Abstract
Background & Aims:
Acute intermittent porphyria (AIP), caused by heterozygous germline mutations of the heme synthesis pathway enzyme HMBS (hydroxymethylbilane synthase), confers a high risk of hepatocellular carcinoma (HCC) development. Yet, the role of HMBS in liver tumorigenesis remains unclear.
Methods:
Herein, we explore HMBS alterations in a large series of 758 HCC cases, including 4 patients with AIP. We quantify the impact of HMBS mutations on heme biosynthesis pathway intermediates and we investigate the molecular and clinical features of HMBS-mutated tumors.
Results:
We identify recurrent bi-allelic HMBS inactivation, both in patients with AIP acquiring a second somatic HMBS mutation and in sporadic HCC with 2 somatic hits. HMBS alterations are enriched in truncating mutations, in particular in splice regions, leading to abnormal transcript structures. Bi-allelic HMBS inactivation results in a massive accumulation of its toxic substrate porphobilinogen and synergizes with CTNNB1-activating mutations, leading to the development of well-differentiated tumors with a transcriptomic signature of Wnt/β-catenin pathway activation and a DNA methylation signature related to ageing. HMBS-inactivated HCC mostly affects females, in the absence of fibrosis and classical HCC risk factors.
Conclusions:
These data identify HMBS as a tumor suppressor gene whose bi-allelic inactivation defines a homogenous clinical and molecular HCC subtype.
Keywords: Hepatocellular carcinoma, Acute intermittent porphyria, Heme biosynthesis pathway, Cancer Genomics
Graphical abstract

Lay summary:
Heme (the precursor to hemoglobin, which plays a key role in oxygen transport around the body) synthesis occurs in the liver and involves several enzymes including hydroxymethylbilane synthase (HMBS). HMBS mutations cause acute intermittent porphyria, a disease caused by the accumulation of toxic porphyrin precursors. Herein, we show that HMBS inactivation is also involved in the development of liver cancers with distinct clinical and molecular characteristics.
Introduction
Hepatocellular carcinoma (HCC) is a diverse, heterogeneous disease with an annual global incidence of almost 1 million. HCC often arises on a background of chronic liver disease which may be caused by viral hepatitis infection (HBV, HCV), alcohol, non-alcoholic steatohepatitis associated with metabolic syndrome, or environmental exposure to toxins such as aflatoxin B and aristolochic acid.1 HCC may also arise in the absence of cirrhosis due to the activation of oncogenes by viral insertions or structural rearrangements, or from the malignant transformation of benign hepatocellular adenomas.1–3 However, ~10% of HCCs cannot be attributed to typical risk factors. At the molecular level, whole-exome/genome sequencing (WES/WGS) studies have uncovered tens of driver genes and 11 cellular pathways recurrently altered in HCC.4–6 These include TERT alterations resulting in telomerase activation, activation of CTNNB1/WNT pathway, or inactivation of TP53, along with alterations of chromatin remodeling (ARID1A), mTOR (mammalian target of rapamycin) signaling, and regulators of oxidative stress.7 However, power estimations show that additional sequencing efforts are needed to unravel rare driver genes involved in small subsets of patients.8 Thus, novel genetic and risk factor associations contributing to HCC tumorigenesis remain to be identified.
Various clinical studies have identified an increased risk of HCC among patients with acute intermittent porphyria (AIP).9–15 AIP is a rare genetic disease caused by germline inactivating mutations in 1 allele of HMBS (hydroxymethylbilane synthase), encoding the enzyme that converts porphobilinogen to hydroxymethylbilane in the heme biosynthesis pathway.16 AIP symptoms include painful neurovisceral attacks with severe abdominal pain, vomiting, tachycardia and hypertension.17,18 Although AIP is an autosomal dominant disease, its penetrance is very low and only ~1% of HMBS mutation carriers will experience acute attacks.19,20 These attacks are triggered by numerous factors (such as alcohol, stress, increased progesterone, and various prescription drugs) which increase demand for heme and expression and/or activity of ALAS1, the enzyme preceding HMBS in the heme synthesis pathway.17,18,21 This results in saturation of the available functional HMBS and causes accumulation of toxic intermediates such as delta-aminolevulinic acid (ALA) and porphobilinogen (PBG).22,23 Several retrospective and prospective cohort studies have identified an increased incidence of HCC among both symptomatic and asymptomatic carriers of germline HMBS-inactivating mutations, with standardized risk ratios ranging from 2 to 38, independent of the traditional HCC risk factors mentioned above.9–12,15,24,25 These tumors often arise in livers without background fibrosis or cirrhosis, although underlying chronic injury cannot be ruled out. It has been proposed that insufficient HMBS activity and accumulation of ALA and PBG could lead to increased levels of oxidative stress and mitochondrial damage, promoting mutagenesis and increasing cancer risk.14,21 However, no definitive mechanism has yet been identified. Interestingly, a case report revealed that a patient with AIP, with 1 defective HMBS allele, developed an HCC tumor which carried an acquired somatic mutation in the second HMBS allele, resulting in complete loss of HMBS activity in the tumor.26 Although isolated, this report raises the question of whether HMBS could act as a tumor suppressor, beyond its role in the pathogenesis of AIP.
To better understand the role of HMBS in HCC development, we herein conducted a large-scale analysis of HMBS mutations in 784 HCC cases from in-house (LICA-FR) and The Cancer Genome Atlas (TCGA) data sets. We identify a subset of HCC with bi-allelic inactivation of HMBS, both in patients with and without AIP. We explore the functional consequences of HMBS mutations on heme pathway metabolites, and we characterize the clinical and molecular characteristics of HMBS-inactivated tumors.
Materials and methods
Description of the cohort
We analyzed WES/WGS data from a total of 758 HCC cases. WGS of 4 HCC (and matched non-tumor liver) cases that developed in 3 patients with AIP was performed for this study. Previously published WGS (n = 60) and WES (n = 347) data from our group (LICA-FR series3,4,49–51) were also reanalyzed for HMBS mutations. HCC samples and their non-tumor counterparts were collected from patients surgically treated in 4 French hospitals located in the Bordeaux and Paris region. The study was approved by institutional review board committees (CCPPRB Paris Saint-Louis IRB00003835). Written informed consent was obtained in accordance with French legislation. All samples were immediately frozen in liquid nitrogen and stored at −80°C. Clinical features of the 411 HCC cases from the LICA-FR series are summarized in Table S1. We also analyzed WES data from TCGA-LIHC project.6 Cholangiocarcinomas and mixed forms of HCC were discarded from TCGA series to keep only pure HCC samples (n = 347).
Patients with AIP
Three patients with HCC diagnosed in a context of acute intermittent porphyria (AIP) were included in this study. Patient #1606 was a 77-year-old female who developed an HCC more than 20 years after AIP diagnosis. Two years after the initial surgical resection, the tumor relapsed with 2 nodules (Edmonson grade 3), one of which (FR-1606T) was sampled and sequenced in this study. Subsequent treatments included radiotherapy, chemoembolization and sorafenib. The patient died from HCC progression 10 years after the initial diagnosis. Patient #4027 (male) was diagnosed with asymptomatic AIP at 33 years-old following a porphyria episode in his sister. He developed a bifocal HCC at age 64 with macrotrabecular massive histology.52 Both nodules were surgically removed and analyzed in this study. This patient relapsed 11 years later and was treated with radiofrequency ablation. Patient #4029 was a 77-year-old female with AIP who developed a well-differentiated HCC. Following surgical resection, no relapse was detected until the patient died 7 years later from another cause. In all cases we also sampled and sequenced adjacent non-tumor liver.
Whole-genome and RNA-sequencing
We performed WGS of 4 HCC cases (and matched non-tumor liver samples) that developed in 3 patients with AIP. We performed RNA-sequencing (RNA-seq) for 3 of them. Sequencing data generation and bioinformatic analyses are described extensively in the supplementary methods.
Mutational signature analysis
We used Palimpsest53 to extract single base substitution (SBS) signatures in the LICA-FR WGS data set. We first performed a de novo extraction to identify potentially new signatures. This analysis revealed 5 signatures, all corresponding to COSMIC signatures known to be operative in liver cancers (Fig. S1). In absence of a new signature, we then estimated the exposures of the 10 known liver cancer signatures (SBS1, 4, 5, 6, 12, 16, 17, 22, 23 and 24) in our series, and we compared their prevalence in HCC with or without HMBS inactivation.
Heme pathway intermediates quantification in frozen liver samples
Three heme biosynthesis pathway intermediates were quantified in frozen HCC and matched non-tumor liver samples: porphobilinogen, protoporphyrin IX (PPIX) and hemin. Heme is not stable in vitro and undergoes spontaneous oxidation to hemin. The level of hemin should be the same as heme. Liver samples were homogenized in water (50 mg tissue in 300 μl water). Then 300 μl of methanol:acetonitrile (v/v, 1:1) was added to a 100 μl aliquot of liver homogenate. After centrifugation, the supernatant was injected into ultra-high performance liquid chromatography with quadrupole time-of-flight mass spectrometry (UPLC-QTOFMS) for the analyses of Hemin and PPIX following previously reported methods.54,55 For PBG analysis, we used acetonitrile to precipitate proteins in liver homogenates, and the porphobilinogen in the supernatant was further derivatized with 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate before detection by UPLC-QTOFMS. For UPLC separation, we used the Acquity UPLC BEH C18 column (2.1 × 100 mm, 1.7 μm) with acetonitrile/water containing 0.1% formic acid as the mobile phase, at a temperature of 50°C. The QTOFMS system was operated in positive mode with electrospray ionization. The detailed mass parameters were published in a previous report.56
Multiomic characterization of HMBS-mutated HCC
To characterize the multiomic features of HMBS-mutated HCC, we analyzed RNA-seq (285 HCC and 29 non-tumor liver samples) and methylation array data (Illumina Infinium HumanMethylation450 BeadChip, 194 HCC and 35 non-tumor liver samples) available for part of the LICA-FR cohort. Except RNA-seq data for 3 HCCs that developed in patients with AIP, all data were previously published.3,29,50,51 T-stochastic neighbor embedding (tSNE) was used to project the expression and methylation data sets into 2 dimensions using the Rtsne package (https://github.com/jkrijthe/Rtsne). Gene expression (G1-G6) and DNA methylation subgroups (M1-M7) were defined as previously described.29,57 Gene expression signatures58,59 and DNA methylation components29 representing coordinated transcriptomic and epigenetic processes recurrently altered in HCC were quantified in each sample and compared between HMBS-mutated and other HCCs using Wilcoxon rank-sum tests.
Results
Molecular history of HCC developed in AIP patients
We sequenced the whole genomes of 4 HCC samples from 3 patients (2 females, 1 male) with diagnosed AIP. We identified, in each patient, a germline HMBS mutation (1 splice, 2 missense) that was documented as pathogenically associated with AIP in the Human Gene Mutation Database27. These mutations were found in both the tumor and non-tumor liver tissues (Fig. 1). The 4 HCC cases had acquired a median of 11,335 somatic point mutations, 673 indels and 26 structural variants, comparable with HCC developed in patients without AIP. Mutational signature analysis revealed a combination of COSMIC signatures 4, 5 and 16, ubiquitous in HCC (Fig. S1). We did not identify any new signature that would be in favor of a genotoxic role of heme pathway intermediates. Interestingly, all 4 tumors had acquired pathogenic somatic mutations of HMBS (1 splice, 1 frameshift, 2 missense), resulting in bi-allelic inactivation of the gene. These mutations were found only in tumor tissues. Other recurrent driver alterations included TERT promoter mutations (n = 4), activating mutations in CTNNB1 exon 3 (n = 3) and FGA frame-shift indels (n = 2). Private mutations were also found in CDKN2A, RPS6KA3, ZNRF3 and KRAS (hotspot mutation G12D). Of note, 2 synchronous HCC nodules were analyzed for patient #4027. These tumors displayed completely different mutation catalogues and thus result from independent tumor initiation events. Each nodule had acquired TERT promoter mutations and a distinct set of mutations in the same driver genes HMBS, CTNNB1 and FGA. Thus, HCC development in patients with AIP systematically involves inactivation of the second HMBS allele and follows a conserved evolutionary path with mutations activating TERT and CTNNB1 oncogenes.
Fig. 1. Molecular history of HCC developed in patients with AIP.

Whole-genome sequencing of HCC and matched non-tumor liver samples from 3 patients with AIP revealed germline HMBS mutations and somatic alterations acquired by cancer cells. Alterations in known liver cancer driver genes are annotated. Patient #4027 developed 2 independent synchronous HCCs. AIP, acute intermittent porphyria; HCC, hepatocellular carcinoma; SVs, structural variants.
Bi-allelic HMBS inactivation in sporadic HCC
To identify sporadic HCC potentially related to HMBS deficiency, we next screened HMBS mutations in 754 HCC cases from inhouse (LICA-FR, n = 60 genomes and 347 exomes, Table S1) and TCGA series (n = 347 exomes6). This data set includes patients with diverse fibrosis stages (39% F0-F1, 17% F2-F3, 44% F4) and diverse risk factors including alcohol intake (34%), HBV (33%) or HCV (18%) infection, and metabolic syndrome (12%). 21% of patients had no identified etiology. We identified 9 somatic HMBS mutations in 7 patients (4 in LICA-FR, 3 in TCGA series, Table 1), a low frequency yet significantly higher than expected by chance given gene size and mutation rate (p = 1.7×10−4, MutSigCV algorithm27), indicating a positive selection of HMBS mutations in HCC. Consistent with a tumor suppressive role, somatic HMBS mutations were mostly truncating, with an enrichment of mutations in splice regions (Fig. 2A). Using matched RNA-seq data, we confirmed the functional impact of splice mutations which all lead to abnormal transcript structures with varying degrees of intron retention or exon shrinkage (Fig. 2B). Of note, 7 of the 9 somatic HMBS mutations were documented as pathogenic mutations associated with AIP in the Human Gene Mutation Database.28 Also consistent with a tumor suppressive role, 5/7 patients with somatic HMBS mutations displayed evidence of bi-allelic inactivation. Two patients (TCGA-EP-A26S and TCGA-ZP-A9D4) harbored rare pathogenic germline mutations known to cause AIP; they may correspond to porphyria cases not annotated as such in TCGA database or healthy carriers of AIP-predisposing variants. Two patients (FR-1731T and FR-429T) harbored 2 different somatic mutations, and FR-1598T displayed a somatic mutation and a focal copy-neutral loss of heterozygosity, leading to the loss of the wild-type copy (Fig. S2). Overall, these data identify a rare subset of HCC with biallelic HMBS inactivation. Although we cannot formally exclude that 2 HMBS alterations may affect the same allele, the co-occurrence of 2 HMBS alterations in 5/7 patients is highly unlikely to occur by chance (p = 1.0×10−11, Fisher’s exact test) and strongly suggests a bi-allelic inactivation of the enzyme in these tumors.
Table 1.
Ten patients harboring HMBS mutations.
| Patient | AIP diagnosis | Sex | Age | HCC risk factors | Fibrosis score | HMBS alterations |
|---|---|---|---|---|---|---|
| #1606 | Yes | F | 77 | Without common etiology | F0 | Germline splice c.e1-1# Somatic missense R26H# |
| #4027 | Yes | M | 64 | Metabolic | F1 | Germline missense L234P# Somatic missense A297P in tumor FR-4027T Somatic splice c.e10-1 in tumor FR-4028T# |
| #4029 | Yes | F | 72 | Without common etiology | F0 | Germline missense L30F# Somatic frameshift DP312fs |
| TCGA-EP-A26S | No | M | 70 | Alcohol | F0 | Germline missense R195C# Somatic frameshift L334Gfs# |
| TCGA-ZP-A9D4 | No | F | 64 | Without common etiology | F0 | Germline splice c.e2+5 Somatic splice c.e13+2# |
| #1731 | No | F | 56 | Without common etiology | F0 | Somatic missense R149L# Somatic missense N169I |
| #428 | No | F | 65 | Without common etiology | F0 | Somatic splice c.e1+5# Somatic splice c.e8-1# |
| #1598 | No | F | 76 | HBV | F0 | Somatic splice c.e12+1# Focal copy-neutral LOH |
| TCGA-DD-A4ND | No | F | 56 | Without common etiology | F0 | Somatic splice c.e1+1# |
| BCM683 | No | M | 75 | Without common etiology | F4 | Somatic missense R175W |
Summary of germline and somatic mutations in each patient and their main clinical characteristics.
Documented pathogenic variant causing AIP.
Fig. 2. Recurrent inactivating HMBS mutations in HCC.

(A) Lollipop plot showing the type and distribution of HMBS mutations along the protein sequence. Amino acid and exon numbering is given with respect to the NCBI Refseq NM_000190 isoform. (B) Impact of splice mutations on RNA structure. RNA-sequencing coverage is shown in grey. Arcs represent split reads indicating splicing events. Mutations are highlighted in each plot. HCC, hepatocellular carcinoma.
HMBS inactivation results in a massive accumulation of PBG in HCC
Next, we investigated the functional consequences of HMBS mutations on the heme biosynthesis pathway. We used UPLC-QTOFMS to quantify the levels of 3 heme synthesis intermediates: PBG (the substrate of HMBS), PPIX and hemin (the oxidative product of heme) (Fig. 3 and Table S2). We analyzed HCC and matched non-tumor liver tissues from 10 patients, including 1 HCC developed in a context of AIP (#1606), 3 sporadic HCCs with bi-allelic somatic inactivation of HMBS (#428, #1598 and #1731), and 6 sporadic HCCs without HMBS inactivation for control comparison. PBG was barely detectable in the non-tumor liver tissues of sporadic HCCs and in HCC samples without HMBS inactivation (median <50 ng per g of tissue, range 0-1,415 ng/g). In contrast, germline HMBS mutation resulted in PBG accumulation in the non-tumor liver of the patient with AIP #1606 (8,022 ng/g), with a striking increase upon inactivation of the second HMBS allele in the tumor (32,547 ng/g). Similarly, the 3 tumors with bi-allelic somatic HMBS inactivation displayed a massive accumulation of PBG (median = 36,223 ng/g, range 25,575-55,289 ng/g). Overall, HCCs with bi-allelic HMBS inactivation displayed a significant increase of PBG compared to both non-tumor liver tissue and tumors without HMBS inactivation (p = 0.0010, Wilcoxon rank-sum test). The late-stage intermediates PPIX and hemin were detected at low levels in all the samples (median = 193 ng/g and 426 ng/g respectively), with a trend towards decreased PPIX levels in HMBS-inactivated HCC with respect to other HCC and non-tumor liver tissues (median = 143 ng/g vs. 237 ng/g, p = 0.10, Wilcoxon rank-sum test). Thus, biallelic HMBS inactivation in HCC results in a blockage of the heme synthesis pathway with major accumulation of its toxic precursor PBG.
Fig. 3. Massive porphobilinogen accumulation in HMBS-mutated HCC.

Three heme biosynthesis pathway intermediates (porphobilinogen, protoporphyrin IX and hemin) were quantified by ultra-high performance liquid chromatography with quadrupole time-of-flight mass spectrometry. Porphobilinogen is the substrate of HMBS, protoporphyrin IX is the last intermediate before heme, and hemin results from the spontaneous oxidation of heme in vitro. These metabolites were quantified in 10 HCC/non-tumor liver pairs from 1 patient with AIP, 3 patients with HCC and somatic HMBS inactivation, and 6 patients with HCC without HMBS inactivation used as controls. AIP, acute intermittent porphyria; HCC, hepatocellular carcinoma.
HMBS inactivation defines a homogenous clinical and molecular HCC subgroup
Patients with HMBS inactivation displayed homogenous clinical characteristics (Fig. 4A), with an enrichment of females (70%, p = 0.0035, Fisher’s exact test), low fibrosis with F0-F1 Metavir fibrosis scores (90%, p = 0.0019, Fisher’s exact test) and an absence of classical HCC risk factors (70%, p = 0.00094, Fisher’s exact test). There was no significant difference in patient age, Edmonson differentiation grade, or tumor size. At the molecular level, HMBS mutations frequently co-occurred with CTNNB1-activating mutations (82%, p = 0.00018, Fisher’s exact test) and to a lesser extent ARID2 (27%, p = 0.026) and CDKN2A mutations (18%, p = 0.048), indicating a synergistic effect on tumorigenesis (Fig. 4B). TERT promoter mutations were also frequent (71%) although not significantly enriched. By contrast, HMBS mutations were exclusive with TP53 mutations (0%, p = 0.040). HMBS-inactivated tumors displayed homogenous gene expression and DNA methylation profiles, evidenced by their tight clustering in t-distributed stochastic neighbor embedding visualizations (Fig. 4C). At the transcriptomic level, HMBS-inactivated HCC mostly belonged to the G5 and G6 subgroups associated with Wnt/β-catenin pathway activation,40,41 displayed high expression of differentiation markers and low expression of proliferation markers compared with other HCCs (Fig. 4D). At the epigenetic level, HMBS-inactivated tumors clustered within methylation subgroup M6 (enriched in well-differentiated CTNNB1-mutated tumors). Compared to other HCC, they displayed higher contribution of age-related signatures of hyper-(MC1) and hypomethylation (MC12) and lower contribution of methylation signatures related to male sex (MC3) and immune infiltration (MC6) (Fig. 4E). This methylation pattern is consistent with the enrichment in female sex and generally advanced age of this tumor subgroup, and with the low levels of immune infiltration in CTNNB1-mutated HCC.29,30 Overall, HMBS inactivation synergizes with Wnt/β-catenin activation and gives rise to a homogenous molecular entity, preferentially developed in the non-fibrotic liver of females without classical risk factors.
Fig. 4. Clinical and molecular features of HMBS-inactivated HCC.

Forest plots showing odds ratios for enrichment of clinical features (A) and driver alterations (B) among HMBS-inactivated HCC. P values were obtained using Fisher’s exact test. (C) t-distributed stochastic neighbor embedding plots showing the unsupervised classification of gene expression (RNA-sequencing) and DNA methylation (Illumina Infinium HumanMethylation450 BeadChip) profiles in the LICA-FR series. Previously defined transcriptomic (G1-G6) and DNA methylation (M1-M7) subgroups are indicated with a color code, and HMBS-mutated HCCs are highlighted with stars. (D) Gene expression signatures showing differential activity in HCC with or without HMBS mutations. (E) DNA methylation components showing differential activity in HCC with or without HMBS mutations. HCC, hepatocellular carcinoma.
Discussion
Our data identify HMBS as a tumor suppressor in HCC with 2 modes of inactivation: (i) germline HMBS mutation carriers, with or without diagnosed AIP, acquiring a second somatic mutation, or (ii) patients with bi-allelic somatic alterations. The fact that these tumors mostly develop in the absence of fibrosis and classical HCC risk factor suggests that HMBS inactivation may be the initiating event that triggers tumorigenesis. The enrichment of truncating mutations (10/18 HMBS mutations), the frequency of bi-allelic inactivation (9/11 tumors) and the massive PBG accumulation that we detected indicate that the heme synthesis pathway is blocked in these tumors. Similarly, a bi-allelic inactivation of protoporphyrin oxidase (PPOX) was previously described in a case of HCC that developed in a patient with variegate porphyria.26 However, complete loss of heme synthesis is not compatible with cell viability, as shown in animal models. We have 2 hypotheses to explain how tumor cells can survive with bi-allelic HMBS mutations. First, missense mutations may lead to enzymes retaining a minor enzymatic activity. For example, in vitro experiments demonstrated that HMBS enzymes with the R26H, L30F and R149L mutations identified in our HCC series retained respectively 3%, 3% and 5% of the wild-type enzyme activity.31 Second, tumor cells may import heme produced by surrounding non-tumor hepatocytes. Although HMBS-mutated HCC displayed higher expression of the heme importer FLVCR2, they also displayed lower expression of the heme importer UNC119 (encoding HRG4) and higher expression of the heme exporter ABCG2. These data do not support an adaptation of HMBS-mutated tumors to increased heme import. However, HMBS-mutated tumor cells do express several heme importers and are certainly able to import heme from their environment.
The tumorigenic effect of heme synthesis pathway blockage may result from the accumulation of porphyrin precursors. The leading hypothesis for acute attacks in AIP is that porphyrin precursors, notably ALA, are neurotoxic, although the precise mechanism of this toxicity remains elusive.32 Hepatotoxicity has not been reported in AIP to our knowledge, but the higher level of PBG resulting from bi-allelic HMBS inactivation may become toxic to hepatocytes and promote tumorigenesis. Alternatively, the tumorigenic effect of heme pathway blockage may be due to the depletion of heme or late pathway intermediates. In particular, PPIX, the last intermediate before heme, is a dual inhibitor of p53 interactions with MDM2 and MDM4.33 Treating human cells with PPIX leads to the overexpression of pro-apoptotic p53 target genes and induces cell death.34 Thus, PPIX depletion in HMBS-inactivated cells may promote p53 inhibition by MDM2/4. Such mechanism would also explain why HMBS and TP53 mutations are exclusive. We found only a non-significant trend towards decreased PPIX in HMBS-mutated tumors, but the baseline concentration in non-tumor liver is so low that it could be challenging to detect the decrease in tumor cells. This hypothesis should be explored further in mechanistic experiments. Finally, HMBS inactivation may have indirect consequences on other metabolic processes.35 We observed an upregulation of several tricarboxylic acid cycle enzymes (IDH3B, OGDH, SUCLA2, SUCLG2, SDHA/B/C/D, FH and CS) in HMBS-mutated HCCs with respect to other HCCs, as well as a downregulation of early glycolysis enzymes (HK1/2 and GPI) and an upregulation of late glycolysis enzymes (PGM1 and PKLR) (Fig. S3 and Table S3). However, these changes were also observed in CTNNB1-mutated HCCs, so it is not clear if they were due to Wnt/β-catenin pathway activation and/or HMBS defects. HMBS-mutated tumors also displayed an upregulation of serine-glycine synthesis pathway enzymes (PSAT1 and SHMT2) that may allow tumor cells to counterbalance the consumption of glycine used to produce of ALA and PBG.
HMBS inactivation defines a homogeneous HCC subgroup with distinct clinical and molecular features. Notably, HMBS-mutated HCCs were strongly enriched in females (7/10 vs. 24% in HCC without HMBS mutation), and this association holds true in patients without AIP (5/7 females). Thus, women are more prone than men to develop HCC upon HMBS inactivation. Similarly, germline HMBS mutations induce more frequent and severe AIP attacks in women.36,37 Indeed, female sex hormones increase the production of porphyrin precursors by inducing ALA-synthase (the first enzyme in the heme synthesis pathway), and GnRH agonists have been shown to prevent cyclical attacks.38,39 Contrary to AIP, which mostly affects young and middle-aged women, HMBS-related HCCs were diagnosed in post-menopausal women. Thus, the role of sex hormones in interaction with heme pathway blockage in liver tumorigenesis remains to be explored. At the molecular level, we identified a striking association between HMBS and CTNNB1 mutations, indicating a synergy between the pathways in liver tumorigenesis. It was previously shown in mice that Ctnnb1 knockout induces downregulation of the heme biosynthesis enzymes ALA-S and ALA-D and protects from porphyria-associated liver injury,40 whereas β-catenin activation enhances the expression of heme synthesis-related genes.41 Consistently, we observed a significant upregulation of 5 heme pathway enzymes (ALAS1, ALAD, UROS, PPOX and FECH) in sporadic HCC with CTNNB1-activating mutations (Fig. S4 and Table S3). HMBS-mutated tumors displayed an even stronger upregulation of ALAS1 and ALAD, suggesting that tumor cells react to pathway blockage by increasing the expression of these early enzymes, reinforcing the accumulation of toxic PBG. Both female sex and CTNNB1 activation may thus promote liver tumorigenesis by intensifying the consequences of HMBS inactivation. Consistently, a recent large-scale study of 1,244 individuals with acute hepatic porphyrias identified a strong association between clinical AIP activity, elevated urinary PBG, and primary liver cancer risk.24
Metabolic reprogramming is considered a hallmark of cancer.42 Frequent metabolic alterations include the shift from oxidative phosphorylation to aerobic glycolysis (Warburg effect), deregulated glutamine uptake or hyperactivation of the serine/glycine biosynthetic pathway.43 Yet, only few metabolic genes have been identified as direct cancer drivers. These include IDH1/2 in glioma and AML,44,45 genes encoding SDH subunits in paraganglioma,46 FH in renal cell cancer47 or FUT9 in colorectal cancer.48 Herein, we identify HMBS as an HCC driver gene and highlight a tumorigenic role of heme biosynthesis pathway blockage in the liver. HMBS mutations should be investigated in patients with HCC without typical risk factors.
Supplementary Material
Highlights.
We identify recurrent HMBS-inactivating mutations in hepatocellular carcinoma.
Bi-allelic HMBS inactivation occurs both in patients with acute intermittent porphyria and sporadic HCC.
HMBS inactivation induces a massive accumulation of its toxic substrate porphobilinogen.
HMBS-mutated HCC mostly develop in females, in the absence of fibrosis and classical HCC risk factors.
HMBS-mutated HCC display activating CTNNB1 mutations and Wnt/β-catenin pathway activation.
Acknowledgements
We thank the clinicians, surgeons, pathologists, hepatologists and oncologists who contributed to the tissue collection and clinical annotations. We also thank the Réseau national CRB Foie (BB-0033-0085), the tumor banks of CHU Bordeaux (BB-0033-00036), Jean Verdier Hospital (APHP) and CHU Henri Mondor (APHP) for contributing to the tissue collection. We warmly thank Lewis Roberts, Renu Dhanasekaran and Mike Torbenson for sharing valuable clinical annotations about HMBS-mutated TCGA cases.
Financial support
This work was supported by France Génomique, ITMO Cancer AVIESAN (Alliance Nationale pour les Sciences de la Vie et de la Santé, National Alliance for Life Sciences & Health) within the framework of the Cancer Plan (“HTE program-HetColi network”), ANRS and the French Liver Biobanks network – INCa, BB-0033-00085. The group is supported by the Ligue Nationale Contre le Cancer (Equipe Labellisée), Labex OncoImmunology (investissement d’avenir), Coup d’Elan de la Fondation Bettencourt-Schueller and the SIRIC CARPEM. LM was supported by the Chateaubriand fellowship from the Embassy of France in the United States. JZ and XM were supported by the National Institute of Diabetes and Digestive and Kidney Diseases (R01DK126875). ET was supported by a Marie Skłodowska-Curie Individual Fellowship from the EU and is a Research Associate of the FRS-FNRS in Belgium. QB was supported by fellowship from the HOB doctoral school, the ministry of Education and Research and Association pour la Recherche contre le Cancer (ARC).
Abbreviations
- AIP
acute intermittent porphyria
- ALA
aminolevulinic acid
- HCC
hepatocellular carcinoma
- HMBS
hydroxymethylbilane synthase
- PBG
porphobilinogen
- PPIX
protoporphyrin IX
- PPOX
protoporphyrin oxidase
- SBS
single base substitution
- RNA-seq
RNA-sequencing
- TCGA
The Cancer Genome Atlas
- UPLC-QTOFMS
ultra-high performance liquid chromatography with quadrupole time-of-flight mass; spectrometry
- WES
whole-exome sequencing
- WGS
whole-genome sequencing
Footnotes
Conflict of interest
The authors declare that they have no competing interests.
Please refer to the accompanying ICMJE disclosure forms for further details.
Supplementary data
Supplementary data to this article can be found online at https://doi.org/10.1016/j.jhep.2022.05.018.
Data availability statement
Whole-genome sequencing and RNA-seq data generated for this study have been deposited to the European Genome Archive (EGA) under study accession number [EGAS00001005986].
References
Author names in bold designate shared co-first authorship
- [1].Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, et al. Hepatocellular carcinoma. Nat Rev Dis Primers 2021;7:1–28. 10.1038/s41572-020-00240-3. [DOI] [PubMed] [Google Scholar]
- [2].Nault J-C, Couchy G, Balabaud C, Morcrette G, Caruso S, Blanc J-F, et al. Molecular classification of hepatocellular adenoma associates with risk factors, bleeding, and malignant transformation. Gastroenterology 2017;152:880–894.e6. 10.1053/j.gastro.2016.11.042. [DOI] [PubMed] [Google Scholar]
- [3].Bayard Q, Meunier L, Peneau C, Renault V, Shinde J, Nault J-C, et al. Cyclin A2/E1 activation defines a hepatocellular carcinoma subclass with a rearrangement signature of replication stress. Nat Commun 2018;9:5235. 10.1038/s41467-018-07552-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Schulze K, Imbeaud S, Letouzé E, Alexandrov LB, Calderaro J, Rebouissou S, et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat Genet 2015;47:505–511. 10.1038/ng.3252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Fujimoto A, Furuta M, Totoki Y, Tsunoda T, Kato M, Shiraishi Y, et al. Whole-genome mutational landscape and characterization of noncoding and structural mutations in liver cancer. Nat Genet 2016;48:500–509. 10.1038/ng.3547. [DOI] [PubMed] [Google Scholar]
- [6].Cancer Genome Atlas Research Network. Electronic address: wheeler@bcm.edu, Cancer Genome Atlas Research Network. Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell 2017;169:1327–1341.e23. 10.1016/j.cell.2017.05.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Zucman-Rossi J, Villanueva A, Nault J-C, Llovet JM. Genetic landscape and biomarkers of hepatocellular carcinoma. Gastroenterology 2015;149:1226–1239.e4. 10.1053/j.gastro.2015.05.061. [DOI] [PubMed] [Google Scholar]
- [8].Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 2014;505:495–501. 10.1038/nature12912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [9].Andersson C, Bjersing L, Lithner F. The epidemiology of hepatocellular carcinoma in patients with acute intermittent porphyria. J Intern Med 1996;240:195–201. 10.1046/j.1365-2796.1996.21847000.x. [DOI] [PubMed] [Google Scholar]
- [10].Linet MS, Gridley G, Nyrén O, Mellemkjaer L, Olsen JH, Keehn S, et al. Primary liver cancer, other malignancies, and mortality risks following porphyria: a cohort study in Denmark and Sweden. Am J Epidemiol 1999;149:1010–1015. 10.1093/oxfordjournals.aje.a009745. [DOI] [PubMed] [Google Scholar]
- [11].Andant C, Puy H, Bogard C, Faivre J, Soulé JC, Nordmann Y, et al. Hepatocellular carcinoma in patients with acute hepatic porphyria: frequency of occurrence and related factors. J Hepatol 2000;32:933–939. 10.1016/s0168-8278(00)80097-5. [DOI] [PubMed] [Google Scholar]
- [12].Innala E, Andersson C. Screening for hepatocellular carcinoma in acute intermittent porphyria: a 15-year follow-up in northern Sweden. J Intern Med 2011;269:538–545. 10.1111/j.1365-2796.2010.02335.x. [DOI] [PubMed] [Google Scholar]
- [13].Sardh E, Wahlin S, Björnstedt M, Harper P, Andersson DEH. High risk of primary liver cancer in a cohort of 179 patients with Acute Hepatic Porphyria. J Inherit Metab Dis 2013;36:1063–1071. 10.1007/s10545-012-9576-9. [DOI] [PubMed] [Google Scholar]
- [14].Peoc’h K, Manceau H, Karim Z, Wahlin S, Gouya L, Puy H, et al. Hepatocellular carcinoma in acute hepatic porphyrias: a Damocles Sword. Mol Genet Metab 2019;128:236–241. 10.1016/j.ymgme.2018.10.001. [DOI] [PubMed] [Google Scholar]
- [15].Saberi B, Naik H, Overbey JR, Erwin AL, Anderson KE, Bissell DM, et al. Hepatocellular carcinoma in acute hepatic porphyrias: results from the longitudinal study of the U.S. Porphyrias Consortium. Hepatology 2021;73:1736–1746. 10.1002/hep.31460. [DOI] [PubMed] [Google Scholar]
- [16].Meyer UA, Strand LJ, Doss M, Rees AC, Marver HS. Intermittent acute porphyria – demonstration of a genetic defect in porphobilinogen metabolism. Https://Doi-OrgProxyInsermbiblioInistFr/101056/NEJM197206152862401 2010. 10.1056/NEJM197206152862401. [DOI] [PubMed] [Google Scholar]
- [17].Puy H, Gouya L, Deybach J-C. Porphyrias. The Lancet 2010;375:924–937. 10.1016/S0140-6736(09)61925-5. [DOI] [PubMed] [Google Scholar]
- [18].Stein PE, Badminton MN, Rees DC. Update review of the acute porphyrias. Br J Haematol 2017;176:527–538. 10.1111/bjh.14459. [DOI] [PubMed] [Google Scholar]
- [19].Nordmann Y, Puy H, Da Silva V, Simonin S, Robreau AM, Bonaiti C, et al. Acute intermittent porphyria: prevalence of mutations in the porphobilinogen deaminase gene in blood donors in France. J Intern Med 1997;242:213–217. 10.1046/j.1365-2796.1997.00189.x. [DOI] [PubMed] [Google Scholar]
- [20].Chen B, Solis-Villa C, Hakenberg J, Qiao W, Srinivasan RR, Yasuda M, et al. Acute intermittent porphyria: predicted pathogenicity of HMBS variants indicates extremely low penetrance of the autosomal dominant disease. Hum Mutat 2016;37:1215–1222. 10.1002/humu.23067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Bonkovsky HL, Dixon N, Rudnick S. Pathogenesis and clinical features of the acute hepatic porphyrias (AHPs). Mol Genet Metab 2019;128:213–218. 10.1016/j.ymgme.2019.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Floderus Y, Sardh E, Möller C, Andersson C, Rejkjaer L, Andersson DEH, et al. Variations in porphobilinogen and 5-aminolevulinic acid concentrations in plasma and urine from asymptomatic carriers of the acute intermittent porphyria gene with increased porphyrin precursor excretion. Clin Chem 2006;52:701–707. 10.1373/clinchem.2005.058198. [DOI] [PubMed] [Google Scholar]
- [23].Sardh E, Harper P, Andersson DEH, Floderus Y. Plasma porphobilinogen as a sensitive biomarker to monitor the clinical and therapeutic course of acute intermittent porphyria attacks. Eur J Intern Med 2009;20:201–207. 10.1016/j.ejim.2008.06.012. [DOI] [PubMed] [Google Scholar]
- [24].Lissing M, Vassiliou D, Floderus Y, Harper P, Bottai M, Kotopouli M, et al. Risk of primary liver cancer in acute hepatic porphyria patients: a matched cohort study of 1244 individuals. J Intern Med 2022. 10.1111/joim.13463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Kauppinen R, Mustajoki P. Acute hepatic porphyria and hepatocellular carcinoma. Br J Cancer 1988;57:117–120. 10.1038/bjc.1988.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Schneider-Yin X, van Tuyll van Serooskerken A-M, Siegesmund M, Went P, Barman-Aksözen J, Bladergroen RS, et al. Biallelic inactivation of proto-porphyrinogen oxidase and hydroxymethylbilane synthase is associated with liver cancer in acute porphyrias. J Hepatol 2015;62:734–738. 10.1016/j.jhep.2014.11.029. [DOI] [PubMed] [Google Scholar]
- [27].Lawrence MS, Stojanov P, Polak P, Kryukov GV, Cibulskis K, Sivachenko A, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 2013;499:214–218. 10.1038/nature12213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Stenson PD, Mort M, Ball EV, Chapman M, Evans K, Azevedo L, et al. The Human Gene Mutation Database (HGMD®): optimizing its use in a clinical diagnostic or research setting. Hum Genet 2020;139:1197–1207. 10.1007/s00439-020-02199-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Meunier L, Hirsch TZ, Caruso S, Imbeaud S, Bayard Q, Roehrig A, et al. DNA methylation signatures reveal the diversity of processes remodeling hepatocellular carcinoma methylomes. Hepatology 2021;74:816–834. 10.1002/hep.31796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Ruiz de Galarreta M, Bresnahan E, Molina-Sánchez P, Lindblad KE, Maier B, Sia D, et al. β-Catenin activation promotes immune escape and resistance to anti-PD-1 therapy in hepatocellular carcinoma. Cancer Discov 2019;9:1124–1141. 10.1158/2159-8290.CD-19-0074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Lenglet H, Schmitt C, Grange T, Manceau H, Karboul N, Bouchet-Crivat F, et al. From a dominant to an oligogenic model of inheritance with environmental modifiers in acute intermittent porphyria. Hum Mol Genet 2018;27:1164–1173. 10.1093/hmg/ddy030. [DOI] [PubMed] [Google Scholar]
- [32].Meyer UA, Schuurmans MM, Lindberg RL. Acute porphyrias: pathogenesis of neurological manifestations. Semin Liver Dis 1998;18:43–52. 10.1055/s-2007-1007139. [DOI] [PubMed] [Google Scholar]
- [33].Jiang L, Malik N, Acedo P, Zawacka-Pankau J. Protoporphyrin IX is a dual inhibitor of p53/MDM2 and p53/MDM4 interactions and induces apoptosis in B-cell chronic lymphocytic leukemia cells. Cell Death Discov 2019;5:77. 10.1038/s41420-019-0157-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Zawacka-Pankau J, Issaeva N, Hossain S, Pramanik A, Selivanova G, Podhajska AJ. Protoporphyrin IX interacts with wild-type p53 protein in vitro and induces cell death of human colon cancer cells in a p53-dependent and -independent manner. J Biol Chem 2007;282:2466–2472. 10.1074/jbc.M608906200. [DOI] [PubMed] [Google Scholar]
- [35].Fiorito V, Chiabrando D, Petrillo S, Bertino F, Tolosano E. The multifaceted role of heme in cancer. Front Oncol 2019;9:1540. 10.3389/fonc.2019.01540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Tschudy DP, Valsamis M, Magnussen CR. Acute intermittent porphyria: clinical and selected research aspects. Ann Intern Med 1975;83:851–864. 10.7326/0003-4819-83-6-851. [DOI] [PubMed] [Google Scholar]
- [37].Anderson KE. LHRH analogues for hormonal manipulation in acute intermittent porphyria. Semin Hematol 1989;26:10–15. [PubMed] [Google Scholar]
- [38].Granick S. The induction in vitro of the synthesis of delta-aminolevulinic acid synthetase in chemical porphyria: a response to certain drugs, sex hormones, and foreign chemicals. J Biol Chem 1966;241:1359–1375. [PubMed] [Google Scholar]
- [39].Anderson KE, Spitz IM, Sassa S, Bardin CW, Kappas A. Prevention of cyclical attacks of acute intermittent porphyria with a long-acting agonist of luteinizing hormone–releasing hormone. Http://DxDoiOrg/101056/NEJM198409063111006 2010. 10.1056/NEJM198409063111006. [DOI] [PubMed] [Google Scholar]
- [40].Saggi H, Maitra D, Jiang A, Zhang R, Wang P, Cornuet P, et al. Loss of hepatocyte β-catenin protects mice from experimental porphyria-associated liver injury. J Hepatol 2019;70:108–117. 10.1016/j.jhep.2018.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Braeuning A, Schwarz M. Zonation of heme synthesis enzymes in mouse liver and their regulation by β-catenin and Ha-ras. Biol Chem 2010;391:1305–1313. 10.1515/BC.2010.115. [DOI] [PubMed] [Google Scholar]
- [42].Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646–674. 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- [43].Pavlova NN, Thompson CB. The emerging hallmarks of cancer metabolism. Cell Metab 2016;23:27–47. 10.1016/j.cmet.2015.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Parsons DW, Jones S, Zhang X, Lin JC-H, Leary RJ, Angenendt P, et al. An integrated genomic analysis of human glioblastoma multiforme. Science 2008;321:1807–1812. 10.1126/science.1164382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Mardis ER, Ding L, Dooling DJ, Larson DE, McLellan MD, Chen K, et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med 2009;361:1058–1066. 10.1056/NEJMoa0903840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Baysal BE, Ferrell RE, Willett-Brozick JE, Lawrence EC, Myssiorek D, Bosch A, et al. Mutations in SDHD, a mitochondrial complex II gene, in hereditary paraganglioma. Science 2000;287:848–851. 10.1126/science.287.5454.848. [DOI] [PubMed] [Google Scholar]
- [47].Kiuru M, Lehtonen R, Arola J, Salovaara R, Järvinen H, Aittomäki K, et al. Few FH mutations in sporadic counterparts of tumor types observed in hereditary leiomyomatosis and renal cell cancer families. Cancer Res 2002;62:4554–4557. [PubMed] [Google Scholar]
- [48].Auslander N, Cunningham CE, Toosi BM, McEwen EJ, Yizhak K, Vizeacoumar FS, et al. An integrated computational and experimental study uncovers FUT9 as a metabolic driver of colorectal cancer. Mol Syst Biol 2017;13:956. 10.15252/msb.20177739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Letouzé E, Shinde J, Renault V, Couchy G, Blanc J-F, Tubacher E, et al. Mutational signatures reveal the dynamic interplay of risk factors and cellular processes during liver tumorigenesis. Nat Commun 2017;8:1315. 10.1038/s41467-017-01358-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Hirsch TZ, Negulescu A, Gupta B, Caruso S, Noblet B, Couchy G, et al. BAP1 mutations define a homogeneous subgroup of hepatocellular carcinoma with fibrolamellar-like features and activated PKA. J Hepatol 2020;72:924–936. 10.1016/j.jhep.2019.12.006. [DOI] [PubMed] [Google Scholar]
- [51].Péneau C, Imbeaud S, La Bella T, Hirsch TZ, Caruso S, Calderaro J, et al. Hepatitis B virus integrations promote local and distant oncogenic driver alterations in hepatocellular carcinoma. Gut 2021. 10.1136/gutjnl-2020-323153.gutjnl-2020-323153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Ziol M, Poté N, Amaddeo G, Laurent A, Nault J-C, Oberti F, et al. Macro-trabecular-massive hepatocellular carcinoma: a distinctive histological subtype with clinical relevance. Hepatology 2018;68:103–112. 10.1002/hep.29762. [DOI] [PubMed] [Google Scholar]
- [53].Shinde J, Bayard Q, Imbeaud S, Hirsch TZ, Liu F, Renault V, et al. Palimpsest: an R package for studying mutational and structural variant signatures along clonal evolution in cancer. Bioinformatics 2018;34:3380–3381. 10.1093/bioinformatics/bty388. [DOI] [PubMed] [Google Scholar]
- [54].Li F, Wang P, Liu K, Tarrago MG, Lu J, Chini EN, et al. A high dose of isoniazid disturbs endobiotic homeostasis in mouse liver. Drug Metab Dispos 2016;44:1742–1751. 10.1124/dmd.116.070920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [55].Wang P, Sachar M, Lu J, Shehu AI, Zhu J, Chen J, et al. The essential role of the transporter ABCG2 in the pathophysiology of erythropoietic protoporphyria. Sci Adv 2019;5:eaaw6127. 10.1126/sciadv.aaw6127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Zhu J, Wang P, Shehu AI, Lu J, Bi H, Ma X. Identification of novel pathways in idelalisib metabolism and bioactivation. Chem Res Toxicol 2018;31:548–555. 10.1021/acs.chemrestox.8b00023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Boyault S, Rickman DS, de Reyniès A, Balabaud C, Rebouissou S, Jeannot E, et al. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology 2007;45:42–52. 10.1002/hep.21467. [DOI] [PubMed] [Google Scholar]
- [58].Nault J-C, Martin Y, Caruso S, Hirsch TZ, Bayard Q, Calderaro J, et al. Clinical impact of genomic diversity from early to advanced hepatocellular carcinoma. Hepatology 2020;71:164–182. 10.1002/hep.30811. [DOI] [PubMed] [Google Scholar]
- [59].Caruso S, Calatayud A-L, Pilet J, La Bella T, Rekik S, Imbeaud S, et al. Analysis of liver cancer cell lines identifies agents with likely efficacy against hepatocellular carcinoma and markers of response. Gastroenterology 2019;157:760–776. 10.1053/j.gastro.2019.05.00. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
Whole-genome sequencing and RNA-seq data generated for this study have been deposited to the European Genome Archive (EGA) under study accession number [EGAS00001005986].
