ArticlePDF Available

Abstract and Figures

PALB2 gene is mutated in about 1-2 % of familial breast cancer as well as in 3-4 % of familial pancreatic cancer cases. Few studies have reported mutations in Italian patients with breast or pancreatic cancer. We evaluate the occurrence of PALB2 mutations in Italian patients affected with hereditary breast and ovarian cancers and define the pathological significance of the putative allelic variants. We recruited 98 patients (F = 93, M = 5) affected with breast and/or ovarian cancer, negative for mutations in BRCA1 and BRCA2 (BRCAX). Genomic DNA was isolated from peripheral blood lymphocytes, PALB2 coding regions and adjacent intronic were sequenced; in silico predictions were carried out using prediction programs. Mutational analysis of PALB2 gene revealed the novel mutation c.1919C>A (p.S640X) in a 29 years old woman with breast cancer. The c.1919C>A (p.S640X) mutation causes the lack of C-terminus region inducing alteration of MORF4L1-PALB2 association and the lack of interaction of PALB2 with RAD51 and BRCA2. In addition, we identified two novel PALB2 variants, c.3047T>C (p.F1016S) and c.*146A>G. In silico analysis conducted for c.*146A>G indicates that this variant does not affect the splicing while c.3047T>C (p.F1016S) was predicted as damaging in three classifier algorithms. The proband carrier of c.3047T>C (p.F1016S) showed two breast cancer cases, two ovarian cancer cases and one pancreatic cancer in mother's family. c.3047T>C (p.F1016S) and c.*146A>G should be considered PALB2 UVs even though the genotype-phenotype correlation for these variants remains still unclear. Our findings indicate that the presence of PALB2 mutation should be routinely investigated in hereditary breast and ovarian cancers families since it could be of clinical relevance for clinical management.
Content may be subject to copyright.
LETTER TO THE EDITOR
Analysis of PALB2 in a cohort of Italian breast cancer patients:
identification of a novel PALB2 truncating mutation
Maria Teresa Vietri Gemma Caliendo Concetta Schiano
Amelia Casamassimi Anna Maria Molinari
Claudio Napoli Michele Cioffi
ÓSpringer Science+Business Media Dordrecht 2015
Abstract PALB2 gene is mutated in about 1–2 % of fa-
milial breast cancer as well as in 3–4 % of familial pan-
creatic cancer cases. Few studies have reported mutations
in Italian patients with breast or pancreatic cancer. We
evaluate the occurrence of PALB2 mutations in Italian
patients affected with hereditary breast and ovarian cancers
and define the pathological significance of the putative
allelic variants. We recruited 98 patients (F =93, M =5)
affected with breast and/or ovarian cancer, negative for
mutations in BRCA1 and BRCA2 (BRCAX). Genomic
DNA was isolated from peripheral blood lymphocytes,
PALB2 coding regions and adjacent intronic were se-
quenced; in silico predictions were carried out using pre-
diction programs. Mutational analysis of PALB2 gene
revealed the novel mutation c.1919C[A (p.S640X) in a
29 years old woman with breast cancer. The c.1919C[A
(p.S640X) mutation causes the lack of C-terminus region
inducing alteration of MORF4L1–PALB2 association and
the lack of interaction of PALB2 with RAD51 and BRCA2.
In addition, we identified two novel PALB2 variants,
c.3047T[C (p.F1016S) and c.*146A[G. In silico analysis
conducted for c.*146A[G indicates that this variant does
not affect the splicing while c.3047T[C (p.F1016S) was
predicted as damaging in three classifier algorithms. The
proband carrier of c.3047T[C (p.F1016S) showed two
breast cancer cases, two ovarian cancer cases and one
pancreatic cancer in mother’s family. c.3047T[C
(p.F1016S) and c.*146A[G should be considered PALB2
UVs even though the genotype–phenotype correlation for
these variants remains still unclear. Our findings indicate
that the presence of PALB2 mutation should be routinely
investigated in hereditary breast and ovarian cancers
families since it could be of clinical relevance for clinical
management.
Keywords PALB2 Hereditary breast cancer Hereditary
ovarian cancer Hereditary pancreatic cancer
Abbreviations
UVs Unclassified variants
HBOC Hereditary breast and ovarian cancer
PALB2 Partner and localizer of BRCA2
MORF4L1 Mortality factor 4 like protein 1
MRG15 MORF4-related gene on chromosome 15
HGVS Human Genome Variation Society
NCBI National Centre for Biotechnology
Information
Hereditary breast cancer genes (BRCA1,BRCA2,CHK2,
TP53,ATM and PALB2) involved in DNA damage re-
sponses suggested that hereditary breast and ovarian cancer
(HBOC) is a consequence of impaired genome stability
control [1]. PALB2, originally identified as a BRCA2-in-
teracting protein, is an essential component for the formation
of the BRCA complex. Indeed, PALB2 acts as a bridge
between BRCA1 and BRCA2 to form a BRCA complex that
then binds RAD51 to initiate homologous recombination
[2]. Mutations in BRCA genes can be found in about 30 % of
M. T. Vietri (&)G. Caliendo A. Casamassimi
A. M. Molinari C. Napoli M. Cioffi
Department of Biochemistry, Biophysics and General Pathology,
Second University of Naples, Via Luigi De Crecchio, 7,
80138 Naples, Italy
e-mail: mariateresa.vietri@unina2.it
C. Schiano C. Napoli
Institute of Diagnostic and Nuclear Development (SDN),
IRCCS, Via E. Gianturco 113, 80143 Naples, Italy
123
Familial Cancer
DOI 10.1007/s10689-015-9786-z
HBOC (mutations in both genes have been described in 1 %
of patients) [3]whereasPALB2 mutations have been iden-
tified about in 1–2 % of familial breast cancer and in 3–4 %
of familial pancreatic cancer [4]. To date, only few studies
have been conducted in Italian patients with breast or pan-
creatic cancer [510]. We aimed to establish the occurrence
and the possible pathological significance of PALB2 muta-
tions and/or variations in a cohort of Italian patients affected
with hereditary breast/ovarian cancers that were negative for
BRCA1 and BRCA2 mutations.
A total of 98 patients (F =93, M =5) were recruited.
Of these, 86 were affected with breast cancer, 2 with breast
and ovarian cancer, 7 with bilateral breast cancer and 3
with ovarian cancer. All subjects were from Campania, a
region of southern Italy. Their age ranged between 26 and
83 years. Patients were selected according to the selection
criteria for hereditary breast cancer based on the Breast
Cancer Linkage Consortium [11]. Informed consent was
obtained from all patients before genetic analysis and
pedigree was traced. PALB2 mutational analysis was con-
ducted in all patients previously screened for BRCA1 and
BRCA2 mutations. Samples from 103 patients (age
40–80 years) affected with breast cancer, collected without
selection for family history of cancer, were used to eval-
uate the frequency of the identified PALB2 variations. As
control, 102 females (age 25–87 years) from the same
geographical area were anonymously enrolled. They were
all cancer-free at the time of blood donation.
The extraction of genomic DNA from peripheral blood
lymphocytes was performed using Wizard Genomic DNA
purification kit (Promega Corporation, Madison, WI,
USA). The presence of germline mutations in PALB2 was
evaluated by direct sequencing of 13 exons and adjacent
intronic regions. One set of primers was used to amplify
each exon, except for exons 4 and 5, which were amplified
in 4 and 2 PCR products respectively, as previously re-
ported [12].
All PCR products were sequenced on both strands using
the ABI PRISM di-Deoxy Terminator Cycle sequencing kit
on ABI 9700 thermal cycle (Applied Biosystems, Fotser
City, CA, USA) and ABI 3100 Genetic Analyser (Applied
Biosystems, Fotser City, CA, USA). The results were
analysed by using the software Mutation Surveyor version
3.24 (Softgenetics, State Collage PA, USA). Mutations
and/or variants were identified as new by referring to the
Human Gene Mutation Database (http://www.biobase-
international.com/product/hgmd) and Leiden Open Varia-
tion Database (https://grenada.lumc.nl/LOVD2/shared1/
home.php?select_db=PALB2). GenBank reference se-
quences used for naming the novel mutation and variants
were NM_024675.3 and NT_010393.15. The sequence
variants and mutation were named and referred in the text
according to the nomenclature used by Human Genome
Variation Society (HGVS; http://www.hgvs.org) and to the
descriptions suggested by den Dunnen and Antonarakis
[13].
Mutation analysis of PALB2 gene showed the presence
of a mutation in 1/98 (1.02 %) breast and/or ovarian cancer
patients. This mutation c.1919C[A (p.S640X) was identi-
fied in a 29 years old woman who was affected with breast
cancer. In addition, we identified two novel PALB2 vari-
ants in two breast cancer patients; the first one was a
missense substitution c.3047T[C (p.F1016S) and the sec-
ond one, c.*146A
[G, was localized in the 30UTR region.
Noteworthy, these variants have not previously reported in
published literature or in the NCBI or in the Ensembl
genome databases. None of 103 sporadic breast cancer
patients and 102 healthy female were found to carry the
nonsense mutation c.1919C[A (p.S640X) or the novel
variants c.3047T[C (p.F1016S) and c.*146A[G. Patients
that were carriers of the novel nonsense mutation
c.1919C[A (p.S640X) or the novel missense variant
c.3047T[C (p.F1016S) were exposed to a second periph-
eral blood sample to confirm the presence of the mutation
also on mRNA. Total RNA was isolated from peripheral
blood lymphocytes using TriZol reagent (Invitrogen,
Carlsbad, CA, USA) and reverse transcribed using Su-
perScript First-Strand Synthesis System (Invitrogen,
Carlsbad, CA), according to the manufacturer’s protocol.
To amplify the region spanning the mutations, we used a
pair of primers complementary to exons 4 and 5 for
c.1919C[A (p.S640X) and to exons 9–10 for c.3047T[C
(p.F1016S). Then, PCR products were sequenced. Besides,
RNA sample analysis confirmed the presence of the iden-
tified mutation and variant.
The novel PALB2 c.1919C[A (p.S640X) mutation is a
nonsense mutation, localized in exon 5, which results in the
introduction of a stop codon at amino acid position 640
(Fig. 1). The proband carrying the c.1919C[A (p.S640X)
mutation was diagnosed with breast cancer at age 29. At
the time of diagnosis, she underwent a right mastectomy.
She reported a family history of breast cancer and other
malignancies, including prostate, colorectal and brain
cancer (Fig. 2). Thus, the major finding of this study is the
novel PALB2 mutation in a group of patients affected with
hereditary breast and/or ovarian cancer and previously
tested for BRCA1 and BRCA2 mutations. Accordingly to
the published literature, which reported PALB2 mutations
in about 1–2 % of hereditary breast cancer [14], also in
Italian series [5,6,10], we found a frequency of 1.02 %.
The nonsense mutation c.1919C[A (p.S640X) was iden-
tified in a patient who was affected with hereditary breast
cancer. Unfortunately, although breast cancers and other
related tumours were reported in both paternal and mater-
nal branches of her family, not all family members/rela-
tives were available for genetic testing. Thus, the lack of
M. T. Vietri et al.
123
useful DNA samples has implicated that transmission of
this mutation is uncertain in the patient’s family.
Several studies have identified different monoallelic
PALB2 truncating in individuals with familial breast cancer
from various geographic areas [1518]. Most of the
pathogenic PALB2 mutations detected in these previous
analyses are truncating, frameshift or stop codon mutations
and they were scattered throughout the entire gene region
with no hot-spot areas [19]. The c.1919C[A (p.S640X)
PALB2 mutation leads to protein truncation and it occurs in
Fig. 1 Partial electropherogram
of PALB2 exon 5 evidencing the
nonsense mutation c.1919C[A
(p.S640X). with forward (a) and
reverse (b) primers. The novel
mutation leads to the
introduction of a stop codon at
amino acid position 640
Fig. 2 Pedigree of a breast cancer family carrying the PALB2 mutation c.1919C[A (pS640X). Age at diagnoses (bold) and age at the present
time or age at exitus (italic) are reported. The proband is marked with an arrow
Novel PALB2 mutation in breast cancer patients
123
a region responsible for binding between PALB2 and
MRG15. The PALB2 protein has a coiled-coil motif at the
N terminus required for interaction with BRCA1 and a
C-terminal domain containing four WD-repeats that me-
diate the interaction with BRCA2 [20]. Moreover, it in-
teracts with RAD51 by two regions, one at the N-terminus
and the other one at the C-terminus. The fourth protein,
which interacts with PALB2, is MORF4L1 that is encoded
by the gene MRG15. MORF4L1 has a binding motif re-
sponsible for the interaction with amino acids 611–764 of
PALB2 [21]. PALB2 links MORF4L1 to the BRCA
complex that is formed in response to DNA damage [22].
The c.1919C[A (p.S640X) mutation causes the absence of
the C-terminus region, thereby inducing an alteration of
MORF4L1–PALB2 association and subsequently the lack
of interaction of PALB2 with RAD51 and BRCA2.
In addition, mutational analysis of our series of patients
showed further two novel variants, c.3047T[C (p.F1016S)
and c.*146A[G. These variants were identified in two
patients affected with hereditary breast cancer but not in
healthy controls or in sporadic breast cancers patients. The
c.3047T[C (p.F1016S) variant was identified in a patient
diagnosed with breast cancer at the age of 28. In mother’s
family there were many relatives with tumour (Fig. 3).
This family showed two breast cancer cases, two ovarian
cancer cases and one pancreatic cancer. The c.*146A[G
variant was identified in a 45 years old woman affected
with breast cancer. The patient showed other malignancies
in her family; particularly, two breast cancer cases, one
hepatic cancer and one Hodgkin lymphoma (Fig. 4).
The novel missense variant, c.3047T[C (p.F1016S),
identified in this study, was analyzed with PolyPhen-2
(http://genetics.bwh.harvard.edu/pph2/), SIFT (http://sift.
bii.a-star.edu.sg/) and A-GVGD (http://agvgd.iarc.fr/
agvgd_input.php). Concordance between these methods
will be therefore a strong predictor. To evaluate potential
splicing effects of the novel c.*146A[G variant in silico
sequence analysis tool NNSPLICE (http://www.frutfly.org/
seq_tools/splice.html) was used. When the splice site is
mutated the program assigns a score considerably lower
Fig. 3 Pedigree of a breast cancer family carrying the novel PALB2 variant c.3047T\C (p.F1016S). Age at diagnoses (bold) and age at the
present time or age at exitus (italic) are reported. The proband is marked with an arrow
M. T. Vietri et al.
123
than the wild-type sequence (\0.40). Overall, in silico
analysis predicted c.3047T[C (p.F1016S) as deleterious in
three classifier algorithms. Moreover, no evidence of a
possible splicing defect for the novel non-coding variant
c.*146A[G was showed by NNSPLICE (Table 1).
The pathogenicity of a variant of unknown clinical
significance is assessed by a number of approaches that
include evaluation of the frequency of the variant in cases
and unaffected controls, mutation segregation analysis with
disease in families, the usage of in silico prediction pro-
grams and in vitro studies. Prediction programs for detec-
tion of splicing defects or protein alterations should not be
considered a replacement for in vitro studies. However, co-
segregation analysis may support the clinical importance of
variants of unknown pathogenetic significance. Unfortu-
nately, in both families of patients with the c.3047T[C
(p.F1016S) missense variant and the untranslated-30UTR
c.*146A[G variant there were not relatives that agreed to
the genetic test. Therefore, the genotype–phenotype cor-
relation for these variants remains unknown and
c.3047T[C (p.F1016S) and c.*146A[G should be con-
sidered UVs of PALB2. Interestingly, in the lineage of
patient with c.3047T[C (p.F1016S), a maternal uncle with
pancreatic cancer was described (Fig. 3). Mutations in
PALB2 occur with a prevalence of 2.1 % in breast cancer
patients when they are selected for a personal and/or family
history of pancreatic cancer [4]. Further studies could
corroborate the pathogenicity of the c.3047T[C
(p.F1016S) variant and confirm the contribution of PALB2
gene mutation in pancreatic cancer.
The analyzed patients also showed ten previously reported
variants, c.-109delG, c.-47G[A, c.212-58A[C(IVS3-58A[C),
c.1676A[G (p.Q559R), c.2014G[C (p.E672Q), c.2590C[T
(p.P864S), c.2794G[A (p.V932M), c.2816T[G (p.L939 W),
c.2993G[A (p.G998E) and c.3300T[G (p.T1100T). The fre-
quencies and their effects are reported in Table 2.Amongthese
variants, -109delG had not yet been previously tested with pre-
diction programs. In silico analysis conducted with NNSPLICE
indicates that also this variant does not affect the splicing
(Table 2). In a previous study, computational analysis for non-
Fig. 4 Pedigree of a breast cancer family carrying the novel PALB2 variant c.*146A[G. Age at diagnoses (bold) and age at the present time or
age at exitus (italic) are reported. The proband is marked with an arrow
Novel PALB2 mutation in breast cancer patients
123
Table 1 In silico study of PALB2 novel variants identified in two patients affected with hereditary breast cancer
Designation Localization Nucleotide
change
Protein
change
HBOC patients
(n =98)
Number
(frequency)
Sporadic breast
cancer (n =103)
Number (frequency)
Healthy female
control (n =102)
Number (frequency)
PolyPhen-2 SIFT A-
GVGD
NNSplice Neoplasia
(age)
a
c.3047T[C
(p.F1016S)
Exon 10 c.3047T[C p.Phe1016Ser 1 (1.02 %) Probably
damaging
Affecting
protein
function
C65 BC (28)
c.*146A[G Non coding
30-UTR
c.*146A[G 1 (1.02 %) No effect BC (45)
a
Age at diagnosis
Table 2 PALB2 genetic variants in hereditary breast and/or ovarian cancer (HBOC), sporadic breast cancer patients and healthy controls
Designation Localization Nucleotide
change
Protein
change
HBOC patients
(n =98)
Number
(frequency)
Sporadic breast
cancer (n =103)
Number (frequency
Healthy female
control (n =102)
Number (frequency
Bioinformatic
prediction
References
Missense
c.1676A[G (p.Q559R) Exon 4 c.1676A[G p.Gln559Arg 21 (21.42 %) 21 (20.38 %) 20 (19.60 %) Benign Zheng et al. [23]
c.2014G[C (p.E672Q) Exon 5 c.2014G[C p.Glu672Gln 14 (14.28 %) 13 (12.62 %) 11 (10.78 %) Benign Zheng et al. [23]
c.2590C[T (p.P864S) Exon 7 c.2590C[T p.Pro864Ser 2 (2.04 %) Possibly benign Zheng et al. [23]
c.2794G[A (p.V932M) Exon 8 c.2794G[A p.Val932Met 1 (1.02 %) Potentially deleterious Wong-Brown et al. [24]
Blanco et al. [25]
c.2816T[G (p.L939W) Exon 8 c.2816T[G p.Leu939Tyr 1 (1.02 %) Deleterious Blanco et al. [25]
c.2993G[A (p.G998E) Exon 9 c.2993G[A p.Gly998Met 9 (9.18 %) 8 (7.76 %) 9 (8.82 %) Deleterious Zheng et al. [23]
Synonymous
c.3300T[G (p.T1100T) Exon 12 c.3300T[G p.Thr1100Thr 13 (13.26 %) 12 (11.65 %) 12 (11.76 %) No effect Blanco et al. [26]
Intronic
c.-109delG Non coding 50UTR c.-109delG 1 (1.02 %) No effect *
c.-47G[A Non coding 50UTR c.-47G[A 6 (6.10 %) 4 (3.88 %) 2 (1.96 %) No effect Blanco et al. [26]
c.212-58A[C IVS3-58 Intron 3 c.212-58A[C 10 (13.20 %) 10 (9.70 %) 12 (11.76 %) No effect Blanco et al. [26]
* Tested in our laboratory using NNSPLICE software
M. T. Vietri et al.
123
synonymous variants predicted c.1676A[G (p.Q559R),
c.2014G[C (p.E672Q) as benign, c.2590C[T (p.P864S) as
possibly benign, whereas, according to all used predictive algo-
rithms, c.2816T[G (p.L939 W) and c.2993G[A (p.G998E)
were reported as deleterious [23]. Moreover, c.2794G[A
(p.V932M) was reported as a variant potentially affecting protein
functionbyinsilicostudies[24,25]. In our series, c.2993G[A
(p.G998E) was reported in 9.18 % of patients, in 8.82 % of
controls and 7.76 % of sporadic breast cancers, in agreement
with the frequencies obtained in the study of Silvestri et al. [7],
suggesting that this variant does not have a pathogenic role. On
the contrary, c.2794G[A (p.V932M) and c.2816T[G
(p.L939 W) were observed only in the group of patients, thus
confirming its possible damaging effect previously reported [23,
24]. Also c.2590C[T (p.P864S) was reported only in HBOC
patients; however, it can be considered benign based on in silico
analysis [24]. Furthermore, no evidence for a possible splicing
defect was provided by analysis with NNSPLICE for c.-47G[A,
c.212-58A[C (IVS3-58A[C) and c.3300T[G (p.T1010T) [26].
Our results show that also c.-109delG does not affect splicing.
Noteworthy, c.-109delG was found in 1.02 % of patients but not
in controls.
Further studies, such as in vitro studies and co-segrega-
tion analyses, are warranted to understand the involvement
of the above-described PALB2 alterations in HBOC and to
elucidate the role of the identified UVs; thus, they have been
planned in our laboratory. In conclusions, PALB2 mutation
testing should be performed routinely to identify mutations
in HBOC families since it may become of clinical relevance
for clinical management. Currently, to optimize molecular
diagnosis of HBOC, which is characterized by genetic
heterogeneity, it is possible to use next generation se-
quencing (NGS). Indeed, this procedure allows evaluating
mutations in several genes including PALB2 [27] and should
be considered as a proper option in the next future.
Conflict of interest The authors declare that they have no conflict
of interest.
References
1. Zhang F, Ma J, Wu J, Ye L, Cai H, Xia B, Yu X (2009) PALB2
links BRCA1 and BRCA2 in the DNA-damage response. Curr
Biol 19:524–529
2. Southey MC, Teo ZL, Winship I (2013) PALB2 and breast
cancer: ready for clinical translation! Appl Clin Genet 6:43–52
3. Vietri MT, Molinari AM, Caliendo G, De Paola ML, Giovanna
D, Gambardella AL, Petronella P, Cioffi M (2013) Double
heterozygosity in the BRCA1 and BRCA2 genes in Italian
family. Clin Chem Lab Med 51:2319–2324
4. Hofstatter EW, Domchek SM, Miron A, Garber J, Wang M,
Componeschi K, Boghossian L, Mitron PL, Nathanson KL, Tung
N (2011) PALB2 mutations in familial breast and pancreatic
cancer. Fam Cancer 10:225–231
5. Balia C, Sensi E, Lombardi G, Roncella M, Bevilacqua G, Caligo
MA (2010) PALB2: a novel inactivating mutation in a Italian
breast cancer family. Fam Cancer 9:531–536
6. Papi L, Putignano AL, Congregati C, Piaceri I, Zanna I, Sera F,
Morrone D, Genuardi M, Palli D (2010) A PALB2 germline
mutation associated with hereditary breast cancer in Italy. Fam
Cancer 9:181–185
7. Silvestri Rizzolo P, Zanna I, Falchetti M, Masala G, Bianchi S,
Papi L, Giannini G, Palli D, Ottini L (2010) PALB2 mutations in
male breast cancer: a population-based study in Central Italy.
Breast Cancer Res Treat 122:299–301
8. Peterlongo P, Catucci I, Pasquini G, Verderio P, Peissel B, Barile
M, Varesco L, Riboni M, Fortuzzi S, Manoukian S, Radice P
(2011) PALB2 germline mutations in familial breast cancer cases
with personal and family history of pancreatic cancer. Breast
Cancer Res Treat 126:825–828
9. Ghiorzo P, Pensotti V, Fornarini G, Sciallero S, Battistuzzi L,
Belli F, Bonelli L, Borgonovo G, Bruno W, Gozza A, Gargiulo S,
Mastracci L, Nasti S, Palmieri G, Papadia F, Pastorino L, Russo
A, Savarino V, Varesco L, Bernard L, Bianchi Scarra
`G, Genoa
Pancreatic Cancer Study Group (2012) Contribution of germline
mutations in the BRCA and PALB2 genes to pancreatic cancer in
Italy. Fam Cancer 11:41–47
10. Catucci I, Peterlongo P, Ciceri S, Colombo M, Pasquini G, Barile
M, Bonanni B, Verderio P, Pizzamiglio S, Foglia C, Falanga A,
Marchetti M, Galastri L, Bianchi T, Corna C, Ravagnani F,
Bernard L, Fortuzzi S, Sardella D, Scuvera G, Peissel B, Man-
oukian S, Tondini C, Radice P (2014) PALB2 sequencing in
Italian familial breast cancer cases reveals a high-risk mutation
recurrent in the province of Bergamo. Genet Med 16:688–694
11. Breast Cancer Linkage Consortium (1997) Pathology of familial
breast cancer: differences between breast cancers in carriers of
BRCA1 or BRCA2 mutations and sporadic cases. Lancet
349:1505–1510
12. Tischkowitz M, Xia B, Sabbaghian N, Reis-Filho JS, Hamel N,
Li G, van Beers EH, Li L, Khalil T, Quenneville LA, Omeroglu
A, Poll A, Lepage P, Wong N, Nederlof PM, Ashworth A, Tonin
PN, Narod SA, Livingston DM, Foulkes WD (2007) Analysis of
PALB2/FANCN-associated breast cancer families. Proc Natl
Acad Sci USA 104:6788–6793
13. den Dunnen JT, Antonarakis SE (2003) Mutation nomenclature.
Curr Protoc Hum Genet. Aug; Chapter 7: Unit 7.13
14. Garcia MJ, Ferna
´ndez V, Osorio A, Barroso A, Llort G, La
´zaro
C, Blanco I, Calde
´s T, de la Hoya M, Ramo
´n Y, Cajal T, Alonso
C, Tejada MI, San Roma
´n C, Robles-Dı
´az L, Urioste M, Benı
´tez
J (2009) Analysis of FANCB and FANCN/PALB2 fanconi ane-
mia genes in BRCA1/2-negative Spanish breast cancer families.
Breast Cancer Res Treat 113:545–1551
15. Rahman N, Seal S, Thompson D, Kelly P, Renwick A, Elliott A,
Reid S, Spanova K, Barfoot R, Chagtai T, Jayatilake H,
McGuffog L, Hanks S, Evans DG, Eccles D, Breast Cancer
Susceptibility Collaboration (UK), Easton DF, Stratton MR
(2007) PALB2, which encodes a BRCA2-interacting protein, is a
breast cancer susceptibility gene. Nat Genet 39:165–167
16. Erkko H, Xia B, Nikkila
¨J, Schleutker J, Syrja
¨koski K, Manner-
maa A, Kallioniemi A, Pylka
¨s K, Karppinen SM, Rapakko K,
Miron A, Sheng Q, Li G, Mattila H, Bell DW, Haber DA, Grip
M, Reiman M, Jukkola-Vuorinen A, Mustonen A, Kere J, Aal-
tonen LA, Kosma VM, Kataja V, Soini Y, Drapkin RI, Livingston
DM, Winqvist R (2007) A recurrent mutation in PALB2 in
Finnish cancer families. Nature 446:316–319
17. Foulkes WD, Ghadirian P, Akbari MR, Hamel N, Giroux S,
Sabbaghian N, Darnel A, Royer R, Poll A, Fafard E, Robidoux A,
Martin G, Bismar TA, Tischkowitz M, Rousseau F, Narod SA
(2007) Identification of a novel truncating PLAB2 mutation and
Novel PALB2 mutation in breast cancer patients
123
analysis of its contribution to early-onset breast cancer in French-
Canadian women. Breast Cancer Res 9:R83
18. Dansonka-Mieszkowska A, Kluska A, Moes J, Dabrowska M,
Nowakowska D, Niwinska A, Derlatka P, Cendrowski K,
Kupryjanczyk J (2010) A novel germline PALB2 deletion in
Polish breast and ovarian cancer patients. BMC Med Genet 11:20
19. Poumpouridou N, Kroupis C (2011) Hereditary breast cancer:
beyond BRCA genetic analysis; PALB2 emerges. Clin Chem Lab
Med 50:423–434
20. Casadei S, Norquist BM, Walsh T, Stray S, Mandell JB, Lee MK,
Stamatoyannopoulos JA, King MC (2011) Contribution of in-
herited mutations in the BRCA2-interacting protein PALB2 to
familial breast cancer. Cancer Res 71:2222–2229
21. Sy SM, Huen MS, Chen J (2009) MRG15 is a novel PALB2-
interacting factor involved in homologous recombination. J Biol
Chem 284:21127–21131
22. Hayakawa T, Zhang F, Hayakawa N, Ohtani Y, Shinmyozu K,
Nakayama J, Andreassen PR (2010) MRG15 binds directly to
PALB2 and stimulates homology-directed repair of chromosomal
breaks. J Cell Sci 123:1124–1130
23. Zheng Y, Zhang J, Niu Q, Huo D, Olopade OI (2012) Novel
germline PALB2 truncating mutations in African American
breast cancer patients. Cancer 118:1362–1370
24. Wong-Brown MW, Avery-Kiejda KA, Bowden NA, Scott RJ
(2014) Low prevalence of germline PALB2 mutations in Aus-
tralian triple-negative breast cancer. Int J Cancer 134:301–305
25. Blanco A, de la Hoya M, Osorio A, Diez O, Miramar MD, Infante
M, Martinez-Bouzas C, Torres A, Lasa A, Llort G, Brunet J,
Gran
˜a B, Perez Segura P, Garcia MJ, Gutie
´rrez-Enrı
´quez S,
Carracedo A
´, Tejada MI, Velasco EA, Calvo MT, Balman
˜aJ,
Benitez J, Calde
´s T, Vega A (2013) Analysis of PALB2 gene in
BRCA1/BRCA2 negative Spanish hereditary breast/ovarian
cancer families with pancreatic cancer cases. PLoS One 8:e67538
26. Blanco A, de la Hoya M, Balman
˜a J, Ramo
´n y Cajal T, Teule
´A,
Miramar MD, Esteban E, Infante M, Benı
´tez J, Torres A, Tejada
MI, Brunet J, Gran
˜a B, Balbı
´nM,Pe
´rez-Segura P, Osorio A,
Velasco EA, Chirivella I, Calvo MT, Feliubadalo
´L, Lasa A,
´ez
O, Carracedo A, Calde
´s T, Vega A (2012) Detection of a large
rearrangement in PALB2 in Spanish breast cancer families with
male breast cancer. Breast Cancer Res Treat 132:307–315
27. Caste
´ra L, Krieger S, Rousselin A, Legros A, Baumann JJ, Bruet
O, Brault B, Fouillet R, Goardon N, Letac O, Baert-Desurmont S,
Tinat J, Bera O, Dugast C, Berthet P, Polycarpe F, Layet V,
Hardouin A, Fre
´bourg T, Vaur D (2014) Next-generation se-
quencing for the diagnosis of hereditary breast and ovarian cancer
using genomic capture targeting multiple candidate genes. Eur J
Hum Genet. doi:10.1038/ejhg.2014.16
M. T. Vietri et al.
123
... Sanger sequencing on the other blood sample was used to confirm the presence of a point mutation, as described [21][22][23]. Molecular analysis in the family members of the probands with mutation was performed by Sanger sequencing [21][22][23]. ...
... Sanger sequencing on the other blood sample was used to confirm the presence of a point mutation, as described [21][22][23]. Molecular analysis in the family members of the probands with mutation was performed by Sanger sequencing [21][22][23]. The results were elaborated using Mutation Surveyor ® software, version 3.24 (Softgenetics, State College, PA, USA). ...
Article
Full-text available
Hereditary cancer syndromes predispose to several types of cancer due to inherited pathogenic variants in susceptibility genes. We describe the case of a 57-year-old woman, diagnosed with breast cancer, and her family. The proband belongs to a family with a suspected tumor syndrome, due to other cancer cases in her family from the paternal and maternal sides. After oncogenetic counseling, she was subjected to mutational analysis with an NGS panel analyzing 27 genes. The genetic analysis showed two monoallelic mutations in low penetrance genes, c.1187G>A (p.G396D) in MUTYH and c.55dup (p.Tyr19Leufs*2) in BRIP1. One of the mutations was inherited from the maternal side and the other from the paternal side, suggesting two different cancer syndrome types in the family. MUTYH mutation was related to the onset of cancers on the paternal side, as confirmed by the occurrence of the same mutation in the proband’s cousin. BRIP1 mutation was found in the proband’s mother, indicating that it was related to the cancer cases observed on the maternal side, including breast cancer and sarcoma. Advances in NGS technologies have allowed the identification of mutations in families with hereditary cancers in genes other than those related to a specific suspected syndrome. A complete oncogenetic counseling, together with molecular tests that enable a simultaneous analysis of multiple genes, is essential for the identification of a correct tumor syndrome and for clinical decision-making in a patient and his/her family. The detection of mutations in multiple susceptibility genes allows the initiation of early risk-reducing measures for identified mutation carriers among family members and to include them in a proper surveillance program for specific syndromes. Moreover, it may enable an adapted treatment for the affected patient, permitting personalized therapeutic options.
... Older males showed a higher mortality rate than females (Mostaza et al., 2020), likely due to sex-specific differences in their immunological background (Klein and Flanagan, 2016). Frail elderly subjects having a general higher prevalence of traditional cardiovascular risk factors and comorbidities, mainly cancer, were more prone to enter in intensive care unit (ICU) during pandemics (Fiorentino et al., 2023;Damayanthi et al., 2021;Wang et al., 2020;Zhou et al., 2020;Napoli et al., 2020a, Suleyman et al., 2020Henry and Vikse, 2020, Niu et al., 2020, Vietri et al., 2015, de Nigris et al., 2013. This is because SARS-CoV-2 virus may inflict a direct attack to the heart and vasculature impacting on cardiac aging and exacerbating the pro-inflammatory state, known as "cytokine storm" which, in turn, was associated to acute respiratory distress syndrome (ARDS) onset and high mortality (Wang et al., 2020;Zhou et al., 2020;Napoli et al., 2020a;Liu et al., 2020;Evans et al., 2020;Hojyo et al., 2020). ...
Article
Full-text available
Immunosenescence contributes to the decline of immune function leading to a reduced ability to respond to severe coronavirus disease 2019 (COVID-19) in elderly patients. Clinical course of COVID-19 is widely heterogeneous and guided by the possible interplay between genetic background and epigenetic-sensitive mechanisms underlying the immunosenescence which could explain, at least in part, the higher percentage of disease severity in elderly individuals. The most convincing evidence regards the hypomethylation of the angiotensin-converting enzyme 2 (ACE2) promoter gene in lungs as well as the citrullination of histone H3 in neutrophils which have been associated with worsening of COVID-19 outcome in elderly patients. In contrast, centenarians who have showed milder symptoms have been associated to a younger “epigenetic age” based on DNA methylation profiles at specific genomic sites (epigenetic clock). Some large prospective studies showed that the acceleration of epigenetic aging as well as the shortening of telomeres were significantly associated with lymphopenia and poor outcome suggesting prognostic biomarkers in elderly COVID-19 patients. Furthermore, randomized clinical trials showed that statins, L-arginine, and resveratrol could mediate anti-inflammatory effects via indirect epigenetic interference and might improve COVID-19 outcome. Here, we discuss the epigenetic-sensitive events which might contribute to increase the risk of severity and mortality in older subjects and possible targeted therapies to counteract immunosenescence.
... Indeed, PALB2 serves as a link between BRCA1 and BRCA2 to induce homologous recombination. It is one of the DNA repair genes; PALB2 mutations are connected with HBOC risk, especially breast and pancreatic malignancies, but few studies have revealed PALB2 variations in PCa patients [38]. ...
Article
Full-text available
The demand for genetic testing (GT) for prostate cancer (PCa) is expanding, but there is limited knowledge about the genetic counseling (GC) needs of men. A strong-to-moderate inherited genetic predisposition causes approximately 5–20% of prostate cancer (PCa). In men with prostate cancer, germline testing may benefit the patient by informing treatment options, and if a mutation is noticed, it may also guide screening for other cancers and have family implications for cascade genetic testing (testing of close relatives for the same germline mutation). Relatives with the same germline mutations may be eligible for early cancer detection strategies and preventive measures. Cascade family testing can be favorable for family members, but it is currently unutilized, and strategies to overcome obstacles like knowledge deficiency, family communication, lack of access to genetic services, and testing expenses are needed. In this review, we will look at the genetic factors that have been linked to prostate cancer, as well as the role of genetic counseling and testing in the early detection of advanced prostate cancer.
... The use of PARP inhibitors has shown great promise in clinical studies in patients with BRCA1/2mutated tumors. In addition to BRCAs, ATM, ATR, CHK2 and PALB2 mutations are potential biomarkers for PARP inhibitor sensitivity [8,9]. Meanwhile, several PARP that have been approved to treat ovarian and breast cancers, also have been proven to be effective in prostate and pancreatic cancer treatments in different clinical settings [10,11]. ...
Article
Full-text available
As homologous recombination deficiency (HRD) is a biomarker to predict the efficiency of PARP inhibitor treatment, this study developed a non-exonic single-nucleotide polymorphism (SNP)-based targeted next-generation sequencing (Tg-NGS) panel and comprehensively examined it both on standards and clinical ovarian cancer tissues. The HRD scores calculated by the panel and whole-genome sequencing (WGS) were consistent, and the analysis by Sequenza was the most reliable. The results on clinical samples revealed that the panel performed better in HRD analysis than SNP microarray. There are several distinctions between this newly developed kit and reported HRD detection panels. First, the panel covers only 52,592 SNPs, which makes it capable of detecting genomic instability. Second, all the SNPs are non-exonic: as a result, the panel can be used cooperatively with any exon panel. Third, all the SNPs selected have a high minor allele frequency (MAF) in Chinese people, making it a better choice for HRD detection in Chinese patients. In summary, this panel is promising in clinical application to guide PARP inhibitors or platinum drugs used in the treatment of ovarian and other cancers.
... Targeted DNA sequencing and circulating free DNA (cfDNA) analysis in oncology are well-established to identify somatic mutations arising in tumors [43][44][45][46]. In contrast, the diagnostic use of cfDNA in transplantation is very different and focuses on detection of germline polymorphic regions that differ between recipient and donor but do not arise spontaneously within the allograft [43,46]. ...
Article
Although decades of the reductionist approach achieved great milestones in optimizing the immunosuppression therapy, traditional clinical parameters still fail in predicting both acute and chronic (mainly) rejection events leading to higher rates across all solid organ transplants. To clarify the underlying immune-related cellular and molecular mechanisms, current biomedical research is increasingly focusing on “transplantomics” which relies on a huge quantity of big data deriving from genomics, transcriptomics, epigenomics, proteomics, and metabolomics platforms. The AlloMap (gene expression) and the AlloSure (donor-derived cell-free DNA) tests represent two successful examples of how omics and liquid biopsy can really improve the precision medicine of heart and kidney transplantation. One of the major challenges in translating big data in clinically useful biomarkers is the integration and interpretation of the different layers of omics datasets. Network Medicine offers advanced bioinformatic-molecular strategies which were widely used to integrate large omics datasets and clinical information in end-stage patients to prioritize potential biomarkers and drug targets. The application of network-oriented approaches to clarify the complex nature of graft rejection is still in its infancy. Here, we briefly discuss the real-life clinical applications derived from omics datasets as well as novel opportunities for establishing predictive tests in solid organ transplantation. Also, we provide an original “graft rejection interactome” and propose network-oriented strategies which can be useful to improve precision medicine of solid organ transplantation.
... The assessment of tumor genetic alterations is currently crucial in oncological patient management and treatment decisions [1][2][3][4][5][6][7][8][9][10]. In fact, in the era of personalized medicine, an accurate genetic tumor assessment allows to identify the more precise treatment, to define the treatment time, and to standardize the follow-up time [11][12][13][14][15][16][17][18][19][20][21]. ...
Article
Full-text available
Purpose: To assess the qualitative relationship between liquid biopsy and conventional tissue biopsy. As a secondary target, we evaluated the relationship between the liquid biopsy results and the T stage, N stage, M stage, and compared to grading. Methods: The Local Ethics Committee of the "Università degli Studi della Campania Luigi Vanvitelli", with the internal resolution number 24997/2020 of 12.11.2020, approved this spontaneous prospective study. According to the approved protocol, patients with lung cancer who underwent Fine-Needle Aspiration Cytology (FNAC), CT-guided biopsy, and liquid biopsy were enrolled. A Yates chi-square test was employed to analyze differences in percentage values of categorical variables. A p-value < 0.05 was considered statistically significant. Data analysis was performed using the Matlab Statistic Toolbox (The MathWorks, Inc., Natick, MA, USA). Results: When a genetic mutation is present on the pathological examination, this was also detected on the liquid biopsy. ROS1 and PDL1 mutations were found in 2/29 patients, while EGFR Exon 21 was identified in a single patient. At liquid biopsy, 26 mutations were identified in the analyzed samples. The mutations with the highest prevalence rate in the study populations were: ALK (Ile1461Val), found in 28/29 patients (96.6%), EML4 (Lys398Arg), identified in 16/29 (55.2%) patients, ALK (Asp1529Glu), found in 14/29 (48.3%) patients, EGFR (Arg521Lys), found in 12/29 (41.4%) patients, ROS (Lys2228Gln), identified in 11/29 (37.9%) patients, ROS (Arg167Gln) and ROS (Ser2229Cys), identified in 10/29 (34.5%) patients, ALK (Lys1491Arg) and PIK3CA (Ile391Met), identified in 8/29 (27.6%) patients, ROS (Thr145Pro), identified in 6/29 (20.7%) patients, and ROS (Ser1109Leu), identified in 4/29 (13.8%) patients. No statistically significant differences can be observed in the mutation rate between the adenocarcinoma population and the squamous carcinoma population (p > 0.05, Yates chi-square test). Conclusions: We showed that, when a genetic mutation was detected in pathological examination, this was always detected by liquid biopsy, demonstrating a very high concordance rate of genomic testing between tissues and their corresponding mutations obtained by liquid biopsy, without cases of false-negative results. In addition, in our study, liquid biopsy highlighted 26 mutations, with the prevalence of ALK mutation in 96.6% of patients, supporting the idea that this approach could be an effective tool in cases with insufficient tumor tissue specimens or in cases where tissue specimens are not obtainable.
Chapter
Prostate cancer (PCa) is the second most diagnosed cancer type globally and is one of the leading causes of death in men. Genetic susceptibility plays a significant role in PCa development with a reported heritability of 57%. Mutations in the different DNA damage repair (DDR) genes (BRCA1, BRCA2, CHEK2, ATM, and PALB2) and in DNA mismatch repair (MMR) genes (MLH1, MSH2, MSH6, and PMS2) are hallmark of hereditary prostate cancer (HPCa) and are included in National Comprehensive Cancer Network (NCCN) guidelines for PCa germline genetic testing. In addition to rare high-risk mutations in susceptibility genes, polygenetic inheritance of low-risk germline variants in the form of single-nucleotide polymorphisms (SNPs) may be utilized to distinguish an individual’s susceptibility to PCa onset and progression. Over the past decade, the number of detected variants has increased to 269, due to the genome-wide association studies (GWAS). The large number of identified variants led to the development of polygenic risk scores (PRS) that aggregates common PCa-associated genetic variants into a single measure. The incorporation of diverse genetic analyses and PRS is highly anticipated to those individuals with positive PCa family history and may lead to improvements in clinical outcomes for this population through early prevention screening efforts.
Article
MORF4-related gene on chromosome 15 (MRG15), a chromatin remodeller, is evolutionally conserved and ubiquitously expressed in mammalian tissues and cells. MRG15 plays vital regulatory roles in DNA damage repair, cell proliferation and division, cellular senescence and apoptosis by regulating both gene activation and gene repression via associations with specific histone acetyltransferase and histone deacetylase complexes. Recently, MRG15 has also been shown to rhythmically regulate hepatic lipid metabolism and suppress carcinoma progression. The unique N-terminal chromodomain and C-terminal MRG domain in MRG15 synergistically regulate its interaction with different cofactors, affecting its functions in various cell types. Thus, how MRG15 elaborately regulates target gene expression and performs diverse functions in different cellular contexts is worth investigating. In this review, we provide an in-depth discussion of how MRG15 controls multiple physiological and pathological processes.
Article
We consider the risk of a disease caused by the presence within the genome of one deleterious mutation or two interacting mutations. For an individual, the probability of being mutated/doubly mutated can be estimated by knowing the phenotype of the other members in the family pedigree. We study the performance of this process as a function of the size, the shape of the family tree and the parameters of the model. We carry out simulations using the parameters pertaining to breast/ovarian cancer in BRCA-mutated families.
Article
Full-text available
Purpose: Monoallelic germ-line deleterious mutations of PALB2 (partner and localizer of BRCA2) are associated with breast cancer risk and have been found in several populations, with carrier frequencies of ~1-2%. Initially, these mutations were considered to have moderate penetrance, but accumulating evidence now indicates that they are associated with much higher risk. Methods: In this study, we sequenced the PALB2 coding regions unlinked to BRCA (breast cancer) genes in 575 probands from Italian breast cancer families recruited in Milan. Results: We found 12 carriers (2.1%) of deleterious mutations, and none of the mutations was found in 784 controls collected in Milan. One of these mutations, the c.1027C>T (p.Gln343X), was found to be recurrent in the province of Bergamo in northern Italy, being detected in 6/113 (5.3%) familial breast cancer cases and 2/477 (0.4%) controls recruited in this area (Fisher's exact test: P < 0.01). Conclusions: Our data provide confirmatory findings that, in the Italian population also, deleterious mutations of PALB2 are relatively frequent predisposing factors for breast cancer and may be associated with high risk of the disease.
Article
Full-text available
To optimize the molecular diagnosis of hereditary breast and ovarian cancer (HBOC), we developed a next-generation sequencing (NGS)-based screening based on the capture of a panel of genes involved, or suspected to be involved in HBOC, on pooling of indexed DNA and on paired-end sequencing in an Illumina GAIIx platform, followed by confirmation by Sanger sequencing or MLPA/QMPSF. The bioinformatic pipeline included CASAVA, NextGENe, CNVseq and Alamut-HT. We validated this procedure by the analysis of 59 patients' DNAs harbouring SNVs, indels or large genomic rearrangements of BRCA1 or BRCA2. We also conducted a blind study in 168 patients comparing NGS versus Sanger sequencing or MLPA analyses of BRCA1 and BRCA2. All mutations detected by conventional procedures were detected by NGS. We then screened, using three different versions of the capture set, a large series of 708 consecutive patients. We detected in these patients 69 germline deleterious alterations within BRCA1 and BRCA2, and 4 TP53 mutations in 468 patients also tested for this gene. We also found 36 variations inducing either a premature codon stop or a splicing defect among other genes: 5/708 in CHEK2, 3/708 in RAD51C, 1/708 in RAD50, 7/708 in PALB2, 3/708 in MRE11A, 5/708 in ATM, 3/708 in NBS1, 1/708 in CDH1, 3/468 in MSH2, 2/468 in PMS2, 1/708 in BARD1, 1/468 in PMS1 and 1/468 in MLH3. These results demonstrate the efficiency of NGS in performing molecular diagnosis of HBOC. Detection of mutations within other genes than BRCA1 and BRCA2 highlights the genetic heterogeneity of HBOC.European Journal of Human Genetics advance online publication, 19 February 2014; doi:10.1038/ejhg.2014.16.
Article
Full-text available
The PALB2 gene, also known as FANCN, forms a bond and co-localizes with BRCA2 in DNA repair. Germline mutations in PALB2 have been identified in approximately 1% of familial breast cancer and 3-4% of familial pancreatic cancer. The goal of this study was to determine the prevalence of PALB2 mutations in a population of BRCA1/BRCA2 negative breast cancer patients selected from either a personal or family history of pancreatic cancer. 132 non-BRCA1/BRCA2 breast/ovarian cancer families with at least one pancreatic cancer case were included in the study. PALB2 mutational analysis was performed by direct sequencing of all coding exons and intron/exon boundaries, as well as multiplex ligation-dependent probe amplification. Two PALB2 truncating mutations, the c.1653T>A (p.Tyr551Stop) previously reported, and c.3362del (p.Gly1121ValfsX3) which is a novel frameshift mutation, were identified. Moreover, several PALB2 variants were detected; some of them were predicted as pathological by bioinformatic analysis. Considering truncating mutations, the prevalence rate of our population of BRCA1/2-negative breast cancer patients with pancreatic cancer is 1.5%. The prevalence rate of PALB2 mutations in non-BRCA1/BRCA2 breast/ovarian cancer families, selected from either a personal or family pancreatic cancer history, is similar to that previously described for unselected breast/ovarian cancer families. Future research directed towards identifying other gene(s) involved in the development of breast/pancreatic cancer families is required.
Article
Full-text available
For almost two decades, breast cancer clinical genetics has operated in an environment where a heritable cause of breast cancer susceptibility is identified in the vast minority of women seeking advice about their personal and/or family history of breast and/or ovarian cancer. A new wave of genetic information is upon us that promises to provide an explanation for the greater proportion of current missing heritability of breast cancer. Whilst researchers refine bioinformatic and analytic methodology necessary to interpret the new genetic data, attention needs to be paid to defining appropriate and coordinated pathways for the translation of this information so that it can be applied in clinical genetic services for the benefit of the majority of women who currently have no explanation for their breast cancer susceptibility. The search for additional breast cancer susceptibility genes remains a very active area of research. Exhausting the power of linkage studies that identified BRCA1 and BRCA2, the research community moved to candidate gene studies that led to the identification of ATM, BRIP1, CHEK2, and PALB2 as so-called "moderate-risk" breast cancer susceptibility genes. Mutations in these genes are rare and although early reports suggested that, on average, they are associated with moderate risks of breast cancer; population-based studies have demonstrated that at least some mutations in these genes are associated with breast cancer risks that are comparable to the average risk associated with BRCA2 mutations. The search for additional breast cancer susceptibility genes has now moved onto research platforms applying massively parallel sequencing capable of sequencing whole human exomes and genomes in single instrument runs. These programs are identifying a large number of additional putative breast cancer susceptibility genes, many of which are currently undergoing validation. It is highly anticipated that the remaining missing heritability of breast cancer will be due to mutations in many different genes, each explaining a small proportion of the currently unexplained heritable breast cancer susceptibility. The characterization of PALB2 as a breast cancer susceptibility gene and subsequent research that has refined our understanding of the prevalence and penetrance of heritable mutations in PALB2 offers a precious opportunity to use the data as a model and develop modes of translation that would be appropriate for the anticipated volume of imminent new information.
Article
Full-text available
Recent reports have shown that mutations in the FANCJ/BRIP1 and FANCN/PALB2 Fanconi Anemia (FA) genes confer a moderate breast cancer risk. Discussion has been raised on the phenotypic characteristics of the PALB2-associated families and tumors. The role of FANCB in breast cancer susceptibility has not been tested to date. Likewise PALB2 mutation frequency has not been studied in Spanish population. We analyzed the complete coding sequence and splicing sites of FANCB and PALB2 in 95 index cases of BRCA1/2-negative Spanish breast cancer families. We also performed an exhaustive screening of three previously described rare but recurrent PALB2 mutations in 725 additional probands. Pathogenic changes were not detected in FANCB. We found a novel PALB2 truncating mutation c.1056_1057delGA (p.K353IfsX7) in one of the 95 screened patients, accounting for a mutation frequency of 1% in our series. Further comprehensive screening of the novel mutation and of previously reported rare but recurrent PALB2 mutations did not reveal any carrier patient. We report the first example of LOH occurring in a PALB2-associated tumor. Our results rule out a major contribution of FANCB to hereditary breast cancer. Our data are consistent with the notion of individually rare PALB2 mutations, lack of mutational hot-spots in the gene and existence of between-population disease-allele heterogeneity. We show evidence that PALB2 loss of function might also conform to the inactivation model of a classic tumor-suppressor gene and present data that adds to the clinically relevant discussion about the existence of a PALB2-breast cancer phenotype.
Article
Full-text available
Abstract Despite the initial enthusiasm following the discovery of the association of BRCA germline mutations with hereditary breast and/or ovarian cancer, in many families affected by the syndrome no pathogenic mutations were detected in the two genes, although exhaustively searched. Many other genes have also been implicated due to their role in the same pathway of DNA repair where the BRCA1/2 genes are involved: homologous recombination (HR). Among them, PALB2 clearly emerges as the third breast cancer susceptibility gene. Its mutations have been detected in most populations investigated so far, albeit rarely: in 1%-4% of families negative for BRCA mutations, with either partial or complete penetrance. In some populations, PALB2 recurrent mutations have been identified and the estimated hazard risks are comparable to those of BRCA mutations. Since new effective targeted therapeutic options are becoming available ("synthetic lethality" with novel PARP inhibitors, etc.) that are applicable to all those patients with a defect in HR pathway, it is imperative to detect all these candidate patients. Data obtained from laboratory tests in the tumor (simple immunohistochemistry, gene expression analysis, etc.) can assist in the recognition of a specific pattern (BRCA1ness, HRless) so that even patients that look "sporadic" could benefit from these targeted therapies. Therefore, a genetic analysis algorithm is proposed, although with the advent of Next Generation Sequencing it is predicted that in the future most germline genetic alterations and also somatic or epigenetic events in the tumor of these genes will be detected.
Article
Full-text available
It has been demonstrated that monoallelic PALB2 (Partner and Localizer of BRCA2) gene mutations predispose to familial breast cancer. Some of the families reported with germline PALB2 mutations presented male breast cancer as a characteristic clinical feature. Therefore, we wanted to investigate the contribution of germline PALB2 mutations in a set of 131 Spanish BRCA1/BRCA2-negative breast/ovarian cancer families with at least one male breast cancer case. The analysis included direct sequencing of all coding exons and intron/exon boundaries as well as a Multiplex Ligation-dependent Probe Amplification-based analysis of genomic rearrangements. For the first time we have identified a genomic rearrangement of PALB2 gene involving a large deletion from exon 7 to 11 in a breast cancer family. We have also identified several PALB2 variants, but no other obvious deleterious PALB2 mutation has been found. Thus, our study does not support an enrichment of PALB2 germline mutations in the subset of breast cancer families with male breast cancer cases. The identification of intronic and exonic variants indicates the necessity of assessing the implications of variants that do not lead to PALB2 truncation in the pathoghenicity of the PALB2 gene.
Article
Background: Double heterozygosity (DH) is an extremely rare event in which both BRCA1 and BRCA2 are mutated simultaneously in a family. To date, few cases of DH have been reported, especially in Ashkenazi populations. In Italy some cases of DH have been reported. In this study, we have described an Italian family with double heterozygosity in the BRCA genes. Methods: The proband is a 43-year-old woman with bilateral breast cancer. She presented two pathogenic mutations in both BRCA genes, IVS8+2T>A (c.547+2T>A;p.Gln148Aspfsx51) in BRCA1, K944X (c.2830A>T;p.Lys944X) in BRCA2 and a novel variant IVS4-57A>G (c.426-57A>G) in BRCA2, not previously described. Both mutations were inherited from the paternal lineage in the proband's family. We investigated all available members of this family and we identified other two family members with DH. Results and conclusions: Our observations support the hypothesis of a non-specific severe phenotype in DH carriers in terms of age of disease onset, cumulative lifetime risk and multiple primary tumours. Furthermore, our findings confirm that in order to identify all cases of DH, it is important not to limit the identification of mutations in a single gene, but extend the analysis to BRCA1 and BRCA2 and other breast cancer susceptibility genes.
Article
Triple-negative breast cancer (TNBC) is a tumour classification that is defined by ER, PR, and HER2 receptor negativity. TNBCs share a similar gene expression profile to BRCA-mutated tumours, have been shown to carry a high proportion of BRCA mutations, and have a more adverse prognosis compared to other types of breast tumours. PALB2 has been shown to be a moderate-penetrance breast cancer susceptibility gene and is involved in the same DNA damage repair pathway as BRCA1 and BRCA2; this raises the possibility that germline PALB2 mutations may be involved in the pathogenesis of TNBCs. In this study, we sequenced the coding regions of PALB2 (including intron/exon boundaries) in genomic DNA from 347 patients diagnosed with TNBC to determine the prevalence of deleterious mutations in this population. Two novel truncating mutations (c.758dup and c.2390del) and one previously-detected truncating mutation (c.3113+5G>C) were found. In addition, five variants predicted to be protein-affecting were also identified. This study shows that the prevalence of PALB2 germline mutations in individuals with TNBC is approximately 1%, similar to the prevalence of PALB2 germline mutation of 1% in familial non-BRCA1/2 breast cancer cohorts. © 2013 Wiley Periodicals, Inc.
Article
Background A few breast cancer cases are attributable to a hereditary predisposition to the disease. We aimed to compare the histological features of breast cancer in women carrying mutations in the susceptibility genes BRCA1 and BRCA2 with controls unselected for family history. Methods The morphological characteristics of specimens from 440 patients with familial breast cancer, including 118 in carriers of BRCA1 mutations and 78 in carriers of BRCA2 mutations, were compared with those from 547 age-matched controls, unselected for family history, by seven pathologists Findings Cancers in carriers of BRCA1 (p<0·0001) and BRCA2 mutations (p=0·04) were, on average, of a higher overall grade than in controls. For example, the proportions in grade 3 were 66% of 139, 41% of 58 and 36% of 368 specimens, respectively. However, when the three grade indices were considered independently, breast cancers in BRCA1-mutation carriers showed more pleomorphism (p=0·006), a higher mitotic count (p<0·0001), and less tubule formation than controls (p=0·006), whereas cancers in BRCA2-mutation carriers showed less tubule formation (p=0·003), but no difference in pleomorphism or mitotic count. The occurrence of invasive lobular carcinoma and invasive ductal carcinoma was not significantly different between carriers of BRCA1 or BRCA2 mutations and controls. Medullary or atypical medullary carcinoma was, however, found more often in BRCA1 (13%, p<0·0001) than in BRCA2-mutation carriers (3%) or controls (2%). Tubular carcinoma was less common in BRCA2-mutation carriers. The few mucoid carcinomas were all in familial cases. Carriers of BRCA1 mutations showed less ductal carcinoma in situ around the invasive lesion than controls (41 vs 56%, p=0·001). Lobular carcinoma in situ was less common in familial cancers (p=0·013), but differences were not significant for BRCA1-mutations or BRCA2-mutation carriers, separately. Interpretation The histology of breast cancers in predisposed women differs from that in sporadic cases, and there are differences between breast cancers in carriers of BRCA1 and BRCA2 mutations. The findings suggest that breast cancer due to BRCA1, has a different natural history to BRCA2 or apparently sporadic disease, which may have implications for screening and management.