Next Article in Journal
Structural and Functional Classification of G-Quadruplex Families within the Human Genome
Previous Article in Journal
Role of Genetic Thrombophilia Markers in Thrombosis Events in Elderly Patients with COVID-19
Previous Article in Special Issue
Endoscopic Surveillance and Treatment of Upper GI Tract Lesions in Patients with Familial Adenomatous Polyposis—A New Perspective on an Old Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

SDHA Germline Mutations in SDH-Deficient GISTs: A Current Update

by
Angela Schipani
1,2,
Margherita Nannini
1,3,
Annalisa Astolfi
1,* and
Maria A. Pantaleo
1,3
1
Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
2
Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CS Utrecht, The Netherlands
3
Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
*
Author to whom correspondence should be addressed.
Genes 2023, 14(3), 646; https://doi.org/10.3390/genes14030646
Submission received: 10 January 2023 / Revised: 17 February 2023 / Accepted: 1 March 2023 / Published: 4 March 2023
(This article belongs to the Special Issue Genetic Predispositions to Tumors of the Digestive System)

Abstract

:
Loss of function of the succinate dehydrogenase complex characterizes 20–40% of all KIT/PDGFRA-negative GIST. Approximately half of SDH-deficient GIST patients lack SDHx mutations and are caused by a hypermethylation of the SDHC promoter, which causes the repression of SDHC transcription and depletion of SDHC protein levels through a mechanism described as epimutation. The remaining 50% of SDH-deficient GISTs have mutations in one of the SDH subunits and SDHA mutations are the most common (30%), with consequent loss of SDHA and SDHB protein expression immunohistochemically. SDHB, SDHC, and SDHD mutations in GIST occur in only 20–30% of cases and most of these SDH mutations are germline. More recently, germline mutations in SDHA have also been described in several patients with loss of function of the SDH complex. SDHA-mutant patients usually carry two mutational events at the SDHA locus, either the loss of the wild type allele or a second somatic event in compound heterozygosis. This review provides an overview of all data in the literature regarding SDHA-mutated GIST, especially focusing on the prevalence of germline mutations in SDH-deficient GIST populations who harbor SDHA somatic mutations, and offers a view towards understanding the importance of genetic counselling for SDHA-variant carriers and relatives.

1. Introduction

Gastrointestinal stromal tumors (GISTs) are mesenchymal tumors of the gastrointestinal tract deriving from the interstitial cells of Cajal (ICC), the mesenchymal cells responsible for the autonomous peristaltic contractions of the gut [1].
Around 10–15% of adult GISTs and all pediatric GISTs do not carry the driver mutations in the tyrosine kinase receptor KIT and/or platelet-derived growth factor receptor-α (PDGFR-α), and they have been called wild type GISTs (WT) [2]. Among the KIT/PDGFRA WT GISTs, about 15% carry mutations in the RAS-pathway (BRAF/RAS or NF1). On the other hand, between 20% and 40% are driven by the loss of function of succinate dehydrogenase (SDH), also defined as SDH-deficient GISTs. The remaining 50% are called quadruple WT (qWT) GISTs since they do not harbor any mutations in KIT/PDGFRA/SDH/RAS-P genes [3,4,5].
SDH-deficient GISTs tend to develop in children and young patients and can be sporadic or associated with Carney-Stratakis syndrome (CSS) and Carney triad (CT). Carney-Stratakis syndrome is a rare hereditary condition characterized by multifocal paragangliomas and GISTs. On the other hand, Carney triad is a rare non-hereditary disease causes by the association of GIST, pulmonary chondroma, and paraganglioma [6,7].
SDH-deficient GISTs exhibit characteristic clinical and pathological features, such as their primary localization in the stomach, multi-nodular growth, an epithelioid phenotype, and a common lymphovascular invasion [8]. This subset of GIST is known for the occurrence of lymph node metastases and an indolent course of the disease [9].
The immunohistochemical loss of succinate dehydrogenase subunit B (SDHB), which indicates the loss of function of the entire SDH-complex, is a representative feature of SDH-deficient GIST [10]. SDHB protein expression by immunohistochemistry is a diagnostic marker for loss of function of the SDH complex, whereas SDHA immunohistochemistry is able to identify SDHA-mutant GISTs [11]. SDH-deficient GISTs are uniformly immunohistochemically positive for KIT and DOG1/Ano1 (Anoctamin-1) [1]. They respond poorly to treatment with imatinib, suggesting that alternative signalling pathways are driving tumor formation [12]. SDH-deficient GISTs present an expression profile extremely different from KIT-mutant GISTs, showing the upregulation of distinctive gene pathways linked to tumor progression. Moreover, they display a depletion of immune competence, suggesting that this GIST subgroup can be considered a non-inflamed tumor [13].
SDH deficiency is mainly caused by the biallelic inactivation of one of the four SDH subunit genes (SDHA, SDHB, SDHC, and SDHD) according to the classic tumor suppressor gene model [14]. Normally, the combination of a first hit, that is, an inactivating germline mutation, with a second hit, represented by another inactivating mutation affecting the second allele or somatic loss of heterozygosity, leads to the loss of function of an SDHx subunit. Less frequently, SDH deficiency is caused by the somatic inactivation of both alleles of a given complex subunit or SDH assembly factor [14]. Approximately half of patients with SDH-mutated GISTs have mutations in one of the SDH subunits and SDHA mutations are the most common (30%) with consequent loss of SDHA and SDHB protein expression immunohistochemically [8,11]. However, it is widely recognized that the promoter hypermethylation and epigenetic silencing of the SDHC gene can result in the inactivation of the SDH complex [15,16]; germline mutations in SDHB, SDHC, and SDHD occur in only 20–30% of SDH-deficient GIST.
More recently, germline mutations in SDHA have also been reported in several patients with loss of function of the SDH complex. This review reports a current comprehensive state-of-the-art study on the germline SDHA mutations in SDH-deficient GIST populations who harbor SDHA somatic mutations by collecting and describing all studies in which these data have been reported.

2. SDH Complex in Cancers

Succinate dehydrogenase, SDH, is a mitochondrial enzyme complex (also known as complex II of the electron transport chain) that is involved in both the citric acid cycle, by catalysing the oxidation of succinate to fumarate, and the electron transport chain, by leading the electron transfer to the terminal acceptor ubiquinone (Figure 1a) [17]. The assembled SDH complex, located in the inner membrane of the mitochondria, consists of four subunits encoded by nuclear genes (SDHA, SDHB, SDHC and SDHD, collectively indicated as SDHx). All of the SDH genes are involved in the tumorigenesis of different types of cancers including GIST, paraganglioma, pheochromocytoma, chronic lymphocytic leukemia, renal cell carcinoma, thyroid cancer, Hodgkin lymphoma, pituitary adenomas, and neuroendocrine tumors of the pancreas [18,19,20,21,22].

3. SDHA Alterations in GIST

Tumor development caused by SDH deficiency results from the complete loss of function of one SDHx subunit, which leads to instability of the entire SDH complex and loss of the enzymatic function of the SDH complex. Disruption of the succinate dehydrogenase complex (SDH), caused by either biallelic mutations or epigenetic silencing, leads to accumulation of succinate, an oncometabolite that promotes tumorigenesis by activating different pathways. Accumulation of succinate leads to the inhibition of prolyl-hydroxylase domain proteins (PHD) activity, causing the stabilization of hypoxia-inducible factor 1 (HIF1), which in turn leads to increased expression of the transcriptional factors as vascular endothelial growth factor (VEGF) and insulin-like growth factor-1 (IGF1), causing increased angiogenic and cell growth signalling (Figure 1b). SDH-deficient GISTs do not show a IGF1R genomic amplification; however, the IGF-1 receptor (IGF1R) is overexpressed both at the mRNA and protein level [23].
Moreover, the accumulation of succinate, which has a similar structure of α-ketoglutarate (α-KG), leads to the inhibition of the α-KG dependent dioxygenases such as JmjC domain containing histone lysine demethylases (KDM) and ten–eleven translocation (TET) enzymes (Figure 1b). The latter are involved in DNA demethylation and transcriptional silencing because they drive the active elimination of 5-methylcytosine (5mC) from methylated CpG sites. Consequently, genome-wide DNA hypermethylation may be related to the disruption of DNA demethylation machinery by downregulated TET enzyme [24]. Intriguingly, it has been recently shown that the characteristic hypermethylation in these tumors is associated with changes in genome topology that can drive an oncogenic program. The hypermethylation is associated with pervasive insulator losses and topological reorganization of the FGF and KIT loci. In particular, the hypermethylated phenotype interrupts the binding of CTCF in regions located in proximity to the FGF3/FGF4 locus, leading to FGF4 overexpression [25].

4. Germline SDH Mutations in GIST

In most SDH-deficient GISTs, the loss of the complex is caused by the presence of mutations in at least one of the four subunits. Mutations in all four SDH genes have been described, but different studies have classified SDHA as the most frequently mutated subunit, with SDHA mutations occurring in ~30% of all SDH-deficient GIST [26]. SDHA patients usually carry two mutational events at the SDHA locus, either the loss of the wild type allele or a second somatic event in compound heterozygosis. SDHB, SDHC and SDHD mutations in GIST are less frequent (20–30%) and most of these mutations are germline [19,27]. Alongside germline mutations of SDHB, SDHC and SDHD, evidence of germline SDHA has been accumulated.

SDHA Germline Mutations

Notably, numerous findings have evaluated that germline mutations in SDHA are highly frequent in SDHA-deficient GIST (Table 1). In 2013, Dwight et al. [28] investigated a sample of 10 SDH-deficient GISTs and displayed that 30% of SDH-deficient GISTs in their series were associated with germline SDHA mutations.
Additionally, Oudijk et al. performed SDHA IHC on 33 WT-GISTs, including nine pediatric/adolescent GISTs, in order to study whether IHC could identify SDHA-mutated GISTs. SDHA-negative samples were sequenced both in corresponding tumor and germline DNA isolated from paraffin-embedded healthy tissue surrounding the tumor; four cases were germline [29].
In a cohort of 11 SDHB-negative WT GIST, five patients (45%) carried SDHA mutation; among them, three patients for which germline DNA was available harbored germline mutation. The presence of a germline mutation accompanied by loss of the allele in the tumor is presumed also in the two patients for which germline DNA was not available; based on tumor sequencing, the data revealed the presence of homozygous missense SDHA mutations [30].
Moreover, heterozygous mutations were observed in DNA from normal tissue of all six out of nine SDHA-deficient GIST patients whose material was available [31].
Furthermore, in a 2016 study, a large sample set of 95 patients was investigated, in which 84 had succinate dehydrogenase-deficient GIST caused either by SDH mutations (75%) or by SDHC promoter hypermethylation (25%), and 18 were syndromic GISTs with chondromas and/or paragangliomas. Of the 63 cases of SDH-mutant GISTs, 16 had mutation in SDHB, 12 in SDHC, and 1 in SDHD, and 34 had mutations in SDHA; among them, 20 cases were germline [32].
SDHA mutations have been observed in an additional seven SDHA/B-deficient cases and six of them were germline mutations [11]. In addition, Carrera et al. identified LOH at the SDHA locus in one GIST patient [33].
The frequency of germline SDHA mutations within the SDH-deficient GISTs is even higher in a recent study that identifies germline mutations in all 14 patients analyzed for which a normal counterpart was available [34,35,36].
Table 1. List of SDHA germline mutations reported in SDH-deficient GISTs.
Table 1. List of SDHA germline mutations reported in SDH-deficient GISTs.
IDAgeSexCytologySiteAssociated TumorICH SDHBIHC SDHAExonGermline Mutation (cDNA, Protein)References
#122M-stomachnonegneg 2c.91C>T; p.R31XItaliano et al., 2012 [37]
#231Mmixedstomach-negneg5c.553C>T; p.Q185XWagner et al., 2013 [31]
#338Fepithelioidstomach-negneg11c.1534C>T; p.R512XWagner et al., 2013
#439Fmixedstomach-negneg8c.688delG; FrameshiftWagner et al., 2013
#522Mmixedstomach-negneg2c.91C>T; p.R31XWagner et al., 2013
#653Fmixedstomach-negneg2c.91C>T; p.R31XWagner et al., 2013
#719Mmixedstomach-negneg2c.91C>T; p.R31XWagner et al., 2013
#845Fepithelioidstomachnoneg neg 15c.1969G>A; p.V657IDwight et al., 2013 [28]
#925Fepithelioidstomachnoneg neg 3c.160C>T; p.Q54XDwight et al., 2013
#1041Mepithelioidstomachnoneg neg 12c.1633+3G>C; splice-siteDwight et al., 2013
#1139Fepitheliod/spindlestomachnoneg -7c.818C>T; p.T2731I Belinsky et al.,2013 [30]
#1252Fepitheliod/spindlestomachnoneg -IVS4/ex5c.457-2_c457delAGC; p.L153Kfs*71Belinsky et al.,2013
#1333Fepitheliod/spindlestomachnoneg -2c.91C>T; p.R31XBelinsky et al.,2013
#1441FepithelioidstomachMTCnegneg2c.91C>T; p.R31XOudijk et al., 2013 [29]
#1553Fspindle cellstomachnonegneg2c.91C>T; p.R31XOudijk et al., 2013
#1647Fmixedstomachnonegneg2c.91C>T; p.R31XOudijk et al., 2013
#1714Mmixedstomachnonegneg2c.91C>T; p.R31XOudijk et al., 2013
#18-- stomachnonegneg2c.91C>T; p.R31XMiettinen et al., 2013 [11]
#19-- stomachnonegneg2c.91C>T; p.R31XMiettinen et al., 2013
#20-- stomachnonegneg2c.91C>T; p.R31XMiettinen et al., 2013
#21-- stomachnonegneg6c.767C>T; p.T256IMiettinen et al., 2013
#22-- stomachnonegneg13c.1794G>C; p.K598NMiettinen et al., 2013
#23-- stomachnonegneg14c.1795–1G>T; Ex 14 5’ splicingMiettinen et al., 2013
#24-- stomachnonegpos5c.562C>T; p.R188WMiettinen et al., 2013
#2523MepitheliodstomachRCC negpos1c.2T>C; p.M1TJiang et al., 2015 [37]
#26---stomachnoneg 10c.1432_1432del; p.478_478delBoikos et al., 2016 [32]
#27---stomachnoneg-11c.1513delA; p.S505fsBoikos et al., 2016
#28---stomachnoneg-14c.1795-1G>T Boikos et al., 2016
#29---stomachnoneg-10c.1340°>G; p.H447RBoikos et al., 2016
#30---stomachnoneg-10c.1367C>T; p.S456LBoikos et al., 2016
#31-- -stomachnoneg-13c.1753C>T; p.R585WBoikos et al., 2016
#3223FepitheliodstomachCHOneg-3c.295C>T; p.H99YBoikos et al., 2016
#33- --stomachnoneg-5c.562C>T; p.R188WBoikos et al., 2016
#34---stomachnoneg-7c.818C>T; p.T273IBoikos et al., 2016
#35---stomachnoneg-2c.91C>T; p.R31XBoikos et al., 2016
#36---stomachnoneg-2c.91C>T; p.R31XBoikos et al., 2016
#3721Mepitheliod/spindlestomachCHOneg-2c.91C>T; p.R31XBoikos et al., 2016
#38-- -stomachnoneg-2c.91C>T; p.R31XBoikos et al., 2016
#39---stomachnoneg-2c.91C>T; p.R31XBoikos et al., 2016
#40---stomachnoneg-2c.91C>T; p.R31XBoikos et al., 2016
#41---stomachnoneg-2c.91C>T; p.R31XBoikos et al., 2016
#42---stomachnoneg-8c.923C>T; p.T308MBoikos et al., 2016
#43- --stomachnoneg-13c.1794G>C; p.K598NBoikos et al., 2016
#4430Mepitheliod/spindlestomachPGL, CHOneg-11c.1532T>C; p.L511PBoikos et al., 2016
#45- --stomach-neg- SDHA deletionBoikos et al.,2016
#4621-- -negneg2c.91C>T; p.R31XGault et al., 2018 [38]
#4727-- -negneg2c.91C>T; p.R31XGault et al., 2018
#4820FepithelioidstomachPGL- - 1c.1A > C; p.(Met1?)Carrera et al., 2019 [33]
#4928F stomachno 9c.1151 C>G; p.S384XPantaleo et al., 2011 [26]
#5030M stomachno 2c.91 C>T; p.R31XPantaleo et al., 2011
#5139F stomachno 13c.1766 G>A; p.R589QPantaleo et al., 2011 [34]
#5237F stomachno 5c.457-3_457-1 delCAG; p.L153splicePantaleo et al., 2014 [35]
#5331F stomachno 9c.1151 C>G; p.S384XPantaleo et al., 2022 [36]
#5461M stomachPGL 6c.698G>T; p.G233VPantaleo et al., 2022
#5521F stomachno 5c.512G>A; p.R171HPantaleo et al., 2022
#5638M stomachno 6c.768G>C; p.G257APantaleo et al., 2022
#5770F stomachno 4c.356G>A; p.W119XPantaleo et al., 2022
#5866F stomachno 14c.1799G>A; p.R600QPantaleo et al., 2022
#5917M stomachno 12c.1663+3 G>C; p.R554splicePantaleo et al., 2022
#6055F stomachno 14c.1799G>A; p.R600QPantaleo et al., 2022
#6150M stomachno 13c.1754G>A; p.R585QPantaleo et al., 2022
#6217M stomachno 6c.628C>T; p.R210XPantaleo et al., 2022

5. SDHA Germline Consideration

Taken together, these data demonstrate that the most common mutation in the analyzed patient cohort occurs in exon 2 of SDHA gene, c.91C>T (Figure 2). This sequence change produces a premature translational stop signal at codon 31 (p.R31X) of the SDHA gene, leading to a truncated protein [8,11,30,37,38].
Interestingly, the high frequency of this mutation in different series suggests the presence of a founder effect, but the incidence of the SDHA p.R31X mutation in different countries makes such founder mutation less likely and might suggest a hotspot mutation.
Another important consideration is the recurrence of the germline SDHA mutation in other cancers. Based on the previously mentioned studies, we can affirm that most SDHA mutations in GISTs are GIST-specific. An association exists between germline SDHA mutations and GIST onset, supporting the idea that GISTs develop alone as the only tumor disease due to SDHA, and not in association with other tumors. To date, only a few cases report SDHA germline mutation in the context of other tumors: paraganglioma in one case, pulmonary chondroma in two cases, and the full Carney triad in one patient [33,38,39].
Additionally, the loss of SDHA expression in tumors consistently predicts the presence of SDHA mutations in tumor cells and associated germline material. Owing to the fact that most of the cases are SDHA-negative at the protein level, the possibility to use immunohistochemistry as a reliable tool for identifying SDHA-mutant GISTs may be considered. All the authors have also clearly showed that the predominant tumor site is the stomach in the totality of cases.
Concerning the issue of the pathogenic effect of different mutations, it is known that genetic variants are usually classified following the American College of Medical Genetics and Genomics (ACMG) guidelines in a five-tiered scheme of pathogenic, likely pathogenic, variant of uncertain significance (VUS), likely benign, or benign. With the increased use of molecular testing and next-generation sequencing (NGS), many SDHA variants of unknown significance (VUS) have been detected. The meaning of VUS is controversial since it is unclear if the variant is related to the disease or not. Up to now, the usage of VUS data in the clinical context is challenging in term of patient management and treatment decisions, and there is a wide agreement on the fact that a VUS should not be used in clinical decision-making. Likewise, if a patient is identified to have a VUS, all clinical decisions should not be based on the presence of the VUS but only on personal and family history.

6. Other SDH Subunit Mutations

SDH-deficient GISTs are the majority of WT GISTs, with germline SDHA mutations as the most frequent mutational event [32,34]. The relative frequency of germline mutations in the SDH subunits has been clearly explored in an observational study of WT GISTs conducted in a large series of 84 SDH-deficient cases. Within the 63 cases of SDH-mutant GIST, 54% had mutations in SDHA subunit, mostly germline, while SDHB/C/D altogether represented the remaining 46% of SDH-mutant GIST [32]. Moreover, while only a minority of GISTs with SDHA mutations (nine of twenty-five available samples) had loss of heterozygosity as the second hit, most tumors with SDHB mutations (12 of 13 available samples) showed loss of heterozygosity as the second oncogenic event [32]. Wild type GIST driven by germline mutations in SDHB, SDHC, or SDHD subunits is a component of the Carney-Stratakis syndrome, an inherited predisposition syndrome characterized by the presence of GIST and paraganglioma [40]. In the same study, 22% of SDH-deficient tumors had hypermethylation of the SDHC promoter sequence [32], leading to loss of SDHC protein expression and functional SDH deficiency [27,41]. These SDH-epimutant tumor patients were younger, overwhelmingly female, had disease of gastric origin, and were often metastatic [32].
The same scenario is evident also in Miettienen et al. [11], who investigated 21 SDH-mutant GISTs, finding 10 SDHA mutations and 11 cases carrying either SDHB, SDHC, or SDHD mutations, mostly germline.
Overall, patients with SDHA-mutant tumors tend to have an older median age compared to the SDH-deficient GISTs due to other subunit mutations [36,42], while SDHB, SDHC, and SDHD mutant GISTs usually also develop paragangliomas [34,43].
Even if germline testing is recommended for all SDH-deficient GISTs, currently, there are no specific surveillance guidelines for these patients, and follow-up should parallel that of the general GIST population.

7. Current Therapies of SDH-Deficient GISTs

To date, the medical management of SDH-deficient GISTs is still controversial because of limited data available, both due to the rarity of this molecular subset of GIST and to the lack of SDH deficiency characterization in most studies [44]. Generally, given the lack of gain-of-function KIT and PDGFRA mutations, SDH-deficient GISTs are widely considered resistant to all tyrosine kinase inhibitors (TKIs), as all other KIT/PDGFRA WT GIST. Thus, there is a consensus to avoid imatinib or any adjuvant treatment in this rare molecular subset of GIST [45].
In the metastatic setting, there are few retrospective data showing some degree of activity of TKIs with anti-angiogenic properties, such as sunitinib, regorafenib, and pazopanib, likely because of the pseudohypoxic phenotype of SDH-deficient GIST [32,46,47,48].
Recent advances on the molecular background of SDH-deficient GISTs have shifted the therapy focus from the standard TKIs to other therapeutic strategies. In particular, given the known overexpression of IGF1R of SDH-deficient GISTs, the oral IGF-1R TKI linsitinib has been tested in a phase II study on adult and pediatric patients with wild type GIST, including 15 SDH-deficient GIST, showing a clinical benefit rate (CBR) and progression-free survival (PFS) at 9 months of 40% and 52%, respectively, suggesting a potential benefit of linsitinib in this patient population, though no objective responses were revealed [49].
Moreover, the O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation markedly prevalent in SDH-deficient GISTs has represented some proof of concept about the potential role of alkylating agents in this subset of GISTs [50], whereas a phase II trial on temozolomide in advanced SDH-GISTs is still ongoing (NCT03556384); a prolonged disease stability after 18 consecutive cycles of temozolomide has been recently reported in a female metastatic and progressive SDH-deficient GIST 45. The same authors presented another anecdotal case with avid disease on DOTATATE PET, experiencing a durable partial response to 177lutetium-DOTA-octreotate PRRT after failing two lines of TKI therapy [51].
Finally, in a recent phase Ib trial, the combination of imatinib and binimetinib has shown encouraging activity in SDH-deficient GIST cohort, suggesting a potential role of targeting the ICC/GIST lineage-specific survival transcription factor, ETV1 [52].
In this evolving scenario, all data should be carefully interpreted in relation to the known frequent indolent behavior of SDH-deficient GISTs which could be underlying most prolonged disease stabilities and the clinical benefit observed in almost all patients.

8. Conclusions

In this review, we have collected data from the literature in order to present the current update of SDHA germline mutations occurring in GISTs. These studies together suggest that there is a high incidence of germline mutations in SDHA-deficient GISTs harboring SDHA somatic mutations. Since the majority of SDHA-mutated GISTs have germline mutations, the possibility of genetic counselling of SDHA mutation carriers and relatives should be evaluated.

Author Contributions

Conceptualization, A.S., M.A.P., A.A. and M.N.; methodology, A.S., M.A.P., A.A. and M.N.; formal analysis, A.S., M.A.P., A.A. and M.N.; investigation, M.A.P. and A.A.; data curation, A.S., M.A.P. and A.A.; writing—original draft preparation, A.S., M.A.P., A.A. and M.N.; writing—review and editing, A.S., M.A.P. and A.A.; visualization, M.A.P., A.A. and M.N.; supervision, M.A.P. and A.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by AIRC IG 26010 grant to Maria Abbondanza Pantaleo.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

GIST, Gastrointestinal stromal tumors; SDH, succinate dehydrogenase; ICC, interstitial cells of Cajal; KIT, tyrosine-protein kinase; PDGFR-α, platelet-derived growth factor receptor-α; CSS, Carney-Stratakis syndrome; CT, Carney triad; Ano1, Anoctamin-1; SDHA succinate dehydrogenase subunit A; SDHB succinate dehydrogenase subunit B; SDHC succinate dehydrogenase subunit C; SDHD succinate dehydrogenase subunit D; PHD, prolyl-hydroxylase domain; HIF1, hypoxia-inducible factor 1; VEGF, vascular endothelial growth factor; IGF1, insulin-like growth factor-1; IGF1R, insulin-like growth factor-1 receptor; α-KG, α-ketoglutarate; KDM histone lysine demethylases; TET, ten–eleven translocation; TKI, tyrosine kinase inhibitor; CBR, clinical benefit rate; PFS, progression-free survival; MGMT, O6-methylguanine-DNA methyltransferase.

References

  1. Kindblom, L.G.; Remotti, H.E.; Aldenborg, F.; Meis-Kindblom, J.M. Gastrointestinal Pacemaker Cell Tumor (GIPACT): Gastrointestinal Stromal Tumors Show Phenotypic Characteristics of the Interstitial Cells of Cajal. Am. J. Pathol. 1998, 152, 1259. [Google Scholar] [PubMed]
  2. Isozaki, K.; Hirota, S. Gain-of-Function Mutations of Receptor Tyrosine Kinases in Gastrointestinal Stromal Tumors. Curr. Genomics 2006, 7, 469–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Gasparotto, D.; Rossi, S.; Polano, M.; Tamborini, E.; Lorenzetto, E.; Sbaraglia, M.; Mondello, A.; Massani, M.; Lamon, S.; Bracci, R.; et al. Quadruple-Negative GIST Is a Sentinel for Unrecognized Neurofibromatosis Type 1 Syndrome. Clin. Cancer Res. 2017, 23, 273–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Nannini, M.; Astolfi, A.; Urbini, M.; Indio, V.; Santini, D.; Heinrich, M.C.; Corless, C.L.; Ceccarelli, C.; Saponara, M.; Mandrioli, A.; et al. Integrated Genomic Study of Quadruple-WT GIST (KIT/PDGFRA/SDH/RAS Pathway Wild-Type GIST). BMC Cancer 2014, 14, 685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Nannini, M.; Urbini, M.; Astolfi, A.; Biasco, G.; Pantaleo, M.A. The Progressive Fragmentation of the KIT/PDGFRA Wild-Type (WT) Gastrointestinal Stromal Tumors (GIST). J. Transl. Med. 2017, 15, 113. [Google Scholar] [CrossRef]
  6. Carney, J.A.; Stratakis, C.A. Familial Paraganglioma and Gastric Stromal Sarcoma: A New Syndrome Distinct from the Carney Triad. Am. J. Med. Genet. 2002, 108, 132–139. [Google Scholar] [CrossRef]
  7. Recht, H.S.; Fishman, E.K. Carney-Stratakis Syndrome: A Dyad of Familial Paraganglioma and Gastrointestinal Stromal Tumor. Radiol. Case Rep. 2020, 15, 2071–2075. [Google Scholar] [CrossRef]
  8. Miettinen, M.; Lasota, J. Succinate Dehydrogenase Deficient Gastrointestinal Stromal Tumors (GISTs)—A Review. Int. J. Biochem. Cell Biol. 2014, 53, 514–519. [Google Scholar] [CrossRef] [Green Version]
  9. Pantaleo, M.A.; Lolli, C.; Nannini, M.; Astolfi, A.; Indio, V.; Saponara, M.; Urbini, M.; La Rovere, S.; Gill, A.; Goldstein, D.; et al. Good Survival Outcome of Metastatic SDH-Deficient Gastrointestinal Stromal Tumors Harboring SDHA Mutations. Genet. Med. 2015, 17, 391–395. [Google Scholar] [CrossRef] [PubMed]
  10. Miettinen, M.; Makhlouf, H.; Sobin, L.H.; Lasota, J. Gastrointestinal Stromal Tumors of the Jejunum and Ileum: A Clinicopathologic, Immunohistochemical, and Molecular Genetic Study of 906 Cases before Imatinib with Long-Term Follow-Up. Am. J. Surg. Pathol. 2006, 30, 477–489. [Google Scholar] [CrossRef]
  11. Miettinen, M.; Killian, J.K.; Wang, Z.F.; Lasota, J.; Lau, C.; Jones, L.; Walker, R.; Pineda, M.; Zhu, Y.J.; Kim, S.Y.; et al. Immunohistochemical Loss of Succinate Dehydrogenase Subunit a (SDHA) in Gastrointestinal Stromal Tumors (GISTs) Signals SDHA Germline Mutation. Am. J. Surg. Pathol. 2013, 37, 234–240. [Google Scholar] [CrossRef] [Green Version]
  12. Murray, M.; Hatcher, H.; Jessop, F.; Williams, D.; Carroll, N.; Bulusu, R.; Judson, I. Treatment of Wild-Type Gastrointestinal Stromal Tumor (WT-GIST) with Imatinib and Sunitinib. Pediatr. Blood Cancer 2008, 50, 386–388. [Google Scholar] [CrossRef] [PubMed]
  13. Indio, V.; Schipani, A.; Nannini, M.; Urbini, M.; Rizzo, A.; De Leo, A.; Altimari, A.; Di Scioscio, V.; Messelodi, D.; Tarantino, G.; et al. Gene Expression Landscape of Sdh-Deficient Gastrointestinal Stromal Tumors. J. Clin. Med. 2021, 10, 1057. [Google Scholar] [CrossRef]
  14. Bannon, A.E.; Kent, J.; Forquer, I.; Town, A.; Klug, L.R.; McCann, K.; Beadling, C.; Harismendy, O.; Sicklick, J.K.; Corless, C.; et al. Biochemical, Molecular, and Clinical Characterization of Succinate Dehydrogenase Subunit A Variants of Unknown Significance. Clin. Cancer Res. 2017, 23, 6733–6743. [Google Scholar] [CrossRef] [Green Version]
  15. Killian, J.K.; Kim, S.Y.; Miettinen, M.; Smith, C.; Merino, M.; Tsokos, M.; Quezado, M.; Smith, W.I.; Jahromi, M.S.; Xekouki, P.; et al. Succinate Dehydrogenase Mutation Underlies Global Epigenomic Divergence in Gastrointestinal Stromal Tumor. Cancer Discov. 2013, 3, 648–657. [Google Scholar] [CrossRef] [Green Version]
  16. Urbini, M.; Astolfi, A.; Indio, V.; Heinrich, M.C.; Corless, C.L.; Nannini, M.; Ravegnini, G.; Biasco, G.; Pantaleo, M.A. SDHC Methylation in Gastrointestinal Stromal Tumors (GIST): A Case Report. BMC Med. Genet. 2015, 16, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Bezawork-Geleta, A.; Rohlena, J.; Dong, L.; Pacak, K.; Neuzil, J. Mitochondrial Complex II: At the Crossroads. Trends Biochem. Sci. 2017, 42, 312–325. [Google Scholar] [CrossRef]
  18. Barletta, J.A.; Hornick, J.L. Succinate Dehydrogenase-Deficient Tumors: Diagnostic Advances and Clinical Implications. Adv. Anat. Pathol. 2012, 19, 193–203. [Google Scholar] [CrossRef]
  19. Janeway, K.A.; Kim, S.Y.; Lodish, M.; Nosé, V.; Rustin, P.; Gaal, J.; Dahia, P.L.M.; Liegl, B.; Ball, E.R.; Raygada, M.; et al. Defects in Succinate Dehydrogenase in Gastrointestinal Stromal Tumors Lacking KIT and PDGFRA Mutations. Proc. Natl. Acad. Sci. USA 2011, 108, 314–318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Baysal, B.E. A Recurrent Stop-Codon Mutation in Succinate Dehydrogenase Subunit B Gene in Normal Peripheral Blood and Childhood T-Cell Acute Leukemia. PLoS ONE 2007, 2, e436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Beamer, L.C. Cowden Syndrome: What Oncology Nurses Need to Know about Increased Risk of Developing Certain Cancers. Oncol. Nurs. Forum 2014, 41, 555–557. [Google Scholar] [CrossRef]
  22. Niemeijer, N.D.; Papathomas, T.G.; Korpershoek, E.; De Krijger, R.R.; Oudijk, L.; Morreau, H.; Bayley, J.P.; Hes, F.J.; Jansen, J.C.; Dinjens, W.N.M.; et al. Succinate Dehydrogenase (SDH)-Deficient Pancreatic Neuroendocrine Tumor Expands the SDH-Related Tumor Spectrum. J. Clin. Endocrinol. Metab. 2015, 19, E1386–E1393. [Google Scholar] [CrossRef]
  23. Pantaleo, M.A.; Astolfi, A.; Di Battista, M.; Heinrich, M.C.; Paterini, P.; Scotlandi, K.; Santini, D.; Catena, F.; Manara, M.C.; Nannini, M.; et al. Insulin-like Growth Factor 1 Receptor Expression in Wild-Type GISTs: A Potential Novel Therapeutic Target. Int. J. Cancer 2009, 125, 2991–2994. [Google Scholar] [CrossRef] [PubMed]
  24. Morin, A.; Goncalves, J.; Moog, S.; Castro-Vega, L.J.; Job, S.; Buffet, A.; Fontenille, M.J.; Woszczyk, J.; Gimenez-Roqueplo, A.P.; Letouzé, E.; et al. TET-Mediated Hypermethylation Primes SDH-Deficient Cells for HIF2α-Driven Mesenchymal Transition. Cell Rep. 2020, 30, 4551–4566.e7. [Google Scholar] [CrossRef] [PubMed]
  25. Flavahan, W.A.; Drier, Y.; Johnstone, S.E.; Hemming, M.L.; Tarjan, D.R.; Hegazi, E.; Shareef, S.J.; Javed, N.M.; Raut, C.P.; Eschle, B.K.; et al. Altered Chromosomal Topology Drives Oncogenic Programs in SDH-Deficient GISTs. Nature 2019, 575, 229–233. [Google Scholar] [CrossRef]
  26. Pantaleo, M.A.; Astolfi, A.; Indio, V.; Moore, R.; Thiessen, N.; Heinrich, M.C.; Gnocchi, C.; Santini, D.; Catena, F.; Formica, S.; et al. SDHA Loss-of-Function Mutations in KIT-PDGFRA Wild-Type Gastrointestinal Stromal Tumors Identified by Massively Parallel Sequencing. J. Natl. Cancer Inst. 2011, 103, 983–987. [Google Scholar] [CrossRef] [Green Version]
  27. Matyakhina, L.; Bei, T.A.; McWhinney, S.R.; Pasini, B.; Cameron, S.; Gunawan, B.; Stergiopoulos, S.G.; Boikos, S.; Muchow, M.; Dutra, A.; et al. Genetics of Carney Triad: Recurrent Losses at Chromosome 1 but Lack of Germline Mutations in Genes Associated with Paragangliomas and Gastrointestinal Stromal Tumors. J. Clin. Endocrinol. Metab. 2007, 92, 2938–2943. [Google Scholar] [CrossRef] [Green Version]
  28. Dwight, T.; Benn, D.E.; Clarkson, A.; Vilain, R.; Lipton, L.; Robinson, B.G.; Clifton-Bligh, R.J.; Gill, A.J. Loss of SDHA Expression Identifies SDHA Mutations in Succinate Dehydrogenase-Deficient Gastrointestinal Stromal Tumors. Am. J. Surg. Pathol. 2013, 37, 226–233. [Google Scholar] [CrossRef]
  29. Oudijk, L.; Gaal, J.; Korpershoek, E.; Van Nederveen, F.H.; Kelly, L.; Schiavon, G.; Verweij, J.; Mathijssen, R.H.J.; Den Bakker, M.A.; Oldenburg, R.A.; et al. SDHA Mutations in Adult and Pediatric Wild-Type Gastrointestinal Stromal Tumors. Mod. Pathol. 2013, 26, 456–463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Belinsky, M.G.; Rink, L.; von Mehren, M. Succinate Dehydrogenase Deficiency in Pediatric and Adult Gastrointestinal Stromal Tumors. Front. Oncol. 2013, 3, 107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Wagner, A.J.; Remillard, S.P.; Zhang, Y.X.; Doyle, L.A.; George, S.; Hornick, J.L. Loss of Expression of SDHA Predicts SDHA Mutations in Gastrointestinal Stromal Tumors. Mod. Pathol. 2013, 26, 289–294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Boikos, S.A.; Pappo, A.S.; Killian, J.K.; LaQuaglia, M.P.; Weldon, C.B.; George, S.; Trent, J.C.; von Mehren, M.; Wright, J.A.; Schiffman, J.D.; et al. Molecular Subtypes of KIT/PDGFRA Wild-Type Gastrointestinal Stromal Tumors. JAMA Oncol. 2016, 2, 922–928. [Google Scholar] [CrossRef] [Green Version]
  33. Carrera, S.; Beristain, E.; Sancho, A.; Iruarrizaga, E.; Rivero, P.; Mañe, J.M.; López Vivanco, G. Germline c.1A>C Heterozygous Pathogenic Variant in SDHA Reported for the First Time in a Young Adult with a Gastric Gastrointestinal Stromal Tumour (GIST): A Case Report. Hered. Cancer Clin. Pract. 2019, 17, 23. [Google Scholar] [CrossRef] [Green Version]
  34. Pantaleo, M.A.; Astolfi, A.; Urbini, M.; Nannini, M.; Paterini, P.; Indio, V.; Saponara, M.; Formica, S.; Ceccarelli, C.; Casadio, R.; et al. Analysis of All Subunits, SDHA, SDHB, SDHC, SDHD, of the Succinate Dehydrogenase Complex in KIT/PDGFRA Wild-Type GIST. Eur. J. Hum. Genet. 2014, 22, 32–39. [Google Scholar] [CrossRef] [Green Version]
  35. Pantaleo, M.A.; Nannini, M.; Astolfi, A.; Biasco, G. A Distinct Pediatric-Type Gastrointestinal Stromal Tumor in Adults: Potential Role of Succinate Dehydrogenase Subunit a Mutations. Am. J. Surg. Pathol. 2011, 35, 1750–1752. [Google Scholar] [CrossRef] [PubMed]
  36. Pantaleo, M.A.; Urbini, M.; Schipani, A.; Nannini, M.; Indio, V.; De Leo, A.; Vincenzi, B.; Brunello, A.; Grignani, G.; Casagrande, M.; et al. SDHA Germline Variants in Adult Patients With SDHA-Mutant Gastrointestinal Stromal Tumor. Front. Oncol. 2022, 11, 5400. [Google Scholar] [CrossRef]
  37. Italiano, A.; Chen, C.L.; Sung, Y.S.; Singer, S.; DeMatteo, R.P.; LaQuaglia, M.P.; Besmer, P.; Socci, N.; Antonescu, C.R. SDHA Loss of Function Mutations in a Subset of Young Adult Wild-Type Gastrointestinal Stromal Tumors. BMC Cancer 2012, 12, 408. [Google Scholar] [CrossRef] [Green Version]
  38. Gault, M.D.; Mandelker, D.; Delair, D.; Stewart, C.R.; Kemel, Y.; Sheehan, M.R.; Siegel, B.; Kennedy, J.; Marcell, V.; Arnold, A.; et al. Germline SDHA Mutations in Children and Adults with Cancer. Cold Spring Harb. Mol. Case Stud. 2018, 4, a002584. [Google Scholar] [CrossRef] [Green Version]
  39. Jiang, Q.; Zhang, Y.; Zhou, Y.H.; Hou, Y.Y.; Wang, J.Y.; Li, J.L.; Li, M.; Tong, H.X.; Lu, W.Q. A Novel Germline Mutation in SDHA Identified in a Rare Case of Gastrointestinal Stromal Tumor Complicated with Renal Cell Carcinoma. Int. J. Clin. Exp. Pathol. 2015, 8, 12188–12197. [Google Scholar]
  40. Pasini, B.; McWhinney, S.R.; Bei, T.; Matyakhina, L.; Stergiopoulos, S.; Muchow, M.; Boikos, S.A.; Ferrando, B.; Pacak, K.; Assie, G.; et al. Clinical and molecular genetics of patients with the Carney-Stratakis syndrome and germline mutations of the genes coding for the succinate dehydrogenase subunits SDHB, SDHC, and SDHD. Eur. J. Hum. Genet. 2008, 16, 79–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Stratakis, C.A.; Carney, J.A. The Triad of Paragangliomas, Gastric Stromal Tumours and Pulmonary Chondromas (Carney Triad), and the Dyad of Paragangliomas and Gastric Stromal Sarcomas (Carney-Stratakis Syndrome): Molecular Genetics and Clinical Implications. Proc. J. Intern. Med. 2009, 266, 43–52. [Google Scholar] [CrossRef] [Green Version]
  42. Miettinen, M.; Wang, Z.F.; Sarlomo-Rikala, M.; Osuch, C.; Rutkowski, P.; Lasota, J. Succinate Dehydrogenase-Deficient GISTs: A Clinicopathologic, Immunohistochemical, and Molecular Genetic Study of 66 Gastric GISTs with Predilection to Young Age. Am. J. Surg. Pathol. 2011, 35, 1712–1721. [Google Scholar] [CrossRef] [Green Version]
  43. McWhinney, S.R.; Pasini, B.; Stratakis, C.A. Familial Gastrointestinal Stromal Tumors and Germ-Line Mutations. N. Engl. J. Med. 2007, 357, 1054–1056. [Google Scholar] [CrossRef]
  44. Nannini, M.; Rizzo, A.; Indio, V.; Schipani, A.; Astolfi, A.; Pantaleo, M.A. Targeted Therapy in SDH-Deficient GIST. Ther. Adv. Med. Oncol. 2021, 13, 17588359211023278. [Google Scholar] [CrossRef] [PubMed]
  45. Casali, P.G.; Blay, J.Y.; Abecassis, N.; Bajpai, J.; Bauer, S.; Biagini, R.; Bielack, S.; Bonvalot, S.; Boukovinas, I.; Bovee, J.V.M.G.; et al. Gastrointestinal Stromal Tumours: ESMO–EURACAN–GENTURIS Clinical Practice Guidelines for Diagnosis, Treatment and Follow-up. Ann. Oncol. 2022, 33, 20–33. [Google Scholar] [CrossRef]
  46. Liu, W.; Zeng, X.; Wu, X.; He, J.; Gao, J.; Shuai, X.; Wang, G.; Zhang, P.; Tao, K. Clinicopathologic Study of Succinate-Dehydrogenase-Deficient Gastrointestinal Stromal Tumors: A Single-Institutional Experience in China. Medicine 2017, 96, e7668. [Google Scholar] [CrossRef]
  47. Ben-Ami, E.; Barysauskas, C.M.; von Mehren, M.; Heinrich, M.C.; Corless, C.L.; Butrynski, J.E.; Morgan, J.A.; Wagner, A.J.; Choy, E.; Yap, J.T.; et al. Long-Term Follow-up Results of the Multicenter Phase II Trial of Regorafenib in Patients with Metastatic and/or Unresectable GI Stromal Tumor after Failure of Standard Tyrosine Kinase Inhibitor Therapy. Ann. Oncol. 2016, 27, 1794–1799. [Google Scholar] [CrossRef] [PubMed]
  48. Ganjoo, K.N.; Villalobos, V.M.; Kamaya, A.; Fisher, G.A.; Butrynski, J.E.; Morgan, J.A.; Wagner, A.J.; D’Adamo, D.; McMillan, A.; Demetri, G.D.; et al. A Multicenter Phase II Study of Pazopanib in Patients with Advanced Gastrointestinal Stromal Tumors (GIST) Following Failure of at Least Imatinib and Sunitinib. Ann. Oncol. 2014, 25, 236–240. [Google Scholar] [CrossRef]
  49. Von Mehren, M.; George, S.; Heinrich, M.C.; Schuetze, S.M.; Yap, J.T.; Yu, J.Q.; Abbott, A.; Litwin, S.; Crowley, J.; Belinsky, M.; et al. Linsitinib (OSI-906) for the Treatment of Adult and Pediatric Wild-Type Gastrointestinal Stromal Tumors, a SARC Phase II Study. Clin. Cancer Res. 2020, 26, 1837–1845. [Google Scholar] [CrossRef] [PubMed]
  50. Lou, L.; Zhang, W.; Li, J.; Wang, Y. Abnormal Mgmt Promoter Methylation in Gastrointestinal Stromal Tumors: Genetic Susceptibility and Association with Clinical Outcome. Cancer Manag. Res. 2020, 12, 9941–9952. [Google Scholar] [CrossRef]
  51. De Silva, M.; Rastogi, S.; Chan, D.; Angel, C.; Prall, O.; Gill, A.; Guminski, A. Succinate Dehydrogenase-Deficient Gastrointestinal Stromal Tumor: From Diagnostic Dilemma to Novel Personalised Therapy in 2 Case Reports. Transl. Cancer Res. 2021, 10, 3588. [Google Scholar] [CrossRef] [PubMed]
  52. Chi, P.; Qin, L.-X.; D’Angelo, S.P.; Dickson, M.A.; Gounder, M.M.; Keohan, M.L.; Shoushtari, A.N.; Condy, M.M.; Konen, T.; Fruauff, A.; et al. A Phase Ib/II Study of MEK162 (Binimetinib [BINI]) in Combination with Imatinib in Patients with Advanced Gastrointestinal Stromal Tumor (GIST). J. Clin. Oncol. 2015, 33, 10507. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the SDH complex in physiological (a) and pathological (b) condition. SDH, succinate dehydrogenase; Q,ubiquinone; QH2,Ubiquinol; Fe–S, iron-sulfur; FAD, flavin adenine dinucleotide; FADH2, dihydroflavine-adenine dinucleotide; e, electron; TET, ten–eleven translocation; Me, methyl group; PHD, prolyl-hydroxylase domain proteins; Ub, ubiquitin; HIF, hypoxia-inducible factor.
Figure 1. Schematic representation of the SDH complex in physiological (a) and pathological (b) condition. SDH, succinate dehydrogenase; Q,ubiquinone; QH2,Ubiquinol; Fe–S, iron-sulfur; FAD, flavin adenine dinucleotide; FADH2, dihydroflavine-adenine dinucleotide; e, electron; TET, ten–eleven translocation; Me, methyl group; PHD, prolyl-hydroxylase domain proteins; Ub, ubiquitin; HIF, hypoxia-inducible factor.
Genes 14 00646 g001
Figure 2. Distribution of reported SDHA gene mutations in GIST. Mutations are annotated at the cDNA (a) and protein level (b).
Figure 2. Distribution of reported SDHA gene mutations in GIST. Mutations are annotated at the cDNA (a) and protein level (b).
Genes 14 00646 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Schipani, A.; Nannini, M.; Astolfi, A.; Pantaleo, M.A. SDHA Germline Mutations in SDH-Deficient GISTs: A Current Update. Genes 2023, 14, 646. https://doi.org/10.3390/genes14030646

AMA Style

Schipani A, Nannini M, Astolfi A, Pantaleo MA. SDHA Germline Mutations in SDH-Deficient GISTs: A Current Update. Genes. 2023; 14(3):646. https://doi.org/10.3390/genes14030646

Chicago/Turabian Style

Schipani, Angela, Margherita Nannini, Annalisa Astolfi, and Maria A. Pantaleo. 2023. "SDHA Germline Mutations in SDH-Deficient GISTs: A Current Update" Genes 14, no. 3: 646. https://doi.org/10.3390/genes14030646

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop