1
|
Moghadamnia M, Dashti-Khavidaki S, Alimadadi H. Role of mTOR Inhibitors in Pediatric Liver Transplant Recipients: A Systematic Review. Paediatr Drugs 2024; 26:673-693. [PMID: 39251556 DOI: 10.1007/s40272-024-00648-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/27/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Immunosuppressive medications play a crucial role in determining both organ and patient survival following liver transplantation (LT). Typically, immunosuppressive protocols for pediatric LT patients rely on calcineurin inhibitors (CNIs). While inhibitors of mammalian target of rapamycin (mTOR) have demonstrated beneficial outcomes in adult recipients of liver allografts, such as improved renal function post-LT, their application in pediatric liver transplant recipients is a subject of debate due to uncertain efficacy and potential adverse effects. OBJECTIVES This review evaluates the potential roles of mTOR inhibitors in the context of pediatric LT patients. METHODS This systematic review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) protocol for conduct and reporting. Databases until 31 August 2023 were searched using specific terms and keywords. All clinical studies focusing on mTOR inhibitors in pediatric LT were included. RESULTS Out of 888 identified articles, 30 studies, involving 386 children who had undergone liver transplantation and received mTOR-inhibitor-based immunosuppressive regimens, met the inclusion criteria. The beneficial impacts of switching from a CNI to an mTOR inhibitor or adding an mTOR inhibitor to CNI-reduced immunosuppression in LT pediatric patients with impaired kidney function are controversial, and high-powered clinical studies are need. It appears that enhancing immunosuppression by adding an mTOR inhibitor to CNI is helpful for pediatric LT recipients who are experiencing refractory acute rejection or chronic rejection. mTOR-inhibitor-containing regimens failed to reduce the occurrence of post-transplant lymphoproliferative disorders (PTLD) among children with LT that may be due to concomitant high CNI concentration among studied patients. The effectiveness of mTOR inhibitors in treating PTLD remains uncertain; however, in patients with PTLD who are at high risk of rejection, mTOR inhibitors may be administered. Conversion to or the addition of mTOR inhibitors to maintenance immunosuppression seems to be suitable for pediatric patients who received a transplant due to hepatic malignancies such as hepatoblastoma or hepatocellular carcinoma or for those with post-transplant primary or recurrent malignancies. Switching to an mTOR inhibitor may improve some CNI-related adverse effects such as gingival hyperplasia, neurotoxicity, nephropathy, hypertrophic cardiomyopathy, or thrombotic microangiopathy. CONCLUSION Although the exact role of mTOR inhibitors among pediatric patients who have received a liver transplant needs further study, two algorithms are presented in this review to guide conversion from CNIs to mTOR inhibitors or the addition of mTOR inhibitor to a CNI-minimization immunosuppressive regimen for pediatric patients who may benefit from this class of drugs. This review mainly consisted of retrospective studies with inadequate sample sizes and lacked a control group, which represents the main limitation of this study.
Collapse
Affiliation(s)
- Marjan Moghadamnia
- Department of Pharmacotherapy, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Simin Dashti-Khavidaki
- Liver Transplantation Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hosein Alimadadi
- Department of Gastroenterology, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| |
Collapse
|
2
|
Cillo U, Carraro A, Avolio AW, Cescon M, Di Benedetto F, Giannelli V, Magistri P, Nicolini D, Vivarelli M, Lanari J. Immunosuppression in liver transplant oncology: position paper of the Italian Board of Experts in Liver Transplantation (I-BELT). Updates Surg 2024; 76:725-741. [PMID: 38713396 DOI: 10.1007/s13304-024-01845-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/31/2023] [Indexed: 05/08/2024]
Abstract
Liver transplant oncology (TO) represents an area of increasing clinical and scientific interest including a heterogeneous group of clinical-pathological settings. Immunosuppressive management after LT is a key factor relevantly impacting result. However, disease-related guidance is still lacking, and many open questions remain in the field. Based on such a substantial lack of solid evidences, the Italian Board of Experts in Liver Transplantation (I-BELT) (a working group including representatives of all national transplant centers), unprecedently promoted a methodologically sound consensus conference on the topic, based on the GRADE approach. The group final recommendations are herein presented and commented. The 18 PICOs and Statements and their levels of evidence and grades of recommendation are reported and grouped into seven areas: (1) risk stratification by histopathological and bio-molecular parameters and role of mTORi post-LT; (2) steroids and HCC recurrence; (3) management of immunosuppression when HCC recurs after LT; (4) mTORi monotherapy; (5) machine perfusion and HCC recurrence after LT; (6) physiopathology of tumor-infiltrating lymphocytes and immunosuppression, the role of inflammation; (7) immunotherapy in liver transplanted patients. The interest in mammalian targets of rapamycin inhibitors (mTORi), for steroid avoidance and the need for a reduction to CNI exposure emerged from the consensus process. A selected list of unmet needs prompting further investigations have also been developed. The so far heterogeneous and granular approach to immunosuppression in oncologic patients deserves greater efforts for a more standardized therapeutic response to the different clinical scenarios. This consensus process makes a first unprecedented step in this direction, to be developed on a larger scale.
Collapse
Affiliation(s)
- Umberto Cillo
- Department of Surgical, Oncological and Gastroenterological Sciences, General Surgery 2 Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Padua University Hospital, Via Giustiniani 2, 34128, Padua, PD, Italy.
| | - Amedeo Carraro
- Liver Transplant Unit, Department of Surgery and Oncology, University Hospital Trust of Verona, Verona, Italy
| | - Alfonso W Avolio
- Department of General Surgery and Liver Transplantation, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Matteo Cescon
- General Surgery and Transplantation Unit, Department of Medical and Surgical Sciences, Azienda Ospedaliero-Universitaria-Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Fabrizio Di Benedetto
- Hepatopancreatobiliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Valerio Giannelli
- Liver Unit, Department of Liver Transplant, Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
| | - Paolo Magistri
- Hepatopancreatobiliary Surgery and Liver Transplantation Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniele Nicolini
- Hepatobiliary and Abdominal Transplantation Surgery, Department of Experimental and Clinical Medicine, Riuniti Hospital, Polytechnic University of Marche, Ancona, Italy
| | - Marco Vivarelli
- Hepatobiliary and Abdominal Transplantation Surgery, Department of Experimental and Clinical Medicine, Riuniti Hospital, Polytechnic University of Marche, Ancona, Italy
| | - Jacopo Lanari
- Department of Surgical, Oncological and Gastroenterological Sciences, General Surgery 2 Hepato-Pancreato-Biliary Surgery and Liver Transplantation, Padua University Hospital, Via Giustiniani 2, 34128, Padua, PD, Italy
| |
Collapse
|
3
|
Abstract
Pediatric precision oncology has provided a greater understanding of the wide range of molecular alterations in difficult-to-treat or rare tumors with the aims of increasing survival as well as decreasing toxicity and morbidity from current cytotoxic therapies. In this article, the authors discuss the current state of pediatric precision oncology which has increased access to novel targeted therapies while also providing a framework for clinical implementation in this unique population. The authors evaluate the targetable mutations currently under investigation-with a focus on pediatric solid tumors-and discuss the key surgical implications associated with novel targeted therapies.
Collapse
Affiliation(s)
- William G Lee
- Department of Surgery, Cedars-Sinai Medical Center, 116 North Robertson Boulevard, Suite PACT 700, Los Angeles, CA 90048, USA. https://twitter.com/william_ghh_lee
| | - Eugene S Kim
- Division of Pediatric Surgery, Department of Surgery, Cedars-Sinai Medical Center, 116 North Robertson Boulevard, Suite PACT 700, Los Angeles, CA 90048, USA.
| |
Collapse
|
4
|
Huang H, Yan J, Xu X, Feng Y, Liu H, Liu J, Xie M, Chen L, Xiang D, Peng W, Zeng L, Zeng Y, Chen F, Zhang S, Liu Q. Everolimus inhibits hepatoblastoma by inducing autophagy-dependent ferroptosis. Drug Dev Res 2024; 85:e22140. [PMID: 38349263 DOI: 10.1002/ddr.22140] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/22/2023] [Accepted: 12/08/2023] [Indexed: 02/15/2024]
Abstract
Everolimus, a known inhibitor of the mammalian target of rapamycin (mTOR), has shown uncertain efficacy in treating hepatoblastoma. This study delves into the potential anti-hepatoblastoma properties of everolimus and its intricate relationship with autophagy and ferroptosis, both in vitro and in vivo. In vivo, tumor tissue from hepatoblastoma patient and human hepatoblastoma cell line HuH-6 were xenografted into nude mice to establish xenograft models for observing the effect of everolimus on tumor growth. In vitro, HuH-6 cells were cultured to evaluate the anti-hepatoblastoma activity of everolimus. Transmission electron microscopy and microtubule-associated proteins 1 light chain 3 (LC3), beclin 1, and p62 protein expressions were employed to investigate autophagy. Additionally, indicators of cell apoptosis, reactive oxygen species (ROS) and proteins associated with ferroptosis were measured to evaluate ferroptosis. The results demonstrate that everolimus treatment effectively induced the formation of autophagosomes in hepatoblastoma cells, upregulated the LC3II/I ratio and beclin 1 expression, and downregulated p62 expression, indicating an enhanced autophagy level both in vitro and in vivo. Furthermore, everolimus treatment induced cell apoptosis, increased ROS level, elevated concentrations of malondialdehyde, 4-hydroxynonenal, and iron content, while reducing the ratio of glutathione/oxidized glutathione, and downregulating the protein expression of glutathione peroxidase 4 and solute carrier family 7 member 11, suggesting its ability to induce ferroptosis in hepatoblastoma cells. Importantly, the induction of ferroptosis by everolimus was significantly reversed in the presence of autophinib, an autophagy inhibitor, indicating the autophagy-dependent of everolimus-induced ferroptosis. Taken together, these findings suggest that everolimus holds promise as an effective anti-hepatoblastoma drug, with its mechanism of action potentially involving the induction of autophagy-dependent ferroptosis in hepatoblastoma cells.
Collapse
Affiliation(s)
- Haijin Huang
- Department of Pediatric Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Jiangxi Provincial Clinical Research Center for Vascular Anomalies, The First Affiliated Hospital of GanNan Medical University, Ganzhou, Jiangxi, China
| | - Jinlong Yan
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xianyun Xu
- Department of Clinical Laboratory, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Yanping Feng
- Department of Neurological Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Haijin Liu
- Department of Pediatric Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jianping Liu
- Department of General Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Mingfeng Xie
- Jiangxi Provincial Clinical Research Center for Vascular Anomalies, The First Affiliated Hospital of GanNan Medical University, Ganzhou, Jiangxi, China
- Chinese & Western Integrative Medicine Discipline, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Jiangxi Key Laboratory of TCM for Prevention and Treatment on Hemangioma, Nanchang, Jiangxi, China
- Integrated Chinese and Western Medicine Institute for Children Health & Drug Innovation, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Leifeng Chen
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Deng Xiang
- Department of General Surgery, The Affiliated Children's Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Wei Peng
- Department of Pediatric Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Linshan Zeng
- Department of Pediatric Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yong Zeng
- Department of Pediatric Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Feng Chen
- Department of Pediatric Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Shouhua Zhang
- Department of General Surgery, The Affiliated Children's Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Qian Liu
- Jiangxi Provincial Clinical Research Center for Vascular Anomalies, The First Affiliated Hospital of GanNan Medical University, Ganzhou, Jiangxi, China
- Chinese & Western Integrative Medicine Discipline, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Jiangxi Key Laboratory of TCM for Prevention and Treatment on Hemangioma, Nanchang, Jiangxi, China
- Integrated Chinese and Western Medicine Institute for Children Health & Drug Innovation, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
5
|
Fang J, Singh S, Cheng C, Natarajan S, Sheppard H, Abu-Zaid A, Durbin AD, Lee HW, Wu Q, Steele J, Connelly JP, Jin H, Chen W, Fan Y, Pruett-Miller SM, Rehg JE, Koo SC, Santiago T, Emmons J, Cairo S, Wang R, Glazer ES, Murphy AJ, Chen T, Davidoff AM, Armengol C, Easton J, Chen X, Yang J. Genome-wide mapping of cancer dependency genes and genetic modifiers of chemotherapy in high-risk hepatoblastoma. Nat Commun 2023; 14:4003. [PMID: 37414763 PMCID: PMC10326052 DOI: 10.1038/s41467-023-39717-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/27/2023] [Indexed: 07/08/2023] Open
Abstract
A lack of relevant genetic models and cell lines hampers our understanding of hepatoblastoma pathogenesis and the development of new therapies for this neoplasm. Here, we report an improved MYC-driven hepatoblastoma-like murine model that recapitulates the pathological features of embryonal type of hepatoblastoma, with transcriptomics resembling the high-risk gene signatures of the human disease. Single-cell RNA-sequencing and spatial transcriptomics identify distinct subpopulations of hepatoblastoma cells. After deriving cell lines from the mouse model, we map cancer dependency genes using CRISPR-Cas9 screening and identify druggable targets shared with human hepatoblastoma (e.g., CDK7, CDK9, PRMT1, PRMT5). Our screen also reveals oncogenes and tumor suppressor genes in hepatoblastoma that engage multiple, druggable cancer signaling pathways. Chemotherapy is critical for human hepatoblastoma treatment. A genetic mapping of doxorubicin response by CRISPR-Cas9 screening identifies modifiers whose loss-of-function synergizes with (e.g., PRKDC) or antagonizes (e.g., apoptosis genes) the effect of chemotherapy. The combination of PRKDC inhibition and doxorubicin-based chemotherapy greatly enhances therapeutic efficacy. These studies provide a set of resources including disease models suitable for identifying and validating potential therapeutic targets in human high-risk hepatoblastoma.
Collapse
Affiliation(s)
- Jie Fang
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shivendra Singh
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Changde Cheng
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sivaraman Natarajan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Heather Sheppard
- Comparative Pathology Core, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ahmed Abu-Zaid
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Adam D Durbin
- Division of Molecular Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ha Won Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Qiong Wu
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jacob Steele
- Center for Advanced Genome Engineering (CAGE), St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jon P Connelly
- Center for Advanced Genome Engineering (CAGE), St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hongjian Jin
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Wenan Chen
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yiping Fan
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering (CAGE), St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jerold E Rehg
- Comparative Pathology Core, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Selene C Koo
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Teresa Santiago
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Joseph Emmons
- VPC Diagnostic Laboratory, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stefano Cairo
- Champions Oncology, 1330 Piccard dr, Rockville, MD, USA
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Disease, Hematology/Oncology & BMT, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Evan S Glazer
- Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, 910 Madison Ave., Suite 325, Memphis, TN, USA
| | - Andrew J Murphy
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, 910 Madison Ave., Suite 325, Memphis, TN, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Andrew M Davidoff
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, 910 Madison Ave., Suite 325, Memphis, TN, USA
- St Jude Graduate School of Biomedical Sciences, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Pathology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Carolina Armengol
- Childhood Liver Oncology Group, Germans Trias i Pujol Research Institute (IGTP), Translational Program in Cancer Research (CARE), Badalona, Spain
- CIBER, Hepatic and Digestive Diseases, Barcelona, Spain
- CIBERehd, Madrid, Spain
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- St Jude Graduate School of Biomedical Sciences, St Jude Children's Research Hospital, Memphis, TN, USA.
| | - Jun Yang
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA.
- St Jude Graduate School of Biomedical Sciences, St Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Pathology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
6
|
Zhu LR, Zheng W, Gao Q, Chen T, Pan ZB, Cui W, Cai M, Fang H. Epigenetics and genetics of hepatoblastoma: Linkage and treatment. Front Genet 2022; 13:1070971. [PMID: 36531231 PMCID: PMC9748487 DOI: 10.3389/fgene.2022.1070971] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 11/14/2022] [Indexed: 09/10/2024] Open
Abstract
Hepatoblastoma is a malignant embryonal tumor with multiple differentiation modes and is the clearest liver malignancy in children. However, little is known about genetic and epigenetic events in Hepatoblastoma. Increased research has recently demonstrated, unique genetic and epigenetic events in Hepatoblastoma, providing insights into its origin and precise treatment. Some genetic disorders and congenital factors are associated with the risk of Hepatoblastoma development, such as the Beckwith-Wiedemann syndrome, Familial Adenomatous polyposis, and Hemihypertrophy. Epigenetic modifications such as DNA modifications, histone modifications, and non-coding RNA regulation are also essential in the development of Hepatoblastoma. Herein, we reviewed genetic and epigenetic events in Hepatoblastoma, focusing on the relationship between these events and cancer susceptibility, tumor growth, and prognosis. By deciphering the genetic and epigenetic associations in Hepatoblastoma, tumor pathogenesis can be clarified, and guide the development of new anti-cancer drugs and prevention strategies.
Collapse
Affiliation(s)
- Li-ran Zhu
- Anhui Institute of Pediatric Research, Anhui Provincial Children’s Hospital, Hefei, China
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei, China
| | - Wanqun Zheng
- Department of Chinese Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qun Gao
- Department of Pediatric Oncology Surgery, Anhui Provincial Children’s Hospital, Hefei, China
| | - Tianping Chen
- Department of Hematology and Oncology, Anhui Provincial Children’s Hospital, Hefei, China
| | - Zhu-bin Pan
- Department of General Surgery, Anhui Provincial Children’s Hospital, Hefei, China
| | - Wei Cui
- Department of Scientific Research and Education, Anhui Provincial Children’s Hospital, Anhui Institute of Pediatric Research, Hefei, China
| | - Ming Cai
- Department of Pharmacy, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Hui Fang
- Anhui Institute of Pediatric Research, Anhui Provincial Children’s Hospital, Hefei, China
| |
Collapse
|
7
|
Li Q, Demir S, Del Río-Álvarez Á, Maxwell R, Wagner A, Carrillo-Reixach J, Armengol C, Vokuhl C, Häberle B, von Schweinitz D, Schmid I, Cairo S, Kappler R. Targeting the Unwindosome by Mebendazole Is a Vulnerability of Chemoresistant Hepatoblastoma. Cancers (Basel) 2022; 14:cancers14174196. [PMID: 36077733 PMCID: PMC9454988 DOI: 10.3390/cancers14174196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Resistance to conventional chemotherapy remains a huge challenge in the clinical management of hepatoblastoma, the most common liver tumor in childhood. By integrating the gene expression data of hepatoblastoma patients into the perturbation prediction tool Connectivity Map, we identified the clinical widely used anthelmintic mebendazole as a drug to circumvent chemoresistance in permanent and patient-derived xenograft cell lines that are resistant to cisplatin, the therapeutic backbone of hepatoblastoma treatment. Viability assays clearly indicated a potent reduction of tumor cell growth upon mebendazole treatment in a dose-dependent manner. The combination of mebendazole and cisplatin revealed a strong synergistic effect, which was comparable to the one seen with cisplatin and doxorubicin, the current treatment for high-risk hepatoblastoma patients. Moreover, mebendazole treatment resulted in reduced colony and tumor spheroid formation capabilities, cell cycle arrest, and induction of apoptosis of hepatoblastoma cells. Mechanistically, mebendazole causes blockage of microtubule formation and transcriptional downregulation of genes encoding the unwindosome, which are highly expressed in chemoresistant tumors. Most importantly, mebendazole significantly reduced tumor growth in a subcutaneous xenograft transplantation mouse model without side effects. In conclusion, our results strongly support the clinical use of mebendazole in the treatment of chemoresistant hepatoblastoma and highlight the potential theranostic value of unwindosome-associated genes.
Collapse
Affiliation(s)
- Qian Li
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
| | - Salih Demir
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
| | - Álvaro Del Río-Álvarez
- Childhood Liver Oncology Group, Health Sciences Research Institute Germans Trias i Pujol IGTP, 08916 Badalona, Spain
| | - Rebecca Maxwell
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
| | - Alexandra Wagner
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
| | - Juan Carrillo-Reixach
- Childhood Liver Oncology Group, Health Sciences Research Institute Germans Trias i Pujol IGTP, 08916 Badalona, Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), 28029 Madrid, Spain
| | - Carolina Armengol
- Childhood Liver Oncology Group, Health Sciences Research Institute Germans Trias i Pujol IGTP, 08916 Badalona, Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), 28029 Madrid, Spain
| | - Christian Vokuhl
- Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany
| | - Beate Häberle
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
| | - Dietrich von Schweinitz
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
| | - Irene Schmid
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
| | | | - Roland Kappler
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, 80337 Munich, Germany
- Correspondence: ; Tel.: +49-89-4400-57810
| |
Collapse
|
8
|
Boster JM, Superina R, Mazariegos GV, Tiao GM, Roach JP, Lovell MA, Greffe BS, Yanni G, Leung DH, Elisofon SA, McDiarmid SV, Gupta NA, Lobritto SJ, Lemoine C, Stoll JM, Vitola BE, Daniel JF, Sayed BA, Desai DM, Martin AE, Amin A, Anand R, Anderson SG, Sundaram SS. Predictors of survival following liver transplantation for pediatric hepatoblastoma and hepatocellular carcinoma: Experience from the Society of Pediatric Liver Transplantation (SPLIT). Am J Transplant 2022; 22:1396-1408. [PMID: 34990053 DOI: 10.1111/ajt.16945] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/28/2021] [Accepted: 12/28/2021] [Indexed: 01/25/2023]
Abstract
Management of unresectable pediatric hepatoblastoma (HB) and hepatocellular carcinoma (HCC) remains challenging. The Society of Pediatric Liver Transplantation (SPLIT) database was used to study survival predictors in pediatric liver transplantation (LT) for HB and HCC. Event-free survival (EFS), associated risk factors, and postoperative complications were studied in children requiring LT for HB/HCC at 16 SPLIT centers. Three-year EFS was 81% for HB (n = 157) and 62% for HCC (n = 18) transplants. Of HB transplants, 6.9% were PRETEXT II and 15.3% were POST-TEXT I/II. Tumor extent did not impact survival (p = NS). Salvage (n = 13) and primary HB transplants had similar 3-year EFS (62% versus 78%, p = NS). Among HCC transplants, 3-year EFS was poorer in older patients (38% in ≥8-year-olds vs 86% <8-year-olds) and those with larger tumors (48% for those beyond versus 83% within Milan criteria, p = NS). Risk of infection (HR 1.5, 95% CI 1.1-2.2, p = .02) and renal injury (HR 2.4, 95% CI 1.7-3.3, p < .001) were higher in malignant versus nonmalignant LT. Survival is favorable for pediatric HB and HCC LT, including outcomes after salvage transplant. Unexpected numbers of LTs occurred in PRE/POST-TEXT I/II tumors. Judicious patient selection is critical to distinguish tumors that are potentially resectable; simultaneously, we must advocate for patients with unresectable malignancies to receive organs.
Collapse
Affiliation(s)
- Julia M Boster
- Department of Pediatrics, Children's Hospital Colorado and University of Colorado, Aurora, Colorado, USA
| | - Riccardo Superina
- Department of Surgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - George V Mazariegos
- Department of Surgery, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gregory M Tiao
- Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jonathan P Roach
- Department of Surgery, Children's Hospital Colorado and University of Colorado, Aurora, Colorado, USA
| | - Mark A Lovell
- Department of Pathology, Children's Hospital Colorado and University of Colorado, Aurora, Colorado, USA
| | - Brian S Greffe
- Department of Pediatric Oncology, Children's Hospital Colorado and University of Colorado, Aurora, Colorado, USA
| | - George Yanni
- Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Daniel H Leung
- Department of Pediatrics, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas, USA
| | - Scott A Elisofon
- Department of Pediatrics, Boston Children's Hospital, Massachusetts, USA
| | - Suzanne V McDiarmid
- Department of Pediatrics, University of California and Los Angeles Mattel Children's Hospital, Los Angeles, California, USA
| | - Nitika A Gupta
- Department of Pediatrics, Children's Healthcare of Atlanta and Emory University School of Medicine, Atlanta, Georgia, USA
| | - Steven J Lobritto
- Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
| | - Caroline Lemoine
- Department of Surgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Janis M Stoll
- Department of Pediatrics, St. Louis Children's Hospital and Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bernadette E Vitola
- Department of Pediatrics, Medical College of Wisconsin and Children's Hospital of Wisconsin, Milwaukee, Wisconsin, USA
| | - James F Daniel
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri, USA
| | - Blayne A Sayed
- Department of Surgery, University Health Network and the Hospital for Sick Children, Toronto, Ontario, Canada
| | - Dev M Desai
- Department of Surgery, University of Texas Southwestern Medical Center and Children's Medical Center, Dallas, Texas, USA
| | - Abigail E Martin
- Department of Surgery, Nemours Children's Hospital Delaware and Sidney Kimmel Medical College of Thomas Jefferson University, Wilmington, Delaware, USA
| | - Arpit Amin
- Department of Surgery, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | - Shikha S Sundaram
- Department of Pediatrics, Children's Hospital Colorado and University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
9
|
Darici S, Alkhaldi H, Horne G, Jørgensen HG, Marmiroli S, Huang X. Targeting PI3K/Akt/mTOR in AML: Rationale and Clinical Evidence. J Clin Med 2020; 9:E2934. [PMID: 32932888 PMCID: PMC7563273 DOI: 10.3390/jcm9092934] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous hematopoietic malignancy characterized by excessive proliferation and accumulation of immature myeloid blasts in the bone marrow. AML has a very poor 5-year survival rate of just 16% in the UK; hence, more efficacious, tolerable, and targeted therapy is required. Persistent leukemia stem cell (LSC) populations underlie patient relapse and development of resistance to therapy. Identification of critical oncogenic signaling pathways in AML LSC may provide new avenues for novel therapeutic strategies. The phosphatidylinositol-3-kinase (PI3K)/Akt and the mammalian target of rapamycin (mTOR) signaling pathway, is often hyperactivated in AML, required to sustain the oncogenic potential of LSCs. Growing evidence suggests that targeting key components of this pathway may represent an effective treatment to kill AML LSCs. Despite this, accruing significant body of scientific knowledge, PI3K/Akt/mTOR inhibitors have not translated into clinical practice. In this article, we review the laboratory-based evidence of the critical role of PI3K/Akt/mTOR pathway in AML, and outcomes from current clinical studies using PI3K/Akt/mTOR inhibitors. Based on these results, we discuss the putative mechanisms of resistance to PI3K/Akt/mTOR inhibition, offering rationale for potential candidate combination therapies incorporating PI3K/Akt/mTOR inhibitors for precision medicine in AML.
Collapse
Affiliation(s)
- Salihanur Darici
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Hazem Alkhaldi
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| | - Gillian Horne
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| | - Heather G. Jørgensen
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| | - Sandra Marmiroli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Xu Huang
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| |
Collapse
|
10
|
Ueno T, Kodama T, Noguchi Y, Deguchi K, Nomura M, Saka R, Watanabe M, Tazuke Y, Bessho K, Okuyama H. Safety and Efficacy of Everolimus Rescue Treatment After Pediatric Living Donor Liver Transplantation. Transplant Proc 2020; 52:1829-1832. [PMID: 32571711 DOI: 10.1016/j.transproceed.2020.01.159] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 01/26/2020] [Indexed: 01/01/2023]
Abstract
PURPOSE Everolimus (EVR) is a derivative of sirolimus with a similar mechanism of action. The safety and efficacy of EVR after pediatric living donor liver transplantation (LDLT) are currently unknown. The purpose of this study was to examine the safety and efficacy of EVR as rescue therapy after pediatric LDLT. METHODS This study included patients younger than 19 years of age who received EVR after LDLT at our institution. EVR was administered as rescue treatment in addition to tacrolimus. In 21 patients, EVR dose, trough level, outcomes, and adverse effects were assessed. RESULTS Original diseases of patients consisted of biliary atresia (n = 11), Alagille syndrome (n = 3), fulminant hepatitis (n = 3), hepatoblastoma (n = 2), and other (n = 2). Mean age at transplant was 2.0 years (range 0.6-6.2 years). Mean age at initial EVR administration was 8.0 years (range 0.9-18.9 years). Indications for EVR use were graft fibrosis (n = 8), refractory acute cellular rejection (n = 5), renal sparing (n = 4), hepatoblastoma (n = 2), and chronic rejection (CR) (n = 2). Mean duration of administration was 17.1 months (range 2.1-60.4 months). Mean dose was 0.5 mg/m2 twice daily. Mean EVR trough level was 2.5 ng/mL (range 1.5-5.0 ng/mL). Liver function improved and fibrosis did not progress in all patients with CR. However, 14 patients (67%) experienced adverse effects that required EVR dose reduction or discontinuation. CONCLUSION EVR is tolerable for pediatric patients after LDLT with dose adjustment. EVR had a certain effect to relieve progression on CR. Further follow-up is required.
Collapse
Affiliation(s)
- Takehisa Ueno
- Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | - Tasuku Kodama
- Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuki Noguchi
- Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Koichi Deguchi
- Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Motonari Nomura
- Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ryuta Saka
- Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Miho Watanabe
- Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuko Tazuke
- Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kazuhiko Bessho
- Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiroomi Okuyama
- Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
11
|
A Transient Pseudosenescent Secretome Promotes Tumor Growth after Antiangiogenic Therapy Withdrawal. Cell Rep 2019; 25:3706-3720.e8. [PMID: 30590043 DOI: 10.1016/j.celrep.2018.12.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/21/2018] [Accepted: 12/05/2018] [Indexed: 01/07/2023] Open
Abstract
VEGF receptor tyrosine kinase inhibitors (VEGFR TKIs) approved to treat multiple cancer types can promote metastatic disease in certain limited preclinical settings. Here, we show that stopping VEGFR TKI treatment after resistance can lead to rebound tumor growth that is driven by cellular changes resembling senescence-associated secretory phenotypes (SASPs) known to promote cancer progression. A SASP-mimicking antiangiogenic therapy-induced secretome (ATIS) was found to persist during short withdrawal periods, and blockade of known SASP regulators, including mTOR and IL-6, could blunt rebound effects. Critically, senescence hallmarks ultimately reversed after long drug withdrawal periods, suggesting that the transition to a permanent growth-arrested senescent state was incomplete and the hijacking of SASP machinery ultimately transient. These findings may account for the highly diverse and reversible cytokine changes observed in VEGF inhibitor-treated patients, and suggest senescence-targeted therapies ("senotherapeutics")-particularly those that block SASP regulation-may improve outcomes in patients after VEGFR TKI failure.
Collapse
|
12
|
Huang Y, Wang Y, Feng Y, Wang P, He X, Ren H, Wang F. Role of Endoplasmic Reticulum Stress-Autophagy Axis in Severe Burn-Induced Intestinal Tight Junction Barrier Dysfunction in Mice. Front Physiol 2019; 10:606. [PMID: 31191335 PMCID: PMC6538921 DOI: 10.3389/fphys.2019.00606] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
Severe burn injury induces intestinal barrier dysfunction; however, the underlying mechanisms remain elusive. Our previous studies have shown that the intestinal epithelial tight junction (TJ) barrier dysfunction is associated with both endoplasmic reticulum (ER) stress and autophagy in severely burned mice, but the precise role of ER stress and autophagy in the burn-induced intestinal TJ barrier dysfunction needs to be determined. In this study, female C57/BL6 mice were assigned randomly to either sham burn or 30% total body surface area (TBSA) full-thickness burn. The effects of ER stress and autophagy on the intestinal epithelial TJ barrier were validated by inducing or inhibiting both ER stress and autophagy in mice treated with sham burn or burn injury. The intestinal permeability, expression, and localization of TJ proteins, ER stress, and autophagy were assessed by physiological, morphological, and biochemical analyses. The results showed that inducing ER stress with tunicamycin or thapsigargin caused the activation of autophagy, the increase of intestinal permeability, as well as the reduction and reorganization of TJ proteins in the sham-burned mice, and aggravated the burn-induced activation of autophagy, increase of intestinal permeability, as well as the reduction and reorganization of TJ proteins. In contrast, inhibiting ER stress with 4-phenylbutyrate alleviated the burn-induced activation of autophagy, increase of intestinal permeability, as well as the reduction and reorganization of TJ proteins. In addition, inducing autophagy with rapamycin resulted in the increase of intestinal permeability, as well as the reduction and reorganization of TJ proteins in the sham-burned mice, and aggravated the burn-induced increase of intestinal permeability as well as the reduction and reorganization of TJ proteins. However, inhibiting autophagy with 3-methyladenine attenuated the burn-induced increase of intestinal permeability, as well as the reduction and reorganization TJ proteins. It is suggested that the ER stress-autophagy axis contributes to the intestinal epithelial TJ barrier dysfunction after severe burn injury.
Collapse
Affiliation(s)
- Yalan Huang
- School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yu Wang
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yanhai Feng
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Pei Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaochong He
- School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hui Ren
- School of Nursing, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fengjun Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
13
|
Hendrickson RJ, Sujka J, Fischer R, Manalang M, Daniel J, Andrews WS. Indications and efficacy of conversion from tacrolimus- to sirolimus-based immunosuppression in pediatric patients who underwent liver transplantation for unresectable hepatoblastoma. Pediatr Transplant 2019; 23:e13369. [PMID: 30719825 DOI: 10.1111/petr.13369] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/19/2018] [Accepted: 12/21/2018] [Indexed: 11/30/2022]
Abstract
SRL-based immunosuppressive strategies in pediatric liver transplantation are not clearly defined, especially within the first year after liver transplant. TAC is the more common, traditional immunosuppressant used. However, SRL may modulate TAC-associated kidney injury and may also have antiproliferative properties that are valuable in the management of patients following liver transplantation for HB. We sought to determine whether early conversion from TAC to SRL was safe, effective, and beneficial in a subset of liver transplant recipients with unresectable HB exposed to CDDP-based chemotherapy. Between 2008 and 2013, six patients were transplanted for unresectable HB. All patients received at least one cycle of CDDP-based chemotherapy prior to transplant. All patients were switched from TAC- to SRL-based immunosuppression within 1 year of transplant. Five patients had improvement in their mGFR, while one patient had a slight decline. The improvement in mGFR was statistically significant. No adverse events were identified. Three patients had BPAR that responded to pulsed steroids. Historical controls showed similar rates of BPAR within the first year after transplant. There were no identified HB recurrences in the follow-up time period. Conversion from TAC to SRL appears to be safe and effective in this selected group of pediatric liver transplant recipients without adverse reaction or HB recurrences.
Collapse
Affiliation(s)
| | - Joseph Sujka
- Department of Surgery, Children's Mercy Hospital, Kansas City, Missouri
| | - Ryan Fischer
- Department of Gastroenterology and Hepatology, Children's Mercy Hospital, Kansas City, Missouri
| | - Michelle Manalang
- Department of Hematology and Oncology, Children's Mercy Hospital, Kansas City, Missouri
| | - James Daniel
- Department of Gastroenterology and Hepatology, Children's Mercy Hospital, Kansas City, Missouri
| | - Walter S Andrews
- Department of Surgery, Children's Mercy Hospital, Kansas City, Missouri
| |
Collapse
|
14
|
Molina L, Yang H, Adebayo Michael AO, Oertel M, Bell A, Singh S, Chen X, Tao J, Monga SP. mTOR inhibition affects Yap1-β-catenin-induced hepatoblastoma growth and development. Oncotarget 2019; 10:1475-1490. [PMID: 30863496 PMCID: PMC6407673 DOI: 10.18632/oncotarget.26668] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 01/22/2019] [Indexed: 12/15/2022] Open
Abstract
Hepatoblastoma (HB) is the most common pediatric liver malignancy. Around 80% of HB demonstrate simultaneous activation of β-catenin and Yes-associated protein 1 (Yap1). The mechanism by which these signaling pathways contribute to HB pathogenesis remain obscure. Recently, mTORC1 activation was reported in human HB cells and in a murine HB model driven by β-catenin and Yap1. Here, we directly investigate the therapeutic impact of mTOR inhibition following HB development in the Yap1-β-catenin model. HB were established by hydrodynamic tail vein injection of Sleeping Beauty transposase and plasmids coding for ΔN90-β-catenin and S127A-Yap1. Five weeks after injection, when HB were evident, mice were randomized into Rapamycin diet-fed or basal-diet-fed groups for 5-weeks. Tumor growth was monitored via ultrasound imaging and mice in both groups were euthanized after 5-weeks for molecular analysis. Transcriptomic analysis showed a strong correlation in gene expression between HB in the Yap1-β-catenin model and HB patient cohorts. Rapamycin treatment decreased HB burden, almost normalizing liver weight to body weight ratio. Ultrasound imaging showed reduction in tumor growth over the duration of Rapamycin treatment as compared to controls. Majority of HB in the controls exhibited crowded fetal or embryonal histology, while remnant tumors in the experimental group showed well-differentiated fetal morphology. Immunohistochemistry confirmed inhibition of mTORC1 in the Rapamycin group. Thus, Rapamycin reduces HB in a clinically relevant model driven by β-catenin and Yap1, supporting use of mTORC1 inhibitors in their therapy. We also show the utility of standard and 3D ultrasound imaging for monitoring liver tumors in mice.
Collapse
Affiliation(s)
- Laura Molina
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hong Yang
- Department of Medical Ultrasonics, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | | | - Michael Oertel
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh PA, USA
| | - Aaron Bell
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sucha Singh
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, CA, USA
| | - Junyan Tao
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Satdarshan P.S. Monga
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh PA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
15
|
Ortiz D, Harden A, Corrales-Medina FF, Saigal G, Tekin A, Garcia J. Pediatric patient with end-stage kidney disease secondary to Eagle-Barrett syndrome and metastatic unresectable hepatoblastoma treated successfully with chemotherapy and liver-kidney transplant. Pediatr Transplant 2018; 22. [PMID: 29356335 DOI: 10.1111/petr.13123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/13/2017] [Indexed: 11/30/2022]
Abstract
HBL is the most common malignant liver neoplasm in children. The etiology of HBL is largely unknown but there are certain syndromes, such as Beckwith-Wiedemann syndrome, that have been clearly associated with an increased incidence of this malignancy. EBS, also known as prune belly syndrome, is a congenital anomaly characterized by lax abdominal musculature, bilateral cryptorchidism requiring, in some cases, hemodialysis due to significant kidney and urinary tract dysfunctions. Despite an improvement on the survival rates of patients with advanced-stage HBL, the presence of concomitant end-stage renal disease that occurs in patients with EBS constitutes a therapeutic challenge for the clinician not only due to the use of nephrotoxic chemotherapy but also due to the potential need for multi-organ transplant. We report case of a 2-year-old male patient with EBS diagnosed with stage IV, metastatic HBL successfully treated with multi-agent chemotherapy while on dialysis whom then underwent a simultaneous liver-kidney transplant followed by adjuvant chemotherapy. Ultimately, the patient achieved cancer remission with normalization of his renal function. Our report emphasizes that patients with HBL in the setting of EBS will not only require careful kidney function monitoring while receiving chemotherapy, but they might also need to undergo multi-organ transplantation in order to achieve adequate cancer control and also normalization of their kidney function. Awareness of this unusual association calls for further investigation to potentially establish a genetic association between these two disease processes.
Collapse
Affiliation(s)
- Daniel Ortiz
- Department of Pediatrics, University of Miami-Holtz Children's Hospital, Miami, FL, USA.,Division of Pediatric Hematology-Oncology, University of Miami-Miller School of Medicine, Miami, FL, USA
| | - Avis Harden
- Department of Pediatrics, University of Miami-Holtz Children's Hospital, Miami, FL, USA
| | - Fernando F Corrales-Medina
- Department of Pediatrics, University of Miami-Holtz Children's Hospital, Miami, FL, USA.,Division of Pediatric Hematology-Oncology, University of Miami-Miller School of Medicine, Miami, FL, USA
| | - Gaurav Saigal
- Section of Pediatric Radiology, Department of Radiology, University of Miami, Miami, FL, USA
| | - Akin Tekin
- Division of Liver/GI Transplantation, Department of Surgery, Miami Transplant Institute, University of Miami, Miami, FL, USA
| | - Jennifer Garcia
- Department of Pediatrics, University of Miami-Holtz Children's Hospital, Miami, FL, USA.,Division of Pediatric Gastroenterology, Miami Transplant Institute, University of Miami, Miami, FL, USA
| |
Collapse
|
16
|
Role of autophagy and its molecular mechanisms in mice intestinal tract after severe burn. J Trauma Acute Care Surg 2017; 83:716-724. [PMID: 28930963 DOI: 10.1097/ta.0000000000001624] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Severe burn can lead to hypoxia/ischemia of intestinal mucosa. Autophagy is the process of intracellular degradation, which is essential for cell survival under stresses, such as hypoxia/ischemia and nutrient deprivation. The present study was designed to investigate whether there were changes in intestinal autophagy after severe burn in mice and further to explore the effect and molecular mechanisms of autophagy on intestinal injury. METHODS This study includes three experiments. Kunming species mice were subjected to 30% total body surface area third-degree burn. First, we determined protein of LC3 (light chain 3), beclin-1, and cleaved-caspase3 by Western blotting and immunohistochemical (paraffin) staining to investigate whether there were changes in intestinal autophagy after severe burn in mice. Then, changes of the status of enteric damage postburn were measured by observing intestinal mucosa morphology under a magnifier, hematoxylin and eosin staining, enzyme-linked immunosorbent assay, Western blotting under the condition that the intestinal autophagy was respectively activated by rapamycin and inhibited by 3-methyladenine. Finally, protein of the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway, LC3-II and beclin-1 were assayed, and mice were treated with compound C before burn. RESULTS The protein of LC3 and beclin-1 were observed at 1 hour postburn and increased to peak-point at 24 hours, reaching the normal level at 96 hours. The cleaved caspase-3 expression increased at 1 hour postburn, but the peak point occurred at 12 hours and had dropped to normal level at 72 hours. In addition, rapamycin enhanced intestinal autophagy and alleviated burn-induced gut damage, while 3-methyladenine showed the against behavior. The AMPK/mTOR pathway which was inhibited decreased the expression of phosphorylated AMPK, LC3-II, and beclin-1, increasing the expression of phosphorylated mTOR. CONCLUSION Intestinal autophagy is activated and response to intestinal apoptosis after serious burn, which alleviated burn-induced intestinal injury. The AMPK/mTOR pathway may involve in the activation of burn-induced autophagy. LEVEL OF EVIDENCE Therapeutic/care management, levels of evidence are not applicable to some studies, such as in vitro work, animal models, cadaver studies.
Collapse
|
17
|
Affiliation(s)
- Matthias Ilmer
- Department of General, Visceral, and Transplantation Surgery, Hospital of the LMU Munich, Munich, Germany
| | - Michael Berger
- Department of Pediatric Surgery, Research Laboratories, Dr. von Hauner Children's Hospital, Ludwig Maximilians University Munich, Munich, Germany.,Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
18
|
Liu P, Calvisi DF, Kiss A, Cigliano A, Schaff Z, Che L, Ribback S, Dombrowski F, Zhao D, Chen X. Central role of mTORC1 downstream of YAP/TAZ in hepatoblastoma development. Oncotarget 2017; 8:73433-73447. [PMID: 29088718 PMCID: PMC5650273 DOI: 10.18632/oncotarget.20622] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 08/21/2017] [Indexed: 11/25/2022] Open
Abstract
Hepatoblastoma (HB) is the most common type of liver malignancy in children. Recent studies suggest that activation of Yes-associated protein (YAP) is a major molecular event in HB development, as activated YAP synergizes with mutant β-catenin to promote HB formation in mice (YAP/β-catenin). However, how YAP regulates HB development remains poorly defined. Similarly, de-regulation of mammalian target of rapamycin complex 1 (mTORC1) signaling has been implicated in multiple tumor types, but its functional role in HB development is scarcely understood. In the present study, we found that mTORC1 is activated in human HB cells and YAP/β-catenin-induced mouse HB tumor tissues. mTOR inhibitor MLN0128 significantly inhibits human HB cell growth in vitro. Furthermore, ablation of Raptor, the unique subunit of mTORC1, strongly delayed YAP/β-catenin-induced HB development in mice. At the molecular level, we found that expression of the amino acid transporter SLC38A1 is induced in mouse HB tissues, and amino acid deprivation leads to mTORC1 suppression in HB cell lines. Silencing of YAP and/or its paralog, transcriptional co-activator with PDZ binding motif (TAZ), decreased SLC38A1 expression as well as mTORC1 activation in HB cells. Furthermore, a frequent and concomitant upregulation of mTORC1 and SLC38A1 was detected in a collection of human HB specimens. Altogether, our study demonstrates the key role of mTORC1 in HB development. YAP and TAZ promote HB development via inducing SLC38A1 expression, whose upregulation leads to mTORC1 activation. Targeting mTOR pathway or amino acid transporters may represent novel therapeutic strategies for the treatment of human HB.
Collapse
Affiliation(s)
- Pin Liu
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.,Department of Bioengineering and Therapeutic Sciences, Liver Center, University of California San Francisco, San Francisco, CA, USA
| | - Diego F Calvisi
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Andras Kiss
- Second Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Antonio Cigliano
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Zsuzsa Schaff
- Second Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences, Liver Center, University of California San Francisco, San Francisco, CA, USA
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Frank Dombrowski
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Dongchi Zhao
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences, Liver Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
19
|
Abstract
There are few treatment options for patients with unresectable or refractory hepatoblastoma which has failed to respond to the standard treatment. The rarity of the disease and lack of experimental materials have hampered the development of new treatments. In this study, the collagen gel droplet-embedded culture drug sensitivity test was used to evaluate the effectiveness of the multikinase inhibitors sorafenib and sunitinib, and other drugs, in relapsed hepatoblastoma tumor tissues. Tumor samples from 6 patients with relapsed hepatoblastoma were tested for drug sensitivity by the collagen gel droplet-embedded culture drug sensitivity test; evaluable results were obtained from 5 of them. All samples were judged to be sensitive to sorafenib with a 50% growth inhibitory concentration (IC50) of 0.5 to 3.1 μg/mL. Sunitinib did not achieve IC50 in 2 of 3 samples within the tested concentration range based on clinically observed serum concentrations. In the drug combination assay using a hepatoblastoma cell line, sorafenib showed synergistic effects with SN-38, an active metabolite of irinotecan. Our results provide the basic science background warranting future clinical trials of a combination of sorafenib and irinotecan for relapsed or refractory hepatoblastoma.
Collapse
|
20
|
Zhang D, Qiu W, Wang P, Zhang P, Zhang F, Wang P, Sun Y. Autophagy can alleviate severe burn-induced damage to the intestinal tract in mice. Surgery 2017. [PMID: 28624177 DOI: 10.1016/j.surg.2017.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND The present study was designed to examine the effect of autophagy and apoptosis on intestinal injury in mice after severe burns. METHODS Kunming mice were subjected to third degree burns over 30% of the total body surface area. Damage to the intestine was assessed by examining changes in intestinal mucosal morphology, enzyme-linked immunosorbent assay of serum d-lactate, diamine oxidase, lipopolysaccharide, interleukin-6, and tumor necrosis factor α (marker of intestinal damage), hematoxylin and eosin staining, and Western blotting under 4 experimental conditions: control group, burn only (burn group), burn and administration of rapamycin to stimulate intestinal autophagy (rapamycin group), or burn and administration of 3-methyladenine to inhibit intestinal autophagy (3-methyladenine group). RESULTS At day 1 postburn, the expression levels of light chain 3 II, beclin-1, and cleaved caspase-3 were significantly greater in all 3 groups of mice subjected to the burn injury than in the control group 1 day postburn; while the levels of light chain 3 II and beclin-1 were significantly greater and those of cleaved caspase-3 were significantly less in the rapamycin group than in the burn group. In contrast, light chain 3 II and beclin-1 levels were significantly less and those of cleaved caspase-3 significantly greater in the 3-methyladenine group. All 3 groups subjected to burn injury showed significantly increased levels of d-lactate, diamine oxidase, lipopolysaccharide, interleukin-6, and tumor necrosis factor α. Of the 3 groups, the rapamycin group exhibited the least observed levels, the 3-methyladenine group the greatest, and the burn group intermediate. Pathologic sections of the intestinal tissue showed that all 3 burn groups exhibited severe intestinal mucosal damage at 1 day postburn. The condition of the 3-methyladenine treatment group was worse than that of the rapamycin treatment group, but better than that of the burn group. CONCLUSION Intestinal autophagy is activated in response to intestinal apoptosis after severe burns and may alleviate burn-induced intestinal injury.
Collapse
Affiliation(s)
- Duanyang Zhang
- Department of Burn Surgery, Huaihai Hospital affiliated to Xuzhou Medical University, Xuzhou, Jiangsu Province, China; and the Department of Burn Surgery, No. 97 Hospital of PLA, Xuzhou, Jiangsu Province, China.
| | - Wei Qiu
- Department of Burn Surgery, Huaihai Hospital affiliated to Xuzhou Medical University, Xuzhou, Jiangsu Province, China; and the Department of Burn Surgery, No. 97 Hospital of PLA, Xuzhou, Jiangsu Province, China
| | - Peng Wang
- Department of Burn Surgery, Huaihai Hospital affiliated to Xuzhou Medical University, Xuzhou, Jiangsu Province, China; and the Department of Burn Surgery, No. 97 Hospital of PLA, Xuzhou, Jiangsu Province, China
| | - Pan Zhang
- Department of Burn Surgery, Huaihai Hospital affiliated to Xuzhou Medical University, Xuzhou, Jiangsu Province, China; and the Department of Burn Surgery, No. 97 Hospital of PLA, Xuzhou, Jiangsu Province, China
| | - Fang Zhang
- Department of Burn Surgery, Huaihai Hospital affiliated to Xuzhou Medical University, Xuzhou, Jiangsu Province, China; and the Department of Burn Surgery, No. 97 Hospital of PLA, Xuzhou, Jiangsu Province, China
| | - PeiP Wang
- Department of Burn Surgery, Huaihai Hospital affiliated to Xuzhou Medical University, Xuzhou, Jiangsu Province, China; and the Department of Burn Surgery, No. 97 Hospital of PLA, Xuzhou, Jiangsu Province, China
| | - Yong Sun
- Department of Burn Surgery, Huaihai Hospital affiliated to Xuzhou Medical University, Xuzhou, Jiangsu Province, China; and the Department of Burn Surgery, No. 97 Hospital of PLA, Xuzhou, Jiangsu Province, China
| |
Collapse
|
21
|
Pivonello C, Negri M, De Martino MC, Napolitano M, de Angelis C, Provvisiero DP, Cuomo G, Auriemma RS, Simeoli C, Izzo F, Colao A, Hofland LJ, Pivonello R. The dual targeting of insulin and insulin-like growth factor 1 receptor enhances the mTOR inhibitor-mediated antitumor efficacy in hepatocellular carcinoma. Oncotarget 2016; 7:9718-31. [PMID: 26756219 PMCID: PMC4891079 DOI: 10.18632/oncotarget.6836] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 12/26/2015] [Indexed: 12/13/2022] Open
Abstract
Deregulation of mTOR and IGF pathways is frequent in hepatocellular carcinoma (HCC), thus mTOR and IGF1R represent suitable therapeutic targets in HCC. The aim of this study was to evaluate the effects of mTOR inhibitors (mTORi) and OSI-906, blocker of IGF1R/IR, on HCC cell proliferation, viability, migration and invasion, and alpha-fetoprotein (α-FP) secretion. In HepG2 and HuH-7 we evaluated, the expression of mTOR and IGF pathway components; the effects of Sirolimus, Everolimus, Temsirolimus and OSI-906 on cell proliferation; the effects of Sirolimus, OSI-906, and their combination, on cell secretion, proliferation, viability, cell cycle, apoptosis, invasion and migration. Moreover, intracellular mechanisms underlying these cell functions were evaluated in both cell lines. Our results show that HepG2 and HuH-7 present with the same mRNA expression profile with high levels of IGF2. OSI-906 inhibited cell proliferation at high concentration, while mTORi suppressed cell proliferation in a dose-time dependent manner in both cell lines. The co-treatment showed an additive inhibitory effect on cell proliferation and viability. This effect was not related to induction of apoptosis, but to G0/G1 phase block. Moreover, the co-treatment prevented the Sirolimus-induced AKT activation as escape mechanism. Both agents demonstrated to be differently effective in inhibiting α-FP secretion. Sirolimus, OSI-906, and their combination, blocked cell migration and invasion in HuH-7. These findings indicate that, co-targeting of IGF1R/IR and mTOR pathways could be a novel therapeutic approach in the management of HCC, in order to maximize antitumoral effect and to prevent the early development of resistance mechanisms.
Collapse
Affiliation(s)
- Claudia Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Università "Federico II" di Napoli, Naples, Italy
| | - Mariarosaria Negri
- IOS & Coleman Medicina Futura Medical Center, Centro Direzionale, Naples, Italy
| | | | - Maria Napolitano
- Immunology Oncology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" (IRCCS), Naples, Italy
| | - Cristina de Angelis
- IOS & Coleman Medicina Futura Medical Center, Centro Direzionale, Naples, Italy
| | | | - Gaia Cuomo
- IRCCS Fondazione SDN, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" (IRCCS), Naples, Italy
| | | | - Chiara Simeoli
- Dipartimento di Medicina Clinica e Chirurgia, Università "Federico II" di Napoli, Naples, Italy
| | - Francesco Izzo
- Hepatobiliary Surgery Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale" (IRCCS), Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Università "Federico II" di Napoli, Naples, Italy
| | - Leo J Hofland
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, Rotterdam, The Netherlands
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Università "Federico II" di Napoli, Naples, Italy
| |
Collapse
|
22
|
Beck A, Eberherr C, Hagemann M, Cairo S, Häberle B, Vokuhl C, von Schweinitz D, Kappler R. Connectivity map identifies HDAC inhibition as a treatment option of high-risk hepatoblastoma. Cancer Biol Ther 2016; 17:1168-1176. [PMID: 27635950 DOI: 10.1080/15384047.2016.1235664] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Hepatoblastoma (HB) is the most common liver tumor of childhood, usually occurring in children under the age of 3 y. The prognosis of patients presenting with distant metastasis, vascular invasion and advanced tumor stages remains poor and children that do survive often face severe late effects from the aggressive chemotherapy regimen. To identify potential new therapeutics for high risk HB we used a 1,000-gene expression signature as input for a Connectivity Map (CMap) analysis, which predicted histone deacetylase (HDAC) inhibitors as a promising therapy option. Subsequent expression analysis of primary HB and HB cell lines revealed a general overexpression of HDAC1 and HDAC2, which has been suggested to be predictive for the efficacy of HDAC inhibition. Accordingly, treatment of HB cells with the HDAC inhibitors SAHA and MC1568 resulted in a potent reduction of cell viability, induction of apoptosis, reactivation of epigenetically suppressed tumor suppressor genes, and the reversion of the 16-gene HB classifier toward the more favorable expression signature. Most importantly, the combination of HDAC inhibitors and cisplatin - a major chemotherapeutic agent of HB treatment - revealed a strong synergistic effect, even at significantly reduced doses of cisplatin. Our findings suggest that HDAC inhibitors skew HB cells toward a more favorable prognostic phenotype through changes in gene expression, thus indicating a targeted molecular mechanism that seems to enhance the anti-proliferative effects of conventional chemotherapy. Thus, adding HDAC inhibitors to the treatment regimen of high risk HB could potentially improve outcomes and reduce severe late effects.
Collapse
Affiliation(s)
- Alexander Beck
- a Department of Pediatric Surgery, Dr. von Hauner Children's Hospital , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Corinna Eberherr
- a Department of Pediatric Surgery, Dr. von Hauner Children's Hospital , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Michaela Hagemann
- a Department of Pediatric Surgery, Dr. von Hauner Children's Hospital , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Stefano Cairo
- b XenTech , 4 rue Pierre Fontaine , Evry , France.,c University of Ferrara, LTTA Center, Department of Morphology , Surgery and Experimental Medicine, Via Fossato di Mortara , Ferrara , Italy
| | - Beate Häberle
- a Department of Pediatric Surgery, Dr. von Hauner Children's Hospital , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Christian Vokuhl
- d Institute of Paidopathology, Pediatric Tumor Registry, Christian-Albrechts-University Kiel , Kiel , Germany
| | - Dietrich von Schweinitz
- a Department of Pediatric Surgery, Dr. von Hauner Children's Hospital , Ludwig-Maximilians-University Munich , Munich , Germany
| | - Roland Kappler
- a Department of Pediatric Surgery, Dr. von Hauner Children's Hospital , Ludwig-Maximilians-University Munich , Munich , Germany
| |
Collapse
|
23
|
Rikhi RR, Spady KK, Hoffman RI, Bateman MS, Bateman M, Howard LE. Hepatoblastoma: A Need for Cell Lines and Tissue Banks to Develop Targeted Drug Therapies. Front Pediatr 2016; 4:22. [PMID: 27047905 PMCID: PMC4800278 DOI: 10.3389/fped.2016.00022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/07/2016] [Indexed: 12/11/2022] Open
Abstract
Limited research exists regarding the most aggressive forms of hepatoblastoma. Cell lines of the rare subtypes of hepatoblastoma with poor prognosis are not only difficult to attain but also challenging to characterize histologically. A community-driven approach to educating parents and families, regarding the need for donated tissue, is necessary for scientists to have access to resources for murine models and drug discovery. Herein, we describe the currently available resources, existing gaps in research, and the path to move forward for uniform cure of hepatoblastoma.
Collapse
Affiliation(s)
- Rishi Raj Rikhi
- Children's Cancer Therapy Development Institute , Beaverton, OR , USA
| | - Kimberlee K Spady
- Faculty of the 2015 Pediatric Cancer Biology Nanocourse, Children's Cancer Therapy Development Institute , Fort Collins, CO , USA
| | - Ruth I Hoffman
- Faculty of the 2015 Pediatric Cancer Biology Nanocourse, Children's Cancer Therapy Development Institute , Fort Collins, CO , USA
| | - Michael S Bateman
- Faculty of the 2015 Pediatric Cancer Biology Nanocourse, Children's Cancer Therapy Development Institute , Fort Collins, CO , USA
| | - Max Bateman
- Faculty of the 2015 Pediatric Cancer Biology Nanocourse, Children's Cancer Therapy Development Institute , Fort Collins, CO , USA
| | - Lisa Easom Howard
- Faculty of the 2015 Pediatric Cancer Biology Nanocourse, Children's Cancer Therapy Development Institute , Fort Collins, CO , USA
| |
Collapse
|
24
|
Ilmer M, Garnier A, Vykoukal J, Alt E, von Schweinitz D, Kappler R, Berger M. 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] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 09/20/2015] [Indexed: 11/16/2022]
|
25
|
Abstract
INTRODUCTION Corticosteroids alone or in combination with azathioprine are the mainstay therapies of autoimmune hepatitis. Suboptimal responses (treatment failure, partial response, drug toxicity), frequent relapse after drug withdrawal, and the emergence of alternative immunosuppressive medications have fueled the pursuit of new treatments. The goals of this review are to present current management strategies and evolving interventions. AREAS COVERED PubMed searches from 1970 - 2014 provide the bases for this review. Corticosteroid regimens should be administered until resolution of symptoms, laboratory tests, and liver tissue abnormalities. Treatment failure warrants high doses of the original regimen, and relapse warrants re-treatment followed by long-term maintenance with azathioprine. The calcineurin inhibitors, budesonide, and mycophenolate mofetil are evolving as frontline therapies, and they may be considered as salvage therapies with the exception of budesonide. Rapamycin, rituximab, and infliximab have also rescued refractory patients but experiences are limited. Anti-oxidants, recombinant molecules, mAbs, and modulators of critical cell populations are key prospects. EXPERT OPINION Autoimmune hepatitis must be managed by multiple medications that supplement or supplant current regimens depending on the clinical situation. Rescue therapies will emerge as adjunctive interventions to minimize tissue damage (prevent fibrosis and hepatocyte apoptosis) and improve immune tolerance (regulatory T cell manipulations).
Collapse
Affiliation(s)
- Albert J Czaja
- Mayo Clinic College of Medicine, From the Division of Gastroenterology and Hepatology , 200 First Street S.W, Rochester, MN 55905 , USA +1 507 284 2691 ; +1 507 284 0538 ;
| |
Collapse
|
26
|
Sakamoto S, Kasahara M, Mizuta K, Kuroda T, Yagi T, Taguchi T, Inomata Y, Umeshita K, Uemoto S, for the Japanese Liver Transplantation Society. Nationwide survey of the outcomes of living donor liver transplantation for hepatoblastoma in Japan. Liver Transpl 2014; 20:333-46. [PMID: 24734315 DOI: 10.1002/lt.23803] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recently, liver transplantation (LT) has been increasingly performed for unresectable hepatoblastoma (HB) with acceptable results. We conducted a national survey of cases undergoing living donor liver transplantation (LDLT) for HB to evaluate their outcomes. Thirty-nine patients (28 males and 11 females with a median age at LDLT of 3.6 years) who had undergone LDLT for HB by the end of 2009 were enrolled in this study. The clinical data were collected from their medical records via a questionnaire survey in 2011 (median follow-up = 4.6 years). Thirteen patients (33.3%) had extrahepatic lesions before LDLT. Thirty-eight patients (97.4%) received chemotherapy, and 15 (38.5%) underwent hepatectomy before LDLT. Twenty-seven patients (69.2%) were alive without recurrence after LDLT, and 12 patients (30.8%) suffered from recurrence. The most common site of recurrence was the lung (9 cases), which was followed by the graft liver (6 cases). The median interval from LDLT to recurrence was 5.5 months. Four of the 9 cases (44.4%) with lung metastasis underwent surgical resection, and 3 were alive without any tumor recurrence. Eight patients died because of tumor recurrence. The multivariate landmark analysis revealed that the independent recurrence risk factors were a high alpha-fetoprotein (AFP) level at diagnosis [≥ 500,000 ng/mL; hazard ratio (HR) = 7.86, P = 0.010], the presence of extrahepatic lesions before LDLT (HR = 3.82, P = 0.042), and a high AFP level at LDLT (≥ 4000 ng/mL; HR = 9.19, P = 0.036). The actuarial 3- and 5-year patient survival rates were 84.3% and 77.3%, respectively. In conclusion, with appropriate timing for scheduled LT, LDLT provides a valuable alternative treatment with excellent results for children with HB.
Collapse
Affiliation(s)
- Seisuke Sakamoto
- Transplantation Center; National Center for Child Health and Development; Tokyo Japan
| | - Mureo Kasahara
- Transplantation Center; National Center for Child Health and Development; Tokyo Japan
| | - Koichi Mizuta
- Department of Transplant Surgery; Jichi Medical University; Tochigi Japan
| | - Tatsuo Kuroda
- Department of Pediatric Surgery; Keio University; Tokyo Japan
| | - Takahito Yagi
- Department of Surgery, Okayama University; Okayama Japan
| | - Tomoaki Taguchi
- Department of Pediatric Surgery; Kyushu University; Fukuoka Japan
| | - Yukihiro Inomata
- Department of Transplant Surgery, Kumamoto University; Kumamoto Japan
| | - Koji Umeshita
- Department of Surgery, Osaka University; Osaka Japan
| | - Shinji Uemoto
- Department of Surgery, Kyoto University; Kyoto Japan
| | | |
Collapse
|
27
|
López-Terrada D, Alaggio R, de Dávila MT, Czauderna P, Hiyama E, Katzenstein H, Leuschner I, Malogolowkin M, Meyers R, Ranganathan S, Tanaka Y, Tomlinson G, Fabrè M, Zimmermann A, Finegold MJ. Towards an international pediatric liver tumor consensus classification: proceedings of the Los Angeles COG liver tumors symposium. Mod Pathol 2014; 27:472-91. [PMID: 24008558 DOI: 10.1038/modpathol.2013.80] [Citation(s) in RCA: 207] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 02/28/2013] [Accepted: 03/05/2013] [Indexed: 02/07/2023]
Abstract
Liver tumors are rare in children, and their diagnoses may be challenging particularly because of the lack of a current consensus classification system. Systematic central histopathological review of these tumors performed as part of the pediatric collaborative therapeutic protocols has allowed the identification of histologic subtypes with distinct clinical associations. As a result, histopathology has been incorporated within the Children's Oncology Group (COG) protocols, and only in the United States, as a risk-stratification parameter and for patient management. Therefore, the COG Liver Tumor Committee sponsored an International Pathology Symposium in March 2011 to discuss the histopathology and classification of pediatric liver tumors, and hepatoblastoma in particular, and work towards an International Pediatric Liver Tumors Consensus Classification that would be required for international collaborative projects. Twenty-two pathologists and experts in pediatric liver tumors, including those serving as central reviewers for the COG, European Société Internationale d'Oncologie Pédiatrique, Gesellschaft für Pädiatrische Onkologie und Hämatologie, and Japanese Study Group for Pediatric Liver Tumors protocols, as well as pediatric oncologists and surgeons specialized in this field, reviewed more than 50 pediatric liver tumor cases and discussed classic and newly reported entities, as well as criteria for their classification. This symposium represented the first collaborative step to develop a classification that may lead to a common treatment-stratification system incorporating tumor histopathology. A standardized, clinically meaningful classification will also be necessary to allow the integration of new biological parameters and to move towards clinical algorithms based on patient characteristics and tumor genetics, which should improve future patient management and outcome.
Collapse
Affiliation(s)
- Dolores López-Terrada
- Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Rita Alaggio
- Division of Pathology, Department of Medicine-DIMED, Pathology Unit, Padova, Italy
| | - Maria T de Dávila
- Departamento de Patologia, Hospital de Pediatría Prof. Dr. J.P. Garrahan, Buenos Aires, Argentina
| | - Piotr Czauderna
- Department of Surgery and Urology for Children and Adolescents, Medical University of Gdansk, Gdansk, Poland
| | - Eiso Hiyama
- Department of Surgery, Natural Science Center for Basic Research and Development, Hiroshima University Hospital, Hiroshima, Japan
| | - Howard Katzenstein
- Aflac Cancer Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Ivo Leuschner
- Institut fur Pathologie, UNI-Klinikum Campus, Kiel, Germany
| | - Marcio Malogolowkin
- Department of Pediatric Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Rebecka Meyers
- Department of Pediatric Surgery, Primary Children's Medical Center, University of Utah, Salt Lake City, UT, USA
| | | | - Yukichi Tanaka
- Division of Pathology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Gail Tomlinson
- Division of Pediatric Hematology-Oncology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Monique Fabrè
- Department of Pathology, Institut de Cancerologie Gustave Roussy, Villejuif, France
| | | | - Milton J Finegold
- Department of Pathology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | | |
Collapse
|
28
|
Morgenstern DA, Marzouki M, Bartels U, Irwin MS, Sholler GLS, Gammon J, Yankanah R, Wu B, Samson Y, Baruchel S. Phase I study of vinblastine and sirolimus in pediatric patients with recurrent or refractory solid tumors. Pediatr Blood Cancer 2014; 61:128-33. [PMID: 23956145 DOI: 10.1002/pbc.24656] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 05/23/2013] [Indexed: 11/10/2022]
Abstract
BACKGROUND The combination of vinblastine and mammalian target of rapamycin (mTOR) inhibitor sirolimus inhibits the growth of neuroblastoma xenografts through pro-apoptotic and anti-angiogenic mechanisms. This phase I study aimed to explore the safety and toxicity of this combination in pediatric patients with advanced solid tumors. PROCEDURE Patients ≤21 years of age with recurrent/refractory solid tumors (including CNS) were eligible. Sirolimus was administered daily by mouth or nasogastric (NG) tube, with doses adjusted to achieve a target trough concentration of 10-15 ng/ml, with weekly intravenous vinblastine (dose escalated 4-6 mg/m(2)/dose according to 3 + 3 phase I design). RESULTS Fourteen patients were enrolled (median age 8.7 years; range 2.3-19) of whom 12 were evaluable for toxicity and 11 for response. One patient experienced a dose-limiting toxicity (grade 3 mucositis) at the highest vinblastine dose level. Myelosuppression was the most common toxicity. Dose-adjusted sirolimus trough concentrations were significantly lower in patients receiving drug via NG tube (1.50 ± 0.75 ng/ml/mg vs. 2.25 ± 1.07 ng/ml/mg for oral administration). Correlative biomarker analysis demonstrated a significant reduction in serum concentration of soluble vascular endothelial growth factor receptor (sVEGFR2) at 28 days compared to baseline consistent with inhibition of angiogenesis. One patient had a partial response and three had stable disease for more than 3 months. CONCLUSIONS The combination of mTOR inhibitor and vinblastine given over an extended continuous schedule is safe, associated with a reduction in circulating angiogenic factor (CAF) VEGFR2 and resulted in clinical responses. Future studies using the intravenously administered mTOR inhibitor temsirolimus are planned.
Collapse
Affiliation(s)
- Daniel A Morgenstern
- Department of Paediatrics, University of Toronto and New Agent and Innovative Therapy Programme, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Bhat M, Sonenberg N, Gores G. The mTOR pathway in hepatic malignancies. Hepatology 2013; 58:810-8. [PMID: 23408390 PMCID: PMC3688698 DOI: 10.1002/hep.26323] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/07/2013] [Indexed: 12/13/2022]
Abstract
The mechanistic/mammalian target of rapamycin (mTOR) pathway plays a critical role in cellular metabolism, growth, and proliferation and has been evaluated as a target for therapy in various malignancies. The mTOR pathway is a major tumor-initiating pathway in hepatocellular carcinoma, with up-regulation seen in up to 50% of tumors. Metformin, which represses mTOR signaling by activating adenosine monophosphate-activated protein kinase, has been shown to decrease liver carcinogenesis in population studies. mTOR inhibitors such as everolimus have been evaluated as adjunctive chemotherapy with some success, although efficacy has been limited by the lack of complete mTOR pathway inhibition. The active site mTOR inhibitors hold greater promise, given that they offer complete mTOR suppression. There is also evidence of mTOR pathway activation in cholangiocarcinoma, although its biological significance in initiating and promoting tumor progression remains ambiguous. This review provides an overview of the complex biochemistry behind the mTOR pathway and its role in carcinogenesis, especially as it pertains to hepatic malignancies.
Collapse
Affiliation(s)
- Mamatha Bhat
- Division of Gastroenterology, McGill University Health Centre, Montreal, Canada,Department of Biochemistry, Goodman Cancer Research Center, McGill University, Montreal, Canada,Division of Gastroenterology and Hepatology, College of Medicine, 200 First Street SW, Mayo Clinic, Rochester, MN, USA
| | - Nahum Sonenberg
- Department of Biochemistry, Goodman Cancer Research Center, McGill University, Montreal, Canada
| | - Gregory Gores
- Division of Gastroenterology and Hepatology, College of Medicine, 200 First Street SW, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
30
|
FAK Inhibition Decreases Hepatoblastoma Survival Both In Vitro and In Vivo. Transl Oncol 2013; 6:206-15. [PMID: 23544173 DOI: 10.1593/tlo.12505] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 01/29/2013] [Accepted: 01/29/2013] [Indexed: 12/31/2022] Open
Abstract
Hepatoblastoma is the most frequently diagnosed liver tumor of childhood, and children with advanced, metastatic or relapsed disease have a disease-free survival rate under 50%. Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase that is important in many facets of tumor development and progression. FAK has been found in other pediatric solid tumors and in adult hepatocellular carcinoma, leading us to hypothesize that FAK would be present in hepatoblastoma and would impact its cellular survival. In the current study, we showed that FAK was present and phosphorylated in human hepatoblastoma tumor specimens. We also examined the effects of FAK inhibition upon hepatoblastoma cells using a number of parallel approaches to block FAK including RNAi and small molecule FAK inhibitors. FAK inhibition resulted in decreased cellular survival, invasion, and migration and increased apoptosis. Further, small molecule inhibition of FAK led to decreased tumor growth in a nude mouse xenograft model of hepatoblastoma. The findings from this study will help to further our understanding of the regulation of hepatoblastoma tumorigenesis and may provide desperately needed novel therapeutic strategies and targets for aggressive, recurrent, or metastatic hepatoblastomas.
Collapse
|
31
|
Zhang SC, Wang WL, Cai WS, Jiang KL, Yuan ZW. Engineered measles virus Edmonston strain used as a novel oncolytic viral system against human hepatoblastoma. BMC Cancer 2012; 12:427. [PMID: 23009685 PMCID: PMC3488522 DOI: 10.1186/1471-2407-12-427] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 09/23/2012] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Hepatoblastoma (HB) is the most common primary, malignant pediatric liver tumor in children. The treatment results for affected children have markedly improved in recent decades. However, the prognosis for high-risk patients who have extrahepatic extensions, invasion of the large hepatic veins, distant metastases and very high alpha-fetoprotein (AFP) serum levels remains poor. There is an urgent need for the development of novel therapeutic approaches. METHODS An attenuated strain of measles virus, derived from the Edmonston vaccine lineage, was genetically engineered to produce carcinoembryonic antigen (CEA). We investigated the antitumor potential of this novel viral agent against human HB both in vitro and in vivo. RESULTS Infection of the Hep2G and HUH6 HB cell lines, at multiplicities of infection (MOIs) ranging from 0.01 to 1, resulted in a significant cytopathic effect consisting of extensive syncytia formation and massive cell death at 72-96 h after infection. Both of the HB lines overexpressed the measles virus receptor CD46 and supported robust viral replication, which correlated with CEA production. The efficacy of this approach in vivo was examined in murine Hep2G xenograft models. Flow cytometry assays indicated an apoptotic mechanism of cell death. Intratumoral administration of MV-CEA resulted in statistically significant delay of tumor growth and prolongation of survival. CONCLUSIONS The engineered measles virus Edmonston strain MV-CEA has potent therapeutic efficacy against HB cell lines and xenografts. Trackable measles virus derivatives merit further exploration in HB treatment.
Collapse
Affiliation(s)
- Shu-Cheng Zhang
- Department of Pediatric Surgery, Major Laboratory of Chinese Health Ministry for Congenital Malformations, Shengjing Hospital of China Medical University, 36 Sanhao Street Heping District, Shenyang 110004, P.R. China
| | - Wei-Lin Wang
- Department of Pediatric Surgery, Major Laboratory of Chinese Health Ministry for Congenital Malformations, Shengjing Hospital of China Medical University, 36 Sanhao Street Heping District, Shenyang 110004, P.R. China
| | - Wei-Song Cai
- Department of Medical Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Street Heping District, Shenyang 110004, P.R. China
| | - Kai-Lei Jiang
- Department of Pediatric Surgery, Major Laboratory of Chinese Health Ministry for Congenital Malformations, Shengjing Hospital of China Medical University, 36 Sanhao Street Heping District, Shenyang 110004, P.R. China
| | - Zheng-Wei Yuan
- Department of Pediatric Surgery, Major Laboratory of Chinese Health Ministry for Congenital Malformations, Shengjing Hospital of China Medical University, 36 Sanhao Street Heping District, Shenyang 110004, P.R. China
| |
Collapse
|
32
|
Abstract
Nonsteroidal medications, previously unfamiliar in the management of autoimmune hepatitis, can supplement or replace conventional corticosteroid regimens, especially in problematic patients. Mycophenolate mofetil is a next-generation purine antagonist that has been useful in treating patients with azathioprine intolerance. It has been less effective in salvaging patients with steroid-refractory disease. Azathioprine is the choice as a corticosteroid-sparing agent in treatment-naive patients and in individuals with corticosteroid intolerance, incomplete response and relapse after drug withdrawal. Tacrolimus is preferred over cyclosporine for recalcitrant disease because of its established preference in organ transplantation, but replacement with cyclosporine should be considered if the disease worsens on treatment. Rapamycin has antiproliferative and proapoptotic actions that warrant further study in autoimmune hepatitis. The nonstandard, nonsteroidal medications are mainly salvage therapies with off-label indications that must be used in highly individualized and well-monitored clinical situations.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN 55905 USA.
| |
Collapse
|