G, Anelli L, Zagaria A, Specchia G, Albano F. TP53 in Myelodysplastic Syndromes: Recent Biological and Clinical Findings. Int J Mol Sci 2020; 21:E3432. [PMID: 32414002 PMCID: PMC7279310 DOI: 10.3390/ijms21103432] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
TP53 dysregulation plays a pivotal role in the molecular pathogenesis of myelodysplastic syndromes (MDS), identifying a subgroup of patients with peculiar features. In this review we report the recent biological and clinical findings of TP53-mutated MDS, focusing on the molecular pathways activation and on its impact on the cellular physiology. In MDS, TP53 mutational status is deeply associated with del(5q) syndrome and its dysregulation impacts on cell cycle, DNA repair and apoptosis inducing chromosomal instability and the clonal evolution of disease. TP53 defects influence adversely the MDS clinical outcome and the treatment response rate, thus new therapeutic approaches are being developed for these patients. TP53 allelic state characterization and the mutational burden evaluation can therefore predict prognosis and identify the subgroup of patients eligible for targeted therapy. For these reasons, in the era of precision medicine, the MDS diagnostic workup cannot do without the complete assessment of TP53 mutational profile.
Collapse
Affiliation(s)
| | | | | | | | | | - Francesco Albano
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy; (C.C.); (G.T.); (L.A.); (A.Z.); (G.S.)
| |
Collapse
|
34
|
|
35
|
Myeloid neoplasms in the setting of sickle cell disease: an intrinsic association with the underlying condition rather than a coincidence; report of 4 cases and review of the literature. Mod Pathol 2019; 32:1712-1726. [PMID: 31371806 DOI: 10.1038/s41379-019-0325-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 12/13/2022]
Abstract
Myeloid neoplasms occasionally occur in patients with sickle cell disease, and the underlying connection between the two diseases is unclear. Herein, we retrospectively analyzed four cases of sickle cell disease patients who developed myeloid neoplasm. Age at time of diagnosis ranged from 27 to 59 years with a median of 35.5 years. Two patients were treated with hydroxyurea and the other two with supportive care alone, with one out of the four patients receiving additional treatment with hematopoietic stem cell transplant. Three patients presented with leukocytosis, and the remaining patient presented with pancytopenia. Two patients were diagnosed with myelodysplastic syndrome/myeloproliferative neoplasm, one with myelodysplastic syndrome, and the other with acute myeloid leukemia. All four cases demonstrated certain degrees of myelodysplasia and complex cytogenetic abnormalities with - 7/7q- and/or - 5/5q- or with 11q23 (KMT2A) rearrangement. This cytogenetic profile resembles that seen in therapy-related myeloid neoplasm, suggesting that myeloid neoplasm in the setting of sickle cell disease may represent a subcategory of the disease distinct from de novo myeloid neoplasm in general. Extensive literature review further demonstrates this similarity in cytogenetic profile, as well as in other associated pathologic features. Potential etiology includes therapy for sickle cell disease, disease-related immunomodulation, or disease-related chronic inflammation. We hypothesize that constant hematopoietic hyperplasia, stimulated by a hemolysis-induced cytokine storm, may increase the chance of somatic mutations/cytogenetic aberrations, resulting in transformation of myeloid precursors. This group of myeloid neoplasms seems to herald a dismal clinical outcome, with median survival <1 year, although the exact pathogenesis and biology of the disease remain to be investigated by large cohorts in future studies.
Collapse
|
36
|
Kim M, Chung YS, Kim KA, Shim HS. Prognostic factors of acinar- or papillary-predominant adenocarcinoma of the lung. Lung Cancer 2019; 137:129-135. [DOI: 10.1016/j.lungcan.2019.09.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/07/2019] [Accepted: 09/28/2019] [Indexed: 10/25/2022]
|
37
|
Karner KH, Kohan JL, Tripp SR, Salama ME. Characterization of myeloid malignancies with TP53 mutations and comparison to P53 expression by immunohistochemical staining methods. Int J Lab Hematol 2019; 42:e35-e37. [PMID: 31390152 DOI: 10.1111/ijlh.13088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 11/26/2022]
Affiliation(s)
- Kristin H Karner
- Department of Pathology, University of Utah, Salt Lake City, Utah.,ARUP Laboratories, Salt Lake City, Utah
| | | | | | | |
Collapse
|
38
|
Hunter AM, Sallman DA. Current status and new treatment approaches in TP53 mutated AML. Best Pract Res Clin Haematol 2019; 32:134-144. [PMID: 31203995 DOI: 10.1016/j.beha.2019.05.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/08/2019] [Indexed: 12/16/2022]
Abstract
Mutations in the essential tumor suppressor gene, TP53, are observed in only 5-10% of acute myeloid leukemia (AML) cases, but are highly associated with therapy-related AML and cases with complex karyotype. The mutational status of TP53 is a critical prognostic indicator, with dismal outcomes consistently observed across studies. Response rates to traditional cytotoxic chemotherapy are poor and long-term survival after allogeneic hematopoietic stem cell transplant is rare. Therapy with hypomethylating agents has resulted in a modest improvement in outcomes over intensive chemotherapy, but durable responses are seldom observed. In view of the intrinsic resistance to standard chemotherapies conferred by mutations in TP53, novel treatment approaches are required. In this review, we examine the current treatment landscape in TP53 mutated AML and discuss emerging therapeutic approaches currently under clinical investigation.
Collapse
Affiliation(s)
- Anthony M Hunter
- Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - David A Sallman
- Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
39
|
Arai N, Homma M, Abe M, Baba Y, Murai S, Watanuki M, Kawaguchi Y, Fujiwara S, Kabasawa N, Tsukamoto H, Uto Y, Ariizumi H, Yanagisawa K, Hattori N, Saito B, Shiozawa E, Harada H, Yamochi-Onizuka T, Nakamaki T, Takimoto M. Impact of CD123 expression, analyzed by immunohistochemistry, on clinical outcomes in patients with acute myeloid leukemia. Int J Hematol 2019; 109:539-544. [PMID: 30847774 DOI: 10.1007/s12185-019-02616-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/12/2019] [Accepted: 02/12/2019] [Indexed: 12/16/2022]
Abstract
Aberrant expression of the interleukin-3 receptor alpha chain (IL3RA or CD123) is frequently observed in patients with a subset of leukemic disorders, including acute myeloid leukemia (AML), particularly in leukemia stem cells. We analyzed the relationships between immunohistochemical (IHC) expression, including that of CD123, and clinical outcomes. This study involved a retrospective analysis of 48 patients diagnosed with de novo AML (M0-M5, n = 48) at our hospital between February 2008 and September 2015. Among patients with de novo AML, CD123 expression was associated with a failure to achieve complete response (CR) to initial induction chemotherapy (P = 0.044) and poor overall survival (OS) (P = 0.036). This is the first study using IHC to demonstrate that CD123 expression is associated with a poor CR rate and poor OS in de novo AML patients. These results support previous reports using flow cytometry (FCM). CD123 expression may thus be useful for assessing AML patients' prognoses. At the time of diagnosis, CD123 expression analysis using IHC may represent a clinically useful assessment for de novo AML patients.
Collapse
Affiliation(s)
- Nana Arai
- Department of Pathology, Showa University School of Medicine, Tokyo, Japan. .,Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan.
| | - Mayumi Homma
- Department of Pathology, Showa University School of Medicine, Tokyo, Japan
| | - Maasa Abe
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Yuta Baba
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - So Murai
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Megumi Watanuki
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Yukiko Kawaguchi
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Shun Fujiwara
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Nobuyuki Kabasawa
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Hiroyuki Tsukamoto
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Yui Uto
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Hirotsugu Ariizumi
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Kouji Yanagisawa
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Norimichi Hattori
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Bungo Saito
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Eisuke Shiozawa
- Department of Pathology, Showa University School of Medicine, Tokyo, Japan
| | - Hiroshi Harada
- Division of Hematology, Department of Medicine, Showa University Fujigaoka Hospital, Kanagawa, Japan
| | | | - Tsuyoshi Nakamaki
- Division of Hematology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-Ku, Tokyo, 142-8666, Japan
| | - Masafumi Takimoto
- Department of Pathology, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
40
|
Prognostic Impact of Immunohistochemical p53 Expression in Bone Marrow Biopsy in Higher Risk MDS: a Pilot Study. Mediterr J Hematol Infect Dis 2019; 11:e2019015. [PMID: 30858953 PMCID: PMC6402551 DOI: 10.4084/mjhid.2019.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 01/24/2019] [Indexed: 12/26/2022] Open
Abstract
Background and objectives Mutations of the TP53 gene have an unfavorable prognosis in Myelodysplastic Syndromes (MDS). The product of the TP53 gene is the p53 protein. Most of the TP53 mutations entail the accumulation of the protein in the nucleus of tumor cells. The immunohistochemical (IHC) staining for p53 can be a surrogate suggesting a mutational status and, if overexpressed, seems to be of prognostic value by itself. The best prognostic cut-off value of overexpression is controversial. The aim of this pilot study is to investigate the correct value from a homogenous group of patients with higher IPSS-R risk MDS. Methods In sixty consecutive patients diagnosed with MDS and categorized as “intermediate,” “high” and “very high” IPSS-risk, the bone marrow biopsies performed at diagnosis were retrospectively re-examined for IHC p53 expression. The result of p53 expression was subsequently related to survival. Results A worse overall survival was observed both in patients whose IHC p53 expression was ≥5% and ≥ 10% compared to patients with a p53 expression below 5% (p= 0.0063) or 10% (p=0.0038) respectively. Conclusions The ICH p53 expression in bone marrow biopsy in higher risk MDS was confirmed to have prognostic value. These results indicate more than 10% expression as the best cut off value.
Collapse
|
41
|
Ruzinova MB, Lee YS, Duncavage EJ, Welch JS. TP53 immunohistochemistry correlates with TP53 mutation status and clearance in decitabine-treated patients with myeloid malignancies. Haematologica 2019; 104:e345-e348. [PMID: 30792212 DOI: 10.3324/haematol.2018.205302] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
| | | | | | - John S Welch
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
42
|
Assi R, Gur HD, Loghavi S, Konoplev SN, Konopleva M, Daver N, Tashakori M, Kadia T, Routbort M, Salem A, Kanagal-Shamanna R, Quesada A, Jabbour EJ, Kornblau SM, Medeiros LJ, Kantarjian H, Khoury JD. P53 protein overexpression in de novo acute myeloid leukemia patients with normal diploid karyotype correlates with FLT3 internal tandem duplication and worse relapse-free survival. Am J Hematol 2018; 93:1376-1383. [PMID: 30117185 DOI: 10.1002/ajh.25255] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/09/2018] [Accepted: 08/13/2018] [Indexed: 12/30/2022]
Abstract
Although ~50% of acute myeloid leukemia (AML) patients have a normal diploid karyotype by conventional cytogenetics at diagnosis, this patient subset has a variable disease course and outcome. Aberrant overexpression of the p53 protein is usually associated with TP53 alterations and a complex karyotype, but the prevalence and impact of p53 overexpression in AML with diploid cytogenetics is unknown. We examined 100 newly diagnosed AML patients to evaluate the impact of p53 expression status quantified in bone marrow core biopsy samples using immunohistochemistry and computer-assisted image analysis. A total of 24 patients had p53 overexpression defined as 3+ staining intensity in ≥5% of cells; this finding correlated with lower platelet counts (P = .002), absence of CD34 expression in blasts (P = .009), higher bone marrow blast counts (P = .04), and a higher frequency of FLT3 internal tandem duplication (P = .007). Overexpression of p53 independently predicted for shorter leukemia-free survival in patients who underwent allogeneic stem cell transplantation by univariate (P = .021) and multivariate analyses (P = .004). There was no correlation between MDM2 and p53 protein expression in this cohort. We conclude that p53 expression evaluated by immunohistochemistry in bone marrow biopsy specimens at the time of AML diagnosis may indicate distinct clinical characteristics in patients with normal diploid cytogenetics and is a potentially valuable tool that can enhance risk-stratification.
Collapse
Affiliation(s)
- Rita Assi
- Departments of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Hatice D. Gur
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Sanam Loghavi
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Sergej N. Konoplev
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Marina Konopleva
- Departments of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Naval Daver
- Departments of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Mehrnoosh Tashakori
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Tapan Kadia
- Departments of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Mark Routbort
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Alireza Salem
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Andres Quesada
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Elias J. Jabbour
- Departments of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Steven M. Kornblau
- Departments of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - L. Jeffrey Medeiros
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Hagop Kantarjian
- Departments of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Joseph D. Khoury
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| |
Collapse
|
43
|
Abstract
TP53 mutated acute myeloid leukemia (AML) responds poorly to chemotherapy and has a short overall survival rate with a median of 5-9 months. Poor outcomes in TP53 mutated AML following chemotherapy have been observed and treatment options remain limited, although the presence of TP53 mutations alone should not be a barrier to therapy. Decitabine is emerging as an alternative treatment option for patients with TP53 mutated AML, although the agent has not been associated with deep molecular remissions and requires additional consolidation. The clinical and genomic characteristics of TP53 mutated AML are reviewed in this paper.
Collapse
Affiliation(s)
- John S Welch
- Department of Internal Medicine, Washington University, 660 Euclid Ave, Box 8007, St. Louis, MO 63110, USA.
| |
Collapse
|
44
|
Kayano H. Histopathology in the diagnosis of high-risk myelodysplastic syndromes. J Clin Exp Hematop 2018; 58:51-60. [PMID: 29998976 DOI: 10.3960/jslrt.18009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
45
|
Kuykendall A, Duployez N, Boissel N, Lancet JE, Welch JS. Acute Myeloid Leukemia: The Good, the Bad, and the Ugly. Am Soc Clin Oncol Educ Book 2018; 38:555-573. [PMID: 30231330 DOI: 10.1200/edbk_199519] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Acute myeloid leukemia (AML) was initially subdivided according to morphology (the French-American-British system), which proved helpful in pathologic categorization. Subsequently, clinical and genomic factors were found to correlate with response to chemotherapy and with overall survival. These included a history of antecedent hematologic disease, a history of chemotherapy or radiation therapy, the presence of various recurrent cytogenetic abnormalities, and, more recently, the presence of specific point mutations. This article reviews the biology and responses of one AML subgroup with consistent response and good outcomes following chemotherapy (core-binding factor leukemia), and two subgroups with persistently bad, and even ugly, outcomes (secondary AML and TP53-mutated AML).
Collapse
MESH Headings
- Alleles
- Biomarkers, Tumor
- Chromosome Aberrations
- Combined Modality Therapy
- Core Binding Factors/genetics
- Core Binding Factors/metabolism
- Gene Frequency
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/mortality
- Mutation
- Neoplasm, Residual/diagnosis
- Neoplasms, Second Primary/diagnosis
- Neoplasms, Second Primary/epidemiology
- Neoplasms, Second Primary/etiology
- Neoplasms, Second Primary/therapy
- Signal Transduction
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Andrew Kuykendall
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| | - Nicolas Duployez
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| | - Nicolas Boissel
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| | - Jeffrey E Lancet
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| | - John S Welch
- From the Moffitt Cancer Center, Tampa, FL; CHU Lille, INSERM, Laboratory of Hematology, University of Lille, Lille, France; Hematology Department, Saint-Louis Hospital, Paris Diderot University, Paris, France; Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
46
|
McGraw KL, Nguyen J, Al Ali NH, Komrokji RS, Sallman D, Zhang X, Song J, Padron E, Lancet JE, Moscinski LC, List AF, Zhang L. Association of EZH2 protein expression by immunohistochemistry in myelodysplasia related neoplasms with mutation status, cytogenetics and clinical outcomes. Br J Haematol 2018; 184:450-455. [PMID: 29359794 DOI: 10.1111/bjh.15099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Kathy L McGraw
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Johnny Nguyen
- Departments of Hematopathology and Clinical Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Najla H Al Ali
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Rami S Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - David Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Xiaohui Zhang
- Departments of Hematopathology and Clinical Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jinming Song
- Departments of Hematopathology and Clinical Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Eric Padron
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jeffrey E Lancet
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Lynn C Moscinski
- Departments of Hematopathology and Clinical Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Alan F List
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Ling Zhang
- Departments of Hematopathology and Clinical Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| |
Collapse
|
47
|
Haider M, Duncavage EJ, Afaneh KF, Bejar R, List AF. New Insight Into the Biology, Risk Stratification, and Targeted Treatment of Myelodysplastic Syndromes. Am Soc Clin Oncol Educ Book 2017; 37:480-494. [PMID: 28561687 DOI: 10.1200/edbk_175397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In myelodysplastic syndromes (MDS), somatic mutations occur in five major categories: RNA splicing, DNA methylation, activated cell signaling, myeloid transcription factors, and chromatin modifiers. Although many MDS cases harbor more than one somatic mutation, in general, there is mutual exclusivity of mutated genes within a class. In addition to the prognostic significance of individual somatic mutations, more somatic mutations in MDS have been associated with poor prognosis. Prognostic assessment remains a critical component of the personalization of care for patient with MDS because treatment is highly risk adapted. Multiple methods for risk stratification are available with the revised International Prognostic Scoring System (IPSS-R), currently considered the gold standard. Increasing access to myeloid gene panels and greater evidence for the diagnostic and predictive value of somatic mutations will soon make sequencing part of the standard evaluation of patients with MDS. In the absence of formal guidelines for their prognostic use, well-validated mutations can still refine estimates of risk made with the IPSS-R. Not only are somatic gene mutations advantageous in understanding the biology of MDS and prognosis, they also offer potential as biomarkers and targets for the treatment of patients with MDS. Examples include deletion 5q, spliceosome complex gene mutations, and TP53 mutations.
Collapse
Affiliation(s)
- Mintallah Haider
- From the Department of Hematology and Medical Oncology, Moffitt Cancer Center and the University of South Florida, Tampa, FL; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO; Moores Cancer Center, Division of Hematology and Oncology, University of California, San Diego, CA; Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Eric J Duncavage
- From the Department of Hematology and Medical Oncology, Moffitt Cancer Center and the University of South Florida, Tampa, FL; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO; Moores Cancer Center, Division of Hematology and Oncology, University of California, San Diego, CA; Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Khalid F Afaneh
- From the Department of Hematology and Medical Oncology, Moffitt Cancer Center and the University of South Florida, Tampa, FL; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO; Moores Cancer Center, Division of Hematology and Oncology, University of California, San Diego, CA; Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Rafael Bejar
- From the Department of Hematology and Medical Oncology, Moffitt Cancer Center and the University of South Florida, Tampa, FL; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO; Moores Cancer Center, Division of Hematology and Oncology, University of California, San Diego, CA; Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| | - Alan F List
- From the Department of Hematology and Medical Oncology, Moffitt Cancer Center and the University of South Florida, Tampa, FL; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO; Moores Cancer Center, Division of Hematology and Oncology, University of California, San Diego, CA; Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
48
|
Peterson LE, Kovyrshina T. Progression inference for somatic mutations in cancer. Heliyon 2017; 3:e00277. [PMID: 28492066 PMCID: PMC5415494 DOI: 10.1016/j.heliyon.2017.e00277] [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: 11/04/2016] [Revised: 03/08/2017] [Accepted: 03/23/2017] [Indexed: 01/05/2023] Open
Abstract
Computational methods were employed to determine progression inference of genomic alterations in commonly occurring cancers. Using cross-sectional TCGA data, we computed evolutionary trajectories involving selectivity relationships among pairs of gene-specific genomic alterations such as somatic mutations, deletions, amplifications, downregulation, and upregulation among the top 20 driver genes associated with each cancer. Results indicate that the majority of hierarchies involved TP53, PIK3CA, ERBB2, APC, KRAS, EGFR, IDH1, VHL, etc. Research into the order and accumulation of genomic alterations among cancer driver genes will ever-increase as the costs of nextgen sequencing subside, and personalized/precision medicine incorporates whole-genome scans into the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Leif E. Peterson
- Center for Biostatistics, Houston Methodist Research Institute, Houston, TX 77030, USA
- Dept. of Healthcare Policy and Research, Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
- Dept. of Biostatistics, School of Public Health, University of Texas – Health Science Center, Houston, TX 77030, USA
- Dept. of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Dept. of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Tatiana Kovyrshina
- Center for Biostatistics, Houston Methodist Research Institute, Houston, TX 77030, USA
- Dept. of Mathematics and Statistics, University of Houston – Downtown, Houston, TX 77002, USA
| |
Collapse
|
49
|
Zhang L, McGraw KL, Sallman DA, List AF. The role of p53 in myelodysplastic syndromes and acute myeloid leukemia: molecular aspects and clinical implications. Leuk Lymphoma 2016; 58:1777-1790. [PMID: 27967292 DOI: 10.1080/10428194.2016.1266625] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
TP53 gene mutations occurring in patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) are associated with high-risk karyotypes including 17p abnormalities, monosomal and complex cytogenetics. TP53 mutations in these disorders portend rapid disease progression and resistance to conventional therapeutics. Notably, the size of the TP53 mutant clone as measured by mutation allele burden is directly linked to overall survival (OS) confirming the importance of p53 as a negative prognostic variable. In nucleolar stress-induced ribosomopathies, such as del(5q) MDS, disassociation of MDM2 and p53 results in p53 accumulation in erythroid precursors manifested as erythroid hypoplasia. P53 antagonism by lenalidomide or other therapeutics such as antisense oligonucleotides, repopulates erythroid precursors and enhances effective erythropoiesis. These findings demonstrate that p53 is an intriguing therapeutic target that is currently under investigation in MDS and AML. This study reviews molecular advances in understanding the role of p53 in MDS and AML, and explores potential therapeutic strategies in this era of personalized medicine.
Collapse
Affiliation(s)
- Ling Zhang
- a Department of Hematopathology and Laboratory Medicine , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| | - Kathy L McGraw
- b Department of Malignant Hematology , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| | - David A Sallman
- b Department of Malignant Hematology , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| | - Alan F List
- b Department of Malignant Hematology , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| |
Collapse
|