Hostname: page-component-54dcc4c588-hp6zs Total loading time: 0 Render date: 2025-09-28T05:23:30.283Z Has data issue: false hasContentIssue false

Mutual Tissue Microchimerism of Hepatoblastomas in Monozygotic Twins From a Familial Adenomatous Polyposis Family

Published online by Cambridge University Press:  18 August 2025

Atsuhiro Arisue
Affiliation:
Department of Surgery, Iwate Medical University School of Medicine, Yahaba, Japan
Kiyoshi Yamaguchi
Affiliation:
Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
Kiyoko Takane
Affiliation:
Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
Yoshiko Asakura
Affiliation:
Department of Pediatrics, Iwate Medical University School of Medicine, Yahaba, Japan
Yasushi Hasegawa
Affiliation:
Department of Surgery, Keio University School of Medicine, Tokyo, Japan
Masaru Mizuno
Affiliation:
Department of Pediatric Surgery, Akita University School of Medicine, Akita, Japan
Hiroyuki Nitta
Affiliation:
Department of Surgery, Iwate Medical University School of Medicine, Yahaba, Japan
Kazuyuki Ishida
Affiliation:
Department of Diagnostic Pathology, Dokkyo Medical University, Mibu, Japan
Takeshi Iwaya
Affiliation:
Department of Clinical Oncology, Iwate Medical University School of Medicine, Yahaba, Japan
Eigo Shimizu
Affiliation:
Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
Seiya Imoto
Affiliation:
Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
Satoru Miyano
Affiliation:
M&D Data Science Center, IIR, Institute of Science Tokyo, Tokyo, Japan
Yoichi Furukawa
Affiliation:
Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
Satoshi S. Nishizuka*
Affiliation:
Division of Biomedical Research and Development, Iwate Medical University Institute for Biomedical Sciences, Yahaba, Japan
*
Corresponding author: Satoshi S. Nishizuka; Email: snishizu@iwate-med.ac.jp

Abstract

Patients with familial adenomatous polyposis (FAP) have increased risk of hepatoblastoma (HB). We report monozygotic twins with HB in a FAP family. To explore genetic alterations in the HBs of the twins, we carried out whole exome sequencing (WES), RNA-seq, and immunohistochemical analyses of the tumors. Additional multiregional digital PCR was performed to profile clonality of each tumor. To determine a pathogenic germline variant in APC, Sanger sequencing was applied for the twins, the father, and the siblings of the father. A pathogenic variant of the APC gene was identified in the father as well as the twins. The WES of the HBs in the twins identified somatic mutations, including an NRAS mutation in the tumor of the first infant (C1), and an ACVR2A mutation in the tumor of the second infant (C2). No somatic mutations were identified in the genes associated with the Wnt signaling pathway. However, accumulation of β-catenin was found in the C1 and C2 tumors by immunohistochemical staining, suggesting activation of the Wnt signaling pathway. Digital PCR analysis revealed that the NRAS mutation was found in multiregional specimens of C1 and those of C2. The ACVR2A mutation was found in multiregional specimens of C2, whereas the mutation was also identified in those of C1. The existence of a shared somatic mutation may suggest that microchimerism took place in the development of HBs through the utero-placental circulatory system. Importantly, the initiation of tumorigenesis is thought to occur during the fetal period after organ development of the liver.

Information

Type
Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2025. Published by Cambridge University Press on behalf of International Society for Twin Studies

Hepatoblastoma (HB) is the most common form of hepatic malignant tumors in children. While several genetic and epidemiologic studies of HB have been conducted, the molecular mechanisms underlying HB in FAP patients have not been fully elucidated. The majority (70%) of HB cases are sporadic and the number of genetic alterations is known to be relatively small compared to adult solid tumors (Eichenmuller et al., Reference Eichenmuller, Trippel, Kreuder, Beck, Schwarzmayr, Haberle, Cairo, Leuschner, von Schweinitz, Strom and Kappler2014; Jia et al., Reference Jia, Dong, Jing, Xu, Wang, Chen, Li, Huang, Zhang, Zhang, Liu, Zheng, Xia, Wang, Dong and He2014; Nagae et al., Reference Nagae, Yamamoto, Fujita, Fujita, Nonaka, Umeda, Fukuda, Tatsuno, Maejima, Hayashi, Kurihara, Kojima, Hishiki, Watanabe, Ida, Yano, Hiyama, Tanaka, Inoue and Hiyama2021). Most HBs are found in infants, for which hepatectomy and/or chemotherapy is considered as standard therapy (Hiyama et al., Reference Hiyama, Hishiki, Watanabe, Ida, Ueda, Kurihara, Yano, Hoshino, Yokoi, Takama, Nogami, Taguchi, Mori, Kihira, Miyazaki, Fuji, Honda, Iehara, Kazama and Yoshimura2020; Katzenstein et al., Reference Katzenstein, Langham, Malogolowkin, Krailo, Towbin, McCarville, Finegold, Ranganathan, Dunn, McGahren, Tiao, O’Neill, Qayed, Furman, Xia, Rodriguez-Galindo and Meyers2019; Perilongo et al., Reference Perilongo, Maibach, Shafford, Brugieres, Brock, Morland, de Camargo, Zsiros, Roebuck, Zimmermann, Aronson, Childs, Widing, Laithier, Plaschkes, Pritchard, Scopinaro, MacKinlay and Czauderna2009; Zsiros et al., Reference Zsiros, Brugieres, Brock, Roebuck, Maibach, Zimmermann, Childs, Pariente, Laithier, Otte, Branchereau, Aronson, Rangaswami, Ronghe, Casanova, Sullivan, Morland, Czauderna and Perilongo2013). If the histology is pure fetal type, then the prognosis is favorable (Malogolowkin et al., Reference Malogolowkin, Katzenstein, Meyers, Krailo, Rowland, Haas and Finegold2011).

Among the limited number of genetic changes, mutations of β-catenin (CTNNB1) have been most frequently observed, followed by APC and LGR6 (Perugorria et al., Reference Perugorria, Olaizola, Labiano, Esparza-Baquer, Marzioni, Marin, Bujanda and Banales2019) in HB. Importantly, it has been reported that the risk of HB is 750 to 7500-fold higher in FAP individuals who carry pathogenic germline variants (Kinzler et al., Reference Kinzler, Nilbert, Su, Vogelstein, Bryan, Levy, Smith, Preisinger, Hedge and McKechnie1991; Nishisho et al., Reference Nishisho, Nakamura, Miyoshi, Miki, Ando, Horii, Koyama, Utsunomiya, Baba and Hedge1991) than in the general population from a population-based registry (Aretz et al., Reference Aretz, Koch, Uhlhaas, Friedl, Propping, von Schweinitz and Pietsch2006; Giardiello et al., Reference Giardiello, Offerhaus, Krush, Booker, Tersmette, Mulder, Kelley and Hamilton1991; Giardiello et al., Reference Giardiello, Petersen, Brensinger, Luce, Cayouette, Bacon, Booker and Hamilton1996; Hughes & Michels, Reference Hughes and Michels1992). These data indicate that abrogation of the Wnt signaling pathway plays a crucial role in HB development (Evers et al., Reference Evers, Gaspar, Kloor, Bozukova, Kadmon, Keller, Sutter and Moog2012; Flanagan et al., Reference Flanagan, Vincan and Phesse2019; Giardiello et al., Reference Giardiello, Petersen, Brensinger, Luce, Cayouette, Bacon, Booker and Hamilton1996; Perugorria et al., Reference Perugorria, Olaizola, Labiano, Esparza-Baquer, Marzioni, Marin, Bujanda and Banales2019; Thomas et al., Reference Thomas, Pritchard, Davidson, McKiernan, Grundy and de Ville de Goyet2003).

Here we report monozygotic twins from a FAP family who suffered from HB asynchronously. Whole exome sequencing and subsequent analysis of their tumors uncovered that microchimerism might occur during the development of HB at a very early stage of life.

Materials and Methods

Clinicopathological Data and Tumor Specimens

A family tree was constructed using a free online pedigree tool. Posttherapeutic (preoperational chemotherapy) specimens (C1) were obtained from Infant-1, whereas only postchemotherapeutic/operational specimens (C2) were obtained from Infant-2 since an emergency operation was performed due to signs of severe hemorrhage during preoperative chemotherapy. The DNA from histologically confirmed tumors were extracted using either a GeneRead DNA FFPE kit (Qiagen, Hilden, Germany) or QIAmp DNA Mini Kit (Qiagen). The tumor cellularity of each tumor specimen was estimated by a certified pathologist (K.I.).

Germline Mutation Analysis

Genomic DNA was extracted from either peripheral blood mononuclear cells (PBMCs) or oral mucosal samples from the twins, their parents, and all siblings (three sisters) of the father using the standard phenol extraction/purification procedure (Sato et al., Reference Sato, Tanigami, Yamakawa, Akiyama, Kasumi, Sakamoto and Nakamura1990). The coding exons in APC were amplified with M13-tailed target-specific primers and the PCR products were sequenced on an Applied Biosystems 3730xl DNA Analyzer using the BigDye Direct Cycle Sequencing Kit (Thermo Fisher Scientific, Waltham, MA) (Yamaguchi et al., Reference Yamaguchi, Nagayama, Shimizu, Komura, Yamaguchi, Shibuya, Arai, Hatakeyama, Ikenoue, Ueno, Miyano, Imoto and Furukawa2016). The primer sequences are available upon request.

Whole Exome Sequencing (WES)

For WES, we isolated DNA located in protein-coding exons using SureSelect Human All Exon v6 kits according to the manufacturer’s instructions (Agilent Technology, Santa Clara, CA). Captured DNA fragments were sequenced using a HiSeq 2500 (Illumina, San Diego, CA) with a standard 125 bp-paired end read protocol. Reads mapping and further analyses were performed using Genomon2 (v.2.6.3), which is an in-house pipeline constructed at the Human Genome Center based at The University of Tokyo (https://github.com/Genomon/genomon). Briefly, sequence reads were aligned to the human reference genome hg19 using Burrows–Wheeler Aligner-MEM (v.0.5.10) and bam files were created for data processing. For the Genomon2 pipeline analysis, the default settings were used for each parameter, and the EBCall (Empirical Baysian mutation Calling) algorithm was used to discriminate somatic mutations from sequencing errors (Shiraishi et al., Reference Shiraishi, Sato, Chiba, Okuno, Nagata, Yoshida, Shiba, Hayashi, Kume, Homma, Sanada, Ogawa and Miyano2013). Finally, SNVs (single nucleotide variants) and short indels were identified using the following procedure: (1) remove reads resulting from strand-specific sequencing errors (0.2 ≤ ‘strandRatio_tumor’ ≤ 0.8); (2) remove intronic and UTR (untranslated region) variants; (3) read depth ‘depth_tumor/normal’ ≥ 50; (4) variant allele frequency ‘misRate_tumor’ ≥ 0.05; and (5) manual review of the aligned reads using the Integrative Genomics Viewer (IGV).

Molecular Assessment of the Wnt Signaling Pathway

RNA-seq analysis was performed using RNA extracted from the tumorous and nontumorous liver tissues in C2. RNA was extracted from the tissues using RNeasy Plus Mini Kit (Qiagen), and the quality and quantity of RNA were calculated using the Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA). RNA sequencing libraries were prepared using the TruSeq RNA Access Library Prep Kit according to the manufacturer’s protocol (Illumina). Sequencing was performed with paired-end reads of 100 bp on an Illumina HiSeq 2500 platform. Read alignment and counting were performed using GenomonExpression (v.0.5.0), which is an in-house pipeline (https://github.com/Genomon-Project/GenomonExpression).

The expression of β-catenin in HBs was evaluated by immunohistochemical staining using an anti-β-catenin antibody (#9582, Cell Signaling Technology, Danvers, MA). The sections were deparaffinized with xylene. After antigen retrieval (10 mM sodium citrate buffer for 10 min at 115ºC), non-specific binding was blocked with goat serum, followed by overnight incubation in the antibodies (dilution 1:200) at 4ºC. After washing, the tissue-antibody reaction was visualized using the EnVision+ System (Agilent Technologies). Hematoxylin was used for nuclear counterstaining.

Digital PCR

Variant allele frequency (VAF) of somatic mutations in tissue and plasma were quantified by means of digital PCR (dPCR). Specific primer pairs of mutations identified by WES from tumors were designed and obtained from OTS-Probes, which is a dPCR probe library that increases the primer and probe annealing specificity with modified bases for amplification of short fragments (Quantdetect Inc., Tokyo, Japan). The probe sequence was labeled either by FAM or VIC fluorescent dyes for wild and mutant types, respectively. Both dyes were quenched by a Blackhole quencher (BQ) dye. The extracted DNA from FFPE and infant plasma were subjected to dPCR reactions in a QuantStudio 3D system (Thermno) and detected with originally developed primer/probe sets (Iwaya et al., Reference Iwaya, Endo, Takahashi, Tokino, Sasaki and Nishizuka2021) (Quantdetect).

Results

Family History

Although two tumor-related deaths and colon polyps were noted in the pedigree, there was no previous history of HB in any family members except for the twins (Figure 1A). All tumor-related lesions were from gastrointestinal origins, including colon and gastric cancer. The paternal grandfather (II-1) passed away due to a traffic accident, but he had been previously treated for colon polyps. The father (III-6) developed gastric and colon cancer in his 20s and underwent curative surgery for these tumors. Periodic follow-up examinations revealed no recurrent disease for the past 7 years.

Figure 1. Genetic background of the twins. (A) Family pedigree. (B) Structure of the APC gene, including functional domains and introns. The position of the identified germline mutation is indicated by the red line. (C) Sanger sequence histogram of the affected infants and their parents. Note: CC, colon cancer; CP, colon polyps; GC, gastric cancer; HB, hepatoblastoma.

Germline Mutations

We carried out genetic analysis of the father’s DNA and identified a 7-base pair-deletion (NM_000038.6, c.3231_3237delTTATACT(p. Val1077fs) in the coding region of APC. Located in the β-catenin binding region of APC, this variation is predicted to cause a frameshift, leading to the production of truncated APC protein lacking the β-catenin binding region (Figure 1B). Subsequent cascade analysis identified the variant in the twins but not in the mother or father’s siblings by Sanger sequencing (Figure 1A, 1C).

Clinical Course of Infant-1 (C1 Tumor)

The twins were born by cesarean section from a 28-year-old mother at 38 weeks gestation. No parental or neonatal problems occurred during the delivery course. They were clinically dichorionic-diamniotic gestational twins. The birth weight of the first baby was 2594 g and that of the second was 2326 g (Figure 2A and 2B). The infants were discharged in stable condition and exhibited a normal developmental course. However, the first of the twins gradually lost appetite and suffered abdominal distention by 12 months of age (Figure 2A). The parents took him to our hospital for consultation of his symptoms at 14 months of age. Physical examinations showed abdominal distention, particularly in the right quarter rib, where an elastic soft mass with the size of approximately 14 cm was palpable. An ultrasound examination detected a liver mass with the consistent size of 14 cm. Because of hyperechoic and nonuniform features of the mass content as well as the size, hepatoblastoma was suspected. Additional computed tomography (CT) scans showed a large, round, and smooth liver mass whose internal density was not uniform, with apparent invasion to the hilar plate that possibly originated from the posterior segment (PRETEXT I) (Brown et al., Reference Brown, Perilongo, Shafford, Keeling, Pritchard, Brock, Dicks-Mireaux, Phillips, Vos and Plaschkes2000). Blood laboratory examinations revealed slight elevation of white blood cells (14,600 /mL), anemia (Hb 8.4 g/dl), and a high level of AFP (33,961 ng/ml), which is a tumor marker of hepatoblastoma. A pathological examination of the tumor biopsy revealed that the tumor histology was a pure fetal type of HB (Supplementary Figure S1A). Based on this diagnosis, two cycles of preoperative chemotherapy (cisplatin/pirarubicin, CITA) were performed, which resulted in a marked reduction of the tumor volume to 9 cm in diameter. A right lobe hepatectomy was subsequently performed at 17 months of age. The surgical margin was negative for cancer and no detectable metastases to other organs were found in his abdominal cavity. The patient had a good postoperative course and underwent adjuvant chemotherapy 20 days after the surgery with one cycle of CITA followed by three cycles of low-CITA. No evidence of metastasis or recurrence has been observed for 7 years since the completion of adjuvant chemotherapy.

Figure 2. Clinical course of the affected infants. The C1 tumor was obtained by biopsy, whereas the C2 tumor was obtained by hepatectomy. (A) Infant-1. The red circle indicates the date of hepatectomy. (B) Infant-2. The red circle indicates the date of hepatectomy.

Clinical Course of Infant-2 (C2 Tumor)

Since the first of the twins (Infant-1) was diagnosed with HB, Infant-2 underwent an abdominal ultrasound examination for HB screening at 15 months of age (Figure 2B) despite not having any symptoms. No neoplastic lesions were detected in his liver at the time of screening. However, he was transported to the emergency department in our hospital at 21 months of age due to a sudden onset of respiratory distress with pallor on his face and subsequent loss of consciousness within a day. On admission, a pallor face and acrocyanosis of the extremities were noticed, and he suffered grade 3 unconsciousness based on the Japan Coma Scale (not awake with external stimulation). In addition, overall abdominal distention was observed, and physical examination detected a palpable and elastic soft mass on his right upper quadrant. Blood laboratory examinations showed a slight increase in the number of white blood cells (12,580 /μL), severe anemia (Hb, 6.9 g/dL), and moderately high AFP (2,127 ng/mL). An emergency CT scan demonstrated that his liver was occupied by a large mass with a diameter of 11 cm that had clear and smooth margins, a capsule-like structure, and heterogenous intensity. The CT scan images additionally depicted a low-density area at the center of the tumor, which strongly indicated tumor hemorrhage. Therefore, we did not perform a tumor biopsy for pathological diagnosis, but instead made a PRETEXT III diagnosis of suspected HB23 according to the images. To manage tumor removal without trisectionectomy, preoperative chemotherapy was given, including one cycle of low-CITA followed by two cycles of CITA. Cycles of the chemotherapy reduced the tumor volume down to 6.6 cm in diameter without additional major hemorrhage from the tumor. The patient then underwent a central bisegmentectomy of the liver at 24 months of age. Pathological examination of surgical specimens confirmed that the tumor was the pure fetal type of HB (Supplementary Figure S1B), which was the same histological type as his brother. Twenty days after hepatectomy, four cycles of CITA postoperative chemotherapy were initiated. No evidence of recurrence or metastasis has been observed over the 7 years since the surgery.

Involvement of Wnt Signaling Pathway in Tumor Development

An immunohistochemical evaluation of the β-catenin protein was performed on the tumor tissue. Both tumors (C1 and C2 tumors) showed positive staining in the cytoplasm of HB cells, but not adjacent normal cells (Supplementary Figure S2). Nuclear localization was occasionally seen in the C1 tumor, but was clearly evident in the C2 tumor. In addition, an RNA-seq analysis of the C2 tumor and the corresponding nontumorous tissues revealed that expression of ODAM, LGR5, and AXIN2, which are three direct target genes in the Wnt pathway, was markedly augmented in the tumor tissue compared to the nontumorous tissue. These data suggest that activation of the Wnt signaling pathway was involved in the development of the HB tumors.

Genetic Analysis of the HBs

We carried out WES on the tumor tissues of the twins (Infant-1 and Infant-2) and nontumorous DNA extracted from their peripheral blood. The average sequence coverage of each exon was 148 (Infant-1) and 182 (Infant-2) for the nontumorous DNA, and 150 (C1) and 196 (C2) for the tumor tissue respectively. Comparison of the germline SNVs between Infant-1 and Infant-2 confirmed that the twins were monozygotic.

We identified a total of eight and one somatic SNVs in C1 and C2 respectively. In the eight SNVs in C1, six were nonsynonymous variants, one was a synonymous variant, and the remaining one was a nonsense mutation. The SNV in C2 was a nonsense mutation.

No somatic mutation was shared in the two tumors. As for driver mutations, we identified an NRAS mutation, NM_002524.5, c.35G>A (p. Gly12Asp) in C1 (with a VAF of 7.1%), and an ACVR2A mutation, NM_001616.5, c.16A>T (p. Lys6Ter) in C2 (with a VAF of 14.2%) (Table 1).

Table 1. Representative somatic mutations that may serve as tumor development trackers

Note: VAF, variant allele frequency; InterVar, https://wintervar.wglab.org/about.php; LP, Likely pathogenic; US, Uncertain significance; COSMIC, Catalogue of Somatic Mutations in Cancer database; NA, Not applicable.

To determine whether the mutations were a late event in the evolution of each HB, we carried out multiregional analysis of the NRAS and ACVR2A mutations by digital PCR analysis. DNA were extracted from six regions of the C1 tumor and seven regions from the C2 tumor, and the mutations were determined using the mutant-specific probe sets. As shown in Figure 3A, the NRAS mutation was detected with different frequencies; 0.2% (region-1), 0.03% (region-2), 0.02% (region-3), 0.2% (region-4), 0.16% (region-5), and 0.14% (region-6) in the C1 tumor, implying intratumorous heterogeneity. Surprisingly, we also found the NRAS mutation in the seven regions of the C2 tumor with the following frequencies: 0.13% (region-5), 0.16% (region-6), 0.09% (region-7), 0.18% (region-8), 0.10% (region-10), 0.09% (region-11), and 0.11% (region-12) (Figure 3B). Similarly, the ACVR2A mutation was identified not only in the C2 tumor (0.42% in region-5, 0.19% in region-6, 2.24% in region-7, 4.06% in region-8, 7.10% in region-10, 1.10% in region-11, and 2.73% in region-12), but also in four of the six regions of the C1 tumor (0.03% in region-1, 0.02% in region-2, 0.4% in region-4, and 0.03% in region-6; Figure 3A and 3B). It is extremely unlikely that the two identical somatic mutations occurred in independent tumor clones after birth. Hence, these results suggest that: (1) implantation of tumor clone(s) occurred through utero-placental blood circulation systems, giving rise to microchimerism of both tumors; and (2) the initiation of HB tumorigenesis likely occurred during the fetal stage after organ development of the liver.

Figure 3. Rare variant detection from multiregional specimens. (A) A macroscopic slice of the Infant-1 tumor (C1). DNA was extracted from six different regions. The variant allele frequency (VAF) of NRAS/ACVR2A in each region is indicated in a bar graph. (B) Macroscopic slices of the Infant-2 tumor (C2). DNA was extracted from seven different regions. The VAF of NRAS/ACVR2A in each region is indicated in a bar graph.

Circulating Tumor DNAs

To investigate the usefulness of the driver mutations for their surveillance, we tested their detection in the ctDNA. Digital PCR analysis identified the ACVR2A mutation in prechemotherapeutic plasma of Infant-2 at a VAF of 1.31%, but not in postoperative plasma. As expected, the NRAS mutation was not detected in postoperative plasma of Infant-1 (no preoperative plasma available). These mutations were not detected in DNA extracted from PBMCs of both infants. Neither mutation was detected in the parents’ plasma DNA (Table 2).

Table 2. Variant allele frequencies (%) of ctDNA

Note: aIn the parenthesis, the numerator is the number of positive reaction units for mutations, whereas the denominator is the number of total (i.e., positive and negative) reaction units for mutations. PBMC, peripheral blood mononuclear cells.

Discussion

HB is a rare embryonal malignant tumor arising from the liver yet is the most common malignant tumor in infants and children. It commonly occurs in the first few years of life and predominantly in boys (Giardiello et al., Reference Giardiello, Offerhaus, Krush, Booker, Tersmette, Mulder, Kelley and Hamilton1991; Giardiello et al., Reference Giardiello, Petersen, Brensinger, Luce, Cayouette, Bacon, Booker and Hamilton1996; Trobaugh-Lotrario et al., Reference Trobaugh-Lotrario, Lopez-Terrada, Li and Feusner2018). FAP is an autosomal dominant disorder forming thousands of colorectal adenomas during adolescence and young adulthood, which is caused by a germline variant of the APC tumor suppressor gene. Although clinical characteristics of HB-affected individuals with APC mutations do not seem to be distinct from those with sporadic HB, it has been estimated that individuals with a FAP background have a 750 to 7500-fold higher risk of developing HB than the general population (Aretz et al., Reference Aretz, Koch, Uhlhaas, Friedl, Propping, von Schweinitz and Pietsch2006; Giardiello et al., Reference Giardiello, Petersen, Brensinger, Luce, Cayouette, Bacon, Booker and Hamilton1996; Hughes & Michels, Reference Hughes and Michels1992). Although investigators have reported APC mutations in HB patients (Aretz et al., Reference Aretz, Koch, Uhlhaas, Friedl, Propping, von Schweinitz and Pietsch2006; Evers et al., Reference Evers, Gaspar, Kloor, Bozukova, Kadmon, Keller, Sutter and Moog2012; Gupta et al., Reference Gupta, Sheridan, Towbin, Geller, Tiao and Bove2013; Ponz de Leon et al., Reference Ponz de Leon, Bianchini, Reggiani-Bonetti, Pedroni, Di Gregorio, Merighi, Rossi, Magnani, Domati and Cacciari2014; Thomas et al., Reference Thomas, Pritchard, Davidson, McKiernan, Grundy and de Ville de Goyet2003; Yang et al., Reference Yang, Sisson, Gupta, Tiao and Geller2018), a concomitant genetic analysis of HB, or HB from twins in FAP pedigrees, has not been fully performed.

It has been reported that HBs have fewer somatic mutations at the whole-genome level than most adult tumors do, including those of adult hepatocellular carcinomas (Eichenmuller et al., Reference Eichenmuller, Trippel, Kreuder, Beck, Schwarzmayr, Haberle, Cairo, Leuschner, von Schweinitz, Strom and Kappler2014; Fujimoto et al., Reference Fujimoto, Furuta, Totoki, Tsunoda, Kato, Shiraishi, Tanaka, Taniguchi, Kawakami, Ueno, Gotoh, Ariizumi, Wardell, Hayami, Nakamura, Aikata, Arihiro, Boroevich, Abe and Nakagawa2016; Jia et al., Reference Jia, Dong, Jing, Xu, Wang, Chen, Li, Huang, Zhang, Zhang, Liu, Zheng, Xia, Wang, Dong and He2014; Nagae et al., Reference Nagae, Yamamoto, Fujita, Fujita, Nonaka, Umeda, Fukuda, Tatsuno, Maejima, Hayashi, Kurihara, Kojima, Hishiki, Watanabe, Ida, Yano, Hiyama, Tanaka, Inoue and Hiyama2021). The activation of the Wnt signaling pathway with an alteration of CTNNB1 has been suggested to be the genetic event observed most frequently (15–87%) in HB (Lopez-Terrada et al., Reference Lopez-Terrada, Gunaratne, Adesina, Pulliam, Hoang, Nguyen, Mistretta, Margolin and Finegold2009; Purcell et al., Reference Purcell, Childs, Maibach, Miles, Turner, Zimmermann and Sullivan2011; Taniguchi et al., Reference Taniguchi, Roberts, Aderca, Dong, Qian, Murphy, Nagorney, Burgart, Roche, Smith, Ross and Liu2002). The majority of alterations are point mutations or deletions in exon 3 of CTNNB1, which results in the impairment of phosphorylation of the β-catenin protein (Rubinfeld et al., Reference Rubinfeld, Albert, Porfiri, Fiol, Munemitsu and Polakis1996). The activity of the β-catenin protein is regulated by the destruction complex composed of AXIN-1, GSK-3β, CK-1, and APC proteins, which degrade other proteins through phosphorylation and subsequent ubiquitination (Clevers, Reference Clevers2006; Ng et al., Reference Ng, Dazert, Boldanova, Coto-Llerena, Nuciforo, Ercan, Suslov, Meier, Bock, Schmidt, Ketterer, Wang, Wieland, Matter, Colombi, Piscuoglio, Terracciano, Hall and Heim2022; Taniguchi et al., Reference Taniguchi, Roberts, Aderca, Dong, Qian, Murphy, Nagorney, Burgart, Roche, Smith, Ross and Liu2002). Although the pathogenic germline variant (APC, c.3231_3237del) was found in C1 and C2 tumors, no second hit mutations were identified in the tumors. Since immunohistochemical analysis revealed accumulated β-catenin protein in the tumors, it is natural to think that the degradation of β-catenin was abrogated in the tumor cells. Thus, there may be undetectable somatic alteration(s) in the regulatory or deep intronic regions, or structural alterations, in the APC gene. Alternatively, other nongenetic events, such as epigenetic changes, could be involved in the loss of APC function in the cells.

In addition to the APC mutation, tumor cells harbored additional driver mutations, including the NRAS mutation in C1 and the ACVR2A mutation in C2. These mutations are not likely to be their early event, since the VAF was not high (i.e., 7.1% for NRAS and 14.2% for ACVR2A). However, tumor cells harboring these mutations might have impairments in growth and/or development. Notably, ACVR2A mutations are frequently identified in NASH-HCC compared to HCC caused by other etiologies (Pinyol et al., Reference Pinyol, Torrecilla, Wang, Montironi, Pique-Gili, Torres-Martin, Wei-Qiang, Willoughby, Ramadori, Andreu-Oller, Taik, Lee, Moeini, Peix, Faure-Dupuy, Riedl, Schuehle, Oliveira, Alves and Llovet2021). In addition, it has been reported that ACVR2A and ACVR1B mutations are frequently detected in colorectal cancer and that these genes play an important role in the carcinogenesis as tumor suppressors (Takeda et al., Reference Takeda, Kataoka, Nakayama, Ali, Oshima, Yamamoto, Park, Takegami, An, Jenkins, Copeland and Oshima2019).

To assess the clonality of each tumor, we decided to use these mutations for multiregional mutational profiling using dPCR. In contrast to the WES approach, dPCR is intended to detect extremely rare variants down to the range of 0.001% VAF, for which an NGS assay would not be able to detect mutations (Diaz & Bardelli, Reference Diaz and Bardelli2014). With the dPCR technology, the multiregional profiling revealed that both mutations were present in both tumors. Interestingly, among a total of 13 regions of tumors, the VAFs of NRAS were consistently higher in C1 tumor regions compared to those of the C2 tumor regions, whereas VAFs of ACVR2A were higher in C2 tumor regions compared to C1. However, the VAF of these mutations seemed too low to be considered a truncal mutation (Frankell et al., Reference Frankell, Dietzen, Al Bakir, Lim, Karasaki, Ward, Veeriah, Colliver, Huebner, Bunkum, Hill, Grigoriadis, Moore, Black, Liu, Thol, Pich, Watkins, Naceur-Lombardelli and Swanton2023), suggesting that these mutations may not play a central role in the development of HBs. A potential explanation for these findings is that the APC germline mutation has impaired degradation of β-catenin, which could drive cell division susceptibility for malignant transformation without additional somatic mutations. However, since no other paternal family member had HB with the same APC germline mutation, there should be nongenetic triggers for HB development in the twins.

As another index of the presence of tumor in the body, a high VAF ctDNA (i.e., 1.31% as ctDNA) for ACVR2A was found in the plasma of prehepatectomy Infant-2 by a dPCR assay. The ACVR2A mutation seemed to have disappeared after chemotherapy and remained undetectable during the postoperative period. These results further support the finding that the ACVR2A mutation is derived from the C2 tumor, and not from the germline. Here, it should again be noted that the ACVR2A mutation was found in the C1 tumor, which was physically separated from the C2 tumor circulation system. It is extremely unlikely that these tumors arose independently with the same somatic mutation. Therefore, as our multiregional dPCR assay demonstrated, it can be reasonably hypothesized that the tumor cells implanted via the maternal and fetal circulation systems during which time the tumors were not yet visible using routine detection modalities (Shatara et al., Reference Shatara, Xavier, Dombkowski, Cukovic, Poulik, Altinok, Ge and Taub2019). The undetectable tumor cells continued to proliferate after birth, and both tumors subsequently became detectable during infancy resulting in two independent chimeric tumors. Indeed, it has been reported that blood chimerism, which is defined as a blood cell genotype, contains different cells derived from two or more distinct zygotes in monochronic dizygotic twins, suggesting that mixed zygotes may result in microchimerism in organs (Lee et al., Reference Lee, Yoon, Ko, Seong, Park, Choi and Oh2014). Although limited to monochorionic dizygotic twins, a systematic review by Peters et al. (Reference Peters, Konig, Verhoeven, Schats, Mijatovic, Ket and Lambalk2017) reported that chimerism was demonstrable in >90% of 31 twins they investigated. The authors suggest that, as most of the chimeric twins were discovered by accident, the phenomena may be far more common than originally speculated. In fact, bidirectional transplacental cell trafficking between mother and fetus during pregnancy has been one of the emerging topics in studying microchimerism, which is defined as an organism being made up of more than one genetically distinct individual (Boddy et al., Reference Boddy, Fortunato, Wilson Sayres and Aktipis2015; Comitre-Mariano et al., Reference Comitre-Mariano, Martinez-Garcia, Garcia-Galvez, Paternina-Die, Desco, Carmona and Gomez-Gaviro2022). In the present study, the microchimerism was confirmed only by tumors of the twins. The fact that both tumors acquired microchimerism consisting of somatic mutations before birth suggests that the HB development initiated during pregnancy. Moreover, the fact that the tumor cells should have been trafficking systemically in the fetus, but only arose in the liver, suggests that some tissue environmental factors may accelerate tumor progression. Although a premature alloimmune system contributing to the establishment of tumor implantation has been suggested (Arakawa et al., Reference Arakawa, Ichikawa, Kubo, Motoi, Kumamoto, Nakajima, Yonemori, Noguchi, Sunami, Shiraishi, Kakishima, Yoshida, Hishiki, Kawakubo, Kuroda, Kiyokawa, Yamada, Yanaihara, Takahashi and Ogawa2021), our findings further imply that an organ-specific cell-trafficking environment may exist during HB development.

Overall, we propose a potential order explaining the development of HBs in the monozygotic twins as follows: (1) the germline APC mutation gave rise to a susceptible genetic background to each tumor; (2) these tumors separately acquired different nongenetic triggers of tumorigenesis during pregnancy; (3) a small portion of each tumor traveled via maternal circulation and implanted in the liver of the counterpart (the implantation continued until birth); and (4) the triggered tumor cells kept proliferating after birth.

This study has the following limitations. Germline mutations were only accessible within two generations since the other three affected individuals (grandparents of the twins) had passed away. Due to clinical priority, only a limited amount of prechemotherapeutic biopsy specimens was available from Infant-1 (C1), whereas posthepatectomy specimens were all postchemotherapeutic tissues (C1). Specimens from Infant-2 were only posthepatectomy (C2). As a result, the quality and reproducibility for sequencing analysis might have been restricted. In fact, nongenetic alterations have not been fully assessed. Blood samples were not periodically obtained due to technical difficulties and infant conditions. Although two SNVs were used for cellular migration tracking, the mechanism of metachronous tumor development remains unknown. Finally, we were unable to examine the microchimerism in tissues other than liver, and we were not able to assess those of their mother.

In conclusion, we have identified somatic driver mutations, an NRAS activating mutation and a loss of function ACVR2A mutation, in HBs that developed in monozygotic twins with a pathogenic germline APC variant. The importance of these driver mutations in the development of HB remains to be elucidated. However, our data suggest that blood and cells of the twins could be mutually transmittable via the utero-placental maternal circulation; and HB tumorigenesis began during the fetal stage after organ development of the liver. We recommend performing periodical surveillance in twins with tumors to assess for malignancy at young ages if one of the twins exhibits a malignant tumor at that age. These findings and recommendations are based on the evidence for tumor cell microchimerism and shared tumor-development in the utero-placental environment.

Supplementary material

To view supplementary material for this article, please visit https://doi.org/10.1017/thg.2025.10019

Acknowledgments

We thank all patients and family members who participated in the study. We also thank Miyuki Ikeda and Seira Hatakeyama (The University of Tokyo) for technical assistance; Taishi Hirai, Mikiya Endo, and Kotaro Oyama (Iwate Medical University) for clinical analysis; and Tamotsu Sugai (Iwate Medical University) for pathological advice. The super-computing resource was provided by the Human Genome Center, The Institute of Medical Science, The University of Tokyo (http://sc.hgc.jp/shirokane.html).

Financial support

This work was supported by JSPS KAKENHI (Grant Numbers of 16H01578 and 21K07223) and a grant from the International Joint Usage/Research Center, The Institute of Medical Science, The University of Tokyo.

Ethical standards

The authors assert that all procedures contributing to this work comply with the ethical standards of the relevant national and institutional committees on human experimentation and with the Helsinki Declaration of 1975, as revised in 2008. This study was approved by both the Iwate Medical University (HGH28-13; HG2020-028) and the University of Tokyo (27-37-1015; 2020-36-1001).

Competing of interests

Nishizuka reports grant/research support from Geninus, LSI Medience, Quantdetect Inc., Iwate Industrial Research Institute, Thermo Fisher Scientific; Honorarium from MSD, Finggal-link; consultation fee from CLEA Japan and Hitachi high-tech; and CEO as well as a stockholder of Quantdetect, Inc.

Publication ethics

All authors have approved the final version of the manuscript and agreed to publish this work. Individual contributions are as follows: AA, SM, YF, SSN desined the study; AA, KY, KT, YA, YH, MM, HN, KI, TI collected clinical data;AA, KY, KT, ES, SI, SM, YF performed sequencing and immunohistochemistry; AA, TI, and SSN performed digital PCR; AA, YF, SSN draft manuscript; AA and SSN prepared Figures 1, 2 and 3; AA, KY, ES, SI prepared Tables 1 and 2; AA, TI, SSN prepared Supplementary Table 1; KI prepared Supplementary Figures 1 and 2; Critical discussion, AA, KY, KT, YF, SSN.

References

Arakawa, A., Ichikawa, H., Kubo, T., Motoi, N., Kumamoto, T., Nakajima, M., Yonemori, K., Noguchi, E., Sunami, K., Shiraishi, K., Kakishima, H., Yoshida, H., Hishiki, T., Kawakubo, N., Kuroda, T., Kiyokawa, T., Yamada, K., Yanaihara, N., Takahashi, K., ¼ Ogawa, C. (2021). Vaginal transmission of cancer from mothers with cervical cancer to infants. New England Journal of Medicine, 384, 4250. https://doi.org/10.1056/NEJMoa2030391 CrossRefGoogle ScholarPubMed
Aretz, S., Koch, A., Uhlhaas, S., Friedl, W., Propping, P., von Schweinitz, D., & Pietsch, T. (2006). Should children at risk for familial adenomatous polyposis be screened for hepatoblastoma and children with apparently sporadic hepatoblastoma be screened for APC germline mutations? Pediatric Blood & Cancer, 47, 811818. https://doi.org/10.1002/pbc.20698 CrossRefGoogle ScholarPubMed
Boddy, A. M., Fortunato, A., Wilson Sayres, M., & Aktipis, A. (2015). Fetal microchimerism and maternal health: A review and evolutionary analysis of cooperation and conflict beyond the womb. Bioessays, 37, 11061118. https://doi.org/10.1002/bies.201500059 CrossRefGoogle ScholarPubMed
Brown, J., Perilongo, G., Shafford, E., Keeling, J., Pritchard, J., Brock, P., Dicks-Mireaux, C., Phillips, A., Vos, A., & Plaschkes, J. (2000). Pretreatment prognostic factors for children with hepatoblastoma ¾- Results from the International Society of Paediatric Oncology (SIOP) study SIOPEL 1. European Journal of Cancer, 36, 14181425. https://doi.org/10.1016/s0959-8049(00)00074-5 CrossRefGoogle ScholarPubMed
Clevers, H. (2006). Wnt/beta-catenin signaling in development and disease. Cell, 127, 469480. https://doi.org/10.1016/j.cell.2006.10.018 CrossRefGoogle ScholarPubMed
Comitre-Mariano, B., Martinez-Garcia, M., Garcia-Galvez, B., Paternina-Die, M., Desco, M., Carmona, S., & Gomez-Gaviro, M. V. (2022). Feto-maternal microchimerism: Memories from pregnancy. iScience, 25, 103664. https://doi.org/10.1016/j.isci.2021.103664 CrossRefGoogle ScholarPubMed
Diaz, L. A Jr., & Bardelli, A. (2014). Liquid biopsies: Genotyping circulating tumor DNA. Journal of Clinical Oncology, 32, 579586. https://doi.org/10.1200/JCO.2012.45.2011 CrossRefGoogle ScholarPubMed
Eichenmuller, M., Trippel, F., Kreuder, M., Beck, A., Schwarzmayr, T., Haberle, B., Cairo, S., Leuschner, I., von Schweinitz, D., Strom, T. M., & Kappler, R. (2014). The genomic landscape of hepatoblastoma and their progenies with HCC-like features. Journal of Hepatology, 61, 13121320. https://doi.org/10.1016/j.jhep.2014.08.009 CrossRefGoogle ScholarPubMed
Evers, C., Gaspar, H., Kloor, M., Bozukova, G., Kadmon, M., Keller, M., Sutter, C., & Moog, U. (2012). Hepatoblastoma in two siblings and familial adenomatous polyposis: Causal nexus or coincidence? Familial Cancer, 11, 529533. https://doi.org/10.1007/s10689-012-9538-2 CrossRefGoogle ScholarPubMed
Flanagan, D. J., Vincan, E., & Phesse, T. J. (2019). Wnt signaling in cancer: Not a binary ON:OFF switch. Cancer Research, 79, 59015906. https://doi.org/10.1158/0008-5472.CAN-19-1362 CrossRefGoogle Scholar
Frankell, A. M., Dietzen, M., Al Bakir, M., Lim, E. L., Karasaki, T., Ward, S., Veeriah, S., Colliver, E., Huebner, A., Bunkum, A., Hill, M. S., Grigoriadis, K., Moore, D. A., Black, J. R. M., Liu, W. K., Thol, K., Pich, O., Watkins, T. B. K., Naceur-Lombardelli, C., ¼ Swanton, C. (2023). The evolution of lung cancer and impact of subclonal selection in TRACERx. Nature, 616, 525533. https://doi.org/10.1038/s41586-023-05783-5 CrossRefGoogle ScholarPubMed
Fujimoto, A., Furuta, M., Totoki, Y., Tsunoda, T., Kato, M., Shiraishi, Y., Tanaka, H., Taniguchi, H., Kawakami, Y., Ueno, M., Gotoh, K., Ariizumi, S., Wardell, C. P., Hayami, S., Nakamura, T., Aikata, H., Arihiro, K., Boroevich, K. A., Abe, T., ¼ Nakagawa, H. (2016). Whole-genome mutational landscape and characterization of noncoding and structural mutations in liver cancer. Nature Genetics, 48, 500509. https://doi.org/10.1038/ng.3547 CrossRefGoogle ScholarPubMed
Giardiello, F. M., Offerhaus, G. J., Krush, A. J., Booker, S. V., Tersmette, A. C., Mulder, J. W., Kelley, C. N., & Hamilton, S. R. (1991). Risk of hepatoblastoma in familial adenomatous polyposis. Journal of Pediatrics, 119, 766768. https://doi.org/10.1016/s0022-3476(05)80297-5 CrossRefGoogle ScholarPubMed
Giardiello, F. M., Petersen, G. M., Brensinger, J. D., Luce, M. C., Cayouette, M. C., Bacon, J., Booker, S. V., & Hamilton, S. R. (1996). Hepatoblastoma and APC gene mutation in familial adenomatous polyposis. Gut, 39, 867869. https://doi.org/10.1136/gut.39.6.867 CrossRefGoogle ScholarPubMed
Gupta, A., Sheridan, R. M., Towbin, A., Geller, J. I., Tiao, G., & Bove, K. E. (2013). Multifocal hepatic neoplasia in 3 children with APC gene mutation. American Journal of Surgical Pathology, 37, 10581066. https://doi.org/10.1097/PAS.0b013e31828aeb18 CrossRefGoogle ScholarPubMed
Hiyama, E., Hishiki, T., Watanabe, K., Ida, K., Ueda, Y., Kurihara, S., Yano, M., Hoshino, K., Yokoi, A., Takama, Y., Nogami, Y., Taguchi, T., Mori, M., Kihira, K., Miyazaki, O., Fuji, H., Honda, S., Iehara, T., Kazama, T., ¼ Yoshimura, K. (2020). Outcome and late complications of hepatoblastomas treated using the Japanese Study Group for Pediatric Liver Tumor 2 Protocol. Journal of Clinical Oncology, 38, 24882498. https://doi.org/10.1200/JCO.19.01067 CrossRefGoogle Scholar
Hughes, L. J., & Michels, V. V. (1992). Risk of hepatoblastoma in familial adenomatous polyposis. American Journal of Medical Genetics, 43, 10231025. https://doi.org/10.1002/ajmg.1320430621 CrossRefGoogle ScholarPubMed
Iwaya, T., Endo, F., Takahashi, F., Tokino, T., Sasaki, Y., & Nishizuka, S. S. (2021). Frequent tumor burden monitoring of esophageal squamous cell carcinoma with circulating tumor DNA using individually designed digital polymerase chain reaction. Gastroenterology, 160, 463465. https://doi.org/10.1053/j.gastro.2020.09.035 CrossRefGoogle ScholarPubMed
Jia, D., Dong, R., Jing, Y., Xu, D., Wang, Q., Chen, L., Li, Q., Huang, Y., Zhang, Y., Zhang, Z., Liu, L., Zheng, S., Xia, Q., Wang, H., Dong, K., & He, X. (2014). Exome sequencing of hepatoblastoma reveals novel mutations and cancer genes in the Wnt pathway and ubiquitin ligase complex. Hepatology, 60, 16861696. https://doi.org/10.1002/hep.27243 CrossRefGoogle ScholarPubMed
Katzenstein, H. M., Langham, M. R., Malogolowkin, M. H., Krailo, M. D., Towbin, A. J., McCarville, M. B., Finegold, M. J., Ranganathan, S., Dunn, S., McGahren, E. D., Tiao, G. M., O’Neill, A. F., Qayed, M., Furman, W. L., Xia, C., Rodriguez-Galindo, C., & Meyers, R. L. (2019). Minimal adjuvant chemotherapy for children with hepatoblastoma resected at diagnosis (AHEP0731): A Children’s Oncology Group, multicentre, phase 3 trial. Lancet Oncology, 20, 719727. https://doi.org/10.1016/S1470-2045(18)30895-7 CrossRefGoogle ScholarPubMed
Kinzler, K. W., Nilbert, M. C., Su, L. K., Vogelstein, B., Bryan, T. M., Levy, D. B., Smith, K. J., Preisinger, A. C., Hedge, P., & McKechnie, D. (1991). Identification of FAP locus genes from chromosome 5q21. Science, 253, 661665. https://doi.org/10.1126/science.1651562 CrossRefGoogle ScholarPubMed
Lee, H. J., Yoon, S. C., Ko, J. M., Seong, M. W., Park, S. S., Choi, J. S., & Oh, S. K. (2014). Monochorionic dizygotic twins with discordant sex and confined blood chimerism. European Journal of Pediatrics, 173, 12491252. https://doi.org/10.1007/s00431-014-2312-8 CrossRefGoogle ScholarPubMed
Lopez-Terrada, D., Gunaratne, P. H., Adesina, A. M., Pulliam, J., Hoang, D. M., Nguyen, Y., Mistretta, T. A., Margolin, J., & Finegold, M. J. (2009). Histologic subtypes of hepatoblastoma are characterized by differential canonical Wnt and Notch pathway activation in DLK+ precursors. Human Pathology, 40, 783794. https://doi.org/10.1016/j.humpath.2008.07.022 CrossRefGoogle ScholarPubMed
Malogolowkin, M. H., Katzenstein, H. M., Meyers, R. L., Krailo, M. D., Rowland, J. M., Haas, J., & Finegold, M. J. (2011). Complete surgical resection is curative for children with hepatoblastoma with pure fetal histology: A report from the Children’s Oncology Group. Journal of Clinical Oncology, 29, 33013306. https://doi.org/10.1200/JCO.2010.29.3837 CrossRefGoogle ScholarPubMed
Nagae, G., Yamamoto, S., Fujita, M., Fujita, T., Nonaka, A., Umeda, T., Fukuda, S., Tatsuno, K., Maejima, K., Hayashi, A., Kurihara, S., Kojima, M., Hishiki, T., Watanabe, K., Ida, K., Yano, M., Hiyama, Y., Tanaka, Y., Inoue, T., ¼ Hiyama, E. (2021). Genetic and epigenetic basis of hepatoblastoma diversity. Nature Communication, 12, 5423. https://doi.org/10.1038/s41467-021-25430-9 CrossRefGoogle ScholarPubMed
Ng, C. K. Y., Dazert, E., Boldanova, T., Coto-Llerena, M., Nuciforo, S., Ercan, C., Suslov, A., Meier, M. A., Bock, T., Schmidt, A., Ketterer, S., Wang, X., Wieland, S., Matter, M. S., Colombi, M., Piscuoglio, S., Terracciano, L. M., Hall, M. N., & Heim, M. H. (2022). Integrative proteogenomic characterization of hepatocellular carcinoma across etiologies and stages. Nature Communications, 13, 2436. https://doi.org/10.1038/s41467-022-29960-8 CrossRefGoogle ScholarPubMed
Nishisho, I., Nakamura, Y., Miyoshi, Y., Miki, Y., Ando, H., Horii, A., Koyama, K., Utsunomiya, J., Baba, S., & Hedge, P. (1991). Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients. Science, 253, 665669. https://doi.org/10.1126/science.1651563 CrossRefGoogle ScholarPubMed
Perilongo, G., Maibach, R., Shafford, E., Brugieres, L., Brock, P., Morland, B., de Camargo, B., Zsiros, J., Roebuck, D., Zimmermann, A., Aronson, D., Childs, M., Widing, E., Laithier, V., Plaschkes, J., Pritchard, J., Scopinaro, M., MacKinlay, G., & Czauderna, P. (2009). Cisplatin versus cisplatin plus doxorubicin for standard-risk hepatoblastoma. New England Journal of Medicine, 361, 16621670. https://doi.org/10.1056/NEJMoa0810613 CrossRefGoogle ScholarPubMed
Perugorria, M. J., Olaizola, P., Labiano, I., Esparza-Baquer, A., Marzioni, M., Marin, J. J. G., Bujanda, L., & Banales, J. M. (2019). Wnt-beta-catenin signalling in liver development, health and disease. Nature Reviews Gastroenterology & Hepatology, 16, 121136. https://doi.org/10.1038/s41575-018-0075-9 CrossRefGoogle ScholarPubMed
Peters, H. E., Konig, T. E., Verhoeven, M. O., Schats, R., Mijatovic, V., Ket, J. C., & Lambalk, C. B. (2017). Unusual twinning resulting in chimerism: A systematic review on monochorionic dizygotic twins. Twin Research and Human Genetics, 20, 161168. https://doi.org/10.1017/thg.2017.4 CrossRefGoogle ScholarPubMed
Pinyol, R., Torrecilla, S., Wang, H., Montironi, C., Pique-Gili, M., Torres-Martin, M., Wei-Qiang, L., Willoughby, C. E., Ramadori, P., Andreu-Oller, C., Taik, P., Lee, Y. A., Moeini, A., Peix, J., Faure-Dupuy, S., Riedl, T., Schuehle, S., Oliveira, C. P., Alves, V. A., ¼ Llovet, J. M. (2021). Molecular characterisation of hepatocellular carcinoma in patients with non-alcoholic steatohepatitis. Journal of Hepatology, 75, 865878. https://doi.org/10.1016/j.jhep.2021.04.049 CrossRefGoogle ScholarPubMed
Ponz de Leon, M., Bianchini, M. A., Reggiani-Bonetti, L., Pedroni, M., Di Gregorio, C., Merighi, A., Rossi, G., Magnani, G., Domati, F., & Cacciari, A. (2014). An unusual case of familial adenomatous polyposis with very early symptom occurrence. Familial Cance, 13, 375380. https://doi.org/10.1007/s10689-014-9718-3 CrossRefGoogle ScholarPubMed
Purcell, R., Childs, M., Maibach, R., Miles, C., Turner, C., Zimmermann, A., & Sullivan, M. (2011). HGF/c-Met related activation of beta-catenin in hepatoblastoma. Journal of Experimental & Clinical Cancer Research, 30, 96. https://doi.org/10.1186/1756-9966-30-96 CrossRefGoogle ScholarPubMed
Rubinfeld, B., Albert, I., Porfiri, E., Fiol, C., Munemitsu, S., & Polakis, P. (1996). Binding of GSK3beta to the APC-beta-catenin complex and regulation of complex assembly. Science, 272, 10231026. https://doi.org/10.1126/science.272.5264.1023 CrossRefGoogle Scholar
Sato, T., Tanigami, A., Yamakawa, K., Akiyama, F., Kasumi, F., Sakamoto, G., & Nakamura, Y. (1990). Allelotype of breast cancer: cumulative allele losses promote tumor progression in primary breast cancer. Cancer Research, 50, 71847189. https://www.ncbi.nlm.nih.gov/pubmed/1977515 Google ScholarPubMed
Shatara, M., Xavier, A. C., Dombkowski, A., Cukovic, D., Poulik, J. M., Altinok, D., Ge, Y., & Taub, J. W. (2019). Monozygotic twins with neuroblastoma MS have a similar molecular profile: A case of twin-to-twin metastasis. British Journal of Cancer, 121, 890893. https://doi.org/10.1038/s41416-019-0594-3 CrossRefGoogle ScholarPubMed
Shiraishi, Y., Sato, Y., Chiba, K., Okuno, Y., Nagata, Y., Yoshida, K., Shiba, N., Hayashi, Y., Kume, H., Homma, Y., Sanada, M., Ogawa, S., & Miyano, S. (2013). An empirical Bayesian framework for somatic mutation detection from cancer genome sequencing data. Nucleic Acids Research, 41, e89. https://doi.org/10.1093/nar/gkt126 CrossRefGoogle ScholarPubMed
Takeda, H., Kataoka, S., Nakayama, M., Ali, M. A. E., Oshima, H., Yamamoto, D., Park, J. W., Takegami, Y., An, T., Jenkins, N. A., Copeland, N. G., & Oshima, M. (2019). CRISPR-Cas9-mediated gene knockout in intestinal tumor organoids provides functional validation for colorectal cancer driver genes. Proceedings of the National Academy of Sciences of the United States of America, 116, 1563515644. https://doi.org/10.1073/pnas.1904714116 CrossRefGoogle ScholarPubMed
Taniguchi, K., Roberts, L. R., Aderca, I. N., Dong, X., Qian, C., Murphy, L. M., Nagorney, D. M., Burgart, L. J., Roche, P. C., Smith, D. I., Ross, J. A., & Liu, W. (2002). Mutational spectrum of beta-catenin, AXIN1, and AXIN2 in hepatocellular carcinomas and hepatoblastomas. Oncogene, 21, 48634871. https://doi.org/10.1038/sj.onc.1205591 CrossRefGoogle ScholarPubMed
Thomas, D., Pritchard, J., Davidson, R., McKiernan, P., Grundy, R. G., & de Ville de Goyet, J. (2003). Familial hepatoblastoma and APC gene mutations: Renewed call for molecular research. European Journal of Cancer, 39, 22002204. https://doi.org/10.1016/s0959-8049(03)00618-x CrossRefGoogle ScholarPubMed
Trobaugh-Lotrario, A. D., Lopez-Terrada, D., Li, P., & Feusner, J. H. (2018). Hepatoblastoma in patients with molecularly proven familial adenomatous polyposis: Clinical characteristics and rationale for surveillance screening. Pediatric Blood & Cancer, 65, e27103. https://doi.org/10.1002/pbc.27103 CrossRefGoogle ScholarPubMed
Yamaguchi, K., Nagayama, S., Shimizu, E., Komura, M., Yamaguchi, R., Shibuya, T., Arai, M., Hatakeyama, S., Ikenoue, T., Ueno, M., Miyano, S., Imoto, S., & Furukawa, Y. (2016). Reduced expression of APC-1B but not APC-1A by the deletion of promoter 1B is responsible for familial adenomatous polyposis. Scientific Reports, 6, 26011. https://doi.org/10.1038/srep26011 CrossRefGoogle Scholar
Yang, A., Sisson, R., Gupta, A., Tiao, G., & Geller, J. I. (2018). Germline APC mutations in hepatoblastoma. Pediatric Blood & Cancer, 65. https://doi.org/10.1002/pbc.26892 CrossRefGoogle ScholarPubMed
Zsiros, J., Brugieres, L., Brock, P., Roebuck, D., Maibach, R., Zimmermann, A., Childs, M., Pariente, D., Laithier, V., Otte, J. B., Branchereau, S., Aronson, D., Rangaswami, A., Ronghe, M., Casanova, M., Sullivan, M., Morland, B., Czauderna, P., Perilongo, G.; International Childhood Liver Tumours Strategy Group (SIOPEL), (2013). Dose-dense cisplatin-based chemotherapy and surgery for children with high-risk hepatoblastoma (SIOPEL-4): A prospective, single-arm, feasibility study. Lancet Oncology, 14, 834842. https://doi.org/10.1016/S1470-2045(13)70272-9 CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Genetic background of the twins. (A) Family pedigree. (B) Structure of the APC gene, including functional domains and introns. The position of the identified germline mutation is indicated by the red line. (C) Sanger sequence histogram of the affected infants and their parents. Note: CC, colon cancer; CP, colon polyps; GC, gastric cancer; HB, hepatoblastoma.

Figure 1

Figure 2. Clinical course of the affected infants. The C1 tumor was obtained by biopsy, whereas the C2 tumor was obtained by hepatectomy. (A) Infant-1. The red circle indicates the date of hepatectomy. (B) Infant-2. The red circle indicates the date of hepatectomy.

Figure 2

Table 1. Representative somatic mutations that may serve as tumor development trackers

Figure 3

Figure 3. Rare variant detection from multiregional specimens. (A) A macroscopic slice of the Infant-1 tumor (C1). DNA was extracted from six different regions. The variant allele frequency (VAF) of NRAS/ACVR2A in each region is indicated in a bar graph. (B) Macroscopic slices of the Infant-2 tumor (C2). DNA was extracted from seven different regions. The VAF of NRAS/ACVR2A in each region is indicated in a bar graph.

Figure 4

Table 2. Variant allele frequencies (%) of ctDNA

Supplementary material: File

Arisue et al. supplementary material

Arisue et al. supplementary material
Download Arisue et al. supplementary material(File)
File 627.8 KB