Bone marrow neoplastic

Bone marrow - neoplastic myeloid

Myelodysplastic syndromes / neoplasms (MDS)

MDS with biallelic TP53 inactivation



Last staff update: 9 July 2024 (update in progress)

Copyright: 2024, PathologyOutlines.com, Inc.

PubMed Search: MDS with biallelic TP53 inactivation

Daniela Vulpe, B.S.
Hatem Kaseb, M.D., Ph.D., M.P.H.
Page views in 2024 to date: 311
Cite this page: Vulpe D, Kaseb H. MDS with biallelic TP53 inactivation. PathologyOutlines.com website. https://www.pathologyoutlines.com/topic/bonemarrowneoplasticMDSbiallelicTP53inactivation.html. Accessed July 15th, 2024.
Definition / general
  • Myelodysplastic neoplasms (MDS), previously called myelodysplastic syndromes, are clonal hematopoietic stem cell neoplasms characterized by ineffective marrow hematopoiesis that results in morphologic dysplasia and peripheral cytopenia; there is increased risk of transformation to acute myeloid leukemia (AML)
  • New MDS subtype per WHO 5th edition and international consensus classification (ICC) 2022
Essential features
  • MDS biTP53 (MDS with biallelic TP53 inactivation) is a new subtype per WHO 5th edition, 2022
  • Detection of ≥ 1 TP53 mutation, meeting molecular diagnostic criteria of biallelic TP53 mutations (Nat Med 2020;26:1549)
  • 33% of TP53 mutated MDS patients had monoallelic mutations whereas 67% had multiple hits (multihit) consistent with biallelic targeting (Nat Med 2020;26:1549)
  • MDS biTP53 is characterized by increased blast counts, higher risk of leukemic transformation and higher risk of mortality
  • TP53 mutated myeloid neoplasms (MN) represent a unique entity with very poor prognosis, irrespective of whether the blast percentage indicates MDS or AML or whether the disease is therapy related or de novo in its ontogeny (Blood Adv 2022;6:2847)
Terminology
  • Per the 5th edition of WHO published in 2022, the terminology of myelodysplastic neoplasms has been introduced to replace the previous term of myelodysplastic syndromes, in parallel to myeloproliferative neoplasm and to emphasize the neoplastic course of the disease; however, the abbreviation of MDS is kept for myelodysplastic neoplasm
ICD coding
  • ICD-10: D46 - myelodysplastic syndromes
Epidemiology
  • In the U.S., the age adjusted incidence of MDS is estimated to be 4 in 100,000 individuals (Blood Rev 2019;34:1)
  • Incidence notably increases with advancing age, male gender, smoking, obesity and history of radiotherapy or chemotherapy (Blood Rev 2019;34:1)
  • Estimated incidence of MDS biTP53 is ~0.03/100,000/year
    • ~10% of MDS patients exhibit TP53 alteration, with 67% of these cases involving biallelic alterations (Leukemia 2019;33:1747)
  • TP53 mutated MDS has lower response to treatment compared to non-TP53 mutated MDS and an overall survival (OS) of 5 - 10 months with prognosis worsening in biallelic TP53 mutations (Cancer Discov 2022;12:2516, Nat Med 2020;26:1549)
  • It is important to note that TP53 has been found to play an important role in both de novo and therapy related MDS; interestingly TP53 mutations were found to be the most common genetic mutation in treatment related MDS (t-MDS) (Nature 2015;518:552)
    • History of cytotoxic treatment is a qualifier that moves the diagnosis to myeloid neoplasm postcytotoxic therapy
Sites
  • Defining characteristic of MDS is bone marrow failure, which occurs as hematopoietic stem cells acquire mutations, resulting in ineffective hematopoiesis, morphological dysplasia and peripheral blood cytopenias (Blood 2019;133:1086)
Pathophysiology
  • TP53 suppressor gene is mutated in about half of human tumors (Nat Med 2020;26:1549)
  • TP53 gene encodes for the p53 protein, which plays a crucial role in regulating various cellular processes, such as the cell cycle and cell death, which is usually triggered by oxidative stress (WHO 5); thus, the inactivation of p53 allows tumor initiation and its evasion from cell death (Nat Rev Cancer 2018;18:89)
  • It is hypothesized that monoallelic TP53 inactivation may contain biallelic TP53 inactivation subclones that can negatively affect disease outcomes, as patients with single TP53 and VAF of > 23% have similar outcomes to biallelic TP53 patients
  • Patients with TP53 mutations were found to have fewer comutated genes; however, they more commonly exhibit del(5q) chromosomal abnormality, monosomal karyotype and complex karyotype (Leukemia 2019;33:1747)
  • Neurofibromin 1 (NF1), which is a negative regulator of Ras signaling, and del(12p), which includes ETV6, were found to be prevalent in TP53 inactivation AML / MDS cases (Blood Adv 2023;7:4586)
  • TP53 mutated MN represents a unique entity with very poor prognosis, irrespective of whether the blast percentage indicates MDS or AML or whether the disease is therapy related or de novo in its ontogeny (Blood Adv 2022;6:2847)
    • Current assignment of these cases into MDS, AML with myelodysplasia related changes (AML MRC) or therapy related myeloid neoplasms (t-MN) appears to divide a biologically similar disease into different diagnostic groups
  • TP53 mutations play an important role in the pathogenesis of many cases of t-AML / t-MDS (Nature 2015;518:552)
    • Loss of TP53 provides a selective advantage for neoplastic growth; TP53 mutations are resistant to chemotherapy and expand preferentially after treatment
Etiology
  • Variable and includes
    • Idiopathic
    • Exposure to certain DNA damaging agents such as chemotherapy or radiation
      • TP53 mutations were found to be more common in t-MN, which was also a poor diagnostic factor (Leukemia 2019;33:2842)
      • Chemotherapy leads to selective expansion of the progenitor cells with TP53 mutations that are resistant to treatment (Nature 2015;518:552)
    • Inherited genetic disorders
      • E.g., Li-Fraumeni syndrome or inherited bone marrow failure syndromes
Diagrams / tables

WHO 4R WHO 5th ICC classification
  • Not an entity
    • TP53 mutation is listed as a one of the common genetic mutations in MDS
    • Prognostic impact is adverse
    • Mutation should be correlated with morphological findings
  • MDS with biallelic TP53 inactivation
    • Fulfilling diagnostic criteria of MDS
    • 2 or more TP53 mutations or 1 mutation with TP53 copy number loss or cnLOH
    • Myeloblasts < 20% bone marrow (BM) and peripheral blood (PB)
  • MDS with mutated TP53
    • Multihit TP53
    • Blasts 0 - 9% in BM / PB
  • AML / MDS with mutated TP53
    • Any somatic TP53 mutation (VAF > 10%)
    • Blasts 10 - 19% in BM / PB


ICC classification of MDS with mutated TP53
MDS with mutated TP53 AML / MDS with mutated TP53
  • Any cytopenia
  • Blasts: 0 - 9%
  • Genetics: multihit TP53, TP53 mutation (VAF > 10%) and complex karyotype often with loss 17p
  • Any cytopenia
  • Blasts: 10 - 19%
  • Genetics: any somatic TP53 mutation (VAF > 10%)


WHO 5th edition, 2022 ICC 2022
Morphologically defined
Lineage Subclassification using dysplastic lineages removed, replaced with MDS LB (MDS with low blasts) MDS, NOS SLD
MDS, NOS MLD
Excessive blast counts in bone marrow
(5 - 9%) or in peripheral blood (2 - 4%)
MDS IB1 MDS EB
(blast count 5 - 9% in bone marrow, 2 - 4% in peripheral blood)
Excessive blast counts in bone marrow
(10 - 19%) or in peripheral blood (5 - 19%)
MDS IB2 MDS AML
(blast count 10 - 19% in bone marrow and 2 - 4% in peripheral blood)
Others cMDS (MDS of childhood)
MDS-h (hypoplastic MDS)
MDS-f (MDS with fibrosis, blasts 5 - 19% in bone marrow, 2 - 19% in peripheral blood)
MDS, NOS (without dysplasia)
Defining genetic abnormalities
SF3B1 mutation MDS SF3B1 (MDS with low blasts and SF3B1 mutation or MDS with ring sideroblasts if SF3B1 wild type) MDS SF3B1 (MDS with mutated SF3B1) or MDS, NOS (with ring sideroblasts and SF3B1 wild type)
del(5q) MDS 5q
(MDS with low blasts and 5q deletion)
[5q deletion alone or with 1 other genetic aberration other than del(7q) / -7]
MDS with del(5q)
[del(5q) is isolated or with up 1 genetic aberration except del(7q) / -7 or multihit TP53]
TP53 MDS biTP53
(MDS with biallelic TP53 inactivation)
MDS with mutated TP53
Multihit TP53
Includes MDS, MDS / AML, AML
Diagnosis
  • Multihit TP53 is approximated by (Blood Adv 2022;6:2847, Nat Med 2020;26:1549)
    • Next generation sequencing analysis or Sanger sequencing covering at least exons 4 - 11 of TP53 gene is required for detection of biallelic TP53 alterations, coupled with a technique to detect copy number status, usually fluorescence in situ hybridization (FISH) with a probe set specific for the TP53 locus on 17p13.1 (Leukemia 2014;28:241, Blood 2013;122:3616)
    • Detection of ≥ 2 TP53 mutations, usually affecting both alleles that can be considered multihit status (Nat Med 2020;26:1549)
    • After exclusion of constitutional changes, a TP53 VAF > 49% may be regarded as presumptive (not definitive) of copy loss on the trans allele or copy neutral LOH
    • In the presence of one TP53 mutation, evidence of TP53 copy loss or copy neutral LOH is required as concurrent 17p LOH, suggestive of biallelic TP53 alterations (Nat Med 2021;27:1239, Nat Commun 2020;11:4980)
  • TP53 mutations are typically associated with complex karyotype; these 2 features are associated with unfavorable risk (N Engl J Med 2016;375:2023, Nat Med 2020;26:1549)
    • Up to 55% of patients with complex karyotype (CK) (> 3 cytogenetic abnormalities) harbor a TP53 mutation (Leukemia 2019;33:1747, Blood Adv 2022;6:2847)
    • Complex karotype TP53 mutated MDS has been shown to predict poor prognosis irrespective of blast percentage and other TP53 molecular characteristics (Blood Adv 2022;6:2847)
  • Presence of TP53 mutation is an independent impact on prognosis that is as significant as severe anemia and more significant than having a bone marrow blast proportion of 10 - 29% (Leukemia 2019;33:1747)
    • To assess patients with TP53 mutations, research has shown that relying on VAF estimates is not sufficient for precise assessment of TP53 allelic state and recommends other approaches for confirmation (Nat Med 2020;26:1549)
Prognostic factors
  • Factors associated with significant decrease in survival in MDS patients with TP53 mutations include (Leukemia 2019;33:1747)
    • Severe anemia (hemoglobin < 8.0 g/dL), NRAS mutation, SF3B1 mutation, TP53 mutation, elevated blast percentage (> 10%), abnormal 3q, abnormal 9 and monosomy 7 as having the greatest survival risk
  • Monosomal karyotype, high complexity and TP53 mutation are individually associated with shorter overall survival (Leukemia 2019;33:1747)
  • Type and abundance of TP53 mutation (VAF) in question may further refine its prognostic impact (Leukemia 2019;33:1747)
  • TP53 mutant CK MDS patients also had significantly higher bone marrow blast proportion and lower platelet counts, 2 factors strongly associated with elevated prognostic risk considered by clinical scoring systems like the IPSS-R (Leukemia 2019;33:1747)
  • Some studies have shown that the effect of TP53 on survival is significant in both TP53 monoallelic and TP53 multihit (Blood Adv 2022;6:2847)
    • No TP53 mutation (median: 33.9 months)
    • TP53 monoallelic (median: 12.5 months)
    • TP53 multihit (median: 9.4 months)
    • No significant influence of TP53 VAF as a continuous variable or using VAF cutoffs (Blood Adv 2022;6:2847)
  • Some studies have shown that only MDS with TP53 multihit shows poor outcomes and response to therapy compared to MDS with TP53 monoallelic mutations (Nat Med 2020;26:1549)
    • Median OS in MDS TP53 multihit state was 8.7 months
    • Median OS in MDS TP53 monoallelic patients was 2.5 years
    • Median OS wild type patients was 3.5 years
    • Patients with monoallelic TP53 mutations and VAF > 22% (n = 38) had increased risk of death compared to wild type patients; monoallelic TP53 mutations and VAF ≤ 22% (n = 87) had OS similar to wild type patients
Case reports
  • 7 year old boy, 44 year old woman and 47 year old woman with Li-Fraumeni syndrome and MDS (Arch Pathol Lab Med 2010;134:1010)
  • 65 year old woman with TP53 germline mutation developed Bowen disease and myelodysplastic syndrome (Intern Med 2005;44:490)
  • 76 year old man presented with refractory anemia with ring sideroblasts, i(17q) and TP53 mutation (Cancer Genet Cytogenet 2002;136:86)
  • 9 patients with TP53 mutant myelodysplastic syndromes managed by allogeneic hematopoietic stem cell transplantation and decitabine containing preconditioning regimen (Front Oncol 2022;12:928324)
Treatment
  • Risk stratification is essential in the clinical care of patients with myelodysplastic syndromes (Blood 2012;120:2454)
    • Types of chromosomal abnormalities present, co-occurring somatic mutations and clinical features all contribute to the actual risk in patients (Leukemia 2019;33:1747)
  • Revised International Prognostic Scoring System (IPSS-R) (Blood 2012;120:2454)
    • MDS are usually classified according to the IPSS-R into 5 prognostic groups
  • Molecular International Prognostic Scoring System (IPSS-M) (NEJM Evid 2022;1:EVIDoa2200008)
    • MDS is classified into 6 IPSS-M risk categories with prognostic differences
    • Compared with the IPSS-R, the IPSS-M shows better prognostic discrimination
  • Patients with TP53 mutations who receive conventional chemotherapy, such as anthracycline based or cytarabine based induction chemotherapy, have poor outcomes; decitabine has been shown to improve overall survival (N Engl J Med 2016;375:2023, Leukemia 2019;33:1747)
    • Possible explanation is that standard chemotherapy leads to outgrowth of preexisting TP53 mutated clones rather than creating TP53 mutations (Nature 2015;518:552)
Microscopic (histologic) description
  • Blast count must be < 19% in bone marrow
  • Bone marrow cellularity is typically hyper / normocellular
  • Significant dysplasia in 1 or more of the myeloid lineages
    • Dysmegakaryopoiesis
    • Dyserythropoiesis
    • Dysgranulopoiesis
  • Reference: Leukemia 2022;36:1703
Microscopic (histologic) images

Contributed by Hatem Kaseb, M.D., Ph.D., M.P.H.
Blast

Blast

Bone marrow with increased blasts

Bone marrow with increased blasts

Dyserythropoiesis

Dyserythropoiesis

Hypercellular bone marrow with dysplasia Hypercellular bone marrow with dysplasia

Hypercellular bone marrow with dysplasia


Hypercellular bone marrow with dysplasia

Hypercellular bone marrow with dysplasia

Increased blasts in bone marrow Increased blasts in bone marrow

Increased blasts in bone marrow

TP53 IHC TP53 IHC

TP53 IHC

Positive stains
  • TP53 (usually)
  • CD34 highlights blasts (variable)
  • CD117 highlights blasts and immature cells (variable)
Negative stains
  • Iron stain for ring sideroblasts
  • CD3 highlights T lymphocytes
  • CD20 highlights B lymphocytes
Flow cytometry description
  • No findings specific to this entity
  • Myeloblasts with aberrant phenotype may be detected
Molecular / cytogenetics description
  • Multihit TP53 is approximated by (Blood Adv 2022;6:2847, Nat Med 2020;26:1549)
    • Detection of ≥ 2 TP53 mutations, usually affecting both alleles that can be considered multihit status (Nat Med 2020;26:1549)
    • After exclusion of constitutional changes, a TP53 VAF > 49% may be regarded as presumptive (not definitive) of copy loss on the trans allele or copy neutral LOH
    • In the presence of one TP53 mutation, evidence of TP53 copy loss or copy neutral LOH is required as concurrent 17p LOH, suggestive of biallelic TP53 alterations (Nat Med 2021;27:1239, Nat Commun 2020;11:4980)
Sample pathology report
  • Bone marrow aspirate, core biopsy, clot section and peripheral blood smear:
    • Myeloid neoplasm with multihit TP53 (see comment)
    • Comment: This neoplasm meets criteria for MDS with increased blasts 2 (MDS IB2) (WHO 5). The presence of TP53 in MDS supersedes other MDS types in classification; therefore, in conjunction with the reported TP53 mutation (VAF: 87%), it is compatible with myelodysplastic syndrome with biallelic TP53 inactivation (MDS biTP53) (WHO 5) and MDS / AML with mutated TP53 (ICC).
Board review style question #1
Which of the following is correct regarding myelodysplastic neoplasm (MDS) with TP53 inactivation / multihit?

  1. Associated with 5q loss
  2. Association with SF3B1 mutation is common
  3. Favorable outcome among MDS types
  4. Rarely transforms to acute myeloid leukemia
  5. TP53 VAF > 49% supports the diagnosis
Board review style answer #1
E. TP53 VAF > 49% supports the diagnosis. This is one of the criteria of MDS with multihit TP53 as laid out by WHO 5th edition. Answer C is incorrect because the disease is associated with unfavorable outcome. Answer D is incorrect because the patients usually progress to acute myeloid leukemia. Answer B is incorrect because the disease is not associated with SF3B1 mutation. Answer A is incorrect because the disease is not associated with 5q loss.

Comment Here

Reference: MDS with biallelic TP53 inactivation
Board review style question #2

Which of the following is correct regarding myelodysplastic neoplasm (MDS) with TP53 inactivation / multihit?

  1. Blast count must be < 10% in bone marrow
  2. Disease shows specific flow cytometry findings
  3. Dysmegakaryopoiesis is the most common dysplastic finding
  4. MDS TP53 monoallelic patients typically show similar prognosis to MDS TP53 multihit patients
  5. Usually associated with complex karyotype
Board review style answer #2
E. Usually associated with complex karyotype. MDS with multihit TP53 is commonly associated with complex karyotype and this association predicts an unfavorable prognosis. Answer B is incorrect because the disease does not show a specific flow cytometry finding. Answer C is incorrect because no distinct dysplasia in a certain lineage has been found to be common in this disease entity. Answer A is incorrect because the blast count must be < 19% in bone marrow. Answer D is incorrect because the majority of papers have shown that the prognosis of MDS TP53 multihit patients is worse than MDS TP53 monoallelic.

Comment Here

Reference: MDS with biallelic TP53 inactivation
Back to top
Image 01 Image 02