Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Med ; 74: 293-306, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36170665

ABSTRACT

Biliary tract cancer (BTC) is the second most common primary liver cancer after hepatocellular carcinoma and accounts for 2% of cancer-related deaths. BTCs are classified according to their anatomical origin into intrahepatic (iCCA), perihilar, or distal cholangiocarcinoma, as well as gall bladder carcinoma. While the mutational profiles in these anatomical BTC subtypes overlap to a large extent, iCCA is notable for the high frequency of IDH1/2 mutations (10-22%) and the nearly exclusive occurrence of FGFR2 fusions in 10-15% of patients. In recent years, FGFR2 fusions have become one of the most promising targets for precision oncology targeting BTC, with FGFR inhibitors already approved in Europe and the United States for patients with advanced, pretreated iCCA. While the therapeutic potential of nonfusion alterations is still under debate, it is expected that the field of FGFR2-directed therapies will be subject to rapid further evolution and optimization. The scope of this review is to provide an overview of oncogenic FGFR signaling in iCCA cells and highlight the pathophysiology, diagnostic testing strategies, and therapeutic promises and challenges associated with FGFR2-altered iCCA.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Precision Medicine , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/diagnosis , Mutation , Bile Ducts, Intrahepatic/pathology , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/therapeutic use
2.
Gut ; 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38857989

ABSTRACT

OBJECTIVE: Intrahepatic cholangiocarcinoma (iCCA) is the second most common primary liver cancer with limited therapeutic options. KRAS mutations are among the most abundant genetic alterations in iCCA associated with poor clinical outcome and treatment response. Recent findings indicate that Poly(ADP-ribose)polymerase1 (PARP-1) is implicated in KRAS-driven cancers, but its exact role in cholangiocarcinogenesis remains undefined. DESIGN: PARP-1 inhibition was performed in patient-derived and established iCCA cells using RNAi, CRISPR/Cas9 and pharmacological inhibition in KRAS-mutant, non-mutant cells. In addition, Parp-1 knockout mice were combined with iCCA induction by hydrodynamic tail vein injection to evaluate an impact on phenotypic and molecular features of Kras-driven and Kras-wildtype iCCA. Clinical implications were confirmed in authentic human iCCA. RESULTS: PARP-1 was significantly enhanced in KRAS-mutant human iCCA. PARP-1-based interventions preferentially impaired cell viability and tumourigenicity in human KRAS-mutant cell lines. Consistently, loss of Parp-1 provoked distinct phenotype in Kras/Tp53-induced versus Akt/Nicd-induced iCCA and abolished Kras-dependent cholangiocarcinogenesis. Transcriptome analyses confirmed preferential impairment of DNA damage response pathways and replicative stress response mediated by CHK1. Consistently, inhibition of CHK1 effectively reversed PARP-1 mediated effects. Finally, Parp-1 depletion induced molecular switch of KRAS-mutant iCCA recapitulating good prognostic human iCCA patients. CONCLUSION: Our findings identify the novel prognostic and therapeutic role of PARP-1 in iCCA patients with activation of oncogenic KRAS signalling.

3.
Hepatology ; 2023 Dec 18.
Article in English | MEDLINE | ID: mdl-38108634

ABSTRACT

Immune-oncology-based regimens have shown efficacy in advanced HCC and have been implemented as standard of care as first-line therapy. Their efficacy, including high response rates, and safety justify their evaluation in earlier disease stages. Following negative results for adjuvant sorafenib in the global STORM trial in 2015, 4 global phase 3 trials, featuring different immune checkpoint inhibitor combinations, entered in parallel the race in the adjuvant setting. The IMbrave050 trial, comparing adjuvant atezolizumab in combination with bevacizumab to active surveillance following curative-intent resection or ablation, was the first to report, fast-tracking the results of the first interim analysis and demonstrating an improvement in recurrence-free survival. The trial has provoked a discussion on the horizon of expectations from adjuvant treatment and the clinical relevance of efficacy endpoints. Moreover, major pathological responses reported from early phase 2 data in the neoadjuvant setting provide a strong rationale for the evaluation of these concepts in phase 3 trials. In this review, we summarize current evidence and outline future directions for systemic therapies in early-stage HCC.

4.
J Hepatol ; 78(3): 614-626, 2023 03.
Article in English | MEDLINE | ID: mdl-36528236

ABSTRACT

BACKGROUND & AIMS: In recent years, intrahepatic cholangiocarcinoma (iCCA) has evolved as a "role model" for precision oncology in gastrointestinal cancers. However, its rarity, paired with its genomic heterogeneity, challenges the development and evolution of targeted therapies. Interrogating large datasets drives better understanding of the characteristics of molecular subgroups of rare cancers and enables the identification of genomic patterns that remain unrecognized in smaller cohorts. METHODS: We performed a retrospective analysis of 6,130 patients diagnosed with iCCA from the FoundationCORE database who received diagnostic panel sequencing on the FoundationOne platform. Short variants/fusion-rearrangements and copy number alterations in >300 tumor-associated genes were evaluated, and the tumor mutational burden (TMB) as well as the microsatellite instability (MSI) status were available for the majority of the cohort. RESULTS: We provide a highly representative cartography of the genomic landscape of iCCA and outline the co-mutational spectra of seven therapeutically relevant oncogenic driver genes: IDH1/2, FGFR2, ERBB2, BRAF, MDM2, BRCA1/2, MET and KRASG12C. We observed a negative selection of RTK/RAS/ERK pathway co-alterations, and an enrichment of epigenetic modifiers such as ARID1A and BAP1 in patients with IDH1/2 and FGFR2 alterations. RNF43 as well as KMT2D occurred with high frequency in MSIhigh and TMBhigh tumors. CONCLUSION: Detailed knowledge of the most prevalent genomic constellations is key to the development of effective treatment strategies for iCCA. Our study provides a valuable resource that could be used to assess the feasibility of clinical trials and subgroup analyses, spurs the development of translationally relevant preclinical models, and serves as a knowledge base to predict potential mechanisms of resistance to targeted therapies in genomically defined subgroups. IMPACT AND IMPLICATIONS: Due to the high frequency of targetable alterations, molecular diagnostics is recommended in patients with biliary tract cancers, and especially in those with iCCA. The identification of an actionable lesion, however, does not guarantee therapeutic success, and the co-mutational spectrum may act as a critical modifier of drug response. Using a large dataset of comprehensive panel sequencing results from 6,130 patients with iCCA, we provide a detailed analysis of the co-mutational spectrum of the most frequent druggable genetic alterations, which is meant to serve as a reference to establish genetically relevant preclinical models, develop hypothesis-driven combination therapies and identify recurrent genetic profiles.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Retrospective Studies , Precision Medicine , Cholangiocarcinoma/pathology , Mutation , Biomarkers, Tumor/genetics , Bile Ducts, Intrahepatic/pathology
5.
Lancet ; 400(10360): 1345-1362, 2022 10 15.
Article in English | MEDLINE | ID: mdl-36084663

ABSTRACT

Hepatocellular carcinoma is one of the most common cancers worldwide and represents a major global health-care challenge. Although viral hepatitis and alcohol remain important risk factors, non-alcoholic fatty liver disease is rapidly becoming a dominant cause of hepatocellular carcinoma. A broad range of treatment options are available for patients with hepatocellular carcinoma, including liver transplantation, surgical resection, percutaneous ablation, and radiation, as well as transarterial and systemic therapies. As such, clinical decision making requires a multidisciplinary team that longitudinally adapts the individual treatment strategy according to the patient's tumour stage, liver function, and performance status. With the approval of new first-line agents and second-line agents, as well as the establishment of immune checkpoint inhibitor-based therapies as standard of care, the treatment landscape of advanced hepatocellular carcinoma is more diversified than ever. Consequently, the outlook for patients with hepatocellular carcinoma has improved. However, the optimal sequencing of drugs remains to be defined, and predictive biomarkers are urgently needed to inform treatment selection. In this Seminar, we present an update on the causes, diagnosis, molecular classification, and treatment of hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Liver Transplantation , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/therapy , Humans , Immune Checkpoint Inhibitors , Liver Neoplasms/drug therapy , Liver Neoplasms/therapy
6.
J Hepatol ; 76(5): 1109-1121, 2022 05.
Article in English | MEDLINE | ID: mdl-35167909

ABSTRACT

BACKGROUND & AIMS: Cholangiocarcinoma (CCA) is a rare and heterogeneous biliary cancer, whose incidence and related mortality is increasing. This study investigates the clinical course of CCA and subtypes (intrahepatic [iCCA], perihilar [pCCA], and distal [dCCA]) in a pan-European cohort. METHODS: The ENSCCA Registry is a multicenter observational study. Patients were included if they had a histologically proven diagnosis of CCA between 2010-2019. Demographic, histomorphological, biochemical, and clinical studies were performed. RESULTS: Overall, 2,234 patients were enrolled (male/female=1.29). iCCA (n = 1,243) was associated with overweight/obesity and chronic liver diseases involving cirrhosis and/or viral hepatitis; pCCA (n = 592) with primary sclerosing cholangitis; and dCCA (n = 399) with choledocholithiasis. At diagnosis, 42.2% of patients had local disease, 29.4% locally advanced disease (LAD), and 28.4% metastatic disease (MD). Serum CEA and CA19-9 showed low diagnostic sensitivity, but their concomitant elevation was associated with increased risk of presenting with LAD (odds ratio 2.16; 95% CI 1.43-3.27) or MD (odds ratio 5.88; 95% CI 3.69-9.25). Patients undergoing resection (50.3%) had the best outcomes, particularly with negative-resection margin (R0) (median overall survival [mOS] = 45.1 months); however, margin involvement (R1) (hazard ratio 1.92; 95% CI 1.53-2.41; mOS = 24.7 months) and lymph node invasion (hazard ratio 2.13; 95% CI 1.55-2.94; mOS = 23.3 months) compromised prognosis. Among patients with unresectable disease (49.6%), the mOS was 10.6 months for those receiving active palliative therapies, mostly chemotherapy (26.2%), and 4.0 months for those receiving best supportive care (20.6%). iCCAs were associated with worse outcomes than p/dCCAs. ECOG performance status, MD and CA19-9 were independent prognostic factors. CONCLUSION: CCA is frequently diagnosed at an advanced stage, a proportion of patients fail to receive cancer-specific therapies, and prognosis remains dismal. Identification of preventable risk factors and implementation of surveillance in high-risk populations are required to decrease cancer-related mortality. LAY SUMMARY: This is, to date, the largest international (pan-European: 26 hospitals and 11 countries) observational study, in which the course of cholangiocarcinoma has been investigated, comparing the 3 subtypes based on the latest International Classification of Diseases 11th Edition (ICD-11) (i.e., intrahepatic [2C12], perihilar [2C18], or distal [2C15] affected bile ducts), which come into effect in 2022. General and tumor-type specific features at diagnosis, risk factors, biomarker accuracy, as well as patient management and outcomes, are presented and compared, outlining the current clinical state of cholangiocarcinoma in Europe.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Bile Duct Neoplasms/diagnosis , Bile Duct Neoplasms/epidemiology , Bile Duct Neoplasms/therapy , Bile Ducts, Intrahepatic/pathology , CA-19-9 Antigen , Cholangiocarcinoma/diagnosis , Cholangiocarcinoma/epidemiology , Cholangiocarcinoma/therapy , Female , Humans , Male , Prognosis , Registries
7.
Hepatology ; 74(3): 1357-1370, 2021 09.
Article in English | MEDLINE | ID: mdl-33709535

ABSTRACT

BACKGROUND AND AIMS: Intrahepatic cholangiocarcinoma (ICC) is the second most common primary liver cancer and a highly lethal malignancy. Chemotherapeutic options are limited, but a considerable subset of patients harbors genetic lesions for which targeted agents exist. Fibroblast growth factor receptor 2 (FGFR2) fusions belong to the most frequent and therapeutically relevant alterations in ICC, and the first FGFR inhibitor was recently approved for the treatment of patients with progressed, fusion-positive ICC. Response rates of up to 35% indicate that FGFR-targeted therapies are beneficial in many but not all patients. Thus far, no established biomarkers exist that predict resistance or response to FGFR-targeted therapies in patients with ICC. APPROACH AND RESULTS: In this study, we use an autochthonous murine model of ICC to demonstrate that FGFR2 fusions are potent drivers of malignant transformation. Furthermore, we provide preclinical evidence that the co-mutational spectrum acts not only as an accelerator of tumor development, but also modifies the response to targeted FGFR inhibitors. Using pharmacologic approaches and RNA-interference technology, we delineate that Kirsten rat sarcoma oncogene (KRAS)-activated mitogen-activated protein kinase signaling causes primary resistance to FGFR inhibitors in FGFR2 fusion-positive ICC. The translational relevance is supported by the observation that a subset of human FGFR2 fusion patients exhibits transcriptome profiles reminiscent of KRAS mutant ICC. Moreover, we demonstrate that combination therapy has the potential to overcome primary resistance and to sensitize tumors to FGFR inhibition. CONCLUSIONS: Our work highlights the importance of the co-mutational spectrum as a significant modifier of response in tumors that harbor potent oncogenic drivers. A better understanding of the genetic underpinnings of resistance will be pivotal to improve biomarker-guided patient selection and to design clinically relevant combination strategies.


Subject(s)
Bile Duct Neoplasms/genetics , Bile Ducts, Intrahepatic , Cell Transformation, Neoplastic/genetics , Cholangiocarcinoma/genetics , Gene Fusion/genetics , Liver Neoplasms, Experimental/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Adenosylhomocysteinase/genetics , Animals , Antigens, Neoplasm/genetics , Antimetabolites, Antineoplastic/pharmacology , Bile Duct Neoplasms/pathology , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cholangiocarcinoma/pathology , Co-Repressor Proteins/genetics , Cyclic AMP Response Element-Binding Protein A/genetics , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Fetal Proteins/genetics , Mice , Microtubule-Associated Proteins/genetics , Mutation , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Vesicular Transport Proteins/genetics , Gemcitabine
8.
Genes Dev ; 28(1): 85-97, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24395249

ABSTRACT

Genetically engineered mouse models (GEMMs) have greatly expanded our knowledge of pancreatic ductal adenocarcinoma (PDAC) and serve as a critical tool to identify and evaluate new treatment strategies. However, the cost and time required to generate conventional pancreatic cancer GEMMs limits their use for investigating novel genetic interactions in tumor development and maintenance. To address this problem, we developed flexible embryonic stem cell (ESC)-based GEMMs that facilitate the rapid generation of genetically defined multiallelic chimeric mice without further strain intercrossing. The ESCs harbor a latent Kras mutant (a nearly ubiquitous feature of pancreatic cancer), a homing cassette, and other genetic elements needed for rapid insertion and conditional expression of tetracycline-controlled transgenes, including fluorescence-coupled shRNAs capable of efficiently silencing gene function by RNAi. This system produces a disease that recapitulates the progression of pancreatic cancer in human patients and enables the study and visualization of the impact of gene perturbation at any stage of pancreas cancer progression. We describe the use of this approach to dissect temporal roles for the tumor suppressor Pten and the oncogene c-Myc in pancreatic cancer development and maintenance.


Subject(s)
Disease Models, Animal , Embryonic Stem Cells , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Animals , Animals, Genetically Modified , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Reproducibility of Results
9.
Gut ; 69(6): 1104-1115, 2020 06.
Article in English | MEDLINE | ID: mdl-31767630

ABSTRACT

OBJECTIVE: Liver fibrosis and cirrhosis resulting from chronic liver injury represent a major healthcare burden worldwide. Growth differentiation factor (GDF) 11 has been recently investigated for its role in rejuvenation of ageing organs, but its role in chronic liver diseases has remained unknown. Here, we investigated the expression and function of GDF11 in liver fibrosis, a common feature of most chronic liver diseases. DESIGN: We analysed the expression of GDF11 in patients with liver fibrosis, in a mouse model of liver fibrosis and in hepatic stellate cells (HSCs) as well as in other liver cell types. The functional relevance of GDF11 in toxin-induced and cholestasis-induced mouse models of liver fibrosis was examined by in vivo modulation of Gdf11 expression using adeno-associated virus (AAV) vectors. The effect of GDF11 on leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5)+ liver progenitor cells was studied in mouse and human liver organoid culture. Furthermore, in vivo depletion of LGR5+ cells was induced by injecting AAV vectors expressing diptheria toxin A under the transcriptional control of Lgr5 promoter. RESULTS: We showed that the expression of GDF11 is upregulated in patients with liver fibrosis and in experimentally induced murine liver fibrosis models. Furthermore, we found that therapeutic application of GDF11 mounts a protective response against fibrosis by increasing the number of LGR5+ progenitor cells in the liver. CONCLUSION: Collectively, our findings uncover a protective role of GDF11 during liver fibrosis and suggest a potential application of GDF11 for the treatment of chronic liver disease.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factors/metabolism , Liver Cirrhosis/metabolism , Liver/metabolism , Stem Cells/metabolism , Animals , Disease Models, Animal , Fluorescent Antibody Technique , Gene Flow , Humans , In Situ Hybridization , Liver/cytology , Male , Mice , Mice, Inbred BALB C , Up-Regulation
10.
Carcinogenesis ; 41(3): 334-344, 2020 05 14.
Article in English | MEDLINE | ID: mdl-31170286

ABSTRACT

Beyond the nearly uniform presence of KRAS mutations, pancreatic cancer is increasingly recognized as a heterogeneous disease. Preclinical in vivo model systems exist, but with the advent of precision oncology, murine models with enhanced genetic flexibility are needed to functionally annotate genetic alterations found in the human malignancy. Here, we describe the generation of focal gene disruptions and large chromosomal deletions via inducible and pancreas-specific expression of Cas9 in adult mice. Experimental mice are derived on demand directly from genetically engineered embryonic stem cells, without the need for further intercrossing. To provide initial validation of our approach, we show that disruption of the E3 ubiquitin ligase Rnf43 accelerates KrasG12D-dependent tumourigenesis. Moreover, we demonstrate that this system can be used to rapidly interrogate the impact of complex cancer-associated alleles through the generation of a previously unstudied 1.2 megabase deletion surrounding the CDKN2A and CDKN2B tumour suppressors. Thus, our approach is capable of reproducibly generating biallelic and precise loss of large chromosomal fragments that, in conjunction with mutant Kras, leads to development of pancreatic ductal adenocarcinoma with full penetrance.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , Gene Editing , Pancreatic Neoplasms/genetics , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Disease Models, Animal , Genome, Human/genetics , Humans , Mice , Mutation/genetics , Pancreas/pathology , Pancreatic Neoplasms/pathology , Precision Medicine , Sequence Deletion/genetics , Pancreatic Neoplasms
11.
Z Gastroenterol ; 58(9): 872-876, 2020 Sep.
Article in German | MEDLINE | ID: mdl-32503059

ABSTRACT

Molecular diagnostics are increasingly important to guide treatment decisions in oncologic patients. For instance, the presence of high-grade microsatellite-instability (MSI-high) is considered to be one of the major positive predictors of therapy response to immune-checkpoint inhibitors in patients with solid tumors. Based on impressive results from several immune-oncology trials, the American Food and Drug Administration (FDA) granted approval to immunotherapy in any previously treated, MSI-high solid cancer in 2017. Here, we report the clinical case of a young patient with MSI-high colorectal cancer. The case illustrates, that insurance companies in Germany are still reluctant to cover the cost of immunotherapy in this specific patient subgroup, which, in our opinion, results in an ethically problematic therapeutic dilemma.


Subject(s)
Colorectal Neoplasms/immunology , Immunotherapy/methods , Colonic Neoplasms/therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , Germany , Humans , Microsatellite Instability , Treatment Outcome
12.
Radiologe ; 60(8): 687-692, 2020 Aug.
Article in German | MEDLINE | ID: mdl-32671491

ABSTRACT

CLINICAL ISSUE: Hepatocellular carcinoma (HCC) is associated with a high mortality rate. STANDARD TREATMENT: For many years, sorafenib was the only, and frequently poorly tolerated systemic treatment option, which lead to the unreflected recurrent use of locoregional treatment modalities, such as transarterial chemoembolization (TACE). INNOVATIONS: Based on recent positive phase III trial results, we now have three systemic therapeutic options available in the first and second line of treatment, respectively. This development enables us to design sequential treatments concepts for patients with advanced HCC. DIAGNOSTIC WORK-UP: Beyond the assessment of tumor burden, the liver function of HCC patients needs to be closely monitored under therapy. PERFORMANCE: High response rates, including complete remissions have been documented for immuno-oncology-based combination regimens in HCC patients. Already today, a median overall survival (mOS) above 20 months can been achieved through the sequential application of systemic therapies in phase II studies in patients with advanced HCC and preserved liver function. ACHIEVEMENTS: Local therapies will remain an integral component of HCC therapy. However, recent advancements will shift the focus towards systemic treatment concepts. PRACTICAL RECOMMENDATIONS: The rigorous implementation of validated scoring systems can contribute towards an improved selection of patients that are suited to locoregional therapies. Longitudinal monitoring of liver function is fundamental to ensure that the optimal point in time for a switch towards systemic therapies is not missed.


Subject(s)
Carcinoma, Hepatocellular/therapy , Immunotherapy , Liver Neoplasms/therapy , Clinical Trials, Phase III as Topic , Humans , Radiology, Interventional
14.
Liver Int ; 39 Suppl 1: 79-97, 2019 05.
Article in English | MEDLINE | ID: mdl-30851232

ABSTRACT

Human cholangiocarcinoma (CCA) is an aggressive tumour entity arising from the biliary tree, whose molecular pathogenesis remains largely undeciphered. Over the last decade, the advent of high-throughput and cell-based techniques has significantly increased our knowledge on the molecular mechanisms underlying this disease while, at the same time, unravelling CCA complexity. In particular, it becomes clear that CCA displays pronounced inter- and intratumoural heterogeneity, which is presumably the consequence of the interplay between distinct tissues and cells of origin, the underlying diseases, and the associated molecular alterations. To better characterize these events and to design novel and more effective therapeutic strategies, a number of CCA experimental and preclinical models have been developed and are currently generated. This review summarizes the current knowledge and understanding of these models, critically underlining their translational usefulness and limitations. Furthermore, this review aims to provide a comprehensive overview on cells of origin, cancers stem cells and their dynamic interplay within CCA tissue.


Subject(s)
Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cholangiocarcinoma/pathology , Disease Models, Animal , Neoplastic Stem Cells/pathology , Animals , Disease Progression , Humans , Tumor Microenvironment
16.
Digestion ; 95(3): 181-185, 2017.
Article in English | MEDLINE | ID: mdl-28288474

ABSTRACT

BACKGROUND: Cholangiocarcinomas (CCAs) are aggressive malignancies that display features of biliary differentiation. According to their anatomical location, CCAs are commonly classified as intrahepatic and extrahepatic tumors, the latter entity being further subdivided into perihilar CCAs, also termed as Klatskin tumors, and distal tumors. While a majority of CCAs occur sporadically, established risk factors such as liver fluke infestation or primary sclerosing cholangitis exist. SUMMARY: Due to lack of efficient early screening markers, CCAs are frequently diagnosed at an advanced stage when curative surgical resection is not an option. Chemotherapy with gemcitabine and cisplatin is currently the standard palliative treatment that prolongs overall survival by 3.6 months as compared to monotherapy with gemcitabine. For CCA patients who progress under gemcitabine/cisplatin, the paucity of prospective, randomized trials is detrimental, and there is currently no recommended second-line regimen with respect to chemotherapy or loco-regional treatment modalities. Molecular profiling of CCAs supports the implementation of targeted approaches, and it is reasonable that personalized therapy will become a mainstay of CCA treatment. In addition, the advent of immunotherapy holds considerable promise, yet, similar to targeted treatment, needs to be prospectively evaluated in clinically and genetically thoroughly characterized patients. KEY POINTS: (1) CCA is a genetically diverse and highly aggressive malignancy. (2) Gemcitabine in combination with cisplatin or oxaliplatin is the current first-line chemotherapy in non-resectable patients. (3) Loco-regional treatment modalities exist but need to be evaluated in prospective randomized trials in the context of systemic chemotherapy. (4) Targeted therapies in molecularly defined subgroups of patients and immunotherapies alone or in combinations will most likely improve survival in the future.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bile Duct Neoplasms/therapy , Cholangiocarcinoma/therapy , Palliative Care/methods , Bile Duct Neoplasms/diagnosis , Bile Duct Neoplasms/epidemiology , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Bile Ducts, Intrahepatic/surgery , Chemotherapy, Adjuvant/methods , Cholangiocarcinoma/diagnosis , Cholangiocarcinoma/epidemiology , Cholangiocarcinoma/pathology , Cisplatin/therapeutic use , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Humans , Incidence , Liver Transplantation , Molecular Targeted Therapy/methods , Neoplasm Staging , Palliative Care/standards , Precision Medicine/methods , Survival Rate , Gemcitabine
17.
EMBO J ; 31(20): 4035-44, 2012 Oct 17.
Article in English | MEDLINE | ID: mdl-22968171

ABSTRACT

Biallelic mutations in the untranslated regions (UTRs) of mRNAs are rare causes for monogenetic diseases whose mechanisms remain poorly understood. We investigated a 3'UTR mutation resulting in a complex immunodeficiency syndrome caused by decreased mRNA levels of p14/robld3 by a previously unknown mechanism. Here, we show that the mutation creates a functional 5' splice site (SS) and that its recognition by the spliceosomal component U1 snRNP causes p14 mRNA suppression in the absence of splicing. Histone processing signals are able to rescue p14 expression. Therefore, the mutation interferes only with canonical poly(A)-site 3' end processing. Our data suggest that U1 snRNP inhibits cleavage or poly(A) site recognition. This is the first description of a 3'UTR mutation that creates a functional 5'SS causative of a monogenetic disease. Moreover, our data endorse the recently described role of U1 snRNP in suppression of intronic poly(A) sites, which is here deleterious for p14 mRNA biogenesis.


Subject(s)
3' Untranslated Regions/genetics , Adaptor Proteins, Signal Transducing/deficiency , Immunologic Deficiency Syndromes/genetics , Neutropenia/congenital , Polyadenylation/genetics , RNA Splice Sites/genetics , RNA, Small Nuclear/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Animals , Base Sequence , Conserved Sequence , Endosomes/ultrastructure , Gene Expression Regulation/drug effects , Genes, Reporter , Histones/physiology , Humans , Introns/genetics , Mammals/genetics , Molecular Sequence Data , Morpholinos/pharmacology , Neutropenia/genetics , Point Mutation , Polyadenylation/drug effects , RNA Splicing/drug effects , RNA Stability , RNA, Messenger/biosynthesis , Sequence Alignment , Sequence Homology, Nucleic Acid , Species Specificity
18.
Proc Natl Acad Sci U S A ; 110(48): 19513-8, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24154728

ABSTRACT

Cholangiocarcinoma is the second most common primary liver cancer and responds poorly to existing therapies. Intrahepatic cholangiocarcinoma (ICC) likely originates from the biliary tree and develops within the hepatic parenchyma. We have generated a flexible orthotopic allograft mouse model of ICC that incorporates common genetic alterations identified in human ICC and histologically resembles the human disease. We examined the utility of this model to validate driver alterations in ICC and tested their suitability as therapeutic targets. Specifically, we showed that the fused-in-glioblastoma-c-ros-oncogene1 (FIG-ROS1(S); FIG-ROS) fusion gene dramatically accelerates ICC development and that its inactivation in established tumors has a potent antitumor effect. Our studies establish a versatile model of ICC that will be a useful preclinical tool and validate ROS1 fusions as potent oncoproteins and therapeutic targets in ICC and potentially other tumor types.


Subject(s)
Cholangiocarcinoma/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic/genetics , Genetic Therapy/methods , Liver Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Animals , Bile Duct Neoplasms , Bile Ducts, Intrahepatic , Cell Line, Tumor , Cholangiocarcinoma/therapy , Gene Transfer Techniques , Genes, p53/genetics , Liver Neoplasms/therapy , Mice , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , RNA Interference , Retroviridae
19.
Proc Natl Acad Sci U S A ; 110(48): 19519-24, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24218589

ABSTRACT

The rapidly growing recognition of the role of oncogenic ROS1 fusion proteins in the malignant transformation of multiple cancers, including lung adenocarcinoma, cholangiocarcinoma, and glioblastoma, is driving efforts to develop effective ROS1 inhibitors for use as molecularly targeted therapy. Using a multidisciplinary approach involving small molecule screening in combination with in vitro and in vivo tumor models, we show that foretinib (GSK1363089) is a more potent ROS1 inhibitor than crizotinib (PF-02341066), an ALK/ROS inhibitor currently in clinical evaluation for lung cancer patients harboring ROS1 rearrangements. Whereas crizotinib has demonstrated promising early results in patients with ROS1-rearranged non-small-cell lung carcinoma, recently emerging clinical evidence suggests that patients may develop crizotinib resistance due to acquired point mutations in the kinase domain of ROS1, thus necessitating identification of additional potent ROS1 inhibitors for therapeutic intervention. We confirm that the ROS1(G2032R) mutant, recently reported in clinical resistance to crizotinib, retains foretinib sensitivity at concentrations below safe, clinically achievable levels. Furthermore, we use an accelerated mutagenesis screen to preemptively identify mutations in the ROS1 kinase domain that confer resistance to crizotinib and demonstrate that these mutants also remain foretinib sensitive. Taken together, our data strongly suggest that foretinib is a highly effective ROS1 inhibitor, and further clinical investigation to evaluate its potential therapeutic benefit for patients with ROS1-driven malignancies is warranted.


Subject(s)
Anilides/pharmacology , Oncogenes/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Quinolines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Base Sequence , Cell Line, Tumor , Cell Survival/drug effects , DNA Primers/genetics , Flow Cytometry , Mice , Molecular Sequence Data , Mutagenesis , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Sequence Analysis, DNA
20.
JCO Precis Oncol ; 8: e2300411, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38394466

ABSTRACT

PURPOSE: Recent studies have provided evidence for a predictive value of RNF43 genetic alterations (GAs) as biomarkers for targeted therapies in microsatellite-stable (MSS) colorectal cancer (CRC). These data have the potential to prioritize treatment strategies in patients with BRAFV600E-mutant CRC and help to identify a subgroup that is more likely to derive benefit versus those patients for whom alternative treatment approaches are needed. We were therefore interested in defining the precise frequency of BRAFV600E and RNF43 GAs and their respective overlap in a large cohort of patients with CRC. METHODS: To address this question, we performed a retrospective analysis that included 52,969 patients diagnosed with CRC from the FoundationCORE database. RESULTS: We observed a striking association of RNF43 GAs with MSI and tumor mutational burden status and BRAFV600E mutations. Overall, 23% of MSS patients with confirmed BRAFV600E mutation harbor an RNF43 GA-which accounts for 1.1% of all patients with CRC and for 15.7% of all CRC BRAFV600E cases. CONCLUSION: Ongoing phase III clinical trials, such as BREAKWATER, should aim to incorporate broader genetic profiling to further validate the superior sensitivity of patients with RNF43-mutant, MSS BRAFV600E CRC to anti-EGFR-/BRAFi-based therapies.


Subject(s)
Colorectal Neoplasms , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Retrospective Studies , Microsatellite Instability , Mutation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL