Advances in the conventional clinical treatment for hepatoblastoma and therapeutic innovation ============================================================================================= * Zhixue Chen * Rui Dong ## Abstract **Background** Hepatoblastoma (HB) is a rare malignancy usually occurring in children under 3 years old. With advancements in surgical techniques and molecular biology, new treatments have been developed. **Data resources** The recent literatures on new treatments, molecular mechanisms and clinical trials for HB were searched and reviewed. **Results** Surgical resection remains the main option for treatment of HB. Although complete resection is recommended, a resection with microscopical positive margins (R1) may have similar 5-year overall survival and 5-year event-free survival (EFS) rates after cisplatin chemotherapy and the control of metastasis, as only once described so far. Indocyanine green-guided surgery can help achieve precise resection. Additionally, associating liver partition and portal vein ligation for staged hepatectomy can rapidly increase future liver remnant volume compared with portal vein ligation or embolization. Cisplatin-containing chemotherapies slightly differ among the guidelines from the International Childhood Liver Tumors Strategy Group (SIOPEL), Children’s Oncology Group (COG) and Chinese Anti-Cancer Association Pediatric Committee (CCCG), and the 3-year EFS rate of patients in SIOPEL and CCCG studies was recently shown to be higher than that in COG studies. Liver transplantation is an option for patients with unresectable HB, and successful cases of autologous liver transplantation have been reported. In addition, effective inhibitors of important targets, such as the mTOR (mammalian target of rapamycin) inhibitor rapamycin, β-catenin inhibitor celecoxib and EpCAM (epithelial cell adhesion molecule) inhibitor catumaxomab, have been demonstrated to reduce the activity of HB cells and to control metastasis in experimental research and clinical trials. **Conclusion** These advances in surgical and medical treatment provide better outcomes for children with HB, and identifying novel targets may lead to the development of future targeted therapies and immunotherapies. * medical oncology * pediatrics ## Introduction Hepatoblastoma (HB) is a rare childhood malignant tumor with an incidence rate of 0.4 per 100 000 people per year.1 It often arises in teenagers under the age of 15 years, especially in children under 3 years old, and is relatively rare in adults,2 with only 47 cases reported in English articles from 1958 to 2016.3 Due to its non-specific clinical manifestations, most patients are diagnosed at an advanced stage. Currently, the international pediatric liver tumor collaboration groups include the Children’s Oncology Group (COG), International Childhood Liver Tumors Strategy Group (SIOPEL),4 Society for Pediatric Oncology and Hematology (GPOH), and Japanese Pediatric Liver Tumors Group (JPLT). Among them, the guidelines of the first two groups are widely accepted due to their rational therapeutic schemes. The Children’s Hepatic Tumor International Collaboration (CHIC) has incorporated their data into a collaborative database.5 In China, a consensus has been reached on the diagnosis and treatment of HB (CCCG-HB-2016)6 based on COG, SIOPEL and their personal experiences. At present, the main treatment strategies for patients with HB include surgical resection, autologous or allogeneic liver transplantation, and chemotherapy. Advances in surgical techniques and therapeutic strategies have improved the precision of hepatectomy and patient prognosis. Additionally, the recent breakthroughs in molecular biology and immunology will facilitate the clinical use of immunotherapies and targeted therapies in HB. In this review, we mainly focused on the progress in conventional clinical treatments and potential laboratory targets or pathways for future therapeutic interventions. ## Clinical treatment ### Staging There are two widely accepted staging systems: the preoperative SIOPEL system4 based on imaging assessments and the postoperative COG staging system7 based on pathology.8 The Chinese Anti-Cancer Association Pediatric Committee (CCCG) accepted the PRE Treatment EXTent of disease (PRETEXT)/POST-Treatment EXTent of disease (POST-TEXT) system and further modified the COG staging system (table 1) and risk stratification system (table 2) on the basis of their personal experience. Analysis of patient data in the CHIC data set verified the reliability and hierarchy of most traditional prognostic indicators, such as old age (especially >8 years), lower or higher alpha fetoprotein (AFP) serum levels (<100 ng/mL or ≥1 000 000 ng/mL), PRETEXT annotation factors and distant metastasis.9 10 The lung is the most common site of metastasis and recurrence site. For patients with HB with lung metastasis, chemotherapy is recommended first, and lung metastasectomy is feasible if it does not achieve complete resection (CR). The outcome of those that achieve CR by chemotherapy only is relatively better resulting from the JPLT-2, SIOPEL-3 and SIOPEL-4 studies.11 12 View this table: [Table 1](http://wjps.bmj.com/content/4/3/e000220/T1) Table 1 Modified COG Evans staging system (CCCG-HB-2016)6 View this table: [Table 2](http://wjps.bmj.com/content/4/3/e000220/T2) Table 2 Risk stratification system (CCCG-HB-2016)6 ### Surgical margins and long-term outcomes The integrity of resection plays a decisive role in the subsequent disease progression and prognosis. R0 resection (microscopically margin negative) is recommended, but the negative impact of R1 resection (microscopically margin positive) on the survival rate of children remains controversial.13 14 In early 2004, Dicken *et al*15 preliminarily observed no significant difference in survival between margins <1 cm of grossly uninvolved liver parenchyma and ≥1 cm after controlling pulmonary metastasis in 23 patients treated for primary HB. Instead, extrahepatic disease and macrovascular involvement were identified as poorer prognosis predictors.15 16 In recent long-term follow-up studies, the 5-year overall survival and 5-year event-free survival (EFS) rates of PRETEXT III/IV stage patients with R1 section were shown to be similar to those in patients with CR,14 16 17 when postoperative chemotherapy was administered, with normal AFP serum levels and imaging manifestations.14 Ren *et al*17 recently stratified the data from 2005 to 2017. They found that R1 resection might be associated with decreased survival in children with mixed epithelial/mesenchymal HB compared with R0 resection and not affect survival outcomes in those with an epithelial subtype without metastasis, confirming the greater importance of conventional prognostic indicators for survival outcomes compared with R0/R1 resection. Therefore, although R0 resection remains the primary aim, R1 resection or surgery with ‘planned close margins’, that is, extreme resections,13 might also have good outcomes and be valuable when orthotopic liver transplantation is not possible. ### Indocyanine green in surgical resection Indocyanine green (ICG) fluorescent imaging18 helps surgeons achieve precise primary resection and metastasectomy. When injected preoperatively, ICG is cleared by normal liver cells and is excreted into the biliary tract but remains in tumor cells for a relatively long time. Clinically, ICG molecular fluorescence imaging is applied to detect primary lesions and metastases that cannot be visualized by the naked eye, making negative resection margins possible. The dose and timing of ICG injections for HB have not been standardized,19 but ICG is generally injected intravenously at a dose of 0.5 mg/kg 72 hours before surgery.18 20–22 Because ICG is only visible in the near-infrared mode, frequent switching between the normal white-light mode and near-infrared mode is required to resect the stained tumor during surgery. To further improve the feasibility of real-time operations, the PINPOINT endoscope fluorescence imaging system with the same focal distance between the white-light mode and the near-infrared mode has been applied. This system overlays both modes and displays HB metastases injected with ICG on the white-light image in high definition, which simplifies the surgical steps without frequent screen switching.21 Furthermore, projection mapping technology has also been used in some cases of HB with lung metastasis. The Medical Imaging Projection System projects the fluorescence image obtained by CT onto the surface of the target organ. With this system, the use of monitors and a dark environment are not required.23 ### Associating liver partition and portal vein ligation for staged hepatectomy Extensive liver resection may lead to severe posthepatectomy liver failure due to an insufficient volume of the future liver remnant (FLR).24 The greatest advantage of associating liver partition and portal vein ligation for staged hepatectomy (ALPPS) is the rapid increase in the FLR volume compared with portal vein ligation or embolization.25 26 ALPPS is a two-stage hepatectomy procedure, including portal vein ligation and staged liver resection.10 27 During the first stage, the liver is completely divided from the FLR with concomitant portal vein ligation of the lobe that will be removed, and the biliary tract system and the blood vessels are well preserved. During the second stage, hepatectomy is performed after achieving adequate FLR (at least 50% of the total liver volume).27 28 ALPPS has shown promising results in adults with hepatobiliary malignancies, but pediatric cases are rarely reported. Chan *et al*29 in 2014, Wiederkehr *et al*27 in 2015 and Hong *et al*24 in 2017 successively performed ALPPS or monosegmental ALPPS in pediatric patients with HB with PRETEXT stage III/IV, and most of them showed a rapid increase in FLR and a decrease in AFP serum levels. In August 2020, Akhaladze *et al*25 reported the first case of pure laparoscopic first-stage ALPPS in a child and showed the feasibility of a laparoscopic approach. In summary, ALPPS provides a better opportunity for CR in patients with primarily unresectable liver tumors, but may also be related to complications such as multifocal recurrence,30 pleural effusion and pneumonia.28 Therefore, the use of ALPPS in pediatric patients deserves further evaluation. ### Liver transplantation The indications for liver transplantation are similar in several groups including COG, SIOPEL, JPLT and CCCG. When the POST-TEXT evaluation of patients is the POST-TEXT IV or P2 (involvement of the main portal vein) or V3 (involvement of all three hepatic veins and/or the inferior vena cava) in POST-TEXT, liver transplantation is recommended.4 6 31 32 In other words, those who respond to chemotherapy but have unresectable tumors and no evidence of persistent extrahepatic disease should be considered for liver transplantation. The CCCG consensus indicates that liver transplantation can be applied after the distant metastasis is removed by surgery or chemotherapy, so the lung metastasis is not the contraindication for orthotopic liver transplantation.6 33 For the unresectable HB, liver transplantation may be effective, with a long-term survival rate of 80% after combination treatment.34–36 Shi *et al*37 successfully implemented ex vivo liver resection and autotransplantation for children with HB. The procedure includes removal of the malignancy in vitro or semi-in vivo, transplanting the remaining liver and anastomosing the superior and inferior vena cava, bile duct and other duct systems to restore the liver blood supply.38 Compared with allogeneic liver transplantation, this approach overcomes the limitations of liver resource scarcity and rejection. Besides, living-related liver transplantation is another better choice to avoid the waiting time for deceased donor liver. The liver graft procured from donor relatives for pediatric recipient always includes the left lateral segment without inferior vena cava,39 40 and the reconstruction of inferior vena cava cannot be avoided. Because the Dacron synthetic blood vessel cannot adapt to the growth of children, it is necessary to find a large lumen vein graft (such as the autogenous iliac vein, recanalized umbilical vein and donor internal jugular vein) to reconstruct the vena cava.41–43 Regarding the timing of liver transplantation, Isono *et al*44 suggested that liver transplantation could be performed when the AFP level remained stable after the last chemotherapy treatment. ### Chemotherapy regimen The SIOPEL and GPOH groups recommend preoperative chemotherapy for HB to achieve smaller resection, whereas the COG and CCCG groups recommend early and primary resection for localized HB and neoadjuvant chemotherapy for advanced stages. Regarding chemotherapy regimens, the COG and CCCG groups use the C5V regimen (cisplatin, fluorouracil and vincristine) as the standard care for low-risk patients and the C5VD regimen (cisplatin, fluorouracil, vincristine and doxorubicin) for intermediate-risk patients. There is a slight difference between COG and CCCG guidelines regarding the high-risk regimen. CCCG recommends ifosfamide and etoposide to decrease the cumulative dose of cisplatin and doxorubicin,6 45 whereas the VI regimen (vincristine+irinotecan) is introduced as upfront window therapy in the COG study AHEP0731.46 47 The 3-year EFS of patients with high-risk HB was 49% in this COG study and 75.7% in a Chinese single-center study that followed the CCCG-HB-2016 consensus.45–47 In comparison with these, the SIOPEL and GPOH groups focused more on the frequency and dose of platinum derivatives with or without doxorubicin.12 48–50 In the SIOPEL-4 study of children with untreated high-risk HB, the children were given cisplatin, doxorubicin and carboplatin and the 3-year EFS was 76%.4 6 10 45 ### Cisplatin-induced ototoxicity Ototoxicity is a severe complication of platinum-containing chemotherapy. Several ototoxicity criteria or grading systems, such as the National Cancer Institute Common Toxicity Criteria for Adverse Events, Brock Ototoxicity Grades and International Society of Pediatric Oncology (SIOP) Boston scale, have been developed. Among them, the SIOP scale may be more sensitive in classifying ototoxicity in children.51 52 The accumulated dosage and single maximum dose of cisplatin have been reported to serve as independent predictors of moderate to severe hearing loss.53 In the SIOPEL-6 trial, sodium thiosulfate (20 g/m2) given 6 hours after cisplatin chemotherapy was demonstrated to effectively reduce hearing loss in patients with standard-risk HB,54 which was consistent with the results of the COG study ACCL0431 which included heterogeneous samples.55 In contrast, amifostine is not recommended because personal advantages or direct benefits were not observed in previous single-center studies or trials.56 57 Other sodium diethyldithiocarbamate and intratympanic middle ear therapies are not currently recommended due to their low evidence quality.56 ## Important targets and their inhibitors Tumor-related proteins and pathways have received increasing attention because understanding their function is essential for the development of novel targeted therapies and immunotherapies. Several key studies on classic signaling pathways and their inhibitors will be discussed in the following section (table 3). View this table: [Table 3](http://wjps.bmj.com/content/4/3/e000220/T3) Table 3 Promising targets and inhibitors for hepatoblastoma ### The PI3K/Akt pathway Phosphatidylinositol 3-kinase (PI3K) and its main downstream target serine-threonine kinase (Akt) constitute the PI3K/Akt pathway, which is a central regulator of numerous signal transduction pathways involved in tumorigenesis. Following activation by factors such as insulin-like growth factor-1 and epidermal growth factor, PI3K inhibits cell apoptosis by phosphorylating Akt.58 PI3K and Akt are highly expressed in HB,58 indicating the importance of the PI3K/Akt pathway in HB. By inhibiting the upstream and downstream proteins in this signaling pathway, the metastasis of tumor cells can be effectively controlled. Classic targeted drugs include Ginkgol C17:1, which is coadministered with cisplatin to stimulate cisplatin-induced apoptosis,59 60 the PI3K inhibitor LY29400258 or the mTOR (mammalian target of rapamycin) inhibitor rapamycin.61 Molina *et al*61 used an HB mouse model to observe the effect of rapamycin and found that rapamycin significantly reduced tumor burden in mice. The Chinese herb polyphyllin VII is an mTOR inhibitor and has been found to inhibit mTOR phosphorylation, promote autophagy and further induce HepG2 cell apoptosis.62 mTOR inhibitors are often used in combination with sorafenib and have been shown to induce cell cycle arrest in the G1 phase and inhibit the invasion ability of tumor cells together with miRNA-378a.63 Akt is another key kinase, and inhibition of its phosphorylation effectively prevents signal transduction. Inhibitory drugs include emodin (a derivative of anthraquinone64), FTY720 (an immunomodulator of multiple sclerosis65) and furazolidone (a broad-spectrum antibiotic66). Among them, the immunosuppressive drug FTY720 (fingolimod) has been found to increase cisplatin sensitivity to effectively reduce the activity of HB cells and induce cell apoptosis in vitro and in vivo,65 which may provide ideas for future clinical tumor treatments. ### The Wnt/β-catenin pathway The Wnt/β-catenin pathway plays an important role in normal development and HB formation. In healthy livers, β-catenin is degraded by the β-catenin destruction complex and proteasome in the absence of extracellular Wnt ligands. However, in the presence of Wnt ligands due to loss of the *APC* gene or mutations in the gene *CTNNB1* (76%–80% of HBs carry mutations in exon 3 of catenin-beta 167), the degradation of β-catenin is inhibited, thus increasing the occurrence of HB.68 β-catenin serves as an important target in the treatment of HB. Indersie *et al*67 established an HB-related microRNA library and screened nine miRNAs. They found that miR-624-5p, let-7i-3p and miR-885-5p inhibited the protein expression of β-catenin, thereby inactivating the Wnt pathway to prolong the G0/G1 phase and shorten the S phase. Among them, as the most effective inhibitor and preferred miRNA replacement therapy, miR-624-5p has been demonstrated to bind to the 3'-untranslated region of β-catenin and target three mRNA variants.67 Other catenin inhibitors include celecoxib (which increases the cytoplasmic translocation and phosphorylation of β-catenin) and ICG001 (which binds cyclic AMP response element-binding protein to block β-catenin transcription). Combined with cisplatin, these inhibitors decreased the viability of the HB cell line HuU6.69 In addition, flavonoids such as epigallocatechin gallate induce cytotoxicity in HB cells by downregulating the Wnt target genes *MYC* and *CCND1*.70 ### Neurokinin receptor Substance P (SP) is widely distributed in the central and peripheral nervous system71 and is produced by self-stimulated tumor cells. Following specific binding to the neurokinin receptor (NK1R), SP promotes cancer cell proliferation, angiogenesis and migration and inhibits apoptosis. SP has been shown to be overexpressed in breast cancer.72 In 2015, Berger *et al*73 first identified overexpression of the truncated NK1R in the human HB cell lines HuU6 and HepG2. Additionally, NK1R antagonists (aprepitant, L-733060 and L-732138) were demonstrated to induce the apoptosis of HB cells and act synergistically with the chemotherapy drug doxorubicin.73 Ilmer *et al*74 further revealed the molecular regulatory mechanism of NK1R antagonists. After treatment of HB cell lines with aprepitant, NK1R inhibition downregulated the Wnt signaling pathway and reduced the expression levels of the β-catenin interaction factor FOXM1, thereby disrupting the interaction between FOXM1 and β-catenin.75 The SP/NK1R complex provides novel therapeutic targets for HB. Aprepitant, one of the drugs used to treat vomiting after chemotherapy, may be applied to future targeted therapy in HB. ### Epithelial cell adhesion molecule (EpCAM/CD326) Epithelial adhesion/activation molecule (EpCAM/CD326) is a type I membrane protein and a potential immunotherapeutic target. Its expression has been observed in epithelial tumor areas in 70%–80% of patients with HB.76 Armeanu-Ebinger *et al*76 suggested that the anti-EpCAM bispecific antibody MT110 could simultaneously bind to EpCAM and CD3 and promote the T cell-mediated lysis of HB tumor cells, thereby impairing tumor development. At present, EpCAM antibodies, such as catumaxomab and IGN101, are being investigated in phase II/III clinical trials related to gastric cancer and small cell lung cancer.77 78 ### Glypican-3 Glypican-3 (GPC3), a surface carcinoembryonic antigen, is often overexpressed in solid malignant tumors in children. For example, the upregulation of GPC3 has been reported in 97% of HB cases.79 GPC3 promotes tumor cell growth and inhibits cell proliferation by activating the Wnt/β-catenin pathway.79 A Japanese team conducted a phase I clinical trial using GPC3-derived peptide vaccines (HLA-A*24:02-restricted GPC3298–306 peptide (EYILSLEEL) and HLA-A*02:01-restricted GPC3144–152 peptide (FVGEFFTDV)) in children with GPC3 overexpression and found that the vaccines induced a GPC3-specific cytotoxic T lymphocytes response.80 In addition, monoclonal antibody immunotherapies, such as GC33 (GPC3 monoclonal antibody, also known as codrituzumab), have been shown to have antitumor effects on GPC3-positive tumors in phase I for hepatocellular carcinoma but less clinical benefit in the Ib/II trials.81–84 These immunological antibodies might be further investigated in clinical trials for HB. ## Conclusion The CCCG-HB-2016 consensus based on the widely accepted SIOPEL and COG guidelines and their personal experiences modified the COG Evans staging system and risk stratification system and deserves mention. Cisplatin-containing chemotherapies differ slightly in SIOPEL, COG and CCCG guidelines, and the 3-year EFS of patients was recently shown to be higher in SIOPEL and CCCG studies compared with that in COG studies. Cisplatin-induced ototoxicity can be effectively reduced with sodium thiosulfate, as demonstrated in SIOPEL and COG studies. ICG-guided resection and ALPPS, which rapidly increase the FLR volume, have gained popularity among pediatric surgeons. These advances in surgical and medical treatment provide better outcomes for children with HB. Additionally, the immunotherapeutic drug catumaxomab (an EpCAM inhibitor) and GPC3-derived peptide vaccines have been investigated in clinical trials. The efficacy of other important inhibitors, such as the mTOR inhibitor rapamycin, Akt inhibitory drugs emodin and fingolimod, β-catenin inhibitor celecoxib and NK1R antagonist aprepitant, have been shown in animal experiments. These inhibitors may provide novel ideas for future targeted therapies and immunotherapies in HB. ## Data availability statement No data are available. ## Ethics statements ### Patient consent for publication Not required. ### Ethics approval Not required. ## Acknowledgments We thank Melissa Crawford, PhD, from Liwen Bianji, Edanz Editing China (www.liwenbianji.cn/ac) for editing the English text of a draft of this manuscript. ## Footnotes * Contributors RD contributed to conceptualization, writing-review and editing. ZXC performed methodology and writing-original draft. Both authors contributed to the article and approved the submitted version. * Funding This work was supported by the Cyrus Tang Foundation, National Natural Science Foundation of China (no. 82072782), the Shanghai Hospital Development Center (Grant No. SHDC12018X22, SHDC2020CR2009A and SHDC12020125). * Competing interests None declared. * Provenance and peer review Not commissioned; externally peer reviewed. * Received October 12, 2020. * Accepted May 21, 2021. * © Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ. [http://creativecommons.org/licenses/by-nc/4.0/](http://creativecommons.org/licenses/by-nc/4.0/) This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See: [http://creativecommons.org/licenses/by-nc/4.0/](http://creativecommons.org/licenses/by-nc/4.0/). ## References 1. Sunil BJ, Palaniappan R, Venkitaraman B, et al. Surgical resection for Hepatoblastoma-Updated survival outcomes. J Gastrointest Cancer 2018;49:493–6.[doi:10.1007/s12029-017-0005-z](http://dx.doi.org/10.1007/s12029-017-0005-z)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28963691 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 2. Allan BJ, Parikh PP, Diaz S, et al. Predictors of survival and incidence of hepatoblastoma in the paediatric population. HPB 2013;15:741–6.[doi:10.1111/hpb.12112](http://dx.doi.org/10.1111/hpb.12112)pmid:http://www.ncbi.nlm.nih.gov/pubmed/23600968 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 3. Duan X-F, Zhao Q. Adult hepatoblastoma: a review of 47 cases. ANZ J Surg 2018;88:E50–4.[doi:10.1111/ans.13839](http://dx.doi.org/10.1111/ans.13839)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27905197 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 4. Czauderna P, Otte JB, Aronson DC, et al. Guidelines for surgical treatment of hepatoblastoma in the modern era--recommendations from the Childhood Liver Tumour Strategy Group of the International Society of Paediatric Oncology (SIOPEL). Eur J Cancer 2005;41:1031–6.[doi:10.1016/j.ejca.2005.02.004](http://dx.doi.org/10.1016/j.ejca.2005.02.004)pmid:http://www.ncbi.nlm.nih.gov/pubmed/15862752 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1016/j.ejca.2005.02.004&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=15862752&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 5. Meyers RL, Maibach R, Hiyama E, et al. Risk-Stratified staging in paediatric hepatoblastoma: a unified analysis from the children's hepatic tumors international collaboration. Lancet Oncol 2017;18:122–31.[doi:10.1016/S1470-2045(16)30598-8](http://dx.doi.org/10.1016/S1470-2045(16)30598-8)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27884679 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1016/S1470-2045(16)30598-8&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=27884679&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 6. Chinese Anti-Cancer Association Pediatric CommitteeChina Medical Association Pediatric Onco-surgrey Group. Expert consensus for Multidiscciplinary management of hepatoblastoma (CCCG-HB-2016). Chin J Pediatr Surg 2017;38:733–9.[doi:10.3760/cma.j.issn.0253-3006.2017.10.003](http://dx.doi.org/10.3760/cma.j.issn.0253-3006.2017.10.003) 7. Ortega JA, Douglass EC, Feusner JH, et al. Randomized comparison of cisplatin/vincristine/fluorouracil and cisplatin/continuous infusion doxorubicin for treatment of pediatric hepatoblastoma: a report from the children's cancer group and the pediatric Oncology Group. J Clin Oncol 2000;18:2665–75.[doi:10.1200/JCO.2000.18.14.2665](http://dx.doi.org/10.1200/JCO.2000.18.14.2665)pmid:http://www.ncbi.nlm.nih.gov/pubmed/10894865 [Abstract/FREE Full Text](http://wjps.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIxOC8xNC8yNjY1IjtzOjQ6ImF0b20iO3M6MjI6Ii93anBzLzQvMy9lMDAwMjIwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 8. Agarwala S, Gupta A, Bansal D, et al. Management of hepatoblastoma: ICMR consensus document. Indian J Pediatr 2017;84:456–64.[doi:10.1007/s12098-017-2301-9](http://dx.doi.org/10.1007/s12098-017-2301-9)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28353129 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 9. Czauderna P, Haeberle B, Hiyama E, et al. The children's hepatic tumors international collaboration (chiC): novel global rare tumor database yields new prognostic factors in hepatoblastoma and becomes a research model. Eur J Cancer 2016;52:92–101.[doi:10.1016/j.ejca.2015.09.023](http://dx.doi.org/10.1016/j.ejca.2015.09.023)pmid:http://www.ncbi.nlm.nih.gov/pubmed/26655560 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1016/j.ejca.2015.09.023&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=26655560&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 10. Yang T, Whitlock RS, Vasudevan SA. Surgical management of hepatoblastoma and recent advances. Cancers 2019;11. doi:[doi:10.3390/cancers11121944](http://dx.doi.org/10.3390/cancers11121944). [Epub ahead of print: 04 Dec 2019].pmid:http://www.ncbi.nlm.nih.gov/pubmed/31817219 11. Hishiki T, Watanabe K, Ida K, et al. The role of pulmonary metastasectomy for hepatoblastoma in children with metastasis at diagnosis: results from the JPLT-2 study. J Pediatr Surg 2017;52:2051–5.[doi:10.1016/j.jpedsurg.2017.08.031](http://dx.doi.org/10.1016/j.jpedsurg.2017.08.031)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28927977 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 12. Zsíros J, Maibach R, Shafford E, et al. Successful treatment of childhood high-risk hepatoblastoma with dose-intensive multiagent chemotherapy and surgery: final results of the SIOPEL-3HR study. J Clin Oncol 2010;28:2584–90.[doi:10.1200/JCO.2009.22.4857](http://dx.doi.org/10.1200/JCO.2009.22.4857)pmid:http://www.ncbi.nlm.nih.gov/pubmed/20406943 [Abstract/FREE Full Text](http://wjps.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIyOC8xNS8yNTg0IjtzOjQ6ImF0b20iO3M6MjI6Ii93anBzLzQvMy9lMDAwMjIwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 13. Fonseca A, Gupta A, Shaikh F, et al. Extreme hepatic resections for the treatment of advanced hepatoblastoma: are planned close margins an acceptable approach? Pediatr Blood Cancer 2018;65. doi:[doi:10.1002/pbc.26820](http://dx.doi.org/10.1002/pbc.26820). [Epub ahead of print: 17 Sep 2017].pmid:http://www.ncbi.nlm.nih.gov/pubmed/28921939 14. Aronson DC, Weeda VB, Maibach R, et al. Microscopically positive resection margin after hepatoblastoma resection: what is the impact on prognosis? A Childhood Liver Tumours Strategy Group (SIOPEL) report. Eur J Cancer 2019;106:126–32.[doi:10.1016/j.ejca.2018.10.013](http://dx.doi.org/10.1016/j.ejca.2018.10.013)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30528797 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 15. Dicken BJ, Bigam DL, Lees GM. Association between surgical margins and long-term outcome in advanced hepatoblastoma. J Pediatr Surg 2004;39:721–5.[doi:10.1016/j.jpedsurg.2004.01.035](http://dx.doi.org/10.1016/j.jpedsurg.2004.01.035)pmid:http://www.ncbi.nlm.nih.gov/pubmed/15137006 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=15137006&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 16. Ren X, Li H, Diao M, et al. Results of surgical resections with positive margins for children with hepatoblastoma: case series from a single Asian center. Pediatr Blood Cancer 2019;66:e27479. [doi:10.1002/pbc.27479](http://dx.doi.org/10.1002/pbc.27479)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30255649 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 17. Ren X, Li H, Diao M, et al. Impact of microscopically margin-positive resection on survival in children with hepatoblastoma after hepatectomy: a retrospective cohort study. Int J Clin Oncol 2020;25:765–73.[doi:10.1007/s10147-019-01573-0](http://dx.doi.org/10.1007/s10147-019-01573-0)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31701290 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 18. Yamada Y, Ohno M, Fujino A, et al. Fluorescence-Guided surgery for hepatoblastoma with indocyanine green. Cancers 2019;11:1215. [doi:10.3390/cancers11081215](http://dx.doi.org/10.3390/cancers11081215) 19. Souzaki R, Kawakubo N, Matsuura T, et al. Navigation surgery using indocyanine green fluorescent imaging for hepatoblastoma patients. Pediatr Surg Int 2019;35:551–7.[doi:10.1007/s00383-019-04458-5](http://dx.doi.org/10.1007/s00383-019-04458-5)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30778701 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 20. Yamamichi T, Oue T, Yonekura T, et al. Clinical application of indocyanine green (ICG) fluorescent imaging of hepatoblastoma. J Pediatr Surg 2015;50:833–6.[doi:10.1016/j.jpedsurg.2015.01.014](http://dx.doi.org/10.1016/j.jpedsurg.2015.01.014)pmid:http://www.ncbi.nlm.nih.gov/pubmed/25783395 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 21. Yamada Y, Hoshino K, Mori T, et al. Metastasectomy of hepatoblastoma utilizing a novel overlay fluorescence imaging system. J Laparoendosc Adv Surg Tech A 2018;28:1152–5.[doi:10.1089/lap.2017.0735](http://dx.doi.org/10.1089/lap.2017.0735) 22. Takahashi N, Yamada Y, Hoshino K, et al. Living donor liver re-transplantation for recurrent hepatoblastoma in the liver graft following complete eradication of peritoneal metastases under indocyanine green fluorescence imaging. Cancers 2019;11. doi:[doi:10.3390/cancers11050730](http://dx.doi.org/10.3390/cancers11050730). [Epub ahead of print: 26 May 2019].pmid:http://www.ncbi.nlm.nih.gov/pubmed/31130716 23. Chen-Yoshikawa TF, Hatano E, Yoshizawa A, et al. Clinical application of projection mapping technology for surgical resection of lung metastasis. Interact Cardiovasc Thorac Surg 2017;25:1010–1.[doi:10.1093/icvts/ivx247](http://dx.doi.org/10.1093/icvts/ivx247)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29049837 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 24. Hong JC, Kim J, Browning M, et al. Modified associating liver partition and portal vein ligation for staged hepatectomy for hepatoblastoma in a small infant: how far can we push the envelope? Ann Surg 2017;266:e16–17.[doi:10.1097/SLA.0000000000002217](http://dx.doi.org/10.1097/SLA.0000000000002217)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28288067 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 25. Akhaladze D, Uskova N, Rabaev G, et al. A minimally invasive first stage of ALPPS for hepatoblastoma in a child. Ann Hepatobiliary Pancreat Surg 2020;24:352–6.[doi:10.14701/ahbps.2020.24.3.352](http://dx.doi.org/10.14701/ahbps.2020.24.3.352)pmid:http://www.ncbi.nlm.nih.gov/pubmed/32843604 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 26. Schadde E, Ardiles V, Slankamenac K, et al. ALPPS offers a better chance of complete resection in patients with primarily unresectable liver tumors compared with conventional-staged hepatectomies: results of a multicenter analysis. World J Surg 2014;38:1510–9.[doi:10.1007/s00268-014-2513-3](http://dx.doi.org/10.1007/s00268-014-2513-3)pmid:http://www.ncbi.nlm.nih.gov/pubmed/24748319 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1007/s00268-014-2513-3&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=24748319&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 27. Wiederkehr JC, Avilla SG, Mattos E, et al. Associating liver partition with portal vein ligation and staged hepatectomy (ALPPS) for the treatment of liver tumors in children. J Pediatr Surg 2015;50:1227–31.[doi:10.1016/j.jpedsurg.2014.10.019](http://dx.doi.org/10.1016/j.jpedsurg.2014.10.019)pmid:http://www.ncbi.nlm.nih.gov/pubmed/25783345 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 28. Ruiz Figueroa EF, Fernández-Placencia RM, Berrospi Espinoza FE, et al. Monosegmental ALPPS: a long-term survival alternative to liver transplant in PRETEXT IV hepatoblastoma. J Surg Case Rep 2019;2019:rjz144. [doi:10.1093/jscr/rjz144](http://dx.doi.org/10.1093/jscr/rjz144)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31086654 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 29. Chan A, Chung PHY, Poon RTP. Little girl who conquered the "ALPPS''. World J Gastroenterol 2014;20:10208–11.[doi:10.3748/wjg.v20.i29.10208](http://dx.doi.org/10.3748/wjg.v20.i29.10208)pmid:http://www.ncbi.nlm.nih.gov/pubmed/25110450 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 30. Qazi AQ, Syed AA, Khan AW, et al. Early multifocal recurrence of hepatoblastoma in the residual liver after R0 liver resection with ALPPS procedure: a case report. Ann Transl Med 2016;4:375.[doi:10.21037/atm.2016.10.01](http://dx.doi.org/10.21037/atm.2016.10.01)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27826578 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 31. Tajiri T, Kimura O, Fumino S, et al. Surgical strategies for unresectable hepatoblastomas. J Pediatr Surg 2012;47:2194–8.[doi:10.1016/j.jpedsurg.2012.09.006](http://dx.doi.org/10.1016/j.jpedsurg.2012.09.006)pmid:http://www.ncbi.nlm.nih.gov/pubmed/23217875 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 32. Meyers RL, Tiao GM, Dunn SP, et al. Liver transplantation in the management of unresectable hepatoblastoma in children. Front Biosci 2012;4:1293–302.[doi:10.2741/e460](http://dx.doi.org/10.2741/e460)pmid:http://www.ncbi.nlm.nih.gov/pubmed/22201955 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 33. Miyamura T, Yoshida R, Yagi T, et al. Successful treatment of unresectable advanced hepatoblastoma: living liver transplantation after surgical removal of lung metastasis. Pediatr Transplant 2011;15:E87–91.[doi:10.1111/j.1399-3046.2009.01262.x](http://dx.doi.org/10.1111/j.1399-3046.2009.01262.x)pmid:http://www.ncbi.nlm.nih.gov/pubmed/20070564 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=20070564&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 34. Fuchs J, Cavdar S, Blumenstock G, et al. POST-TEXT III and IV hepatoblastoma: extended hepatic resection avoids liver transplantation in selected cases. Ann Surg 2017;266:318–23.[doi:10.1097/SLA.0000000000001936](http://dx.doi.org/10.1097/SLA.0000000000001936)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27501172 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 35. Filin AV, Kim EF, Charchyan ER, et al. [Liver transplantation for hepatoblastoma in children]. Khirurgiia 2020:5–13.[doi:10.17116/hirurgia20201115](http://dx.doi.org/10.17116/hirurgia20201115)pmid:http://www.ncbi.nlm.nih.gov/pubmed/33210501 36. Ramos-Gonzalez G, LaQuaglia M, O'Neill AF, et al. Long-Term outcomes of liver transplantation for hepatoblastoma: a single-center 14-year experience. Pediatr Transplant 2018:e13250. [doi:10.1111/petr.13250](http://dx.doi.org/10.1111/petr.13250)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29888545 37. Shi S-J, Wang D-L, Hu W, et al. Ex vivo liver resection and autotransplantation with cardiopulmonary bypass for hepatoblastoma in children: a case report. Pediatr Transplant 2018;22:e13268. [doi:10.1111/petr.13268](http://dx.doi.org/10.1111/petr.13268)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30003622 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 38. Zhang MM, Ning Y, YC L. Applicationantologous liver transplantation in complex infantile hepatoblastoma: a report of one case. Chin J Pediatr Surg 2018;39:44–9.[doi:10.3760/cma.j.issn.0253-3006.2018.01.010](http://dx.doi.org/10.3760/cma.j.issn.0253-3006.2018.01.010) 39. Chardot C, Saint Martin C, Gilles A, et al. Living-Related liver transplantation and vena cava reconstruction after total hepatectomy including the vena cava for hepatoblastoma. Transplantation 2002;73:90–2.[doi:10.1097/00007890-200201150-00017](http://dx.doi.org/10.1097/00007890-200201150-00017)pmid:http://www.ncbi.nlm.nih.gov/pubmed/11792985 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=11792985&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 40. Tannuri ACA, Gibelli NEM, Ricardi LRS, et al. Living related donor liver transplantation in children. Transplant Proc 2011;43:161–4.[doi:10.1016/j.transproceed.2010.11.013](http://dx.doi.org/10.1016/j.transproceed.2010.11.013)pmid:http://www.ncbi.nlm.nih.gov/pubmed/21335177 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=21335177&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 41. Namgoong J-M, Choi J-U, Hwang S, et al. Pediatric living donor liver transplantation with homograft replacement of retrohepatic inferior vena cava for advanced hepatoblastoma. Ann Hepatobiliary Pancreat Surg 2019;23:178–82.[doi:10.14701/ahbps.2019.23.2.178](http://dx.doi.org/10.14701/ahbps.2019.23.2.178)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31225421 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 42. Shinkai M, Mochizuki K, Kitagawa N, et al. Usefulness of a Recanalized umbilical vein for vascular reconstruction in pediatric hepatic surgery. Pediatr Surg Int 2016;32:553–8.[doi:10.1007/s00383-016-3893-2](http://dx.doi.org/10.1007/s00383-016-3893-2)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27083897 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 43. Hort A, Karpelowsky J, Shun A, et al. Use of a donor iliac vein graft for reconstruction of the inferior vena cava in liver transplantation for hepatoblastoma with caval extension. Pediatr Transplant 2019;23:e13409. [doi:10.1111/petr.13409](http://dx.doi.org/10.1111/petr.13409)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30946509 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 44. Isono K, Ohya Y, Lee K-J, et al. Pretransplant trends in α-fetoprotein levels as a predictor of recurrence after living donor liver transplantation for unresectable hepatoblastoma: a single-institution experience. Pediatr Transplant 2018;22:e13221. [doi:10.1111/petr.13221](http://dx.doi.org/10.1111/petr.13221)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29761829 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 45. Wang TY, Han YL, Gao YJ, et al. Retrospective analysis of childhood hepatoblastoma in a single centre in China. Clin Oncol 2019;31:471–8.[doi:10.1016/j.clon.2019.03.044](http://dx.doi.org/10.1016/j.clon.2019.03.044)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31000431 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 46. Katzenstein HM, Furman WL, Malogolowkin MH, et al. Upfront window vincristine/irinotecan treatment of high-risk hepatoblastoma: a report from the children's Oncology Group AHEP0731 study Committee. Cancer 2017;123:2360–7.[doi:10.1002/cncr.30591](http://dx.doi.org/10.1002/cncr.30591)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28211941 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 47. O'Neill AF, Towbin AJ, Krailo MD, et al. Characterization of pulmonary metastases in children with hepatoblastoma treated on children's Oncology Group protocol AHEP0731 (the treatment of children with all stages of hepatoblastoma): a report from the children's Oncology Group. J Clin Oncol 2017;35:3465–73.[doi:10.1200/JCO.2017.73.5654](http://dx.doi.org/10.1200/JCO.2017.73.5654)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28892430 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 48. Hackl C, Schlitt HJ, Kirchner GI, et al. Liver transplantation for malignancy: current treatment strategies and future perspectives. World J Gastroenterol 2014;20:5331–44.[doi:10.3748/wjg.v20.i18.5331](http://dx.doi.org/10.3748/wjg.v20.i18.5331)pmid:http://www.ncbi.nlm.nih.gov/pubmed/24833863 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=24833863&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 49. Czauderna P, Lopez-Terrada D, Hiyama E, et al. Hepatoblastoma state of the art: pathology, genetics, risk stratification, and chemotherapy. Curr Opin Pediatr 2014;26:19–28.[doi:10.1097/MOP.0000000000000046](http://dx.doi.org/10.1097/MOP.0000000000000046)pmid:http://www.ncbi.nlm.nih.gov/pubmed/24322718 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1097/MOP.0000000000000046&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=24322718&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 50. Zsiros J, Brugieres L, Brock P, et al. Dose-Dense cisplatin-based chemotherapy and surgery for children with high-risk hepatoblastoma (SIOPEL-4): a prospective, single-arm, feasibility study. Lancet Oncol 2013;14:834–42.[doi:10.1016/S1470-2045(13)70272-9](http://dx.doi.org/10.1016/S1470-2045(13)70272-9)pmid:http://www.ncbi.nlm.nih.gov/pubmed/23831416 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1016/S1470-2045(13)70272-9&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=23831416&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 51. Brock PR, Knight KR, Freyer DR, et al. Platinum-induced ototoxicity in children: a consensus review on mechanisms, predisposition, and protection, including a new International Society of pediatric oncology Boston ototoxicity scale. J Clin Oncol 2012;30:2408–17.[doi:10.1200/JCO.2011.39.1110](http://dx.doi.org/10.1200/JCO.2011.39.1110)pmid:http://www.ncbi.nlm.nih.gov/pubmed/22547603 [Abstract/FREE Full Text](http://wjps.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIzMC8xOS8yNDA4IjtzOjQ6ImF0b20iO3M6MjI6Ii93anBzLzQvMy9lMDAwMjIwLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 52. Knight KR, Chen L, Freyer D, et al. Group-Wide, prospective study of ototoxicity assessment in children receiving cisplatin chemotherapy (ACCL05C1): a report from the children's Oncology Group. J Clin Oncol 2017;35:440–5.[doi:10.1200/JCO.2016.69.2319](http://dx.doi.org/10.1200/JCO.2016.69.2319)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27937095 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1200/JCO.2016.69.2319&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 53. Wei M, Yuan X. Cisplatin-Induced ototoxicity in children with solid tumor. J Pediatr Hematol Oncol 2019;41:e97–100.[doi:10.1097/MPH.0000000000001282](http://dx.doi.org/10.1097/MPH.0000000000001282)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30095690 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 54. Brock PR, Maibach R, Childs M, et al. Sodium thiosulfate for protection from cisplatin-induced hearing loss. N Engl J Med 2018;378:2376–85.[doi:10.1056/NEJMoa1801109](http://dx.doi.org/10.1056/NEJMoa1801109)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29924955 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1056/NEJMoa1801109&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 55. Freyer DR, Chen L, Krailo MD, et al. Effects of sodium thiosulfate versus observation on development of cisplatin-induced hearing loss in children with cancer (ACCL0431): a multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol 2017;18:63–74.[doi:10.1016/S1470-2045(16)30625-8](http://dx.doi.org/10.1016/S1470-2045(16)30625-8)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27914822 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1016/S1470-2045(16)30625-8&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 56. Freyer DR, Brock PR, Chang KW, et al. Prevention of cisplatin-induced ototoxicity in children and adolescents with cancer: a clinical practice guideline. Lancet Child Adolesc Health 2020;4:141–50.[doi:10.1016/S2352-4642(19)30336-0](http://dx.doi.org/10.1016/S2352-4642(19)30336-0)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31866182 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 57. Katzenstein HM, Chang KW, Krailo M, et al. Amifostine does not prevent platinum-induced hearing loss associated with the treatment of children with hepatoblastoma: a report of the intergroup hepatoblastoma study P9645 as a part of the children's Oncology Group. Cancer 2009;115:5828–35.[doi:10.1002/cncr.24667](http://dx.doi.org/10.1002/cncr.24667)pmid:http://www.ncbi.nlm.nih.gov/pubmed/19813275 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1002/cncr.24667&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=19813275&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) [Web of Science](http://wjps.bmj.com/lookup/external-ref?access_num=000272545400028&link_type=ISI) 58. Zhu HT, Xiao XM. Research progress of PI3K/Akt signaling pathway and malignant solid tumors in children. Chin J Pediatr Surg 2009;8:57–9.[doi:10.3969/j.issn.1671-6353.2009.06.020](http://dx.doi.org/10.3969/j.issn.1671-6353.2009.06.020) 59. Liu J, Li Y, Yang X, et al. Effects of ginkgol C17:1 on cisplatin-induced autophagy and apoptosis in HepG2 cells. Oncol Lett 2018;15:1021–9.[doi:10.3892/ol.2017.7398](http://dx.doi.org/10.3892/ol.2017.7398)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29399162 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.3892/ol.2017.7398&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 60. Li Y, Zhang X, Yang X, et al. Differential effects of ginkgol C17:1 on cisplatin-induced cytotoxicity: protecting human normal L02 hepatocytes versus sensitizing human hepatoma HepG2 cells. Oncol Lett 2019;17:3181–90.[doi:10.3892/ol.2019.9974](http://dx.doi.org/10.3892/ol.2019.9974)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30867748 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 61. Molina L, Yang H, Adebayo Michael AO, et al. mTOR inhibition affects Yap1-β-catenin-induced hepatoblastoma growth and development. Oncotarget 2019;10:1475–90.[doi:10.18632/oncotarget.26668](http://dx.doi.org/10.18632/oncotarget.26668)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30863496 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.18632/oncotarget.26668&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=30863496&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 62. Zhang C, Jia X, Wang K, et al. Polyphyllin VII induces an autophagic cell death by activation of the JNK pathway and inhibition of PI3K/Akt/mTOR pathway in HepG2 cells. PLoS One 2016;11:e0147405. [doi:10.1371/journal.pone.0147405](http://dx.doi.org/10.1371/journal.pone.0147405)pmid:http://www.ncbi.nlm.nih.gov/pubmed/26808193 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1371/journal.pone.0147405&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 63. Fu H, Zhang J, Pan T, et al. miR‑378a enhances the sensitivity of liver cancer to sorafenib by targeting VEGFR, Pdgfrβ and c‑Raf. Mol Med Rep 2018;17:4581–8.[doi:10.3892/mmr.2018.8390](http://dx.doi.org/10.3892/mmr.2018.8390)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29328399 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 64. Cui Y, Lu P, Song G, et al. Involvement of PI3K/Akt, ERK and p38 signaling pathways in emodin-mediated extrinsic and intrinsic human hepatoblastoma cell apoptosis. Food Chem Toxicol 2016;92:26–37.[doi:10.1016/j.fct.2016.03.013](http://dx.doi.org/10.1016/j.fct.2016.03.013)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27032576 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1016/j.fct.2016.03.013&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=27032576&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 65. Stafman LL, Williams AP, Marayati R, et al. Pp2A activation alone and in combination with cisplatin decreases cell growth and tumor formation in human HUH6 hepatoblastoma cells. PLoS One 2019;14:e0214469. [doi:10.1371/journal.pone.0214469](http://dx.doi.org/10.1371/journal.pone.0214469)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30969990 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 66. Deng S, Tang S, Zhang S, et al. Furazolidone induces apoptosis through activating reactive oxygen species-dependent mitochondrial signaling pathway and suppressing PI3K/Akt signaling pathway in HepG2 cells. Food Chem Toxicol 2015;75:173–86.[doi:10.1016/j.fct.2014.11.019](http://dx.doi.org/10.1016/j.fct.2014.11.019)pmid:http://www.ncbi.nlm.nih.gov/pubmed/25434308 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1016/j.fct.2014.11.019&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 67. Indersie E, Lesjean S, Hooks KB, et al. MicroRNA therapy inhibits hepatoblastoma growth in vivo by targeting β-catenin and Wnt signaling. Hepatol Commun 2017;1:168–83.[doi:10.1002/hep4.1029](http://dx.doi.org/10.1002/hep4.1029)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29404451 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 68. Russell JO, Monga SP. Wnt/β-Catenin Signaling in Liver Development, Homeostasis, and Pathobiology. Annu Rev Pathol 2018;13:351–78.[doi:10.1146/annurev-pathol-020117-044010](http://dx.doi.org/10.1146/annurev-pathol-020117-044010)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29125798 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1146/annurev-pathol-020117-044010&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 69. Ellerkamp V, Lieber J, Nagel C, et al. Pharmacological inhibition of beta-catenin in hepatoblastoma cells. Pediatr Surg Int 2013;29:141–9.[doi:10.1007/s00383-012-3237-9](http://dx.doi.org/10.1007/s00383-012-3237-9)pmid:http://www.ncbi.nlm.nih.gov/pubmed/23266718 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1007/s00383-012-3237-9&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=23266718&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 70. Gödeke J, Maier S, Eichenmüller M, et al. Epigallocatechin-3-Gallate inhibits hepatoblastoma growth by reactivating the Wnt inhibitor SFRP1. Nutr Cancer 2013;65:1200–7.[doi:10.1080/01635581.2013.828085](http://dx.doi.org/10.1080/01635581.2013.828085)pmid:http://www.ncbi.nlm.nih.gov/pubmed/24127655 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1080/01635581.2013.828085&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=24127655&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 71. Muñoz M, Rosso M, Coveñas R. The NK-1 receptor: a new target in cancer therapy. Curr Drug Targets 2011;12:909–21.[doi:10.2174/138945011795528796](http://dx.doi.org/10.2174/138945011795528796)pmid:http://www.ncbi.nlm.nih.gov/pubmed/21226668 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.2174/138945011795528796&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=21226668&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) [Web of Science](http://wjps.bmj.com/lookup/external-ref?access_num=000291640000014&link_type=ISI) 72. Coveñas R, Muñoz M. Cancer progression and substance P. Histol Histopathol 2014;29:881–90.[doi:10.14670/HH-29.881](http://dx.doi.org/10.14670/HH-29.881)pmid:http://www.ncbi.nlm.nih.gov/pubmed/24535838 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.14670/HH-29.881&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 73. Berger M, Neth O, Ilmer M, et al. Hepatoblastoma cells express truncated neurokinin-1 receptor and can be growth inhibited by aprepitant in vitro and in vivo. J Hepatol 2014;60:985–94.[doi:10.1016/j.jhep.2013.12.024](http://dx.doi.org/10.1016/j.jhep.2013.12.024)pmid:http://www.ncbi.nlm.nih.gov/pubmed/24412605 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1016/j.jhep.2013.12.024&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=24412605&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 74. Ilmer M, Garnier A, Vykoukal J, et al. Targeting the neurokinin-1 receptor compromises canonical Wnt signaling in hepatoblastoma. Mol Cancer Ther 2015;14:2712–21.[doi:10.1158/1535-7163.MCT-15-0206](http://dx.doi.org/10.1158/1535-7163.MCT-15-0206)pmid:http://www.ncbi.nlm.nih.gov/pubmed/26516161 [Abstract/FREE Full Text](http://wjps.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6Im1vbGNhbnRoZXIiO3M6NToicmVzaWQiO3M6MTA6IjE0LzEyLzI3MTIiO3M6NDoiYXRvbSI7czoyMjoiL3dqcHMvNC8zL2UwMDAyMjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 75. Garnier A, Ilmer M, Kappler R, et al. Therapeutic innovations for targeting hepatoblastoma. Anticancer Res 2016;36:5577–92.[doi:10.21873/anticanres.11143](http://dx.doi.org/10.21873/anticanres.11143)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27793881 [Abstract/FREE Full Text](http://wjps.bmj.com/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImFudGljYW5yZXMiO3M6NToicmVzaWQiO3M6MTA6IjM2LzExLzU1NzciO3M6NDoiYXRvbSI7czoyMjoiL3dqcHMvNC8zL2UwMDAyMjAuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 76. Armeanu-Ebinger S, Hoh A, Wenz J, et al. Targeting EpCAM (CD326) for immunotherapy in hepatoblastoma. Oncoimmunology 2013;2:e22620. [doi:10.4161/onci.22620](http://dx.doi.org/10.4161/onci.22620)pmid:http://www.ncbi.nlm.nih.gov/pubmed/23482411 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=23482411&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 77. Baeuerle PA, Gires O, EpCAM GO. Epcam (CD326) finding its role in cancer. Br J Cancer 2007;96:417–23.[doi:10.1038/sj.bjc.6603494](http://dx.doi.org/10.1038/sj.bjc.6603494)pmid:http://www.ncbi.nlm.nih.gov/pubmed/17211480 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.1038/sj.bjc.6603494&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=17211480&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) [Web of Science](http://wjps.bmj.com/lookup/external-ref?access_num=000244100300003&link_type=ISI) 78. CM O, Chon HJ, Kim C. Combination immunotherapy using oncolytic virus for the treatment of advanced solid tumors. Int J Mol Sci 2020;21.[doi:10.3390/ijms21207743](http://dx.doi.org/10.3390/ijms21207743) 79. Ortiz MV, Roberts SS, Glade Bender J, et al. Immunotherapeutic targeting of GPC3 in pediatric solid embryonal tumors. Front Oncol 2019;9:108. [doi:10.3389/fonc.2019.00108](http://dx.doi.org/10.3389/fonc.2019.00108)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30873384 [CrossRef](http://wjps.bmj.com/lookup/external-ref?access_num=10.3389/fonc.2019.00108&link_type=DOI) [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=30873384&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 80. Tsuchiya N, Hosono A, Yoshikawa T, et al. Phase I study of glypican-3-derived peptide vaccine therapy for patients with refractory pediatric solid tumors. Oncoimmunology 2017;7:e1377872. [doi:10.1080/2162402X.2017.1377872](http://dx.doi.org/10.1080/2162402X.2017.1377872)pmid:http://www.ncbi.nlm.nih.gov/pubmed/29296538 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 81. Shimizu Y, Suzuki T, Yoshikawa T, et al. Next-Generation cancer immunotherapy targeting glypican-3. Front Oncol 2019;9:248. [doi:10.3389/fonc.2019.00248](http://dx.doi.org/10.3389/fonc.2019.00248)pmid:http://www.ncbi.nlm.nih.gov/pubmed/31024850 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 82. Abou-Alfa GK, Puig O, Daniele B, et al. Randomized phase II placebo controlled study of codrituzumab in previously treated patients with advanced hepatocellular carcinoma. J Hepatol 2016;65:289–95.[doi:10.1016/j.jhep.2016.04.004](http://dx.doi.org/10.1016/j.jhep.2016.04.004)pmid:http://www.ncbi.nlm.nih.gov/pubmed/27085251 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 83. Pradier MF, Reis B, Jukofsky L, et al. Case-Control Indian buffet process identifies biomarkers of response to Codrituzumab. BMC Cancer 2019;19:278. [doi:10.1186/s12885-019-5472-0](http://dx.doi.org/10.1186/s12885-019-5472-0)pmid:http://www.ncbi.nlm.nih.gov/pubmed/30922327 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom) 84. Abou-Alfa GK, Yen C-J, Hsu C-H, et al. Phase Ib study of codrituzumab in combination with sorafenib in patients with non-curable advanced hepatocellular carcinoma (HCC). Cancer Chemother Pharmacol 2017;79:421–9.[doi:10.1007/s00280-017-3241-9](http://dx.doi.org/10.1007/s00280-017-3241-9)pmid:http://www.ncbi.nlm.nih.gov/pubmed/28120036 [PubMed](http://wjps.bmj.com/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fwjps%2F4%2F3%2Fe000220.atom)