A mutation in the gene encoding the small subunit-associated ribosomal protein RPS19, leading to RPS19 haploinsufficiency, is one of the ribosomal protein gene defects responsible for the rare inherited bone marrow failure syndrome Diamond Blackfan anemia (DBA). Additional inherited and acquired defects in ribosomal proteins (RPs) continue to be identified and are the basis for a new class of diseases called the ribosomopathies. Acquired RPS14 haploinsufficiency has been found to be causative of the bone marrow failure found in 5q– myelodysplastic syndromes. Both under- and overexpression of RPs have also been implicated in several malignancies. This review will describe the somatic ribosomopathies that have been found to be associated with a variety of solid tumors as well as leukemia and will review cancers in which over- or underexpression of these proteins seem to be associated with outcome.

Learning Objectives
  • To review the leukemias and solid tumors found to have an acquired ribosomal protein defect

  • To review the solid tumors in which over- and underexpression of ribosomal proteins seem to be associated with outcome

Each ribosome that constitutes the cellular translational machinery is composed of a small (40S) subunit consisting of an 18S RNA and 33 ribosomal proteins (RPs) and a large (60S) subunit with a 5S RNA, a 28S RNA, a 5.8S RNA, and 46 RPs. This complex of structural ribosomal RNAs and associated proteins is carefully regulated and omnipresent. Thus, the initial discovery in 1997 by Gustavsson et al1  that Diamond-Blackfan anemia (DBA), a rare inherited bone marrow failure syndrome, was the result of a defect in the gene encoding the small subunit-associated ribosomal protein RPS19 was met with considerable skepticism. DBA now stands as the founding member of the class of disorders known as ribosomopathies, and pathogenic mutations have been described in at least 19 other RP genes.2-11  In DBA, these mutated RP genes are estimated to be inherited in an autosomal dominant manner in about half the cases, the remaining cases being de novo mutations. DBA has also recently been clarified as a cancer predisposition syndrome.12  Patients with DBA are predisposed to a variety of solid tumors as well as myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Adult patients in particular have a significantly increased risk of luminal gastrointestinal cancers.13  Notably, there seems to be no genotypic predilection to MDS, AML, or solid tumors, which suggests that cancers result from downstream events and are not related to the specific RP haploinsufficiency. Another inherited ribosomopathy, Shwachman-Diamond syndrome (SDS), results most commonly from mutations in SBDS leading to faulty ribosome subunit joining.14  SDS generally presents with neutropenia but other cytopenias and pancytopenia are not uncommon. Solid tumors are rarely reported in SDS,15  but the disorder clearly predisposes to MDS and AML, although the exact mechanisms remain unknown.16 

In 2006, Ebert et al17  reported that the 5q– syndrome, a subtype of MDS, was the result of an acquired somatic RPS14 deletion. Although typically presenting in the seventh decade of life, 5q– syndrome was soon thereafter reported in 2 children with anemia who had been misdiagnosed as having DBA.18  Over the past 10 years, somatic RP mutations have been found in a variety of cancers in patients without evidence of DBA or signs of another inherited bone marrow failure syndrome. Somatic mutations in genes encoding RPs seem to be a common feature of many cancers, suggesting their importance in oncogenesis.19  Furthermore, over- and underexpression of some of the RPs are present in a variety of malignancies and have been postulated to be predictive of outcome.20  This article summarizes the recent published findings relevant to acquired ribosomopathies (Table 1) and suggests areas of future inquiry. We describe how aberrations in RP expression that act by overexpression as oncogenes or by haploinsufficiency as possible tumor suppressors serve as heretofore underappreciated drivers of malignancy. The concordance of inactivating RP mutations with TP53 inactivation suggests that the latter is an interdicting mutation to the selective pressure of diminished translational capacity, nucleolar stress, and growth retardation as a consequence of RP loss of function.

Table 1.

Cancer types and associated RP mutations or deletions

Cancer typeRP mutation/deletionReference
T-cell acute lymphoblastic leukemia RPL5, RPL10, RPL11, RPL22 27, 31 
Chronic lymphoblastic leukemia RPS15, RPSA, RPS20 32, 33 
5q- syndrome RPS14 19 
Glioblastoma multiforme RPL5 34, 35 
Gastric adenocarcinoma RPSA, RPS5, RPL22 35, 41, 42 
Endometrial carcinoma RPS20, RPL22 35, 37, 39 
Melanoma RPL5, RPL11, RPS27 35, 36 
Breast cancer RPL5 35 
Cancer typeRP mutation/deletionReference
T-cell acute lymphoblastic leukemia RPL5, RPL10, RPL11, RPL22 27, 31 
Chronic lymphoblastic leukemia RPS15, RPSA, RPS20 32, 33 
5q- syndrome RPS14 19 
Glioblastoma multiforme RPL5 34, 35 
Gastric adenocarcinoma RPSA, RPS5, RPL22 35, 41, 42 
Endometrial carcinoma RPS20, RPL22 35, 37, 39 
Melanoma RPL5, RPL11, RPS27 35, 36 
Breast cancer RPL5 35 

An acquired deletion of the short arm of chromosome 5 has been known to lead to MDS, which may subsequently progress to AML in some patients. In particular, the 5q– syndrome has been noted mostly, but not exclusively, in women older than age 75 years who present with macrocytic anemia and erythroid hypoplasia and have a recurrent somatic 1.5-Mb commonly deleted region (CDR) in 5q.21  Ebert et al17  used short hairpin RNAs to target each of the 40 genes within the CDR and identified RPS14 haploinsufficiency as the predominant cause of erythroid hypoplasia in 5q– syndrome. Roles for CSNK1A1 and microRNA-145/146a have also been demonstrated to contribute to the overall phenotype.22  Unique to this situation and not specifically related to RP haploinsufficiency, deletions in the common deleted region (CDR) in 5q– that also result in IRAK/TRAF activation may play a role in malignant transformation.23  Interestingly, RPS14 deletions have not been identified in patients with DBA, but a growing number of other RP gene germline deletions have been noted.

Since Ebert’s seminal contribution, several hematologic malignancy–associated RP mutations have been identified. RPL22 has been shown to be essential in T-cell development through the T-cell receptor signaling pathway.24  In fact, a murine RPl22 knockout blocked the development of αβ-lineage T cells selectively by activating a p53-dependent checkpoint.25  Rao et al26  identified mutations of RPL22 that led to monoallelic inactivation in 4 (∼10%) of 47 patients with T-cell acute lymphoblastic leukemia (T-ALL). Two of these 4 patients had induction failure, and 1 responded to chemotherapy but then relapsed. In this limited population, of the 9 patients with induction failure, 2 (22%) had deletions involving the region of RPL22. Of the 38 patients who achieved induction remission, only 2 had a deletion containing the RPL22 gene. On further investigation, 6 (∼30%) of 19 T-ALL cell lines and 1 of 20 primary patient samples taken at relapse had a deletion in RPL22 that resulted in a frameshift and a subsequent truncated RPL22 protein. Haploinsufficiency of RPL22 in these patients portended aggressive disease, which prompted these authors to postulate that RPL22 is a tumor suppressor. The authors then showed that RPL22 inactivation resulted in an increase in LIN28B, which had previously been demonstrated to be a direct transcriptional target of NF-κB. The increase in LIN28B had been shown to be associated with an increase in cell proliferation and tumor growth.27  Subsequently in 2013, De Keersmaecker et al28  reported somatic RP gene mutations and deletions in RPL5, RPL10, and RPL22 in 20% of children with acute T-ALL with RPL10 R98S, a missense mutation, found exclusively in 7.9% of the pediatric patients with T-ALL. Defects in RPL11 were also found, but less frequently.

Landau and colleagues29  identified an RPS15 mutation as a novel driver mutation of chronic lymphoblastic leukemia (CLL) through whole-exome sequencing of 278 patient and germline samples and 2 other previously published whole-exome sequencing cohorts. RPS15 was recurrently mutated in 23 of the patients sequenced (4.3%), and these mutations were associated with a shorter progression-free survival. This was further confirmed by an analysis of another group of 41 patients from Europe30  in whom RPS15 mutations were found pretreatment (17.1%) and at relapse (19.5%). Of the 8 patients, 3 had additional mutations in TP53 and 3 had deletions in 11q. Similar to the whole cohort, 6 patients (75%) achieved complete remission and 5 patients (63%) relapsed within 3 years of diagnosis. Targeted resequencing of RPS15 performed on a larger CLL cohort revealed that 6% of patients had mutations. RPS15 mutations were also noted exclusively in the more aggressive forms of CLL. Concurrent mutations in TP53 were found more commonly in patients with RPS15-mutated CLL vs patients with nonmutated RPS15 CLL (36% vs 18%; P < .01). Overall survival was poor for patients with RPS15 mutations and even worse for patients with concurrent RPS15 and TP53 mutations. In addition, of the patients without RPS15 mutations, 3 had RPSA and RPS20 mutations.

RPL5 has been found to be mutated in glioblastoma multiforme (GBM) and other tumors.31  Further investigation of The Cancer Genome Atlas (TCGA) database by Fancello and colleagues32  identified 5 RP genes that were mutated in 4 different cancer types: RPL5 in cutaneous melanoma and GBM, RPL11 also in cutaneous melanoma, RPS5 in gastric adenocarcinoma, RPS20 in uterine corpus endometrial carcinoma, and RPSA also in gastric adenocarcinoma. Precise analysis of RPL5 mutations has documented heterozygous deletions in GBM (11%), melanoma (28%), and breast cancer (34%) samples associated with lower RPL5 expression. The possible role of RPL5 as a tumor suppressor gene in GBM was noted, and underexpression of RPL5 was associated with lower overall survival in these patients. Regarding cutaneous melanoma, a previously reported recurrent mutation has been noted in the 5′ untranslated region of RPS27 in about 10% of samples, and it was the most frequent mutation in the melanoma samples studied.33 

Mutations in RPL22 have also been discovered in 10.9% of uterine corpus or endometrioid endometrial carcinoma through the efforts of TCGA, which used sequencing to document somatic variants across different tumor types.34  Loss of DNA mismatch repair and subsequent tumor microsatellite instability (MSI) are found in 30% of these types of endometrial cancers.35  Further characterization of RPL22 mutations in these tumors found a heterozygous nucleotide deletion in 116 (52%) of 226 tumors.36  Interestingly, in smaller cohorts, only the MSI-high tumors carried this mutation, and it was not seen in any MSI-stable tumors. In this study, the females with the RPL22 mutations were older than mutation-negative females but had similar progression-free survival. The significance of this mutation in the progression of these tumors and its relevance with other known mutations such as PTEN and TP53 are areas that need to be investigated.

Gastric cancer is also known to have MSI. A study by Nagarajan et al37  revealed recurrent deletions of RPL22 in 64% of MSI-positive gastric cancer tumors, but no mutations were found in MSI-negative tumors. A subsequent report on MSI-unstable endometrial and colorectal tumors confirmed a heterozygous RPL22 mutation in 50% and 77% of tumors, respectively, further implicating this gene in MSI cancers.38 

Yang et al39  demonstrated downregulation of RPL22 messenger RNA (mRNA) and protein levels in non–small-cell lung cancer tissue compared with levels found in normal controls. RPL15 underexpression with lower mRNA and protein levels has been reported in pancreatic ductal adenocarcinoma tumor tissue compared with noncancerous tissues.40  Correlation with tumor characteristics in 2 cohorts of patients revealed association of low levels of RPL15 expression with poor histology and vascular invasion. Positive correlation was also noted with increased overall patient survival in those with high levels of RPL15 expression in the tumor. Further investigation with RPL15 short interfering RNAs (siRNAs) showed that RPL15 overexpression blocked the invasiveness of the pancreatic cancer cells. In a murine model, RPL15 overexpression led to fewer pulmonary metastases than in controls.

Yong et al41  reviewed the data from the TCGA database and from primary, secondary, and recurrent GBM tumor specimens to identify RPS11, RPS20, and VEGF-A as possible markers for prognosis. Overexpression of RPS11 in all GBM tumors was associated with more than a fourfold increase in death; overexpression of RPS20 had a twofold increase. This hazard rate remained the same if only primary GBMs were analyzed. However, in secondary GBMs (those who had progressed from low-grade gliomas) only increased expression of RPS11 was found to be associated with poor prognosis; in recurrent tumors, overexpression of RPS20 was more predictive of poor survival. In the TCGA database with 578 GBM reported, patients with high RPS11 or RPS20 mRNA expression had a 20% increased hazard of death when compared with patients with low-level expression. However, patients with both RPS11 and RPS20 overexpression had a 43% increase in the hazard of death when compared with those with low levels of both. These authors further postulate that RPS11 and RPS20 may be therapeutic targets.

Investigators have also compared patients’ gastric cancer tumor tissue with non-tumor tissue and found increased expression of RPL15 in the tumors compared with the normal gastric tissue from controls, but this did not correlate with cancer stage.42  This overexpression was also seen in different gastric cancer cell lines. The authors demonstrated siRNA knockdown of RPL15 leading to decrease in tumor size in mice, thus deeming RPL15 a potential therapeutic target. Overexpression of full-length RPL15 complementary DNA has also been noted in esophageal tumors.43  Kasai and colleagues44  studied expression profiles of many RPs in human normal colorectal mucosa and colorectal adenocarcinoma cells. They found that RPS11 and RPL7 were significantly overexpressed in the cancer cells but 10 other RPs were underexpressed. RPL13 was found to be overexpressed in 28% of gastric cancer, 41% of colorectal cancer, and 20% of liver cancer tissues compared with normal tissue.45  Inhibition of RPL13 by siRNA transfection showed reduction in cancer cell growth. RPL13 mRNA overexpression correlated with advanced stage in gastric cancer and in colorectal cancer, but it was not statistically significant in the latter, and with no other pathologic or clinical features.

RP genes were found to be both up- and downregulated in human hepatocellular carcinoma (HCC).46  Further investigations were performed on RPL36 because of its overexpression in hepatoma cell lines.47  Overexpression of RPL36 was noted in human HCC tumor tissue compared with the adjacent non-tumor tissue. Interestingly, high RPL36 levels correlated with hepatic synthetic function and lower α-fetoprotein levels whereas low RPL36 levels were noted with increased portal vein invasion and higher stage of HCC. In fact, patients with high levels in their tumors were found to have longer survival. Thus, according to these findings, levels of RPL36 may be decreased with tumor progression and could be used as a marker of disease state.

Overexpression of RPL39 (along with MLF2) was found in breast cancer stem cells derived from patient biopsies and lung metastases from these patients.48  Overexpression was also associated with cell migration and proliferation, and treatment with RPL39 siRNA showed significant decrease in tumor volume. The gain-of-function mutation found in RPL39 was not found in the primary tumors in the TCGA database but was present in more than 10% of the lung metastases. Clinically, patients with this mutation had a significantly shorter median time to relapse compared with those without this mutation.

RPL19 has been reported to also be overexpressed in malignant prostate cancer cell lines, 4.9 to 6.7-fold higher than in benign prostate cancer cell lines.49  This was recapitulated in malignant and benign human prostate tissues. In fact, when assessed with regard to the grade of the malignancy, intensity of RPL19 staining positively correlated with the higher grade of the tumor. Kaplan-Meier analysis confirmed that patients who expressed higher levels of RPL19 in the tumor had significantly poorer survival, thus making this a possible tumor marker in the future.

Strong evidence now exists that overexpression of RPs that results in disrupted translation can be an oncogenic driver that confers malignant growth potential to tissues with such acquired mutations. It also seems certain that RP haploinsufficiency, both germline (DBA) and somatic (5q– syndrome and other cancers), creates a selective pressure predisposing to malignancy. The importance of RP hemizygosity as a driver of malignancy is strongly supported by the observation that 43% of several tumor specimens and cancer cell lines are RP haploinsufficient.19  Furthermore, the strong association of somatic acquired RP hemizygosity with inactivating mutations of TP53, as described by Ajore et al,50  imply that the TP53 inactivation may result from inactivating interdicting mutations in TP53 as a consequence of the growth suppressive properties of RP haploinsufficiency. We suspect that although the notion is hypothetical, altered translation of tissue-specific transcripts acting as either oncogenes or tumor suppressors will be identified as drivers of oncogenesis as a consequence of RP haploinsufficiency. RP haploinsufficiency almost certainly also results in selective expression of preferred transcripts perhaps by favoring canonical translation over internal ribosome entry site–mediated translation. Whether this mechanism is functional in malignancy in a tissue-specific manner is currently unknown.

Both over- and underexpression of RPs suggest that they are potential therapeutic targets. The role of RP genes, presumably as both oncogenes and tumor suppressors, is an emerging science. There is much to be learned about how aberrations in the translational machinery and its components can lead to the development of both hematologic and nonhematologic cancers. It seems probable that influencing RP expression will emerge as an important therapeutic target strategy.

Adrianna Vlachos, The Feinstein Institute for Medical Research, 350 Community Dr, Manhasset, NY 11030; e-mail: avlachos@northwell.edu.

1.
Gustavsson
P
,
Willing
TN
,
van Haeringen
A
, et al
.
Diamond-Blackfan anaemia: genetic homogeneity for a gene on chromosome 19q13 restricted to 1.8 Mb
.
Nat Genet
.
1997
;
16
(
4
):
368
-
371
.
2.
Vlachos
A
,
Blanc
L
,
Lipton
JM
.
Diamond Blackfan anemia: a model for the translational approach to understanding human disease
.
Expert Rev Hematol
.
2014
;
7
(
3
):
359
-
372
.
3.
Farrar
JE
,
Vlachos
A
,
Atsidaftos
E
, et al
.
Ribosomal protein gene deletions in Diamond-Blackfan anemia
.
Blood
.
2011
;
118
(
26
):
6943
-
6951
.
4.
Landowski
M
,
O’Donohue
MF
,
Buros
C
, et al
.
Novel deletion of RPL15 identified by array-comparative genomic hybridization in Diamond-Blackfan anemia
.
Hum Genet
.
2013
;
132
(
11
):
1265
-
1274
.
5.
Farrar
JE
,
Quarello
P
,
Fisher
R
, et al
.
Exploiting pre-rRNA processing in Diamond Blackfan anemia gene discovery and diagnosis
.
Am J Hematol
.
2014
;
89
(
10
):
985
-
991
.
6.
Gripp
KW
,
Curry
C
,
Olney
AH
, et al
;
UW Center for Mendelian Genomics
.
Diamond-Blackfan anemia with mandibulofacial dystostosis is heterogeneous, including the novel DBA genes TSR2 and RPS28
.
Am J Med Genet A
.
2014
;
164A
(
9
):
2240
-
2249
.
7.
Mirabello
L
,
Macari
ER
,
Jessop
L
, et al
.
Whole-exome sequencing and functional studies identify RPS29 as a novel gene mutated in multicase Diamond-Blackfan anemia families
.
Blood
.
2014
;
124
(
1
):
24
-
32
.
8.
Bhar
S
,
Mirabello
L
,
Chatterjee
N
, et al
.
De novo RPS20 mutations in Diamond Blackfan anemia [abstract]
.
Blood
.
2014
;
124
(
21
).
Abstract 2667
.
9.
Wang
R
,
Yoshida
K
,
Toki
T
, et al
.
Loss of function mutations in RPL27 and RPS27 identified by whole-exome sequencing in Diamond-Blackfan anaemia
.
Br J Haematol
.
2015
;
168
(
6
):
854
-
864
.
10.
Mirabello
L
,
Khincha
PP
,
Ellis
SR
, et al
.
Novel and known ribosomal causes of Diamond-Blackfan anaemia identified through comprehensive genomic characterisation
.
J Med Genet
.
2017
;
54
(
6
):
417
-
425
.
11.
Ikeda
F
,
Yoshida
K
,
Toki
T
, et al
.
Exome sequencing identified RPS15A as a novel causative gene for Diamond-Blackfan anemia
.
Haematologica
.
2017
;
102
(
3
):
e93
-
e96
.
12.
Vlachos
A
,
Rosenberg
PS
,
Atsidaftos
E
,
Alter
BP
,
Lipton
JM
.
Incidence of neoplasia in Diamond Blackfan anemia: a report from the Diamond Blackfan Anemia Registry
.
Blood
.
2012
;
119
(
16
):
3815
-
3819
.
13.
Vlachos
A
,
Rosenberg
PS
,
Kang
J
,
Atsidaftos
E
,
Alter
BP
,
Lipton
JM
.
Myelodysplastic syndrome and gastrointestinal carcinomas characterize the cancer risk in Diamond Blackfan anemia: a report from the Diamond Blackfan Anemia Registry [abstract]
.
Blood
.
2016
;
128
(
22
).
Abstract 333
.
14.
Boocock
GR
,
Morrison
JA
,
Popovic
M
, et al
.
Mutations in SBDS are associated with Shwachman-Diamond syndrome
.
Nat Genet
.
2003
;
33
(
1
):
97
-
101
.
15.
Singh
SA
,
Vlachos
A
,
Morgenstern
NJ
, et al
.
Breast cancer in a case of Shwachman Diamond syndrome
.
Pediatr Blood Cancer
.
2012
;
59
(
5
):
945
-
946
.
16.
Savage
SA
,
Dufour
C
.
Classical inherited bone marrow failure syndromes with high risk for myelodysplastic syndrome and acute myelogenous leukemia
.
Semin Hematol
.
2017
;
54
(
2
):
105
-
114
.
17.
Ebert
BL
,
Pretz
J
,
Bosco
J
, et al
.
Identification of RPS14 as a 5q- syndrome gene by RNA interference screen
.
Nature
.
2008
;
451
(
7176
):
335
-
339
.
18.
Vlachos
A
,
Farrar
JE
,
Atsidaftos
E
, et al
.
Diminutive somatic deletions in the 5q region lead to a phenotype atypical of classical 5q- syndrome
.
Blood
.
2013
;
122
(
14
):
2487
-
2490
.
19.
Ajore
R
,
Raiser
D
,
McConkey
M
, et al
.
Deletion of ribosomal protein genes is a common vulnerability in human cancer, especially in concert with TP53 mutations
.
EMBO Mol Med
.
2017
;
9
(
4
):
498
-
507
.
20.
Goudarzi
KM
,
Lindström
MS
.
Role of ribosomal protein mutations in tumor development (Review)
.
Int J Oncol
.
2016
;
48
(
4
):
1313
-
1324
.
21.
Boultwood
J
,
Fidler
C
,
Strickson
AJ
, et al
.
Narrowing and genomic annotation of the commonly deleted region of the 5q- syndrome
.
Blood
.
2002
;
99
(
12
):
4638
-
4641
.
22.
Sperling
AS
,
Gibson
CJ
,
Ebert
BL
.
The genetics of myelodysplastic syndrome: from clonal haematopoiesis to secondary leukaemia
.
Nat Rev Cancer
.
2017
;
17
(
1
):
5
-
19
.
23.
Schneider
RK
,
Schenone
M
,
Ferreira
MV
, et al
.
Rps14 haploinsufficiency causes a block in erythroid differentiation mediated by S100A8 and S100A9
.
Nat Med
.
2016
;
22
(
3
):
288
-
297
.
24.
Jarboui
MA
,
Wynne
K
,
Elia
G
,
Hall
WW
,
Gautier
VW
.
Proteomic profiling of the human T-cell nucleolus
.
Mol Immunol
.
2011
;
49
(
3
):
441
-
452
.
25.
Anderson
SJ
,
Lauritsen
JP
,
Hartman
MG
, et al
.
Ablation of ribosomal protein L22 selectively impairs alphabeta T cell development by activation of a p53-dependent checkpoint
.
Immunity
.
2007
;
26
(
6
):
759
-
772
.
26.
Rao
S
,
Lee
SY
,
Gutierrez
A
, et al
.
Inactivation of ribosomal protein L22 promotes transformation by induction of the stemness factor, Lin28B
.
Blood
.
2012
;
120
(
18
):
3764
-
3773
.
27.
King
CE
,
Wang
L
,
Winograd
R
, et al
.
LIN28B fosters colon cancer migration, invasion and transformation through let-7-dependent and -independent mechanisms
.
Oncogene
.
2011
;
30
(
40
):
4185
-
4193
.
28.
De Keersmaecker
K
,
Atak
ZK
,
Li
N
, et al
.
Exome sequencing identifies mutation in CNOT3 and ribosomal genes RPL5 and RPL10 in T-cell acute lymphoblastic leukemia
.
Nat Genet
.
2013
;
45
(
2
):
186
-
190
.
29.
Landau
DA
,
Tausch
E
,
Taylor-Weiner
AN
, et al
.
Mutations driving CLL and their evolution in progression and relapse
.
Nature
.
2015
;
526
(
7574
):
525
-
530
.
30.
Ljungström
V
,
Cortese
D
,
Young
E
, et al
.
Whole-exome sequencing in relapsing chronic lymphocytic leukemia: clinical impact of recurrent RPS15 mutations
.
Blood
.
2016
;
127
(
8
):
1007
-
1016
.
31.
Lawrence
MS
,
Stojanov
P
,
Mermel
CH
, et al
.
Discovery and saturation analysis of cancer genes across 21 tumour types
.
Nature
.
2014
;
505
(
7484
):
495
-
501
.
32.
Fancello
L
,
Kampen
KR
,
Hofman
IJ
,
Verbeeck
J
,
De Keersmaecker
K
.
The ribosomal protein gene RPL5 is a haploinsufficient tumor suppressor in multiple cancer types
.
Oncotarget
.
2017
;
8
(
9
):
14462
-
14478
.
33.
Dutton-Regester
K
,
Gartner
JJ
,
Emmanuel
R
, et al
.
A highly recurrent RPS27 5'UTR mutation in melanoma
.
Oncotarget
.
2014
;
5
(
10
):
2912
-
2917
.
34.
Kandoth
C
,
McLellan
MD
,
Vandin
F
, et al
.
Mutational landscape and significance across 12 major cancer types
.
Nature
.
2013
;
502
(
7471
):
333
-
339
.
35.
Hause
RJ
,
Pritchard
CC
,
Shendure
J
,
Salipante
SJ
.
Classification and characterization of microsatellite instability across 18 cancer types
.
Nat Med
.
2016
;
22
(
11
):
1342
-
1350
.
36.
Novetsky
AP
,
Zighelboim
I
,
Thompson
DM
Jr
,
Powell
MA
,
Mutch
DG
,
Goodfellow
PJ
.
Frequent mutations in the RPL22 gene and its clinical and functional implications
.
Gynecol Oncol
.
2013
;
128
(
3
):
470
-
474
.
37.
Nagarajan
N
,
Bertrand
D
,
Hillmer
AM
, et al
.
Whole-genome reconstruction and mutational signatures in gastric cancer
.
Genome Biol
.
2012
;
13
(
12
):
R115
.
38.
Ferreira
AM
,
Tuominen
I
,
van Dijk-Bos
K
, et al
.
High frequency of RPL22 mutations in microsatellite-unstable colorectal and endometrial tumors
.
Hum Mutat
.
2014
;
35
(
12
):
1442
-
1445
.
39.
Yang
M
,
Sun
H
,
Wang
H
,
Zhang
S
,
Yu
X
,
Zhang
L
.
Down-regulation of ribosomal protein L22 in non-small cell lung cancer
.
Med Oncol
.
2013
;
30
(
3
):
646
.
40.
Yan
TT
,
Fu
XL
,
Li
J
, et al
.
Downregulation of RPL15 may predict poor survival and associate with tumor progression in pancreatic ductal adenocarcinoma
.
Oncotarget
.
2015
;
6
(
35
):
37028
-
37042
.
41.
Yong
WH
,
Shabihkhani
M
,
Telesca
D
, et al
.
Ribosomal proteins RPS11 and RPS20, two stress-response markers of glioblastoma stem cells, are novel predictors of poor prognosis in glioblastoma patients
.
PLoS One
.
2015
;
10
(
10
):
e0141334
.
42.
Wang
H
,
Zhao
LN
,
Li
KZ
,
Ling
R
,
Li
XJ
,
Wang
L
.
Overexpression of ribosomal protein L15 is associated with cell proliferation in gastric cancer
.
BMC Cancer
.
2006
;
6
:
91
.
43.
Wang
Q
,
Yang
C
,
Zhou
J
,
Wang
X
,
Wu
M
,
Liu
Z
.
Cloning and characterization of full-length human ribosomal protein L15 cDNA which was overexpressed in esophageal cancer
.
Gene
.
2001
;
263
(
1-2
):
205
-
209
.
44.
Kasai
H
,
Nadano
D
,
Hidaka
E
, et al
.
Differential expression of ribosomal proteins in human normal and neoplastic colorectum
.
J Histochem Cytochem
.
2003
;
51
(
5
):
567
-
574
.
45.
Kobayashi
T
,
Sasaki
Y
,
Oshima
Y
, et al
.
Activation of the ribosomal protein L13 gene in human gastrointestinal cancer
.
Int J Mol Med
.
2006
;
18
(
1
):
161
-
170
.
46.
Lee
YC
,
Hur
W
,
Choi
JE
, et al
.
[Analysis of gene expression in primary hepatocellular carcinoma using differentially displayed reverse transcriptase polymerase chain reaction] [in Korean]
.
Korean J Gastroenterol
.
2009
;
53
(
6
):
361
-
368
.
47.
Song
MJ
,
Jung
CK
,
Park
CH
, et al
.
RPL36 as a prognostic marker in hepatocellular carcinoma
.
Pathol Int
.
2011
;
61
(
11
):
638
-
644
.
48.
Dave
B
,
Granados-Principal
S
,
Zhu
R
, et al
.
Targeting RPL39 and MLF2 reduces tumor initiation and metastasis in breast cancer by inhibiting nitric oxide synthase signaling
.
Proc Natl Acad Sci USA
.
2014
;
111
(
24
):
8838
-
8843
.
49.
Bee
A
,
Ke
Y
,
Forootan
S
, et al
.
Ribosomal protein l19 is a prognostic marker for human prostate cancer
.
Clin Cancer Res
.
2006
;
12
(
7 Pt 1
):
2061
-
2065
.
50.
Ajore
R
,
Raiser
D
,
McConkey
M
, et al
.
Deletion of ribosomal protein genes is a common vulnerability in human cancer, especially in concert with TP53 mutations
.
EMBO Mol Med
.
2017
;
9
:
498
-
507
.

Competing Interests

Conflict-of-interest disclosure: The author declares no competing financial interests.

Author notes

Off-label drug use: None disclosed.

This article appears only in the electronic edition of Hematology 2017.