Вы находитесь на странице: 1из 14

review

Li-Fraumeni syndrome: a paradigm for the understanding of


hereditary cancer predisposition

Jessica M. Valdez, Kim E. Nichols and Chimene Kesserwan


Division of Cancer Predisposition, St. Jude Childrens Research Hospital, Memphis, TN, USA

spectrum of other benign and malignant neoplasms is also


Summary
observed (Nichols et al, 2001).
Li-Fraumeni syndrome (LFS) is a rare cancer predisposing A breakthrough in the understanding of LFS occurred in
condition caused by germline mutations in TP53, the gene 1990 when Malkin et al (1990) demonstrated that individuals
encoding the TP53 transcription factor. LFS is typified by the with LFS harbour germline mutations in TP53 the gene
development of a wide spectrum of childhood and adult- encoding the TP53 transcription factor (tumor protein p53,
onset malignancies, which includes, among others, the lym- TP53; previously termed p53). Also known as the guardian
phoid and myeloid leukaemias, myelodysplastic syndrome of the genome, TP53 plays a central role in cellular growth
and, to a lesser extent, lymphoma. Accordingly, it is impor- control by regulating the expression of numerous down-
tant that haematologists/oncologists be familiar with this stream genes involved in cell cycle arrest, apoptosis, DNA
pleiotropic hereditary cancer syndrome. The high cancer risk repair and senescence, particularly in response to DNA dam-
and variability in type and age of cancer onset have raised age and other cellular stressors (Vousden & Prives, 2009;
questions about the underlying biology and optimal treat- Reinhardt & Schumacher, 2012). TP53 was examined as an
ment approaches for individuals with LFS. Since its descrip- LFS candidate gene based on observations that somatic TP53
tion almost 50 years ago, many clinical and basic research mutations were prevalent in the majority of human cancers,
investigations have provided insights into the pathogenesis, including those seen in LFS (Nigro et al, 1989). Furthermore,
manifestations, genetic testing and management strategies for transgenic mice expressing mutant Trp53 alleles developed
individuals with LFS. Here we provide an update on the cur- LFS-type cancers, including OS, STS and lymphoid neo-
rent state of knowledge regarding LFS with an emphasis, plasms (Lavigueur et al, 1989). A heterozygous germline
where possible, on information relevant to practicing haema- TP53 mutation was identified in each of the five families
tologists. analysed by Malkin et al (1990), and in one family, the
mutation co-segregated with tumour formation. At the same
Keywords: Li-Fraumeni syndrome, cancer predisposition, time, an unrelated kindred harbouring a germline TP53
germline, TP53, mutation. mutation was reported by Srivastava et al (1990). Taken
together, these formative observations confirmed that muta-
Li-Fraumeni syndrome (LFS) is a prototypical cancer predis- tions in TP53 occur not only as common somatic events in
posing syndrome first described in 1969 by Drs. Frederick Li cancer, but also as rare germline events in individuals with
and Joseph Fraumeni following their retrospective analysis of LFS.
children with rhabdomyosarcoma (RMS) (Li & Fraumeni,
1969a,b). Since their initial report, many studies by these and
The spectrum of cancers observed in LFS
other investigators have revealed that LFS is a rare autosomal
dominant disorder that confers an increased risk to develop One of the most striking features of LFS is the remarkable
six common core cancers, including soft tissue sarcomas tendency for affected individuals to develop solid and haema-
(STS), osteosarcoma (OS), premenopausal breast cancer, cen- tological cancers. The risk for germline TP53 mutation carri-
tral nervous system (CNS) tumours, adrenocortical carci- ers to develop one or more neoplasms is estimated to be
noma (ACC) and leukaemia (Malkin et al, 1990). A wide 50% by age 3031 years for females and age 46 years for
males (Mai et al, 2016). Remarkably, this risk nears 70100%
by age 70 years for both sexes (Mai et al, 2016). Collectively,
Correspondence: Chimene Kesserwan, Division of Cancer the core cancers comprise the majority of LFS-related
Predisposition, St. Jude Childrens Research Hospital, Chilis Care tumours (Table I) (Bougeard et al, 2015; Mai et al, 2016).
Center I3312, MS1170, 262 Danny Thomas Place, Memphis, TN Among these, breast cancer is the most common and
38105, USA. accounts for approximately 2530% of all cancers in LFS
E-mail: chimene.kesserwan@stjude.org families (Gonzalez et al, 2009b). Unlike BRCA1- and BRCA2-

2016 John Wiley & Sons Ltd, British Journal of Haematology doi: 10.1111/bjh.14461
Review

Table I. LFS core cancer type known characteristics.

Core cancer type Prevalence in LFS Age distribution (Median) Tumour phenotype

Adrenocortical 613% (Gonzalez et al, 2009a; First 5 years of life (Rodriguez- ACC harbouring somatic ATRX
carcinoma Bougeard et al, 2015; Galindo et al, 2005) (3 years) mutations are larger, more likely to be
Id Said et al, 2016; Fewer than 10% of patients advanced stage and associated with a
Mai et al, 2016) are 15 years or older at diagnosis poorer prognosis (Pinto et al, 2015)
(Ribeiro et al, 2000)
Breast cancer 2731% (Gonzalez et al, 2009a; Pre-menopausal women Invasive ductal carcinoma commonly
Id Said et al, 2016) (33 years) (Olivier et al, 2003) ER and PR positive and/or HER2/neu
amplified (Masciari et al, 2012;
Melhem-Bertrandt et al, 2012;
Bougeard et al, 2015)

CNS tumour (choroid 914% (Gonzalez et al, 2009a; Biphasic with first peak in childhood Medulloblastoma of Sonic hedgehog
plexus carcinoma, Bougeard et al, 2015; and second peak at 2040 years subtype (SHH) (Zhukova et al, 2013)
medulloblastoma, Wasserman et al, 2015; of age (Palmero et al, 2010) and presence of chromothripsis
gliomas) Id Said et al, 2016) (16 years) (Olivier et al, 2003;
Tabori et al, 2010;
McBride et al, 2014)
Soft tissue sarcomas 17827% Prior to age 5 years Non-alveolar, anaplastic
(rhabdomyosarcoma) (Bougeard et al, 2015; (Olivier et al, 2002; rhabdomyosarcoma (Hettmer et al,
Id Said et al, 2016) Palmero et al, 2010) (36 years) 2014), mostly embryonal
(Hettmer et al, 2014) rhabdomyosarcoma (ERMS)

Osteosarcoma 13416% Prior to age 18 years in children No clear phenotypic differences between
(Bougeard et al, 2015; (14 years). Few reported cases OS occurring in individuals with LFS
Id Said et al, 2016) in adults up to age 55 years versus those without LFS
(Bougeard et al, 2015)
Leukaemia 24% (Gonzalez et al, 2009a; Children and young adults Hypodiploid phenotype (Pui et al, 1987;
Bougeard et al, 2015) (12 years) (Bougeard et al, 2015) Nachman et al, 2007; Holmfeldt et al,
2013; Malkin et al, 2014)

ACC, adrenocortical carcinoma; CNS, central nervous system; ER, oestrogen-receptor; LFS, Li-Fraumeni syndrome; OS, osteosarcoma; PR,
progesterone receptor.

associated hereditary breast cancer, which can affect men as of 13416% (Bougeard et al, 2015). RMS often occurs
well as women, breast cancer appears to occur exclusively in before the age of 5 years (Diller et al, 1995; Ognjanovic et al,
women with LFS (Malkin, 1994). In female germline TP53 2012; Hettmer et al, 2014) and OS before the age of 30 (Mir-
mutation carriers, the breast cancer risk increases at approxi- abello et al, 2015). In a study of 15 children with non-alveo-
mately 20 years of age and continues into adulthood, with lar anaplastic RMS, Hettmer et al (2014) found that all five
the median age of breast cancer onset being 33 years (Sorrell children diagnosed under 3 years of age, and six of eight
et al, 2013; Bougeard et al, 2015). Consistent with this find- (75%) diagnosed between 3 and 7 years of age, harboured a
ing, 38% of women with apparently sporadic breast cancer germline TP53 mutation. Thus, children with non-alveolar
who present under the age of 30 years harbour a germline anaplastic RMS, or individuals with early onset OS warrant
TP53 mutation (Mouchawar et al, 2010; McCuaig et al, consideration for LFS, particularly if there is a positive family
2012). Many LFS-associated breast cancers exhibit oestrogen history of cancer. Other STS are rarely encountered, includ-
and progesterone receptor positivity, and HER2/neu amplifi- ing, liposarcoma, leiomyosarcoma and several less well
cation (Masciari et al, 2012; Melhem-Bertrandt et al, 2012; defined subtypes (Ognjanovic et al, 2012).
Bougeard et al, 2015). Malignant phyllodes tumours, uncom- Neoplasms of the CNS account for 914% (Gonzalez et al,
mon fibroepithelial lesions of the breast, are also infrequently 2009a; Sorrell et al, 2013; Bougeard et al, 2015) of LFS
reported (Birch et al, 2001; Villani et al, 2016). tumours with the most common type being glioblastoma/as-
Together, STS and OS comprise 2538% of LFS-related trocytoma. Medulloblastoma, ependymoma, supratentorial
malignancies (Gonzalez et al, 2009a; Ognjanovic et al, 2012) primitive neuroectodermal tumours and choroid plexus car-
and are the most common cancers in children and adoles- cinomas (CPC) are also reported (Sorrell et al, 2013). The
cents. The prevalence of STS has been reported to range age of onset of CNS cancers is biphasic with the first peak in
from 178% to 27%, while OS has a slightly lower prevalence childhood and a second peak at 2040 years of age (median

2 2016 John Wiley & Sons Ltd, British Journal of Haematology


Review

16 years) (Olivier et al, 2003; Tabori et al, 2010; McBride phenotype and that paediatric, but not adult low hypodi-
et al, 2014). There is a strong relationship between LFS and ploid, ALL may be considered a rare manifestation of LFS.
CPC, with up to 100% of children with CPC harbouring a To date, one family presenting with leukaemia as a pri-
germline TP53 mutation, regardless of whether or not there mary manifestation of LFS has been reported. Powell et al
is also a positive family history of cancer (Krutilkova et al, (2013) described a Hispanic kindred in which five individuals
2005; Gonzalez et al, 2009a; Tabori et al, 2010). Among chil- were diagnosed with leukaemia. One case of ALL exhibited
dren with medulloblastoma, those with aberrant activation of doubling of a hypodiploid clone; the cytogenetic findings
the Sonic Hedgehog pathway are most likely to harbour related to the other cases was not discussed. Exome sequenc-
germline TP53 mutations (Zhukova et al, 2013) as are those ing of this kindred revealed a p.R306X germline mutation,
whose tumours contain somatic TP53 mutations or evidence which has been previously identified in both somatic and
of chromothripsis (catastrophic DNA rearrangements) germline samples, generally in association with the more typ-
(Rausch et al, 2012; Zhukova et al, 2013). ical cancers seen in LFS. The unique leukaemia predilection
Adrenocortical carcinoma adevelops in 613% (Kleihues in this kindred suggests the potential existence of an as yet
et al, 1997; Gonzalez et al, 2009a; Bougeard et al, 2015; Mai unidentified genetic modifier associated with the develop-
et al, 2016) of LFS patients with a bi-modal age distribution ment of haematopoietic cancers in the TP53 mutation carri-
peaking before 5 years of age and again in the fourth to fifth ers in this family (Powell et al, 2013).
decades of life. Studies of children with what appears to be In addition to the core cancers, LFS patients develop addi-
sporadic ACC reveal a 3080% prevalence of germline TP53 tional malignancies, many of which occur at earlier than
mutations (Wagner et al, 1994; Libe & Bertherat, 2005; Pinto expected ages, including colorectal, lung, pancreatic, mela-
et al, 2015; Wasserman et al, 2015). The R337H TP53 foun- noma, prostate, kidney, testicular, laryngeal, head and neck
der mutation (see below, Genotype-Phenotype Correlations), and ovarian cancers (Varley et al, 1997; Chompret et al,
which is present in up to 03% of the Southern Brazilian 2000; Nichols et al, 2001; Olivier et al, 2003; Wong et al,
population, is well-recognized to be associated with the 2006; Gonzalez et al, 2009a; Ruijs et al, 2010).
development of ACC in Brazilian children (Ribeiro et al,
2001; Custodio et al, 2012).
LFS and the risk for second primary and
Leukaemia, including acute lymphoblastic leukaemia
therapy-associated cancers
(ALL), acute myeloid leukaemia (AML) and myelodysplastic
syndrome (MDS), exhibit a prevalence of 24% in germline Li-Fraumeni syndrome patients are more likely to develop
TP53 mutation carriers (Gonzalez et al, 2009a; Ruijs et al, multiple primary and therapy-related cancers, a proportion
2010; Bougeard et al, 2015). Lymphoma occurs slightly less of which are haematological. Among 200 patients from 24
often, with a prevalence of 2% (Bougeard et al, 2015). The LFS kindreds reported by Hisada et al (1998), 30 (15%)
median age of onset is 12 (range 235) for leukaemia and developed a second cancer, eight (4%) a third cancer and
13 years (range 242) for lymphoma (Bougeard et al, 2015). four (2%) a fourth cancer. In this study, the relative risk of
There is a paucity of published literature detailing the AML, occurrence of a second cancer was 53 times greater than that
MDS and lymphoma subtypes that occur in individuals with of the general population, with a cumulative probability of
LFS. However, when ALL occurs in the context of LFS, it second cancer development of 57% at 30 years following the
often exhibits a pre-B cell low hypodiploid phenotype in diagnosis of a first cancer. Remarkably, the relative risk of a
which the leukaemic cells contain 3239 chromosomes, second cancer was 83-times higher for individuals with LFS
although other ALL subtypes have also been described who developed a first cancer during childhood. Mai et al
(Holmfeldt et al, 2013; Malkin et al, 2014). In a large geno- (2016) recently reported similar results where about 49% of
mic profiling study involving 124 cases of childhood hypodi- germline TP53 mutation carriers with a first cancer devel-
ploid ALL, Holmfeldt et al (2013) reported that oped at least one other cancer after a median time period of
approximately 912% of the low hypodiploid samples exhib- 10 years.
ited somatic TP53 mutations, the majority of which were Data describing the factors contributing to development
homozygous as a result of loss of the wild-type TP53 allele. of therapy-related cancer in LFS are limited; however, it is
Remarkably, almost half of the studied cases demonstrated probable that these partly result from the carcinogenic effects
presence of the same TP53 mutations within non-tumour of the radiation (RT) and/or chemotherapy used to treat the
cells, suggesting that these were germline alterations. Zhang previous cancer(s). Findings supporting this notion include
et al (2015) also described that 19% of children with hypodi- laboratory data in which RT-treated fibroblasts from LFS
ploid ALL harbour germline TP53 mutations. In the report patients exhibit compromised cell cycle arrest and increased
by Holmfeldt et al (2013), somatic TP53 mutations were also chromosomal aberrations compared to wild-type fibroblasts
observed in 10 of 11 (909%) low hypodiploid ALL samples (Boyle et al, 2001; De Moura et al, 2010). Following a single
obtained from adult patients; however, none had evidence of exposure to 1 or 4 Gy of irradiation, Trp53 heterozygous
a germline TP53 mutation. Together, these findings suggest mice exhibit a marked reduction in tumour latency when
that TP53 mutations are a hallmark of the low hypodiploid compared to wild-type Trp53 animals (Kemp et al, 1994).

2016 John Wiley & Sons Ltd, British Journal of Haematology 3


Review

Consistent with these findings, several case reports and small However, Baker et al (1989) showed that there was muta-
case series describe an increased prevalence of solid cancers tional inactivation of one TP53 allele with loss of the second
in LFS patients who receive RT versus those who do not, wild-type allele in two colorectal cancers studied. Around
with some tumours arising within the radiation field (Hey- this time, Finlay et al (1989) demonstrated that oncogene-
mann et al, 2010; Pierce & Haffty, 2011). Despite these driven cellular transformation could be inhibited by forced
observations, little remains known about the true prevalence expression of wild-type TP53. Collectively these and other
of RT-induced cancers, as well as their relationship to radia- observations revealed that TP53 served as a tumour suppres-
tion dose, age at treatment with RT, and/or the tissue being sor and not as an oncogene as originally proposed.
radiated. In unstressed cells, TP53 levels are kept low via a nega-
Germline TP53 mutations may also contribute to therapy- tive-regulatory feedback mechanism mediated by the E3
associated leukaemias and MDS. Towards this end, a study ubiquitin ligase Mouse Double Minute 2 homolog (MDM2),
of 53 patients with therapy-associated myeloid neoplasms whose expression is induced by TP53. MDM2 binds to the
revealed four (75%) harboring germline TP53 mutations N-terminal domain of TP53 and covalently attaches ubiqui-
(Schulz et al, 2012). Similarly, a study of 47 breast cancer tin, thus marking TP53 for degradation by nuclear and cyto-
survivors with secondary leukaemia identified three (6%) plasmic proteasomes (Fig 2). Accordingly, in normal cells,
individuals with germline TP53 alterations (Churpek et al, TP53 is very unstable, with a half-life ranging from 5 to
2016). In the latter study, two of these three individuals 30 min (Moll & Petrenko, 2003). Following exposure to
developed ALL, an unusual treatment-related haematopoietic genotoxic stressors, such as ionizing radiation, oxidative
malignancy. Compared to primary ALL developing in chil- stress and nucleotide depletion, TP53 and MDM2 become
dren with germline TP53 mutations, these two cases of pre- phosphorylated. As a result, the MDM2-TP53 interaction is
sumed therapy-related leukaemia did not exhibit a weakened. This weakening lessens TP53 degradation, allow-
hypodiploid phenotype. Hence, for cancer patients who ing TP53 to accumulate, self-associate and bind to specific
develop secondary ALL, regardless of its cytogenetic make- DNA sequences. Functional TP53 tetramers then induce the
up, consideration should be given to germline TP53 testing expression of numerous downstream target genes that regu-
(for further discussion see below, Clinical Management). late critical cellular processes, such as cell cycle arrest, apop-
tosis and DNA repair.
The TP53 gene and its encoded protein
The role of TP53 in the haematopoietic
TP53 is located at chromosome 17p13.1 and spans approxi-
compartment
mately 20 kb of genomic DNA. It is composed of 14 exons
including ten that encode the full length 393-amino acid The haematopoietic compartment exhibits a remarkable and
TP53 protein, one non-coding exon (exon 1) and three alter- dynamic capacity for regeneration. Under normal conditions,
native exons (exons 2/3, 9b, 9c) (Hainaut & Pfeifer, 2016). haematopoietic stem cells (HSC), the precursors from which
The TP53 functional domains consist of an N-terminal tran- all blood cells arise, remain quiescent with only a small frac-
scriptional activation domain, proline-rich region, DNA tion entering the cell cycle and giving rise to differentiating
binding domain, tetramerization domain and C-terminal reg- progenitors that replenish cells that are lost due to normal
ulatory domain (Fig 1) (Beckerman & Prives, 2010). It was processes, such as aging. In contrast, under conditions of
initially proposed that TP53 functioned as an oncogene given stress, such as trauma or exposure to cytotoxic therapies,
the finding that many tumours expressed increased levels of HSC must proliferate to maintain and/or expand their pool
the TP53 protein (Rotter, 1983; Eliyahu et al, 1984). while replacing those cells that are actively damaged or lost.

Transcriptional activation TP53 DNA-binding Nuclear Regulation


domain domain localization signal domain

50 100 150 200 250 300 350

Proline-rich region Zinc binding site Oligomerization domain

Fig 1. Schematic depicting the TP53 protein (Zhou et al, 2016). TP53 is comprised of 393 amino acids and contains several functional domains,
including: (i) N-terminal transcriptional activation domain; (ii) Proline rich region; (iii) DNA binding domain; (iv) Oligomerization domain,
which facilitates the formation of TP53 dimers and tetramers; and (v) C-terminal domain involved in down-regulation of DNA binding. TP53
also contains critical zinc binding sites, which enables the proper protein folding that is required for site-specific DNA binding and transcriptional
activation. The nuclear localization signal facilitates movement into the nucleus, where TP53 can exert its effects. Numbers represent position of
amino acid residues from N- to C-terminus. (Figure re-drawn from https://pecan.stjude.org/proteinpaint/TP53.)

4 2016 John Wiley & Sons Ltd, British Journal of Haematology


Review

UV Radiation Hypoxia

DNA Damage Oncogenes

TP53 Degradation

Wt- Wt-
TP53 TP53
Wt- Wt- Mut- Mut-
TP53 TP53 TP53 TP53
Wt- Wt-
TP53 TP53
Wt- Wt- Wt- Wt-
TP53 TP53 TP53 TP53
Wt- Mut-
TP53 TP53

MDM2 MDM2
DNA Repair
Wt- Wt- Mut- Wt-
TP53 TP53 TP53 TP53
Normal Impaired
Cell Cycle Arrest Function Function
Wt- Wt- Wt- Mut-
TP53 TP53 TP53 TP53

Apoptosis

(A) (B)

Fig 2. Functions of TP53. (A) In response to cellular stress signals, wild-type TP53 tetramers bind to DNA (in green) and trigger the activation
of genes responsible for a myriad of functions including DNA repair, cell cycle arrest, apoptosis and senescence. During the course of these activi-
ties, TP53 induces expression of MDM2, leading to TP53 degradation through proteasomal degradation. (B) In contrast to wild-type TP53,
mutant TP53 proteins cannot bind effectively to DNA (in grey), resulting in impaired functions, including poor induction of MDM2 and hence
TP53 accumulation.

Within the haematopoietic compartment, TP53 is mainly In addition to its role as a regulator of HSC quiescence
expressed in HSC, where it is essential for maintaining HSC and self-renewal capacity, TP53 is important for regulating
quiescence, self-renewal and genomic integrity (Asai et al, HSC fitness, where too little or too much TP53 activity can
2011; Pant et al, 2012). Evidence for the importance of compromise the ability of HSC to repopulate the marrow. As
TP53 in HSC quiescence comes from the study of mice in noted above, Trp53-null HSC exhibit increased proliferative
which Trp53 or the genes encoding TP53 regulators such as and short term repopulating capacity. Nonetheless, they are
Mdm2 have been deleted, overexpressed or mutated. inferior to their Trp53-sufficient counterparts when it comes
Towards this end, Trp53-null HSC enter the cell cycle more to long-term repopulation (Asai et al, 2011). Similarly, too
readily than wild-type cells (Liu et al, 2009). Consistent much TP53 is detrimental. The latter is made evident in
with these findings, Trp53-null mice exhibit a two- to mice with high basal levels of TRP53 activity, such as those
three-fold increase in Lineage Sca-1+ c-kit+ (LSK) cells (a harbouring additional copies of the Trp53 gene or animals
bone marrow population containing HSC) as well as haploinsufficient for the TP53 inhibitor Mdm2, which exhibit
CD150+ CD48 LSK, which are enriched for long term enhanced radiosensitivity and poor haematopoietic recovery
repopulating cells (Liu et al, 2009; Asai et al, 2011). Addi- following exposure to low levels of ionizing radiation. Collec-
tional proof that TP53 is important in HSC comes from tively, these and other studies reveal that TP53 is a critical
bone marrow transplantation studies in which TP53-defi- protein whose levels and activity must be carefully regulated
cient bone marrow cells out compete TP53-expressing cells within HSC to prevent leukemogenesis and promote home-
in competitive repopulation assays (Liu et al, 2009). Based ostasis of the haematopoietic compartment under normal
on these findings, it has been suggested that the transient and stressful conditions (Asai et al, 2011; Pant et al, 2012).
inhibition of TP53 function could be used to promote bone
marrow recovery following chemo- or radiation therapy by
Mechanism by which TP53 contributes to
allowing for increased HSC proliferation and/or self-
tumour formation
renewal. Indeed, small molecule inhibitors known as
pififthrins are under development for these purposes (Asai There are several proposed mechanisms by which the mutant
et al, 2011). TP53 protein promotes tumour formation. These

2016 John Wiley & Sons Ltd, British Journal of Haematology 5


Review

mechanisms include the loss of its tumour suppressor func- regulation of the ATP binding cassette subfamily B member
tion (LoF) or conversely, the acquisition of dominant nega- 1 gene (ABCB1; also termed multidrug resistance 1 gene,
tive (DN) or gain of function (GoF) effects. The loss of MDR1) by mutant TP53, which confers drug resistance to
tumour suppressor function is caused by reduced TP53 cancer cells harbouring somatic TP53 mutations (Chin et al,
expression or altered TP53 function. Reduced expression can 1992). Additional genes that are aberrantly regulated by
result from genomic deletions that remove all or part of mutant TP53 include NFKB1, MYC, EGR1 and TERT, among
TP53, while altered function stems from missense mutations others (Brosh & Rotter, 2009). Therefore, whereas wild-type
that affect its functional domains. Consistent with the latter, TP53 regulates the expression of genes that mediate growth
many LFS-associated TP53 mutations reside within the criti- suppression, apoptosis and DNA repair, mutant TP53,
cal DNA binding domain. These mutations produce mutant through mechanisms that are not completely defined, regu-
TP53 proteins with impaired DNA binding and thus altered lates the expression of genes that enable cell proliferation,
regulation of downstream target genes. drug-resistance, survival and metastasis.
Missense mutations in TP53 can also exert a DN effect in Regardless of the mechanism of action, mutations affecting
which the mutant TP53 protein physically interacts with and TP53 perturb its many functions, leading to aberrant tran-
inhibits the function of wild-type TP53. This property was scription, altered MDM2-TP53 interactions, mutant TP53
initially shown by Milner et al (1991), where in vitro co- accumulation, deregulated cell cycle progression and
translation of wild type and mutant TP53 enabled the forma- impaired mechanisms of DNA repair (Fig 2).
tion of complexes containing both proteins. Willis et al
(2004) provided further insights into the properties of these
The spectrum of TP53 mutations in individuals
aberrant complexes. By examining the functions of cell lines
with LFS
expressing both wild-type and mutant TP53, these investiga-
tors demonstrated that the presence of mutant TP53 greatly The distribution of TP53 germline mutations in LFS is simi-
reduced the ability of the wild-type protein to bind to pro- lar those identified in tumours, with the majority clustered
moters of TP53-responsive target genes. Indeed, the DN within the DNA binding domain where there are six recur-
effect may explain why some TP53 mutation carriers exhibit rent hotspot mutations involving codons 175 (R175H), 245
retention of the wild-type TP53 allele within their tumours. (G245S), 248 (R248Q, R248W), 273 (R273H) and 282
Towards this end, Varley et al (1997) demonstrated that only (R282W) (Wasserman et al, 2015). Functional studies reveal
57% of tumours from patients with germline TP53 muta- that, among the mutations most commonly examined, the
tions showed loss of the wild-type allele and retention of the majority exerts a DN effect (Petitjean et al, 2007). Mutations
mutated allele. A subset of these patients was also analysed in intronic or regulatory regions are also reported; however,
by Birch et al (1998), who noted that all tumours that their functional significance remains to be elucidated. The
retained both TP53 alleles developed in individuals who har- International Agency for Research on Cancer (IARC) main-
boured germline missense mutations within the DNA bind- tains a well-established repository of somatic and germline
ing domain. This was in contrast to individuals harbouring TP53 mutations. The current release (R18, April 2016, http://
null mutations, where loss of both alleles was invariably seen p53.iarc.fr) includes 885 families harbouring 360 unique
in the tumours (Birch et al, 1998). TP53 mutations. Among the germline TP53 mutations
The GOF effect is characterized by acquisition of novel included, missense alterations constitute about 73%, non-
properties by mutant TP53 independent of the presence of sense 889%, splice site 816%, frameshift 566%, silent
the wild-type TP53 protein. Early evidence supporting the 055%, large deletions 073%, intronic 061% and other
oncogenic function of mutant TP53 was derived from the mutations (complex rearrangement, insertions, deletions)
observation that introduction of mutant TP53 into Trp53- 207%.
null cells allowed for tumour outgrowth in mice. Cells com-
pletely devoid of TP53 form tumours that spontaneously
Genotypephenotype correlations in LFS
regress. In contrast, cells expressing mutant TP53 exhibit
lethal outgrowth (Wolf et al, 1984). Brosh and Rotter (2009) In keeping with experimental data that not all TP53 mutants
elegantly review the properties by which mutant TP53 medi- function equally, several genotype-phenotype studies involv-
ates the GOF effect. One key mechanism involves the inacti- ing LFS families have suggested that the type of germline
vation of TP63/TP73 (p63/p73) proteins by mutant TP53; TP53 mutation may serve as a predictor of tumour type and
TP63 and TP73 are considered TP53-related proteins. When age of cancer onset. Birch et al (1998) examined 34 families
overexpressed, TP63/TP73 induce growth arrest and apopto- meeting classic clinical criteria for LFS and observed that
sis in a TP53-like manner. Mutant TP53 binds to TP63/TP73 families harbouring missense mutations within the TP53
and aborts their ability to dampen cell proliferation and pro- DNA binding domain had a more highly penetrant pheno-
mote apoptosis. Another important mechanism involves the type than families with other (nonsense or truncating) or no
modulation of expression specific target genes by mutant mutations. Most recently, Bougeard et al (2015) observed a
TP53. Once such example includes transcriptional up significantly earlier age of tumour onset for patients with

6 2016 John Wiley & Sons Ltd, British Journal of Haematology


Review

missense mutations (mean age of tumour onset 238 years) individuals harbouring the heterozygous G/T or homozygous
compared to those with frameshift or in-frame deletions/in- G/G genotype developed STS an average of 12 years earlier
sertions, nonsense mutations and genomic rearrangements and breast cancer 10 years earlier than those carrying the
(285 years). The difference was even greater when missense homozygous wild-type T/T genotype. In LFS patients with
mutations were compared to genomic rearrangements corre- STS, the number of primary tumours was also higher in
sponding to extreme forms of null alterations (358 years). those with SNP309G/T or G/G. Mechanistic studies have
Additionally, patients whose mutations were known to exert revealed that the G allele for SNP309 results in higher levels
a DN effect exhibited an even earlier age at cancer onset of MDM2 mRNA and protein, resulting in increased degra-
(213 years). Interestingly, ACC appear to be the only type of dation of TP53 and thus defective TP53-mediated apoptosis
LFS-associated cancer consistently associated with germline (Bond et al, 2004, 2006). The early age at cancer onset con-
mutations occurring outside the DNA-binding domain, with ferred by MDM2 SNP309 is further amplified by presence of
the majority of patients carrying mutations that are not a common polymorphism in codon 72 of TP53
dominant-negative missense mutations (Bougeard et al, (NM_000546.5 (TP53): c.215C>G ;rs1042522) in which the
2015). proline (P) at codon 72 is replaced by arginine (R). Thus,
The concept that different germline TP53 alterations con- compared to TP53 containing P72, TP53 R72 binds MDM2
fer variable oncogenic potential is further illustrated in the with higher affinity, resulting in enhanced degradation. The
unique phenotype of carriers of the TP53 R337H mutation, presence of MDM2 SNP309 and TP53 R72 was shown to
which exhibits a high prevalence as a founder mutation in have a cumulative effect and associated with earlier age of
Southern Brazil (Ribeiro et al, 2001). This mutation is cancer onset (Malkin, 2011).
located within the tetramerization domain and was first iden- Recently, a second MDM2 polymorphism, denoted
tified by its strong association with an increased risk for SNP285G>C (rs117039649), was reported to be in linkage
childhood ACC. A recent study showed that among 292 chil- with the SNP309G allele. In a large study of 195 LFS
dren diagnosed with paediatric cancer in Southern Brazil, 11 patients, this haplotype was shown to influence cancer onset
(37%) carried TP53 R337H, nine of whom were diagnosed with cancers occurring 5 years earlier in those with the
with ACC and two with CPC (Giacomazzi et al, 2013). The MDM2 285309 GG haplotype compared to other haplo-
fact that none of the carrier parents in this study had cancer types (Renaux-Petel et al, 2014). In this study, the interaction
at the time of writing of this report suggested that the TP53 between this MDM2 haplotype and the TP53 codon 72 poly-
R337H mutation was associated with a lower penetrance morphism was not examined. Additional genetic factors have
compared to other germline TP53 mutations. Although ini- been reported as potential modifiers of the LFS phenotype,
tial reports suggested that the main cancer risk associated including a 16-base pair insertion/duplication in intron 3 of
with the TP53 R337H mutation was the development of TP53 (rs17878362) (Marcel et al, 2009), a SNP within a
childhood ACC, more recent studies reveal that this muta- microRNA known as miR-605 (rs2043556) (Id Said et al,
tion can be also be identified in patients with CPC, OS, 2016), and the presence of germline DNA copy number vari-
breast cancer and, more recently, neuroblastoma (Giacomazzi ants (Malkin, 2011). The extent to which these modifiers
et al, 2013; Seidinger et al, 2015; Andrade et al, 2016). All reliably and consistently influence LFS phenotype remains to
told, it is likely that TP53 R337H predisposes to the more be determined.
typical cancers seen in LFS, albeit with a lower penetrance. To date, there are no reports of genetic modifiers linked
specifically to the development of haematopoietic cancers in
individuals with LFS.
Genetic modifiers influencing the phenotype in
germline TP53 mutation carriers
Genetic testing for LFS
The variability in age of onset and type of cancers occurring
in TP53 mutation carriers, even those within the same fam- Following the discovery of TP53 as the causative gene in LFS
ily, has supported the notion that factors other than the in 1990, there was caution regarding the provision of germ-
germline TP53 mutation influence disease presentation. line genetic testing because, at the time, there was limited
Indeed, many studies have sought specific genetic modifiers understanding of how genetic test results would influence
and examined their effect on disease penetrance and expres- clinical management and significant concerns regarding the
sivity, as well as on the levels or function of mutant TP53. In psychological effects associated with a positive test result.
this regard, a single nucleotide polymorphism (SNP) within With time, however, a better understanding has emerged
the first intron of MDM2 (NM_002392.3:c.14+309T>G; regarding the natural history of LFS and the possible
SNP309; rs2279744) has been associated with an earlier age approaches to tumour surveillance and cancer risk reduction.
of tumour onset in LFS (Bond et al, 2004; Bougeard et al, In addition, studies of the psychological and emotional con-
2006; Ruijs et al, 2007). Bond et al (2004, 2006) studied 88 sequences of testing, while limited, have revealed that adults
individuals with LFS and demonstrated an accelerated age of undergoing LFS testing do not experience long-term adverse
cancer onset in association with SNP309. Specifically, psychological consequences (Lammens et al, 2010a).

2016 John Wiley & Sons Ltd, British Journal of Haematology 7


Review

Accordingly, pre-symptomatic and diagnostic germline TP53 fact that some individuals with LFS do not necessarily need a
testing are now more widely accepted by families and health- positive family of cancer in order to warrant consideration of
care providers. Given the clinical and psychological complex- germline TP53 testing. The revised Chompret criteria recom-
ities of LFS, it is strongly recommended that genetic testing mend testing for a proband with: (i) an LFS-type tumour
for this condition be completed by haematologists, oncolo- diagnosed before the age of 46 years and at least one-first or
gists, genetics professionals or other providers who are famil- second degree relative with an LFS-type tumour before the
iar with this condition and only in the context of pre- and age of 56 years or with multiple primary tumours; (ii) multi-
post-test genetic counselling (Malkin, 2011). ple primary tumours diagnosed before the age of 46 years,
One of the key issues to be contemplated when offering two of which are LFS component tumours, regardless of the
germline TP53 genetic analysis to a patient with leukaemia family history; or (iii) an ACC or a CPC at any age, regard-
or MDS is that testing cannot always be performed using a less of the family history (Tinat et al, 2009).
sample of blood. In patients with active disease or positive Currently, germline TP53 testing is most commonly con-
minimal residual disease (MRD) status, it may be difficult to sidered for individuals meeting classic or Chompret criteria
determine whether an identified mutation is constitutional or or to any females with early-onset breast cancer who lack a
originating from circulating neoplastic cells. Indeed, with the BRCA1 or BRCA2 mutation. As noted above, children with
increased sensitivity of next generation sequencing, it is now low hypodiploid ALL or any individuals with therapy-
possible to detect mutations present at a frequency of 10%
or less. Therefore, presence of a TP53 mutation could reflect Table II. Li-Fraumeni syndrome diagnostic and testing criteria.
germline mosaicism or alternatively, presence of a leukaemic
Classic LFS criteria (Li et al, 1988)
or MDS clone. Saliva samples and cells obtained by buccal
Proband diagnosed with sarcoma before age 45 years
swabbing are often considered as alternative sources of DNA AND first degree relative with a cancer diagnosed before age
for genetic testing. Unfortunately, neoplastic cells may also 45 years
contaminate these samples. Ideally, testing should involve the AND a first-degree or second-degree relative with any cancer
analysis of skin fibroblasts obtained from a punch skin with onset before age 45 years
biopsy. This procedure is more invasive and requires 6 OR a sarcoma at any age
8 weeks to generate cells for testing; however, it provides the 70% probability of identifying a germline TP53 mutation
(Varley, 2003)
best chances for the most accurate diagnosis.
Birch LFL criteria (Birch et al, 1998)
Proband with any childhood cancer, sarcoma, brain tumour or
LFS classification criteria adrenocortical carcinoma with onset less than 45 years of age
AND a first-degree relative with either breast cancer, sarcoma,
To facilitate the identification of individuals and families brain tumour, adrenocortical carcinoma or leukaemia with onset at
with suspected LFS and provide guidelines for germline TP53 any age
genetic testing, several classification systems have been devel- AND a first-degree relative with any cancer with onset before
60 years of age
oped (Table II). According to the classic and most stringent
2040% probability of identifying a germline TP53 mutation
criteria, LFS is defined by the presence of: (i) a proband with (Varley, 2003)
a sarcoma diagnosed before the age of 45 years; (ii) a first Eeles LFL criteria (Eeles, 1995)
degree relative with any cancer before the age of 45 years; Two first-degree or second-degree relatives with either breast
and (iii) a first- or second-degree relative with any cancer cancer, sarcoma, brain tumour, or leukaemia at any age
before the age of 45 years or a sarcoma at any age (Li et al, 2040% probability of identifying a germline TP53 mutation
1988). More inclusive classification systems have been created (Varley, 2003)
for those families that do not meet classic criteria, but 2009 Chompret criteria (Tinat et al, 2009)
Proband with core LFS tumour (premenopausal breast cancer,
nonetheless exhibit an LFS-like or LFL phenotype. The first
soft tissue sarcoma, osteosarcoma, brain tumour, leukaemia
was reported by Birch et al (1994), and includes families in or adrenocortical carcinoma) before 46 years of age
which the proband has: (i) any childhood cancer or a sar- AND at least one-first-degree or second-degree relative with
coma, CNS tumour or ACC diagnosed before the age of LFS-core tumour (except breast cancer if the proband has
45 years; (ii) a first or second degree relative with an LFS- breast cancer) before 56 years of age
type tumour at any age; and (iii) a first or second degree rel- OR with multiple tumours
ative with any cancer before the age of 60 years. The second Proband with multiple tumours (except multiple breast tumours),
two of which are core LFS tumour types, first of which had to
was reported 1 year later (Eeles, 1995) and includes families
occur prior to age 46 years
with two-first or second degree relatives with an LFS tumour Any patient with adrenocortical carcinoma or choroid plexus
at any age. A more recent classification scheme, was put tumour irrespective of family history.
forth by Chompret et al (2001) and revised in 2009 (Tinat 8295% sensitivity and 4758% specificity (Gonzalez et al, 2009a;
et al, 2009). Although more restrictive than the Birch or Tinat et al, 2009; Ruijs et al, 2010)
Eeles criteria in terms of the cancer types and ages of onset
in the proband, the Chompret criteria take into account the LFL, Li-Fraumeni syndrome-like; LFS, Li-Fraumeni syndrome.

8 2016 John Wiley & Sons Ltd, British Journal of Haematology


Review

associated leukaemia or MDS also warrant strong considera- in conjunction with frequent blood work and imaging studies
tion, especially when there is a personal or family history of (Table III). A total of 40 asymptomatic tumours were detected
LFS-associated cancers. As might be expected, the chances of in 19 (32%) of 59 patients who underwent surveillance, while
finding a germline TP53 mutation differ depending upon the 61 symptomatic tumours were detected in 43 (88%) of the 49
criteria used to guide testing, with about 6080% of individ- patients who initially declined surveillance (19 of these subse-
uals meeting classic and 2030% meeting Chompret criteria quently crossed over to the surveillance arm). Notably, the 5-
harbouring a germline TP53 mutation (Olivier et al, 2003; year survival was 88% for those undergoing and 596% for
Varley, 2003; Mai et al, 2012). Most LFS mutations are those declining surveillance (Villani et al, 2011). The results of
inherited with the frequency of de novo mutations at around this study are promising and provide the first evidence in sup-
720% (Gonzalez et al, 2009b). port of the potential survival benefits of surveillance for carri-
Given the observation that a sizable proportion of individu- ers of germline TP53 mutations. Nonetheless, this screening
als with an LFS or LFS-like phenotype lack identifiable germ- protocol is not without its limitations, with one patient having
line TP53 mutations, it is very likely that other genetic false negative and two patients false positive results. Further-
aberrations exist to explain cancer development. These aberra- more, there were a number of incidental findings identified by
tions might include mutations in non-coding regions of the whole body magnetic resonance imaging (WBMRI; a compo-
genome that negatively influence TP53 expression or, alterna- nent of the screening protocol), the majority of which required
tively, mutations in genes other than TP53 itself. Therefore, dedicated follow-up imaging. Currently, several trials are
families who test negative for germline TP53 mutations, ongoing to further investigate the utility of WBMRI with or
including those presenting with leukaemia as the sole manifes- without other screening modalities as a means of tumour
tation, should be carefully evaluated and offered testing for surveillance in LFS and to identify the optimal types and fre-
other cancer predisposing conditions, where appropriate. quencies of screening procedures (Villani et al, 2016).
Familial testing for LFS may identify asymptomatic indi- In the report by Villani et al (2016), two individuals devel-
viduals who carry a germline TP53 mutation. Such informa- oped AML and two MDS. Only one of these survived: an adult
tion has significant health implications and these individuals with MDS who chose not to undergo surveillance. Currently,
should be offered surveillance and for women, the possibility it remains unknown whether surveillance for haematopoietic
of prophylactic mastectomy, as would be done for TP53 malignancies confers a medical benefit for those affected with
mutation carriers who have already developed one or more LFS. Nonetheless, consideration should be given to regular
cancers (Villani et al, 2016). monitoring of peripheral blood counts. In addition, patients
For individuals of reproductive age, genetic counselling should undergo periodic physical examinations to evaluate for
should be offered to discuss recurrence risks. Parents with an the signs and symptoms of a haematopoietic cancer, such as
identified germline TP53 mutation can consider prenatal test- fever, pallor, petechiae, easy bruising, lymphadenopathy and
ing, which includes analysing DNA extracted from fetal cells hepatosplenomegaly. If any of these are present, or if there are
obtained by amniocentesis at 1518 weeks gestation or by persistent changes in the blood counts from baseline, a bone
chorionic villus sampling at ten to 12 weeks gestation, as well marrow aspirate and biopsy should be performed.
as pre-implantation genetic diagnosis. It is important to recognize the emotional, psychological
and financial burdens that frequent screening evaluations cre-
ate. These tests require multiple visits to the clinic and are
Cancer surveillance in LFS
fraught with the possibility of false positive results. In a study
The goals of cancer surveillance are to improve overall out- of high-risk women who were proven germline TP53 muta-
comes by enabling the early detection and treatment of tion carriers and those at 50% risk, Lammens et al (2010b)
tumours. In general, surveillance is best suited for solid neo- observed that 90% of participants surveyed believed in the
plasms, where prognosis may be better following complete value of surveillance to detecting early stage tumours. Fur-
resection of the presenting tumour. Indeed, such is the case thermore, 84% stated that surveillance gave them a sense of
with many LFS-associated solid tumours including certain sar- control and 70% indicated a sense of security. In this study,
coma subtypes, glioma, medulloblastoma, CPC and ACC there were no significant differences in the levels of distress
(McBride et al, 2014). Smaller completely resected tumours or worry between women who chose and those who did not
may require reduced or even no chemo- or radiation therapy. choose to undergo surveillance as recommended by their
In theory, this could minimize the risk for therapy-associated health care providers. While limitations exist in terms of
second cancers. Although the diversity of cancer types and age sample size, these findings are reassuring. As pertains to chil-
at cancer onset make screening in LFS challenging, Villani et al dren, there is concern that frequent screening may increase
(2016) recently reported that a multi-modal screening proto- anxiety and reduce the quality of life (McBride et al, 2014).
col is feasible and associated with a significantly improved sur- Further research is needed to objectively evaluate these con-
vival for individuals with LFS. Using what is now referred to cerns, as children and adolescents are at a formative, yet psy-
as the Toronto protocol, these investigators monitored carri- chologically vulnerable period in their lives and may require
ers of a germline TP53 mutation using physical examination life-long cancer monitoring.

2016 John Wiley & Sons Ltd, British Journal of Haematology 9


Review

Table III. Villani et al (2016) surveillance protocol based on tumour prophylactic mastectomy (Thull & Vogel, 2004). For those
type. who develop a breast cancer, mastectomy should be encour-
Adrenocortical carcinoma aged over lumpectomy to lessen the risks for development of
 Children (birth to 18 years old) and adults (1840 years old) new primary breast cancers or radiation-induced secondary
Ultrasound abdomen and pelvis every 34 months malignancies (Schneider et al, 1993).
Blood/urine tests to be completed every 34 months There are no consensus recommendations regarding the
acute management of solid cancers in patients with germline
17 OH-progesterone, total testosterone, dehydroepiandros-
terone sulphate, androstendione, 24-h urine cortisol TP53 mutations. When possible, it is prudent to reduce
Brain tumour
exposure to ionizing radiation to help decrease the risk for
secondary tumours within the radiation field. Concern also
 Children and Adults
exists regarding the choice of chemotherapeutic agents, par-
Annual Brain MRI
ticularly alkylating agents, whose use is associated with the
Breast cancer possible risk of inducing leukaemia or myelodysplasia (Felix
 Adults et al, 1996; Schulz et al, 2012; Kamihara et al, 2014). Regard-
Monthly breast self-examination (18 years +) less of these risks, it is currently recommended that cure of
Clinical breast examination, annual mammography and breast the existing malignancy must remain the top priority.
MRI screening* In terms of LFS-associated leukaemia, the treatment of
Twice a year starting at age 20 OR hypodiploid ALL has been challenging, with survival rates of
510 years before earliest known breast cancer in family 50% or less despite the use of intensive chemotherapeutic
Soft tissue and bone sarcoma regimens (Pui et al, 1987; Nachman et al, 2007). Further-
 Children more, until recently, there were no known prognostic mark-
Annual rapid whole body MRI ers to help direct therapy. Mullighan et al (2015) reported
that the outcome of children with hypodiploid ALL could be
 Adults (18 years+) stratified based on the MRD level at the end of leukaemia
Annual rapid whole body MRI induction (Mullighan et al, 2015). In this study, 20 children
Ultrasound of abdomen and pelvis every 34 months
with hypodiploid ALL, including 12 with the low hypodi-
Leukaemia or lymphoma ploid subtype (only one of whom had a documented germ-
 Children and adults line TP53 mutation) were treated according to the St Jude
Blood tests to be completed every 34 months TOTAL Therapy Studies. Patients with MRD 1% after com-
Complete blood counts, erythrocyte sedimentation rate, pletion of induction were offered the option of allogeneic
lactate dehydrogenase haematopoietic stem cell transplantation (allo-SCT).
Colorectal cancer Although all 20 patients achieved a clinical remission at the
 Adults end of induction, only 14 were found to have a negative
Colonoscopies every 2 years starting at age 25 years OR

MRD status. Survival was significantly better for those with
10 years before earliest known colon cancer in the family negative MRD (855%) versus those with a positive result
Melanoma (444%). The one patient with a germline TP53 mutation
 Adults experienced a relapse. Although numbers are small, this
Annual dermatological examination study suggests that, as with other forms of ALL, MRD serves
an important prognostic indicator for childhood hypodiploid
General assessment
ALL, an aggressive and very difficult-to-treat leukaemia sub-
 Adults and children type.
Complete physical examination every 34 months The published literature as to whether the presence of a
Prompt medical assessment with primary care doctor for any germline TP53 mutation confers a poorer prognosis in
concerns patients with haematopoietic cancers is limited. Similarly,
there is little information regarding the optimal treatment
MRI, magnetic resonance imaging.
approaches for primary or therapy-related disease in germ-
*Breast MRI to alternate with annual rapid whole-body MRI.
line TP53 mutation carriers. At present, it is not clear
whether treatment regimens should be altered to avoid or
lessen exposure to DNA damaging chemotherapeutic agents,
Recommendations for cancer prevention and
as is done with patients who have Fanconi anaemia or Ataxia
treatment
Telangiectasia. In addition, it remains to be determined
Currently there are limited options for cancer prevention in whether allo-SCT will improve the outcomes for patients
LFS. Given the high risk for breast cancer development in with relapsed or refractory leukaemia. When allo-SCT is
women with LFS, females harbouring a germline TP53 muta- being considered, it is important to offer relatives targeted
tion should be counselled about the option of risk-reducing TP53 genetic testing prior to harvesting to avoid the

10 2016 John Wiley & Sons Ltd, British Journal of Haematology


Review

possibility of collecting and inadvertently transplanting TP53 phenotype may permit the development of individualized
mutation-containing stem cells. Finally, it was recently monitoring protocols. Finally, with the advent of genome
reported that low hypodiploid ALL cells exhibit activation of editing and induced pluripotent stem cell technologies, it is
Ras and PI3K pathways and are susceptible to inhibition of now possible to generate and possibly reverse specific germ-
PI3K and/or mTOR signalling (Holmfeldt et al, 2013). These line TP53 mutations. These technologies will allow for the
data suggest that therapeutic targeting of these pathways modelling of tumour formation in LFS and the dissection of
could prove beneficial as a treatment for patients with low critical cellular pathways and processes that can be then tar-
hypodiploid ALL, positive MRD and/or recalcitrant disease. geted to treat or even prevent tumour formation. Looking to
the future, there is no doubt that LFS will remain as a para-
digm for the exploration and understanding of TP53-asso-
Conclusions and future directions
ciated solid and blood cancers and heritable cancer risk.
Much has been learned about TP53 and its central role in
maintaining genomic stability and suppressing malignant
Acknowledgements
transformation. Nonetheless, it is impossible to predict with
certainty when and which TP53 mutation carriers will We gratefully acknowledge Brandon Stelter from the Depart-
develop a first or subsequent cancer, how to delay or prevent ment of Biomedical Communications at St. Jude, who helped
these cancers from occurring, and how to treat them most create the images in Figs 1 and 2. We also thank Gerry Zam-
effectively. Future collaborative studies that integrate clinical, betti, Emilia Pinto, Emily Quinn, Rose McGee, Regina Nuc-
genomic and biological information will undoubtedly shed cio, Kayla Hamilton, Victoria Liddle and Stacy Hines-Dowell
light onto this challenging cancer predisposition syndrome. for their careful review of this manuscript.
Towards this end, the increased application of high through-
put molecular approaches will provide new insights into the
Author contributions
genetic and epigenetic events that drive tumour formation.
This information may also help to explain the diverse mani- All authors contributed equally to the design of the study
festations of LFS, including the differences in disease pene- and writing of this review. All authors critically reviewed
trance, heterogeneity of tumours formed and the disparity in the manuscript and agreed to submit the paper for publi-
age of cancer onset among germline TP53 mutation carriers. cation.
A better understanding of how these factors influence disease

References Birch, J.M., Blair, V., Kelsey, A.M., Evans, D.G., polymorphism on age of tumour onset in Li-
Harris, M., Tricker, K.J. & Varley, J.M. (1998) Fraumeni syndrome. Journal of Medical Genetics,
Andrade, K.C., Santiago, K.M., Fortes, F.P., Mam- Cancer phenotype correlates with constitutional 43, 531533.
belli, L.I., Nobrega, A.F. & Achatz, M.I. (2016) TP53 genotype in families with the Li-Fraumeni Bougeard, G., Renaux-Petel, M., Flaman, J.M.,
Early-onset breast cancer patients in the South syndrome. Oncogene, 17, 10611068. Charbonnier, C., Fermey, P., Belotti, M., Gau-
and Southeast of Brazil should be tested for the Birch, J.M., Alston, R.D., McNally, R.J., Evans, thier-Villars, M., Stoppa-Lyonnet, D., Con-
TP53 p. R337H mutation. Genetics and Molecu- D.G., Kelsey, A.M., Harris, M., Eden, O.B. & solino, E., Brugieres, L., Caron, O., Benusiglio,
lar Biology, 39, 199202. Varley, J.M. (2001) Relative frequency and mor- P.R., Bressac-de Paillerets, B., Bonadona, V.,
Asai, T., Liu, Y., Bae, N. & Nimer, S.D. (2011) phology of cancers in carriers of germline TP53 Bonaiti-Pellie, C., Tinat, J., Baert-Desurmont, S.
The p53 tumor suppressor protein regulates mutations. Oncogene, 20, 46214628. & Frebourg, T. (2015) Revisiting Li-Fraumeni
hematopoietic stem cell fate. Journal Cell Physi- Bond, G.L., Hu, W., Bond, E.E., Robins, H., Lutz- syndrome from TP53 mutation carriers. Journal
ology, 226, 22152221. ker, S.G., Arva, N.C., Bargonetti, J., Bartel, F., of Clinical Oncology, 33, 23452352.
Baker, S.J., Fearon, E.R., Nigro, J.M., Hamilton, Taubert, H., Wuerl, P., Onel, K., Yip, L., Boyle, J.M., Spreadborough, A., Greaves, M.J.,
S.R., Preisinger, A.C., Jessup, J.M., vanTuinen, Hwang, S.J., Strong, L.C., Lozano, G. & Levine, Birch, J.M., Varley, J.M. & Scott, D. (2001) The
P., Ledbetter, D.H., Barker, D.F., Nakamura, Y., A.J. (2004) A single nucleotide polymorphism in relationship between radiation-induced G(1)ar-
White, R. & Vogelstein, B. (1989) Chromosome the MDM2 promoter attenuates the p53 tumor rest and chromosome aberrations in Li-Frau-
17 deletions and p53 gene mutations in colorec- suppressor pathway and accelerates tumor for- meni fibroblasts with or without germline TP53
tal carcinomas. Science, 244, 217221. mation in humans. Cell, 119, 591602. mutations. British Journal of Cancer, 85, 293
Beckerman, R. & Prives, C. (2010) Transcriptional Bond, G.L., Hirshfield, K.M., Kirchhoff, T., Alexe, 296.
regulation by p53. Cold Spring Harbor Perspec- G., Bond, E.E., Robins, H., Bartel, F., Taubert, Brosh, R. & Rotter, V. (2009) When mutants gain
tives in Biology, 2, a000935. H., Wuerl, P., Hait, W., Toppmeyer, D., Offit, new powers: news from the mutant p53 field.
Birch, J.M., Hartley, A.L., Tricker, K.J., Prosser, K. & Levine, A.J. (2006) MDM2 SNP309 accel- Nature Reviews Cancer, 9, 701713.
J., Condie, A., Kelsey, A.M., Harris, M., Jones, erates tumor formation in a gender-specific and Chin, K.V., Ueda, K., Pastan, I. & Gottesman,
P.H., Binchy, A., Crowther, D., Craft, A.W., hormone-dependent manner. Cancer Research, M.M. (1992) Modulation of activity of the pro-
Eden, O.B., Evans, E.T., Mann, J.R., Martin, 66, 51045110. moter of the human MDR1 gene by Ras and
J., Mitchell, L.D. & Santibanez-Koref, M.F. Bougeard, G., Baert-Desurmont, S., Tournier, I., p53. Science, 255, 459462.
(1994) Prevalence and diversity of constitu- Vasseur, S., Martin, C., Brugieres, L., Chompret, Chompret, A., Brugieres, L., Ronsin, M., Gardes,
tional mutations in the p53 gene among 21 A., Bressac-de Paillerets, B., Stoppa-Lyonnet, D., M., Dessarps-Freichey, F., Abel, A., Hua, D.,
Li-Fraumeni families. Cancer Research, 54, Bonaiti-Pellie, C. & Frebourg, T. (2006) Impact Ligot, L., Dondon, M.G., Bressac-de Paillerets,
12981304. of the MDM2 SNP309 and p53 Arg72Pro B., Frebourg, T., Lemerle, J., Bonaiti-Pellie, C. &

2016 John Wiley & Sons Ltd, British Journal of Haematology 11


Review

Feunteun, J. (2000) P53 germline mutations in Gonzalez, K.D., Buzin, C.H., Noltner, K.A., Gu, strengthens the link between childhood choroid
childhood cancers and cancer risk for carrier D., Li, W., Malkin, D. & Sommer, S.S. (2009b) plexus carcinoma and germline TP53 mutations.
individuals. British Journal of Cancer, 82, 1932 High frequency of de novo mutations in Li- European Journal of Cancer, 41, 15971603.
1937. Fraumeni syndrome. Journal of Medical Genetics, Lammens, C.R., Aaronson, N.K., Wagner, A., Sij-
Chompret, A., Abel, A., Stoppa-Lyonnet, D., Bru- 46, 689693. mons, R.H., Ausems, M.G., Vriends, A.H., Ruijs,
gieres, L., Pages, S., Feunteun, J. & Bonaiti-Pel- Hainaut, P. & Pfeifer, G.P. (2016) Somatic TP53 M.W., van Os, T.A., Spruijt, L., Gomez Garcia,
lie, C. (2001) Sensitivity and predictive value of mutations in the era of genome sequencing. E.B., Kluijt, I., Nagtegaal, T., Verhoef, S. & Bleiker,
criteria for p53 germline mutation screening. Cold Spring Harbor Perspectives in Medicine, 6, E.M. (2010a) Genetic testing in Li-Fraumeni syn-
Journal Medical Genetics, 38, 4347. 123. doi: 10.1101/cshperspect.a026179 drome: uptake and psychosocial consequences.
Churpek, J.E., Marquez, R., Neistadt, B., Claussen, Hettmer, S., Archer, N.M., Somers, G.R., Novok- Journal of Clinical Oncology, 28, 30083014.
K., Lee, M.K., Churpek, M.M., Huo, D., Weiner, met, A., Wagers, A.J., Diller, L., Rodriguez- Lammens, C.R., Bleiker, E.M., Aaronson, N.K.,
H., Bannerjee, M., Godley, L.A., Le Beau, M.M., Galindo, C., Teot, L.A. & Malkin, D. (2014) Wagner, A., Sijmons, R.H., Ausems, M.G.,
Pritchard, C.C., Walsh, T., King, M.C., Olopade, Anaplastic rhabdomyosarcoma in TP53 germline Vriends, A.H., Ruijs, M.W., van Os, T.A.,
O.I. & Larson, R.A. (2016) Inherited mutations mutation carriers. Cancer, 120, 10681075. Spruijt, L., Gomez Garcia, E.B., Cats, A., Nagte-
in cancer susceptibility genes are common Heymann, S., Delaloge, S., Rahal, A., Caron, O., gaal, T. & Verhoef, S. (2010b) Regular surveil-
among survivors of breast cancer who develop Frebourg, T., Barreau, L., Pachet, C., Mathieu, lance for Li-Fraumeni syndrome: advice,
therapy-related leukemia. Cancer, 122, 304311. M.C., Marsiglia, H. & Bourgier, C. (2010) adherence and perceived benefits. Familial Can-
Custodio, G., Komechen, H., Figueiredo, F.R., Radio-induced malignancies after breast cancer cer, 9, 647654.
Fachin, N.D., Pianovski, M.A. & Figueiredo, postoperative radiotherapy in patients with Li- Lavigueur, A., Maltby, V., Mock, D., Rossant, J.,
B.C. (2012) Molecular epidemiology of adreno- Fraumeni syndrome. Radiation Oncology, 5, 104. Pawson, T. & Bernstein, A. (1989) High inci-
cortical tumors in southern Brazil. Molecular Hisada, M., Garber, J.E., Fung, C.Y., Fraumeni, Jr, dence of lung, bone, and lymphoid tumors in
and Cellular Endocrinology, 351, 4451. J.F. & Li, F.P. (1998) Multiple primary cancers transgenic mice overexpressing mutant alleles of
De Moura, J., Kavalec, F.L., Doghman, M., Rosati, in families with Li-Fraumeni syndrome. Journal the p53 oncogene. Molecular and Cellular Biol-
R., Custodio, G., Lalli, E., Cavallari, G.M., Santa of the National Cancer Institute, 90, 606611. ogy, 9, 39823991.
Maria, J. & Figueiredo, B.C. (2010) Heterozy- Holmfeldt, L., Wei, L., Diaz-Flores, E., Walsh, M., Li, F.P. & Fraumeni, Jr, J.F. (1969a) Rhab-
gous TP53stop146/R72P fibroblasts from a Li- Zhang, J., Ding, L., Payne-Turner, D., Church- domyosarcoma in children: epidemiologic study
Fraumeni syndrome patient with impaired man, M., Andersson, A., Chen, S.C., McCastlain, and identification of a familial cancer syndrome.
response to DNA damage. International Journal K., Becksfort, J., Ma, J., Wu, G., Patel, S.N., Journal of the National Cancer Institute, 43,
of Oncology, 36, 983990. Heatley, S.L., Phillips, L.A., Song, G., Easton, J., 13651373.
Diller, L., Sexsmith, E., Gottlieb, A., Li, F.P. & Parker, M., Chen, X., Rusch, M., Boggs, K., Li, F.P. & Fraumeni, Jr, J.F. (1969b) Soft-tissue
Malkin, D. (1995) Germline p53 mutations are Vadodaria, B., Hedlund, E., Drenberg, C., Baker, sarcomas, breast cancer, and other neoplasms. A
frequently detected in young children with rhab- S., Pei, D., Cheng, C., Huether, R., Lu, C., Ful- familial syndrome? Annals of Internal Medicine,
domyosarcoma. Journal of Clinical Investigation, ton, R.S., Fulton, L.L., Tabib, Y., Dooling, D.J., 71, 747752.
95, 16061611. Ochoa, K., Minden, M., Lewis, I.D., To, L.B., Li, F.P., Fraumeni, Jr, J.F., Mulvihill, J.J., Blattner,
Eeles, R.A. (1995) Germline mutations in the TP53 Marlton, P., Roberts, A.W., Raca, G., Stock, W., W.A., Dreyfus, M.G., Tucker, M.A. & Miller,
gene. Cancer Surveys, 25, 101124. Neale, G., Drexler, H.G., Dickins, R.A., Ellison, R.W. (1988) A cancer family syndrome in
Eliyahu, D., Raz, A., Gruss, P., Givol, D. & Oren, D.W., Shurtleff, S.A., Pui, C.H., Ribeiro, R.C., twenty-four kindreds. Cancer Research, 48,
M. (1984) Participation of p53 cellular tumour Devidas, M., Carroll, A.J., Heerema, N.A., 53585362.
antigen in transformation of normal embryonic Wood, B., Borowitz, M.J., Gastier-Foster, J.M., Libe, R. & Bertherat, J. (2005) Molecular genetics
cells. Nature, 312, 646649. Raimondi, S.C., Mardis, E.R., Wilson, R.K., of adrenocortical tumours, from familial to spo-
Felix, C.A., Hosler, M.R., Provisor, D., Salhany, K., Downing, J.R., Hunger, S.P., Loh, M.L. & Mul- radic diseases. European Journal of Endocrinology,
Sexsmith, E.A., Slater, D.J., Cheung, N.K., lighan, C.G. (2013) The genomic landscape of 153, 477487.
Winick, N.J., Strauss, E.A., Heyn, R., Lange, B.J. hypodiploid acute lymphoblastic leukemia. Nat- Liu, Y., Elf, S.E., Miyata, Y., Sashida, G., Liu, Y.,
& Malkin, D. (1996) The p53 gene in pediatric ure Genetics, 45, 242252. Huang, G., Di Giandomenico, S., Lee, J.M.,
therapy-related leukemia and myelodysplasia. Id Said, B., Kim, H., Tran, J., Novokmet, A. & Deblasio, A., Menendez, S., Antipin, J., Reva, B.,
Blood, 87, 43764381. Malkin, D. (2016) Super-transactivation TP53 Koff, A. & Nimer, S.D. (2009) p53 regulates
Finlay, C.A., Hinds, P.W. & Levine, A.J. (1989) variant in the germline of a family with Li-Frau- hematopoietic stem cell quiescence. Cell Stem
The p53 proto-oncogene can act as a suppressor meni syndrome. Human Mutation, 37, 889892. Cell, 4, 3748.
of transformation. Cell, 57, 10831093. Kamihara, J., Rana, H.Q. & Garber, J.E. (2014) Mai, P.L., Malkin, D., Garber, J.E., Schiffman, J.D.,
Giacomazzi, J., Selistre, S.G., Rossi, C., Alemar, B., Germline TP53 mutations and the changing Weitzel, J.N., Strong, L.C., Wyss, O., Locke, L.,
Santos-Silva, P., Pereira, F.S., Netto, C.B., Cos- landscape of Li-Fraumeni syndrome. Human Means, V., Achatz, M.I., Hainaut, P., Frebourg,
sio, S.L., Roth, D.E., Brunetto, A.L., Zagonel- Mutation, 35, 654662. T., Evans, D.G., Bleiker, E., Patenaude, A., Sch-
Oliveira, M., Martel-Planche, G., Goldim, J.R., Kemp, C.J., Wheldon, T. & Balmain, A. (1994) neider, K., Wilfond, B., Peters, J.A., Hwang,
Hainaut, P., Camey, S.A. & Ashton-Prolla, P. p53-deficient mice are extremely susceptible to P.M., Ford, J., Tabori, U., Ognjanovic, S., Den-
(2013) Li-Fraumeni and Li-Fraumeni-like syn- radiation-induced tumorigenesis. Nature Genet- nis, P.A., Wentzensen, I.M., Greene, M.H., Frau-
drome among children diagnosed with pediatric ics, 8, 6669. meni, Jr, J.F. & Savage, S.A. (2012) Li-Fraumeni
cancer in Southern Brazil. Cancer, 119, 4341 Kleihues, P., Schauble, B., zur Hausen, A., Esteve, syndrome: report of a clinical research workshop
4349. J. & Ohgaki, H. (1997) Tumors associated with and creation of a research consortium. Cancer
Gonzalez, K.D., Noltner, K.A., Buzin, C.H., Gu, p53 germline mutations: a synopsis of 91 fami- Genetics, 205, 479487.
D., Wen-Fong, C.Y., Nguyen, V.Q., Han, J.H., lies. American Journal Pathology, 150, 113. Mai, P.L., Best, A.F., Peters, J.A., DeCastro, R.M.,
Lowstuter, K., Longmate, J., Sommer, S.S. & Krutilkova, V., Trkova, M., Fleitz, J., Gregor, V., Khincha, P.P., Loud, J.T., Bremer, R.C., Rosen-
Weitzel, J.N. (2009a) Beyond Li Fraumeni syn- Novotna, K., Krepelova, A., Sumerauer, D., berg, P.S. & Savage, S.A. (2016) Risks of first and
drome: clinical characteristics of families with Kodet, R., Siruckova, S., Plevova, P., Bendova, subsequent cancers among TP53 mutation carri-
p53 germline mutations. Journal of Clinical S., Hedvicakova, P., Foreman, N.K. & Sedlacek, ers in the National Cancer Institute Li-Fraumeni
Oncology, 27, 12501256. Z. (2005) Identification of five new families syndrome cohort. Cancer, 205, 479487.

12 2016 John Wiley & Sons Ltd, British Journal of Haematology


Review

Malkin, D. (1994) Germline p53 mutations and & Southey, M.C. (2010) Population-based esti- Pinto, E.M., Chen, X., Easton, J., Finkelstein, D.,
heritable cancer. Annual Review of Genetics, 28, mate of the contribution of TP53 mutations to Liu, Z., Pounds, S., Rodriguez-Galindo, C.,
443465. subgroups of early-onset breast cancer: Aus- Lund, T.C., Mardis, E.R., Wilson, R.K., Boggs,
Malkin, D. (2011) Li-fraumeni syndrome. Genes tralian Breast Cancer Family Study. Cancer K., Yergeau, D., Cheng, J., Mulder, H.L., Manne,
Cancer, 2, 475484. Research, 70, 47954800. J., Jenkins, J., Mastellaro, M.J., Figueiredo, B.C.,
Malkin, D., Li, F.P., Strong, L.C., Fraumeni, Jr, Mullighan, C.G., Jeha, S., Pei, D., Payne-Turner, D., Dyer, M.A., Pappo, A., Zhang, J., Downing, J.R.,
J.F., Nelson, C.E., Kim, D.H., Kassel, J., Gryka, Coustan-Smith, E., Roberts, K.G., Waanders, E., Ribeiro, R.C. & Zambetti, G.P. (2015) Genomic
M.A., Bischoff, F.Z., Tainsky, M.A. & Friend, Choi, J.K., Ma, X., Raimondi, S.C., Fan, Y., Yang, landscape of paediatric adrenocortical tumours.
S.H. (1990) Germ line p53 mutations in a famil- W., Song, G., Yang, J.J., Inaba, H., Downing, J.R., Nature Communications, 6, 6302.
ial syndrome of breast cancer, sarcomas, and Leung, W.H., Bowman, W.P., Relling, M.V., Powell, B.C., Jiang, L., Muzny, D.M., Trevino,
other neoplasms. Science, 250, 12331238. Evans, W.E., Zhang, J., Campana, D. & Pui, C.H. L.R., Dreyer, Z.E., Strong, L.C., Wheeler, D.A.,
Malkin, D., Nichols, K.E., Zelley, K. & Schiffman, (2015) Outcome of children with hypodiploid Gibbs, R.A. & Plon, S.E. (2013) Identification of
J.D. (2014) Predisposition to pediatric and ALL treated with risk-directed therapy based on TP53 as an acute lymphocytic leukemia suscep-
hematologic cancers: a moving target. American MRD levels. Blood, 126, 28962899. tibility gene through exome sequencing. Pedi-
Society of Clinical Oncology Educational Book, 34, Nachman, J.B., Heerema, N.A., Sather, H., atric Blood & Cancer, 60, E1E3.
e44e55. Camitta, B., Forestier, E., Harrison, C.J., Das- Pui, C.H., Williams, D.L., Raimondi, S.C., Rivera,
Marcel, V., Palmero, E.I., Falagan-Lotsch, P., Mar- tugue, N., Schrappe, M., Pui, C.H., Basso, G., G.K., Look, A.T., Dodge, R.K., George, S.L.,
tel-Planche, G., Ashton-Prolla, P., Olivier, M., Silverman, L.B. & Janka-Schaub, G.E. (2007) Behm, F.G., Crist, W.M. & Murphy, S.B. (1987)
Brentani, R.R., Hainaut, P. & Achatz, M.I. Outcome of treatment in children with hypodi- Hypodiploidy is associated with a poor progno-
(2009) TP53 PIN3 and MDM2 SNP309 poly- ploid acute lymphoblastic leukemia. Blood, 110, sis in childhood acute lymphoblastic leukemia.
morphisms as genetic modifiers in the Li-Frau- 11121115. Blood, 70, 247253.
meni syndrome: impact on age at first diagnosis. Nichols, K.E., Malkin, D., Garber, J.E., Fraumeni, Rausch, T., Jones, D.T., Zapatka, M., Stutz, A.M.,
Journal of Medical Genetics, 46, 766772. Jr, J.F. & Li, F.P. (2001) Germ-line p53 muta- Zichner, T., Weischenfeldt, J., Jager, N., Remke,
Masciari, S., Dillon, D.A., Rath, M., Robson, M., tions predispose to a wide spectrum of early- M., Shih, D., Northcott, P.A., Pfaff, E., Tica, J.,
Weitzel, J.N., Balmana, J., Gruber, S.B., Ford, onset cancers. Cancer Epidemiology, Biomarkers Wang, Q., Massimi, L., Witt, H., Bender, S., Ple-
J.M., Euhus, D., Lebensohn, A., Telli, M., and Prevention, 10, 8387. ier, S., Cin, H., Hawkins, C., Beck, C., von
Pochebit, S.M., Lypas, G. & Garber, J.E. (2012) Nigro, J.M., Baker, S.J., Preisinger, A.C., Jessup, Deimling, A., Hans, V., Brors, B., Eils, R.,
Breast cancer phenotype in women with TP53 J.M., Hostetter, R., Cleary, K., Bigner, S.H., Scheurlen, W., Blake, J., Benes, V., Kulozik,
germline mutations: a Li-Fraumeni syndrome Davidson, N., Baylin, S., Devilee, P., Glover, T., A.E., Witt, O., Martin, D., Zhang, C., Porat, R.,
consortium effort. Breast Cancer Research and Collins, F.S., Weston, A., Modali, R., Harris, Merino, D.M., Wasserman, J., Jabado, N., Fon-
Treatment, 133, 11251130. C.C. & Vogelstein, B. (1989) Mutations in the tebasso, A., Bullinger, L., Rucker, F.G., Dohner,
McBride, K.A., Ballinger, M.L., Killick, E., Kirk, J., p53 gene occur in diverse human tumour types. K., Dohner, H., Koster, J., Molenaar, J.J., Ver-
Tattersall, M.H., Eeles, R.A., Thomas, D.M. & Nature, 342, 705708. steeg, R., Kool, M., Tabori, U., Malkin, D., Kor-
Mitchell, G. (2014) Li-Fraumeni syndrome: can- Ognjanovic, S., Olivier, M., Bergemann, T.L. & shunov, A., Taylor, M.D., Lichter, P., Pfister,
cer risk assessment and clinical management. Hainaut, P. (2012) Sarcomas in TP53 germline S.M. & Korbel, J.O. (2012) Genome sequencing
Nature Reviews Clinical Oncology, 11, 260271. mutation carriers: a review of the IARC TP53 of pediatric medulloblastoma links catastrophic
McCuaig, J.M., Armel, S.R., Novokmet, A., Gins- database. Cancer, 118, 13871396. DNA rearrangements with TP53 mutations. Cell,
burg, O.M., Demsky, R., Narod, S.A. & Malkin, Olivier, M., Eeles, R., Hollstein, M., Khan, M.A., 148, 5971.
D. (2012) Routine TP53 testing for breast cancer Harris, C.C. & Hainaut, P. (2002) The IARC Reinhardt, H.C. & Schumacher, B. (2012) The p53
under age 30: ready for prime time? Familial TP53 database: new online mutation analysis network: cellular and systemic DNA damage
Cancer, 11, 607613. and recommendations to users. Human Muta- responses in aging and cancer. Trends in Genet-
Melhem-Bertrandt, A., Bojadzieva, J., Ready, K.J., tion, 19, 607614. ics, 28, 128136.
Obeid, E., Liu, D.D., Gutierrez-Barrera, A.M., Olivier, M., Goldgar, D.E., Sodha, N., Ohgaki, H., Renaux-Petel, M., Sesboue, R., Baert-Desurmont,
Litton, J.K., Olopade, O.I., Hortobagyi, G.N., Kleihues, P., Hainaut, P. & Eeles, R.A. (2003) S., Vasseur, S., Fourneaux, S., Bessenay, E., Fre-
Strong, L.C. & Arun, B.K. (2012) Early onset Li-Fraumeni and related syndromes: correlation bourg, T. & Bougeard, G. (2014) The MDM2
HER2-positive breast cancer is associated with between tumor type, family structure, and TP53 285G-309G haplotype is associated with an ear-
germline TP53 mutations. Cancer, 118, 908913. genotype. Cancer Research, 63, 66436650. lier age of tumour onset in patients with Li-Frau-
Milner, J., Medcalf, E.A. & Cook, A.C. (1991) Palmero, E.I., Achatz, M.I., Ashton-Prolla, P., Oli- meni syndrome. Familial Cancer, 13, 127130.
Tumor suppressor p53: analysis of wild-type vier, M. & Hainaut, P. (2010) Tumor protein 53 Ribeiro, R.C., Michalkiewicz, E.L., Figueiredo,
and mutant p53 complexes. Molecular Cell Biol- mutations and inherited cancer: beyond Li-Frau- B.C., DeLacerda, L., Sandrini, F., Pianovsky,
ogy, 11, 1219. meni syndrome. Current Opinion in Oncology, M.D., Sampaio, G. & Sandrini, R. (2000)
Mirabello, L., Yeager, M., Mai, P.L., Gastier-Foster, 22, 6469. Adrenocortical tumors in children. Brazilian
J.M., Gorlick, R., Khanna, C., Patino-Garcia, A., Pant, V., Quintas-Cardama, A. & Lozano, G. Journal of Medical and Biological Research, 33,
Sierrasesumaga, L., Lecanda, F., Andrulis, I.L., (2012) The p53 pathway in hematopoiesis: les- 12251234.
Wunder, J.S., Gokgoz, N., Barkauskas, D.A., sons from mouse models, implications for Ribeiro, R.C., Sandrini, F., Figueiredo, B., Zam-
Zhang, X., Vogt, A., Jones, K., Boland, J.F., Cha- humans. Blood, 120, 51185127. betti, G.P., Michalkiewicz, E., Lafferty, A.R.,
nock, S.J. & Savage, S.A. (2015) Germline TP53 Petitjean, A., Mathe, E., Kato, S., Ishioka, C., DeLacerda, L., Rabin, M., Cadwell, C., Sampaio,
variants and susceptibility to osteosarcoma. Tavtigian, S.V., Hainaut, P. & Olivier, M. (2007) G., Cat, I., Stratakis, C.A. & Sandrini, R. (2001)
Journal of the National Cancer Institute, 107, Impact of mutant p53 functional properties on An inherited p53 mutation that contributes in a
14. TP53 mutation patterns and tumor phenotype: tissue-specific manner to pediatric adrenal corti-
Moll, U.M. & Petrenko, O. (2003) The MDM2- lessons from recent developments in the IARC cal carcinoma. Proceedings of the National Acad-
p53 interaction. Molecular Cancer Research, 1, TP53 database. Human Mutation, 28, 622629. emy of Sciences of the United States of America,
10011008. Pierce, L.J. & Haffty, B.G. (2011) Radiotherapy in 98, 93309335.
Mouchawar, J., Korch, C., Byers, T., Pitts, T.M., the treatment of hereditary breast cancer. Semi- Rodriguez-Galindo, C., Figueiredo, B.C., Zambetti,
Li, E., McCredie, M.R., Giles, G.G., Hopper, J.L. nars in Radiation Oncology, 21, 4350. G.P. & Ribeiro, R.C. (2005) Biology, clinical

2016 John Wiley & Sons Ltd, British Journal of Haematology 13


Review

characteristics, and management of adrenocorti- with Li-Fraumeni syndrome. Nature, 348, 747 pediatric adrenocortical carcinoma: a childrens
cal tumors in children. Pediatric Blood Cancer, 749. oncology group study. Journal of Clinical Oncol-
45, 265273. Tabori, U., Shlien, A., Baskin, B., Levitt, S., Ray, ogy, 33, 602609.
Rotter, V. (1983) p53, a transformation-related cel- P., Alon, N., Hawkins, C., Bouffet, E., Pien- Willis, A., Jung, E.J., Wakefield, T. & Chen, X.
lular-encoded protein, can be used as a bio- kowska, M., Lafay-Cousin, L., Gozali, A., Zhu- (2004) Mutant p53 exerts a dominant negative
chemical marker for the detection of primary kova, N., Shane, L., Gonzalez, I., Finlay, J. & effect by preventing wild-type p53 from binding
mouse tumor cells. Proceedings of the National Malkin, D. (2010) TP53 alterations determine to the promoter of its target genes. Oncogene, 23,
Academy of Sciences of the United States of Amer- clinical subgroups and survival of patients with 23302338.
ica, 80, 26132617. choroid plexus tumors. Journal of Clinical Wolf, D., Harris, N. & Rotter, V. (1984) Reconsti-
Ruijs, M.W., Schmidt, M.K., Nevanlinna, H., Tom- Oncology, 28, 19952001. tution of p53 expression in a nonproducer Ab-
miska, J., Aittomaki, K., Pruntel, R., Verhoef, S. Thull, D.L. & Vogel, V.G. (2004) Recognition and MuLV-transformed cell line by transfection of a
& Vant Veer, L.J. (2007) The single-nucleotide management of hereditary breast cancer syn- functional p53 gene. Cell, 38, 119126.
polymorphism 309 in the MDM2 gene con- dromes. Oncologist, 9, 1324. Wong, P., Verselis, S.J., Garber, J.E., Schneider, K.,
tributes to the Li-Fraumeni syndrome and Tinat, J., Bougeard, G., Baert-Desurmont, S., Vas- DiGianni, L., Stockwell, D.H., Li, F.P. & Syngal,
related phenotypes. European Journal of Human seur, S., Martin, C., Bouvignies, E., Caron, O., S. (2006) Prevalence of early onset colorectal
Genetics, 15, 110114. Bressac-de Paillerets, B., Berthet, P., Dugast, C., cancer in 397 patients with classic Li-Fraumeni
Ruijs, M.W., Verhoef, S., Rookus, M.A., Pruntel, Bonaiti-Pellie, C., Stoppa-Lyonnet, D. & Fre- syndrome. Gastroenterology, 130, 7379.
R., van der Hout, A.H., Hogervorst, F.B., Kluijt, bourg, T. (2009) 2009 version of the Chompret Zhang, J., Walsh, M.F., Wu, G., Edmonson, M.N.,
I., Sijmons, R.H., Aalfs, C.M., Wagner, A., criteria for Li Fraumeni syndrome. Journal of Gruber, T.A., Easton, J., Hedges, D., Ma, X.,
Ausems, M.G., Hoogerbrugge, N., van Asperen, Clinical Oncology, 27, e108e109; author reply Zhou, X., Yergeau, D.A., Wilkinson, M.R.,
C.J., Gomez Garcia, E.B., Meijers-Heijboer, H., e110. Vadodaria, B., Chen, X., McGee, R.B., Hines-
Ten Kate, L.P., Menko, F.H. & vant Veer, L.J. Varley, J.M. (2003) Germline TP53 mutations and Dowell, S., Nuccio, R., Quinn, E., Shurtleff, S.A.,
(2010) TP53 germline mutation testing in 180 Li-Fraumeni syndrome. Human Mutation, 21, Rusch, M., Patel, A., Becksfort, J.B., Wang, S.,
families suspected of Li-Fraumeni syndrome: 313320. Weaver, M.S., Ding, L., Mardis, E.R., Wilson,
mutation detection rate and relative frequency Varley, J.M., Evans, D.G. & Birch, J.M. (1997) Li- R.K., Gajjar, A., Ellison, D.W., Pappo, A.S., Pui,
of cancers in different familial phenotypes. Jour- Fraumeni syndrome a molecular and clinical C.H., Nichols, K.E. & Downing, J.R. (2015)
nal of Medical Genetics, 47, 421428. review. British Journal of Cancer, 76, 114. Germline mutations in predisposition genes in
Schneider, K., Zelley, K., Nichols, K.E. & Garber, J. Villani, A., Tabori, U., Schiffman, J., Shlien, A., pediatric cancer. New England Journal of Medi-
(1993) Li-Fraumeni syndrome. In: GeneReviews Beyene, J., Druker, H., Novokmet, A., Finlay, J. cine, 373, 23362346.
(R) (ed. by R.A. Pagon, M.P. Adam, H.H. & Malkin, D. (2011) Biochemical and imaging Zhou, X., Edmonson, M.N., Wilkinson, M.R.,
Ardinger, S.E. Wallace, A. Amemiya, L.J.H. surveillance in germline TP53 mutation carriers Patel, A., Wu, G., Liu, Y., Li, Y., Zhang, Z.,
Bean, T.D. Bird, C.T. Fong, H.C. Mefford, with Li-Fraumeni syndrome: a prospective Rusch, M.C., Parker, M., Becksfort, J., Downing,
R.J.H. Smith & K. Stephens), University of observational study. Lancet Oncology, 12, 559 J.R. & Zhang, J. (2016) Exploring genomic alter-
Washington, Seattle, WA. 567. ation in pediatric cancer using ProteinPaint.
Schulz, E., Valentin, A., Ulz, P., Beham-Schmid, Villani, A., Shore, A., Wasserman, J.D., Stephens, Nature Genetics, 48, 46.
C., Lind, K., Rupp, V., Lackner, H., Wolfler, A., D., Kim, R.H., Druker, H., Gallinger, B., Nau- Zhukova, N., Ramaswamy, V., Remke, M., Pfaff,
Zebisch, A., Olipitz, W., Geigl, J., Berghold, A., mer, A., Kohlmann, W., Novokmet, A., Tabori, E., Shih, D.J., Martin, D.C., Castelo-Branco, P.,
Speicher, M.R. & Sill, H. (2012) Germline muta- U., Tijerin, M., Greer, M.C., Finlay, J.L., Schiff- Baskin, B., Ray, P.N., Bouffet, E., von Bueren,
tions in the DNA damage response genes man, J.D. & Malkin, D. (2016) Biochemical and A.O., Jones, D.T., Northcott, P.A., Kool, M.,
BRCA1, BRCA2, BARD1 and TP53 in patients imaging surveillance in germline TP53 mutation Sturm, D., Pugh, T.J., Pomeroy, S.L., Cho, Y.J.,
with therapy related myeloid neoplasms. Journal carriers with Li-Fraumeni syndrome: 11 year Pietsch, T., Gessi, M., Rutkowski, S., Bognar, L.,
of Medical Genetics, 49, 422428. follow-up of a prospective observational study. Klekner, A., Cho, B.K., Kim, S.K., Wang, K.C.,
Seidinger, A.L., Fortes, F.P., Mastellaro, M.J., Car- Lancet Oncology, 17, 12951305. Eberhart, C.G., Fevre-Montange, M., Fouladi,
dinalli, I.A., Zambaldi, L.G., Aguiar, S.S. & Vousden, K.H. & Prives, C. (2009) Blinded by the M., French, P.J., Kros, M., Grajkowska, W.A.,
Yunes, J.A. (2015) Occurrence of Neuroblas- light: the growing complexity of p53. Cell, 137, Gupta, N., Weiss, W.A., Hauser, P., Jabado, N.,
toma among TP53 p. R337H Carriers. PLoS 413431. Jouvet, A., Jung, S., Kumabe, T., Lach, B., Leo-
ONE, 10, e0140356. Wagner, J., Portwine, C., Rabin, K., Leclerc, J.M., nard, J.R., Rubin, J.B., Liau, L.M., Massimi, L.,
Sorrell, A.D., Espenschied, C.R., Culver, J.O. & Narod, S.A. & Malkin, D. (1994) High fre- Pollack, I.F., Shin Ra, Y., Van Meir, E.G., Zitter-
Weitzel, J.N. (2013) Tumor protein p53 (TP53) quency of germline p53 mutations in childhood bart, K., Schuller, U., Hill, R.M., Lindsey, J.C.,
testing and Li-Fraumeni syndrome: current sta- adrenocortical cancer. Journal of the National Schwalbe, E.C., Bailey, S., Ellison, D.W., Haw-
tus of clinical applications and future directions. Cancer Institute, 86, 17071710. kins, C., Malkin, D., Clifford, S.C., Korshunov,
Molecular Diagnosis & Therapy, 17, 3147. Wasserman, J.D., Novokmet, A., Eichler-Jonsson, A., Pfister, S., Taylor, M.D. & Tabori, U. (2013)
Srivastava, S., Zou, Z.Q., Pirollo, K., Blattner, W. C., Ribeiro, R.C., Rodriguez-Galindo, C., Zam- Subgroup-specific prognostic implications of
& Chang, E.H. (1990) Germ-line transmission betti, G.P. & Malkin, D. (2015) Prevalence and TP53 mutation in medulloblastoma. Journal of
of a mutated p53 gene in a cancer-prone family functional consequence of TP53 mutations in Clinical Oncology, 31, 29272935.

14 2016 John Wiley & Sons Ltd, British Journal of Haematology

Вам также может понравиться