1
|
Oh DY, He AR, Qin S, Chen LT, Okusaka T, Kim JW, Suksombooncharoen T, Lee MA, Kitano M, Burris HA, Bouattour M, Tanasanvimon S, Zaucha R, Avallone A, Cundom J, Kuzko A, Wang J, Xynos I, Vogel A, Valle JW. Durvalumab plus chemotherapy in advanced biliary tract cancer: 3-year overall survival update from the phase III TOPAZ-1 study. J Hepatol 2025:S0168-8278(25)02201-9. [PMID: 40381735 DOI: 10.1016/j.jhep.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/29/2025] [Accepted: 05/01/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND At the TOPAZ-1 (NCT03875235) primary analysis, durvalumab plus gemcitabine and cisplatin (GemCis) significantly improved overall survival (OS) in advanced biliary tract cancer (aBTC). We report updated exploratory analyses of OS and safety, characterisation of extended long-term survivors (eLTS), and subsequent anticancer therapy (SAT) use. METHODS Participants with aBTC received durvalumab+GemCis or placebo+GemCis every 3 weeks (≤8 cycles), then durvalumab or placebo monotherapy every 4 weeks until progressive disease or other discontinuation criteria were met. OS and serious adverse events (SAEs) were assessed in the full analysis and safety analysis sets (FAS/SAS), respectively. eLTS outcomes were assessed (FAS participants alive ≥30 months after randomisation). RESULTS 685 participants were randomised: durvalumab+GemCis (n = 341); placebo+GemCis (n = 344). After a median 41.3 (95% confidence interval [CI] 39.3-44.1) months' follow-up in all participants, median OS (95% CI) for durvalumab+GemCis versus placebo+GemCis was 12.9 (11.6-14.1) versus 11.3 (10.1-12.5) months (hazard ratio, 0.74 [95% CI 0.63-0.87]); 36-month OS rate was 14.6% versus 6.9%, respectively. In participants who achieved disease control (566/685; 82.6%), the 36-month OS rate was higher for durvalumab+GemCis (17.0%) versus placebo+GemCis (7.6%). Overall, 12.8% were eLTS, with more eLTS in the durvalumab+GemCis (17.0%) versus placebo+GemCis (8.7%) arms; eLTS included all clinically relevant subgroups. Durvalumab+GemCis improved OS regardless of SAT use. In eLTS, SAEs were comparable between arms and less frequent than in the full SAS. CONCLUSIONS Survival benefit and manageable safety continued with durvalumab+GemCis versus placebo+GemCis approximately 3 years after the last participant was randomised. All clinically relevant subgroups were represented in eLTS, supporting standard-of-care status for durvalumab+GemCis in aBTC. LAY SUMMARY The TOPAZ-1 study found that treatment with durvalumab plus gemcitabine and cisplatin (chemotherapy, also known as GemCis), helped people with advanced biliary tract cancer (BTC) to live longer on average than those treated with a placebo plus GemCis. The latest results from TOPAZ-1 showed that these benefits continued for over 3 years in participants treated with durvalumab plus GemCis and side effects continued to be manageable. At an updated analysis, carried out 3 years after the last participant started the study, twice as many participants treated with durvalumab plus GemCis were alive compared to those treated with placebo plus GemCis. Results also showed that the positive effect of durvalumab plus GemCis compared with placebo plus GemCis was not affected by the use of other therapies some participants received after they finished the study treatment. These results continue to support durvalumab plus GemCis as a standard first-line treatment for people with advanced BTC. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT03875235; https://clinicaltrials.gov/study/NCT03875235.
Collapse
Affiliation(s)
- Do-Youn Oh
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Aiwu Ruth He
- Division of Hematology and Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA
| | - Shukui Qin
- Cancer Center of Nanjing, Jinling Hospital, Nanjing, China
| | - Li-Tzong Chen
- Kaohsiung Medical University Hospital and Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Jin Won Kim
- Medical Oncology, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | | | - Myung Ah Lee
- Seoul St. Mary's Hospital, College of Medicine, Cancer Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Masayuki Kitano
- Second Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| | - Howard A Burris
- Drug Development Department, Sarah Cannon Research Institute, Nashville, Tennessee, USA
| | | | - Suebpong Tanasanvimon
- Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Renata Zaucha
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Antonio Avallone
- Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples, Italy
| | - Juan Cundom
- Medical Oncology, Instituto de Investigaciones Metabólicas, Buenos Aires, Argentina
| | | | - Julie Wang
- Oncology R&D, AstraZeneca, New York, USA
| | | | - Arndt Vogel
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada; Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Medical School Hannover, Hannover, Germany
| | - Juan W Valle
- Research, Cholangiocarcinoma Foundation, Herriman, Utah, USA; Division of Cancer Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
2
|
Bekric D, Kiesslich T, Ocker M, Winklmayr M, Ritter M, Dobias H, Beyreis M, Neureiter D, Mayr C. The efficacy of ferroptosis-inducing compounds IKE and RSL3 correlates with the expression of ferroptotic pathway regulators CD71 and SLC7A11 in biliary tract cancer cells. PLoS One 2024; 19:e0302050. [PMID: 38603713 PMCID: PMC11008848 DOI: 10.1371/journal.pone.0302050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
INTRODUCTION Biliary tract cancer (BTC) is a lethal disease with a bad overall survivability, partly arising from inadequate therapeutic alternatives, detection at a belated stage, and a resistance to common therapeutic approaches. Ferroptosis is a form of programmed cell death that depends on reactive oxygen species (ROS) and iron, causing excessive peroxidation of polyunsaturated fatty acids (PUFAs). Therefore, the objective of this investigation is, whether ferroptosis can be induced in BTC in vitro and whether this induction is dependent on specific molecular markers. METHODS The study conducted resazurin assay and IC25/50 calculation to explore the possible cytotoxic outcomes of different classes of ferroptosis-inducing substances (FINs) on a comprehensive in vitro model of 11 BTC cell lines. Combinatory treatments with different cell death inhibitors were performed to evaluate the magnitude of ferroptosis induction. To ascertain whether ferroptotic cell death occurred, liperfluo and iron assay kits were employed to evaluate lipid ROS and intracellular iron abundance. Potential biomarkers of ferroptosis sensitivity were then assessed via western blot analysis, a rtPCR panel and functional assay kits. RESULTS The study found that different FINs reduced cell viability in a cell line-dependent manner. In addition, we measured increased lipid ROS and intracellular Fe2+ levels upon exposure to FINs in BTC cells. Combining FINs with inhibitors of ferroptosis, necroptosis or apoptosis suggests the occurrence of ferroptotic events in BTC cell lines CCC-5, HuH-28 and KKU-055. Furthermore, we found that BTC cells display a heterogeneous profile regarding different molecular genes/markers of ferroptosis. Subsequent analysis revealed that sensitivity of BTC cells towards IKE and RSL3 positively correlated with CD71 and SLC7A11 protein expression. CONCLUSION Our results demonstrate that induction of ferroptosis is a promising approach to inhibit BTC cell growth and that the sensitivity of BTC cells towards ferroptosis induction might be dependent on molecular markers such as CD71 and SLC7A11.
Collapse
Affiliation(s)
- Dino Bekric
- Center of Physiology, Pathophysiology and Biophysics, Institute of Physiology and Pathophysiology Salzburg, Paracelsus Medical University, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Tobias Kiesslich
- Center of Physiology, Pathophysiology and Biophysics, Institute of Physiology and Pathophysiology Salzburg, Paracelsus Medical University, Salzburg, Austria
- Department of Internal Medicine I, University Clinics Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Matthias Ocker
- Division of Hematology, Oncology, and Cancer Immunology, Medical Department, Charité University Medicine Berlin, Berlin, Germany
- Tacalyx GmbH, Berlin, Germany
| | - Martina Winklmayr
- Ludwig Boltzmann Institute for Arthritis und Rehabilitation, Paracelsus Medical University, Salzburg, Austria
| | - Markus Ritter
- Center of Physiology, Pathophysiology and Biophysics, Institute of Physiology and Pathophysiology Salzburg, Paracelsus Medical University, Salzburg, Austria
- Ludwig Boltzmann Institute for Arthritis und Rehabilitation, Paracelsus Medical University, Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, Salzburg, Austria
- Kathmandu Medical School of Medical Sciences, Dhulikhel, Nepal
| | - Heidemarie Dobias
- Center of Physiology, Pathophysiology and Biophysics, Institute of Physiology and Pathophysiology Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Marlena Beyreis
- Center of Physiology, Pathophysiology and Biophysics, Institute of Physiology and Pathophysiology Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Daniel Neureiter
- Cancer Cluster Salzburg, Salzburg, Austria
- Institute of Pathology, University Clinics Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Christian Mayr
- Center of Physiology, Pathophysiology and Biophysics, Institute of Physiology and Pathophysiology Salzburg, Paracelsus Medical University, Salzburg, Austria
- Department of Internal Medicine I, University Clinics Salzburg, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
3
|
Requeijo C, Bracchiglione J, Meza N, Acosta-Dighero R, Salazar J, Santero M, Meade AG, Quintana MJ, Rodríguez-Grijalva G, Selva A, Solà I, Urrútia G, Bonfill Cosp X, On behalf of Appropriateness of Systemic Oncological Treatments for Advanced Cancer (ASTAC) Research Group. Anticancer Drugs Compared to No Anticancer Drugs in Patients with Advanced Hepatobiliary Cancer: A Mapping Review and Evidence Gap Map. Clin Epidemiol 2023; 15:1069-1085. [PMID: 38025841 PMCID: PMC10644842 DOI: 10.2147/clep.s431498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/14/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Despite being commonly recommended, the impact of anticancer drugs (ACDs) on patient-important outcomes beyond survival for advanced hepatobiliary cancers (HBCs) may not have been sufficiently assessed. We aim to identify and map the evidence regarding ACDs versus best supportive care (BSC) for advanced HBCs, considering patient-centered outcomes. Methods In this mapping review, we included systematic reviews, randomized controlled trials, quasi-experimental, and observational studies comparing ACDs (chemotherapy, immunotherapy, biological/targeted therapy) versus BSC for advanced HBCs. We searched MEDLINE (PubMed), EMBASE (Ovid), Cochrane Library, Epistemonikos, PROSPERO and clinicaltrials.gov for eligible studies. Two reviewers performed the screening and data extraction processes. We developed evidence maps for each type of cancer. Results We included 87 studies (60 for advanced liver cancer and 27 for gallbladder or bile duct cancers). Most of the evidence favored ACDs for survival outcomes, and BSC for toxicity. We identified several evidence gaps for non-survival outcomes, including quality of life or quality of end-of-life care. Discussion Patient-important outcomes beyond survival in advanced HBCs are insufficiently assessed by the available evidence. Future studies need to address these gaps to better inform decision-making processes.
Collapse
Affiliation(s)
- Carolina Requeijo
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Javier Bracchiglione
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- Interdisciplinary Centre for Health Studies (CIESAL), Valparaiso University, Viña del Mar, Chile
- CIBER of Epidemiology and Public Health (CIBERESP), Barcelona, Spain
| | - Nicolás Meza
- Interdisciplinary Centre for Health Studies (CIESAL), Valparaiso University, Viña del Mar, Chile
| | - Roberto Acosta-Dighero
- Interdisciplinary Centre for Health Studies (CIESAL), Valparaiso University, Viña del Mar, Chile
| | - Josefina Salazar
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Marilina Santero
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Adriana-G Meade
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - María Jesús Quintana
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | | | - Anna Selva
- CIBER of Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- Clinical Epidemiology and Cancer Screening, Parc Taulí Hospital Universitari, Parc Taulí Research and Innovation Institute Foundation (I3PT-CERCA), Autonomous University of Barcelona, Sabadell, Spain
| | - Ivan Solà
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Barcelona, Spain
| | - Gerard Urrútia
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Barcelona, Spain
| | - Xavier Bonfill Cosp
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - On behalf of Appropriateness of Systemic Oncological Treatments for Advanced Cancer (ASTAC) Research Group
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
- Interdisciplinary Centre for Health Studies (CIESAL), Valparaiso University, Viña del Mar, Chile
- CIBER of Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
- Clinical Epidemiology and Cancer Screening, Parc Taulí Hospital Universitari, Parc Taulí Research and Innovation Institute Foundation (I3PT-CERCA), Autonomous University of Barcelona, Sabadell, Spain
| |
Collapse
|
4
|
Bekric D, Tornesello ML, Ocker M, Mayr C, Kiesslich T, Neureiter D. Editorial: Novel therapeutic approaches for biliary tract cancer and hepatocellular carcinoma. Front Cell Dev Biol 2023; 11:1320084. [PMID: 38020902 PMCID: PMC10656678 DOI: 10.3389/fcell.2023.1320084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Affiliation(s)
- Dino Bekric
- Center of Physiology, Pathophysiology and Biophysics, Institute of Physiology and Pathophysiology Salzburg, Paracelsus Medical University, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| | - Maria Lina Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Naples, Italy
| | - Matthias Ocker
- Medical Department, Division of Hematology, Oncology, and Cancer Immunology, Campus Charité Mitte, Charité University Medicine Berlin, Germany and Tacalyx GmbH, Berlin, Germany
| | - Christian Mayr
- Center of Physiology, Pathophysiology and Biophysics, Institute of Physiology and Pathophysiology Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Tobias Kiesslich
- Center of Physiology, Pathophysiology and Biophysics, Institute of Physiology and Pathophysiology Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Daniel Neureiter
- Cancer Cluster Salzburg, Salzburg, Austria
- Institute of Pathology, University Clinics Salzburg, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
5
|
Doleschal B, Taghizadeh H, Webersinke G, Piringer G, Schreil G, Decker J, Aichberger KJ, Kirchweger P, Thaler J, Petzer A, Schmitt CA, Prager GW, Rumpold H. Real world evidence reveals improved survival outcomes in biliary tract cancer through molecular matched targeted treatment. Sci Rep 2023; 13:15421. [PMID: 37723192 PMCID: PMC10507096 DOI: 10.1038/s41598-023-42083-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/05/2023] [Indexed: 09/20/2023] Open
Abstract
Biliary tract cancers are rare cancers with poor prognosis due to a lack of therapeutic options, especially after the failure of first-line systemic treatment. Targeted treatments for this clinical situation are promising and have entered clinical practice. We aimed to describe the overall survival of matched targeted treatment after first-line treatment in patients with biliary tract cancers in an Austrian real-world multicenter cohort. We performed a multicenter retrospective chart review of patients with biliary tract cancer between September 2015 and January 2022. Data, including comprehensive molecular characteristics-next generation sequencing (NGS) and immunohistochemistry (IHC), clinical history, surgical procedures, ablative treatments, patient history, and systemic chemotherapy, were extracted from the records of the participating institutions. Targeted treatment was matched according to the ESMO scale for the clinical actionability of molecular targets (ESCAT). We identified 159 patients with the available molecular characteristics. A total of 79 patients underwent second-line treatment. Of these, 36 patients received matched targeted treatment beyond the first-line and were compared with 43 patients treated with cytotoxic chemotherapy in terms of efficacy outcomes. For Tier I/II alterations, we observed a progression free survival ratio (PFStargeted/PFSpre-chemotherapy) of 1.86, p = 0.059. The overall survival for patients receiving at least two lines of systemic treatment significantly favored the targeted approach, with an overall survival of 22.3 months (95% CI 14.7-29.3) vs. 17.5 months (95% CI 1.7-19.8; p = 0.048). Our results underscore the value of targeted treatment approaches based on extended molecular characterization of biliary tract cancer to improve clinical outcomes.
Collapse
Affiliation(s)
- Bernhard Doleschal
- Department of Internal Medicine I for Hematology with Stem Cell Transplantation, Hemostaseology, and Medical Oncology, Ordensklinikum Linz, Seilerstaette 4, 4010, Linz, Austria.
| | - Hossein Taghizadeh
- Department of Internal Medicine, University Hospital St. Pölten, St. Pölten, Austria
| | - Gerald Webersinke
- Laboratory for Molecular Genetic Diagnostics, Ordensklinikum Linz, Linz, Austria
| | - Gudrun Piringer
- Department of Oncology and Hematology, Kepler University Clinic Linz, Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Georg Schreil
- Department of Internal Medicine, State Hospital Pyhrn Eisenwurzen, Steyr, Austria
| | - Jörn Decker
- Department of Internal Medicine, State Hospital Rohrbach, Rohrbach, Austria
| | - Karl J Aichberger
- Department of Internal Medicine, State Hospital Rohrbach, Rohrbach, Austria
| | - Patrick Kirchweger
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
- Department of General and Visceral Surgery, Ordensklinikum Linz, Linz, Austria
- Gastrointestinal Cancer Center, Ordensklinikum Linz, Linz, Austria
| | - Josef Thaler
- Department of Internal Medicine IV, Hospital Wels-Grieskirchen, Wels, Austria
| | - Andreas Petzer
- Department of Internal Medicine I for Hematology with Stem Cell Transplantation, Hemostaseology, and Medical Oncology, Ordensklinikum Linz, Seilerstaette 4, 4010, Linz, Austria
| | - Clemens A Schmitt
- Department of Oncology and Hematology, Kepler University Clinic Linz, Linz, Austria
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Gerald W Prager
- Division of Oncology, Department of Medicine I, Medical University Vienna, Vienna, Austria
| | - Holger Rumpold
- Medical Faculty, Johannes Kepler University Linz, Linz, Austria
- Gastrointestinal Cancer Center, Ordensklinikum Linz, Linz, Austria
| |
Collapse
|
6
|
Evaluation of Tazemetostat as a Therapeutically Relevant Substance in Biliary Tract Cancer. Cancers (Basel) 2023; 15:cancers15051569. [PMID: 36900361 PMCID: PMC10000745 DOI: 10.3390/cancers15051569] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Biliary tract cancer (BTC) is a gastrointestinal malignancy associated with a poor survival rate. Current therapies encompass palliative and chemotherapeutic treatment as well as radiation therapy, which results in a median survival of only one year due to standard therapeutic ineffectiveness or resistance. Tazemetostat is an FDA-approved inhibitor of enhancer of Zeste homolog 2 (EZH2), a methyltransferase involved in BTC tumorigenesis via trimethylation of histone 3 at lysine 27 (H3K27me3), an epigenetic mark associated with silencing of tumor suppressor genes. Up to now, there are no data available regarding tazemetostat as a possible treatment option against BTC. Therefore, the aim of our study is a first-time investigation of tazemetostat as a potential anti-BTC substance in vitro. In this study, we demonstrate that tazemetostat affects cell viability and the clonogenic growth of BTC cells in a cell line-dependent manner. Furthermore, we found a strong epigenetic effect at low concentrations of tazemetostat, which was independent of the cytotoxic effect. We also observed in one BTC cell line that tazemetostat increases the mRNA levels and protein expression of the tumor suppressor gene Fructose-1,6-bisphosphatase 1 (FBP1). Interestingly, the observed cytotoxic and epigenetic effects were independent of the mutation status of EZH2. To conclude, our study shows that tazemetostat is a potential anti-tumorigenic substance in BTC with a strong epigenetic effect.
Collapse
|
7
|
Khankhel ZS, Goring S, Bobiak S, Lamy FX, Nayak D, Garside J, Reese ES, Schoenherr N. Second-line treatments in advanced biliary tract cancer: systematic literature review of efficacy, effectiveness and safety. Future Oncol 2022; 18:2321-2338. [PMID: 35387496 DOI: 10.2217/fon-2021-1302] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: A systematic review was conducted to understand clinical, economic and health-related quality-of-life outcomes in second-line biliary tract cancer. Materials & methods: The review followed established recommendations. The feasibility of network meta-analysis revealed limited networks, thus synthesis was limited to a summary of reported ranges, percentiles and medians. Results: The review included 62 trials and observational studies highly variable with respect to key baseline characteristics. Commonly evaluated second-line treatments included fluoropyrimidine-, gemcitabine- and S-1-based regimens. Across active treatment arms, median overall survival ranged from 3.5 to 15.0 months (median: 6.9), median progression-free survival from 1.4 to 6.5 months (median: 2.9) and objective response from 0 to 36.4%. Outcomes were similar between study types, with a few notable outliers. Treatment-related/-emergent adverse events were infrequently reported; no studies reported economic or health-related quality-of-life outcomes. Conclusions: Biliary tract cancer is a difficult-to-treat disease with poor prognosis. Despite evolving treatment landscapes, more recent studies did not show clinical outcome improvement, highlighting an unmet need among advanced/metastatic patients.
Collapse
Affiliation(s)
| | - Sarah Goring
- SMG Outcomes Research, Vancouver, BC, V6T0C2, Canada
| | - Sarah Bobiak
- EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, USA, an affiliate of Merck KGaA
| | | | | | | | - Emily S Reese
- EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, USA, an affiliate of Merck KGaA
| | | |
Collapse
|
8
|
Benefit of metastasectomy in renal cell carcinoma: A propensity score analysis. Clin Genitourin Cancer 2022; 20:344-353. [DOI: 10.1016/j.clgc.2022.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/15/2021] [Accepted: 03/21/2022] [Indexed: 12/14/2022]
|
9
|
Gonzalez-Carmona MA, Möhring C, Mahn R, Zhou T, Bartels A, Sadeghlar F, Bolch M, Vogt A, Kaczmarek DJ, Heling DJ, Dold L, Nattermann J, Branchi V, Matthaei H, Manekeller S, Kalff JC, Strassburg CP, Mohr RU, Weismüller TJ. Impact of regular additional endobiliary radiofrequency ablation on survival of patients with advanced extrahepatic cholangiocarcinoma under systemic chemotherapy. Sci Rep 2022; 12:1011. [PMID: 35046437 PMCID: PMC8770452 DOI: 10.1038/s41598-021-04297-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022] Open
Abstract
Prognosis of patients with advanced extrahepatic cholangiocarcinoma (eCCA) is poor. The current standard first-line treatment is systemic chemotherapy (CT) with gemcitabine and a platinum derivate. Additionally, endobiliary radiofrequency ablation (eRFA) can be applied to treat biliary obstructions. This study aimed to evaluate the additional benefit of scheduled regular eRFA in a real-life patient cohort with advanced extrahepatic cholangiocarcinoma under standard systemic CT. All patients with irresectable eCCA treated at University Hospital Bonn between 2010 and 2020 were eligible for inclusion. Patients were stratified according to treatment: standard CT (n = 26) vs. combination of eRFA with standard CT (n = 40). Overall survival (OS), progression free survival (PFS), feasibility and toxicity were retrospectively analyzed using univariate and multivariate approaches. Combined eRFA and CT resulted in significantly longer median OS (17.3 vs. 8.6 months, p = 0.004) and PFS (12.9 vs. 5.7 months, p = 0.045) compared to the CT only group. While groups did not differ regarding age, sex, tumor stage and chemotherapy treatment regimen, mean MELD was even higher (10.1 vs. 6.7, p = 0.015) in the eRFA + CT group. The survival benefit of concomitant eRFA was more evident in the subgroup with locally advanced tumors. Severe hematological toxicities (CTCAE grades 3 - 5) did not differ significantly between the groups. However, therapy-related cholangitis occurred more often in the combined treatment group (p = 0.031). Combination of eRFA and systemic CT was feasible, well-tolerated and could significantly prolong survival compared to standard CT alone. Thus, eRFA should be considered during therapeutic decision making in advanced eCCA.
Collapse
Affiliation(s)
- Maria A Gonzalez-Carmona
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| | - Christian Möhring
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Robert Mahn
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Taotao Zhou
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Alexandra Bartels
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Farsaneh Sadeghlar
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Maximilian Bolch
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Annabelle Vogt
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Dominik J Kaczmarek
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Dominik J Heling
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Leona Dold
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Jacob Nattermann
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Vittorio Branchi
- Department of Surgery, University Hospital of Bonn, Bonn, Germany
| | - Hanno Matthaei
- Department of Surgery, University Hospital of Bonn, Bonn, Germany
| | | | - Jörg C Kalff
- Department of Surgery, University Hospital of Bonn, Bonn, Germany
| | - Christian P Strassburg
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Raphael U Mohr
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Tobias J Weismüller
- Department of Internal Medicine I, University Hospital of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
10
|
Möhring C, Feder J, Mohr RU, Sadeghlar F, Bartels A, Mahn R, Zhou T, Marinova M, Feldmann G, Brossart P, von Websky M, Matthaei H, Manekeller S, Glowka T, Kalff JC, Weismüller TJ, Strassburg CP, Gonzalez-Carmona MA. First Line and Second Line Chemotherapy in Advanced Cholangiocarcinoma and Impact of Dose Reduction of Chemotherapy: A Retrospective Analysis. Front Oncol 2021; 11:717397. [PMID: 34858809 PMCID: PMC8631360 DOI: 10.3389/fonc.2021.717397] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 10/22/2021] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Prognosis of patients with irresectable cholangiocarcinoma is still poor. The ABC-02 trial established the current first line (1L) standard systemic chemotherapy (CT) with gemcitabine/platinum derivate for advanced cholangiocarcinoma. However, the majority of patients needed therapy adaptions. Thus, the aim of this study was to evaluate 1L and second line (2L) therapy regimens and the impact of therapy adaptions in an unselected real-life cohort of patients with advanced cholangiocarcinoma. MATERIALS AND METHODS This is a single institution retrospective analysis of patients with irresectable cholangiocarcinoma who were treated with gemcitabine/platinum derivate from 2010 to 2018. Overall survival (OS), progression-free survival (PFS) and toxicity were analyzed for all patients, especially with regard to CT de-escalation. RESULTS Fifty-eight patients receiving gemcitabine/platinum derivate were included in the analysis. Median OS and PFS were 12.2 and 6.9 months. Interestingly, 41 patients (71%) needed therapy de-escalation. However, despite reduced CT exposition, there was no-significant difference in OS (10.8 months vs. 15.6 months, p = 0.127), and patients suffered from less adverse events during CT. 21 (36%) patients reached 2L CT, most often with FOLFIRI (57%). Survival beyond the end of 1L CT was 7.1 months with 2L CT vs. 2.9 months with BSC. CONCLUSION In our study, the combination of gemcitabine/platinum derivate showed similar OS and PFS as randomized prospective phase II/III trials. Therapy regimen adaptions were needed in the majority of patients. However, individualized modifications of the therapy regimen allowed better tolerance as well as continuation of therapy and did not significantly influence median OS. Furthermore, our study revealed a potential survival benefit with 2L CT for selected patients.
Collapse
Affiliation(s)
- Christian Möhring
- Department of Internal Medicine I, University Hospital, Bonn, Germany
| | - Jan Feder
- Department of Internal Medicine I, University Hospital, Bonn, Germany
| | - Raphael U. Mohr
- Department of Internal Medicine I, University Hospital, Bonn, Germany
| | | | - Alexandra Bartels
- Department of Internal Medicine I, University Hospital, Bonn, Germany
| | - Robert Mahn
- Department of Internal Medicine I, University Hospital, Bonn, Germany
| | - Taotao Zhou
- Department of Internal Medicine I, University Hospital, Bonn, Germany
| | - Milka Marinova
- Department of Radiology, University Hospital, Bonn, Germany
| | - Georg Feldmann
- Department of Internal Medicine III, University Hospital, Bonn, Germany
| | - Peter Brossart
- Department of Internal Medicine III, University Hospital, Bonn, Germany
| | - Martin von Websky
- Department of Internal Medicine III, University Hospital, Bonn, Germany
| | - Hanno Matthaei
- Department of Internal Medicine III, University Hospital, Bonn, Germany
| | | | - Tim Glowka
- Department of Internal Medicine III, University Hospital, Bonn, Germany
| | - Jörg C. Kalff
- Department of Internal Medicine III, University Hospital, Bonn, Germany
| | | | | | | |
Collapse
|
11
|
Zhu AX, Macarulla T, Javle MM, Kelley RK, Lubner SJ, Adeva J, Cleary JM, Catenacci DVT, Borad MJ, Bridgewater JA, Harris WP, Murphy AG, Oh DY, Whisenant JR, Lowery MA, Goyal L, Shroff RT, El-Khoueiry AB, Chamberlain CX, Aguado-Fraile E, Choe S, Wu B, Liu H, Gliser C, Pandya SS, Valle JW, Abou-Alfa GK. Final Overall Survival Efficacy Results of Ivosidenib for Patients With Advanced Cholangiocarcinoma With IDH1 Mutation: The Phase 3 Randomized Clinical ClarIDHy Trial. JAMA Oncol 2021; 7:1669-1677. [PMID: 34554208 PMCID: PMC8461552 DOI: 10.1001/jamaoncol.2021.3836] [Citation(s) in RCA: 296] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Question Does ivosidenib treatment improve overall survival outcomes vs placebo among patients with chemotherapy-refractory cholangiocarcinoma with IDH1 mutation? Findings In this phase 3 randomized clinical trial including 187 previously treated patients with advanced cholangiocarcinoma with IDH1 mutation, ivosidenib treatment resulted in numerically improved overall survival benefits vs placebo, despite a high rate of crossover. Ivosidenib preserved certain quality of life subscales and was well tolerated. Meaning The combined efficacy data and tolerable safety profile, as well as corroborating quality of life data, support the clinical benefit of ivosidenib relative to placebo in cholangiocarcinoma with IDH1 mutation, which has an unmet need for new treatments. Importance Isocitrate dehydrogenase 1 (IDH1) variations occur in up to approximately 20% of patients with intrahepatic cholangiocarcinoma. In the ClarIDHy trial, progression-free survival as determined by central review was significantly improved with ivosidenib vs placebo. Objective To report the final overall survival (OS) results from the ClarIDHy trial, which aimed to demonstrate the efficacy of ivosidenib (AG-120)—a first-in-class, oral, small-molecule inhibitor of mutant IDH1—vs placebo for patients with unresectable or metastatic cholangiocarcinoma with IDH1 mutation. Design, Setting, and Participants This multicenter, randomized, double-blind, placebo-controlled, clinical phase 3 trial was conducted from February 20, 2017, to May 31, 2020, at 49 hospitals across 6 countries among patients aged 18 years or older with cholangiocarcinoma with IDH1 mutation whose disease progressed with prior therapy. Interventions Patients were randomized 2:1 to receive ivosidenib, 500 mg, once daily or matched placebo. Crossover from placebo to ivosidenib was permitted if patients had disease progression as determined by radiographic findings. Main Outcomes and Measures The primary end point was progression-free survival as determined by blinded independent radiology center (reported previously). Overall survival was a key secondary end point. The primary analysis of OS followed the intent-to-treat principle. Other secondary end points included objective response rate, safety and tolerability, and quality of life. Results Overall, 187 patients (median age, 62 years [range, 33-83 years]) were randomly assigned to receive ivosidenib (n = 126; 82 women [65%]; median age, 61 years [range, 33-80 years]) or placebo (n = 61; 37 women [61%]; median age, 63 years [range, 40-83 years]); 43 patients crossed over from placebo to ivosidenib. The primary end point of progression-free survival was reported elsewhere. Median OS was 10.3 months (95% CI, 7.8-12.4 months) with ivosidenib vs 7.5 months (95% CI, 4.8-11.1 months) with placebo (hazard ratio, 0.79 [95% CI, 0.56-1.12]; 1-sided P = .09). When adjusted for crossover, median OS with placebo was 5.1 months (95% CI, 3.8-7.6 months; hazard ratio, 0.49 [95% CI, 0.34-0.70]; 1-sided P < .001). The most common grade 3 or higher treatment-emergent adverse event (≥5%) reported in both groups was ascites (11 patients [9%] receiving ivosidenib and 4 patients [7%] receiving placebo). Serious treatment-emergent adverse events considered ivosidenib related were reported in 3 patients (2%). There were no treatment-related deaths. Patients receiving ivosidenib reported no apparent decline in quality of life compared with placebo. Conclusions and Relevance This randomized clinical trial found that ivosidenib was well tolerated and resulted in a favorable OS benefit vs placebo, despite a high rate of crossover. These data, coupled with supportive quality of life data and a tolerable safety profile, demonstrate the clinical benefit of ivosidenib for patients with advanced cholangiocarcinoma with IDH1 mutation. Trial Registration ClinicalTrials.gov Identifier: NCT02989857
Collapse
Affiliation(s)
- Andrew X Zhu
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston.,Jiahui International Cancer Center, Jiahui Health, Shanghai, China
| | | | - Milind M Javle
- Department of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - R Kate Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco
| | - Sam J Lubner
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison
| | - Jorge Adeva
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - James M Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Mitesh J Borad
- Department of Hematology-Oncology, Mayo Clinic, Scottsdale, Arizona
| | - John A Bridgewater
- Department of Medical Oncology, University College London Cancer Institute, London, United Kingdom
| | | | - Adrian G Murphy
- Department of Oncology-Gastrointestinal Cancer, Johns Hopkins University, Baltimore, Maryland
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | | | - Maeve A Lowery
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston.,Trinity St James Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Lipika Goyal
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston
| | - Rachna T Shroff
- Department of Medicine, University of Arizona Cancer Center, Tucson
| | - Anthony B El-Khoueiry
- Department of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles
| | - Christina X Chamberlain
- Agios Pharmaceuticals Inc, Cambridge, Massachusetts.,Now with Servier Pharmaceuticals, LLC, Boston, Massachusetts
| | - Elia Aguado-Fraile
- Agios Pharmaceuticals Inc, Cambridge, Massachusetts.,Repare Therapeutics, Cambridge, Massachusetts
| | - Sung Choe
- Agios Pharmaceuticals Inc, Cambridge, Massachusetts.,Now with Servier Pharmaceuticals, LLC, Boston, Massachusetts
| | - Bin Wu
- Agios Pharmaceuticals Inc, Cambridge, Massachusetts.,Bristol Myers Squibb, Cambridge, Massachusetts
| | - Hua Liu
- Agios Pharmaceuticals Inc, Cambridge, Massachusetts.,Now with Servier Pharmaceuticals, LLC, Boston, Massachusetts
| | - Camelia Gliser
- Agios Pharmaceuticals Inc, Cambridge, Massachusetts.,Now with Servier Pharmaceuticals, LLC, Boston, Massachusetts
| | - Shuchi S Pandya
- Agios Pharmaceuticals Inc, Cambridge, Massachusetts.,Now with Servier Pharmaceuticals, LLC, Boston, Massachusetts
| | - Juan W Valle
- Division of Cancer Sciences, University of Manchester, Department of Medical Oncology, The Christie National Health Service Foundation Trust, Manchester, United Kingdom
| | - Ghassan K Abou-Alfa
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Medical College at Cornell University, New York, New York
| |
Collapse
|
12
|
Müller M, Posch F, Kiem D, Barth D, Horvath L, Stotz M, Schaberl-Moser R, Pichler M, Greil R, Jost PJ, Seeber A, Amann A, Schlick K, Gerger A, Riedl JM. Benefit of second-line therapy for advanced esophageal squamous cell carcinoma: a tri-center propensity score analysis. Ther Adv Med Oncol 2021; 13:17588359211039930. [PMID: 34616490 PMCID: PMC8488508 DOI: 10.1177/17588359211039930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 07/26/2021] [Indexed: 12/24/2022] Open
Abstract
Background The level of evidence for palliative second-line therapy in advanced esophageal squamous cell carcinoma (aESCC) is limited. This is the first study that reports efficacy data comparing second-line therapy + active symptom control (ASC) versus ASC alone in aESCC. Methods We conducted a tri-center retrospective cohort study (n = 166) including patients with aESCC who had experienced disease progression on palliative first-line therapy. A propensity score model using inverse probability of treatment weighting (IPTW) was implemented for comparative efficacy analysis of overall survival (OS) in patients with second-line + ASC (n = 92, 55%) versus ASC alone (n = 74, 45%). Results The most frequent second-line regimens used were docetaxel (36%) and paclitaxel (18%). In unadjusted primary endpoint analysis, second-line + ASC was associated with significantly longer OS compared with ASC alone [hazard ratio (HR) = 0.49, 95% confidence interval (CI): 0.35-0.69, p < 0.0001]. However, patients in the second-line + ASC group were characterized by more favorable baseline features including a better Eastern Cooperative Oncology Group (ECOG) performance status, a longer first-line treatment duration and lower C-reactive protein levels. After rigorous adjusting for baseline confounders by re-weighting the data with the IPTW the favorable association between second-line and longer OS weakened but prevailed. The median OS was 6.1 months in the second-line + ASC group and 3.2 months in the ASC group, respectively (IPTW-adjusted HR = 0.40, 95% CI: 0.24-0.69, p = 0.001). Importantly, the benefit of second-line was consistent across several clinical subgroups, including patients with ECOG performance status ⩾1 and age ⩾65 years. The most common grade 3 or 4 adverse events associated with palliative second-line therapy were hematological toxicities. Conclusion This real-world study supports the concept that systemic second-line therapy prolongs survival in patients with aESCC.
Collapse
Affiliation(s)
- Moritz Müller
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Florian Posch
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Dominik Kiem
- IIIrd Medical Department of Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectious Disease, Salzburg Cancer Research Institute, Paracelsus Medical University, Salzburg, Austria
| | - Dominik Barth
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Lena Horvath
- Department of Internal Medicine V: Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Stotz
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Renate Schaberl-Moser
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Richard Greil
- IIIrd Medical Department of Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectious Disease, Salzburg Cancer Research Institute, Paracelsus Medical University, Salzburg, Austria
| | - Philipp J Jost
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Andreas Seeber
- Department of Internal Medicine V: Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Arno Amann
- Department of Internal Medicine V: Hematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Konstantin Schlick
- IIIrd Medical Department of Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectious Disease, Salzburg Cancer Research Institute, Paracelsus Medical University, Salzburg, Austria
| | - Armin Gerger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Jakob M Riedl
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, Graz 8036, Austria
| |
Collapse
|
13
|
Dandachi N, Posch F, Graf R, Suppan C, Klocker EV, Müller HD, Lindenmann J, Terbuch A, Heitzer E, Balic M. Longitudinal tumor fraction trajectories predict risk of progression in metastatic HR + breast cancer patients undergoing CDK4/6 treatment. Mol Oncol 2021; 15:2390-2400. [PMID: 33264486 PMCID: PMC8410553 DOI: 10.1002/1878-0261.12870] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/29/2020] [Accepted: 11/30/2020] [Indexed: 12/26/2022] Open
Abstract
Despite improved clinical outcomes, intrinsic or acquired resistance to CDK4/6 inhibitor treatment has limited the success of this treatment in HR+ HER2- metastatic breast cancer patients. Biomarkers are urgently needed, and longitudinal biomarker measurements may harbor more dynamic predictive and prognostic information compared to single time point measurements. The aim of this study was to explore the longitudinal evolution of circulating tumor fractions within cell-free DNA assessed by an untargeted sequencing approach during CDK4/6 therapy and to quantify the potential association between longitudinal z-score measurements and clinical outcome by using joint models. Forty-nine HR+ HER2- metastatic breast cancer patients were enrolled, and z-score levels were measured at baseline and during 132 follow-up visits (median number of measurements per patient = 3, 25th -75th percentile: 3-5, range: 1-8). We observed higher baseline z-score levels (estimated difference 0.57, 95% CI: 0.147-0.983, P-value = 0.008) and a constant increase of z-score levels over follow-up time (overall P-value for difference in log z-score over time = 0.024) in patients who developed progressive disease. Importantly, the joint model revealed that elevated z-score trajectories were significantly associated with higher progression risk (HR of log z-score at any time of follow-up = 3.3, 95% CI, 1.44-7.55, P = 0.005). In contrast, single z-score measurement at CDK4/6 inhibitor treatment start did not predict risk of progression. In this prospective study, we demonstrate proof-of-concept that longitudinal z-score trajectories rather than single time point measurements may harbor important dynamic information on the development of disease progression in HR+ HER2- breast cancer patients undergoing CDK4/6 inhibitor treatment.
Collapse
Affiliation(s)
- Nadia Dandachi
- Division of OncologyDepartment of Internal MedicineMedical University of GrazAustria
- Research Unit Epigenetic and Genetic Cancer BiomarkersMedical University of GrazAustria
| | - Florian Posch
- Division of OncologyDepartment of Internal MedicineMedical University of GrazAustria
| | - Ricarda Graf
- Institute of Human GeneticsDiagnostic and Research Center for Molecular BioMedicineChristian Doppler Laboratory for Liquid Biopsies for early Detection of CancerMedical University of GrazAustria
| | - Christoph Suppan
- Division of OncologyDepartment of Internal MedicineMedical University of GrazAustria
| | - Eva Valentina Klocker
- Division of OncologyDepartment of Internal MedicineMedical University of GrazAustria
| | - Hannah Deborah Müller
- Division of OncologyDepartment of Internal MedicineMedical University of GrazAustria
| | - Jörg Lindenmann
- Divison of Thoracic and Hyperbaric SurgeryDepartment of SurgeryMedical University of GrazAustria
| | - Angelika Terbuch
- Division of OncologyDepartment of Internal MedicineMedical University of GrazAustria
| | - Ellen Heitzer
- Institute of Human GeneticsDiagnostic and Research Center for Molecular BioMedicineChristian Doppler Laboratory for Liquid Biopsies for early Detection of CancerMedical University of GrazAustria
| | - Marija Balic
- Division of OncologyDepartment of Internal MedicineMedical University of GrazAustria
- Research Unit Circulating Tumor Cells and Cancer Stem CellsMedical University of GrazAustria
| |
Collapse
|
14
|
Gemcitabine/nab-Paclitaxel versus FOLFIRINOX for palliative first-line treatment of advanced pancreatic cancer: A propensity score analysis. Eur J Cancer 2021; 151:3-13. [PMID: 33951545 DOI: 10.1016/j.ejca.2021.03.040] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/13/2021] [Accepted: 03/24/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Gemcitabine/nab-paclitaxel (GN) and FOLFIRINOX are standard first-line treatment options for advanced pancreatic ductal adenocarcinoma (aPDAC), but currently no prospective randomised head-to-head comparison between these treatments has yet been performed. METHODS We conducted a comparative propensity score (PS) analysis of overall (OS) and progression-free survival (PFS) in a tri-centre cohort of patients with aPDAC undergoing palliative first-line treatment with either GN or FOLFIRINOX. RESULTS In unadjusted analysis, OS and PFS were highly similar between patients treated with GN (n = 297) and FOLFIRINOX (n = 158). In detail, median, 1- and 2-year OS estimates were 10.1 months, 42% and 18% in the GN group, as compared to 11.2 months, 45% and 12% in the FOLFIRINOX group, respectively (log-rank p = 0.783). Accordingly, median (4.6 versus 4.8 months), 6-month (40% versus 43%) and 1-year (9% versus 9%) PFS estimates did not significantly differ (log-rank p = 0.717). However, patients treated with FOLFIRINOX were significantly younger, had fewer comorbidities, and a better Eastern Cooperative Oncology Group performance status. These imbalances were accounted for by weighting the data with the PS. In PS analysis of survival outcomes, OS and PFS remained comparable between the two treatment groups. In detail, PS-weighted median, 1- and 2-year OS estimates were 10.1 months, 42% and 18% in the GN group, as compared to 10.1 months, 40% and 13% in the FOLFIRINOX group (PS-weighted log-rank p = 0.449). PS-weighted PFS estimates again did not differ (PS-weighted log-rank p = 0.329). CONCLUSION This real-world comparative effectiveness study indicates that FOLFIRINOX and GN have similar effectiveness in the palliative first-line treatment of aPDAC.
Collapse
|
15
|
Chakrabarti S, Kamgar M, Mahipal A. Targeted Therapies in Advanced Biliary Tract Cancer: An Evolving Paradigm. Cancers (Basel) 2020; 12:2039. [PMID: 32722188 PMCID: PMC7465131 DOI: 10.3390/cancers12082039] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
Biliary tract cancers (BTCs) are a heterogeneous group of adenocarcinomas that originate from the epithelial lining of the biliary tree. BTCs are characterized by presentation with advanced disease precluding curative surgery, rising global incidence, and a poor prognosis. Chemotherapy is the mainstay of the current treatment, which results in a median overall survival of less than one year, underscoring the need for novel therapeutic agents and strategies. Next-generation sequencing-based molecular profiling has shed light on the underpinnings of the complex pathophysiology of BTC and has uncovered numerous actionable targets, leading to the discovery of new therapies tailored to the molecular targets. Therapies targeting fibroblast growth factor receptor (FGFR) fusion, isocitrate dehydrogenase (IDH) mutations, the human epidermal growth factor receptor (HER) family, DNA damage repair (DDR) pathways, and BRAF mutations have produced early encouraging results in selected patients. Current clinical trials evaluating targeted therapies, as monotherapies and in combination with other agents, are paving the way for novel treatment options. Genomic profiling of cell-free circulating tumor DNA that can assist in the identification of an actionable target is another exciting area of development. In this review, we provide a contemporaneous appraisal of the evolving targeted therapies and the ongoing clinical trials that will likely transform the therapeutic paradigm of BTC.
Collapse
Affiliation(s)
- Sakti Chakrabarti
- Department of Hematology-Oncology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; (S.C.); (M.K.)
| | - Mandana Kamgar
- Department of Hematology-Oncology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA; (S.C.); (M.K.)
| | - Amit Mahipal
- Department of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
16
|
Abou-Alfa GK, Macarulla T, Javle MM, Kelley RK, Lubner SJ, Adeva J, Cleary JM, Catenacci DV, Borad MJ, Bridgewater J, Harris WP, Murphy AG, Oh DY, Whisenant J, Lowery MA, Goyal L, Shroff RT, El-Khoueiry AB, Fan B, Wu B, Chamberlain CX, Jiang L, Gliser C, Pandya SS, Valle JW, Zhu AX. Ivosidenib in IDH1-mutant, chemotherapy-refractory cholangiocarcinoma (ClarIDHy): a multicentre, randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol 2020; 21:796-807. [PMID: 32416072 PMCID: PMC7523268 DOI: 10.1016/s1470-2045(20)30157-1] [Citation(s) in RCA: 697] [Impact Index Per Article: 139.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Isocitrate dehydrogenase 1 (IDH1) mutations occur in approximately 13% of patients with intrahepatic cholangiocarcinoma, a relatively uncommon cancer with a poor clinical outcome. The aim of this international phase 3 study was to assess the efficacy and safety of ivosidenib (AG-120)-a small-molecule targeted inhibitor of mutated IDH1-in patients with previously treated IDH1-mutant cholangiocarcinoma. METHODS This multicentre, randomised, double-blind, placebo-controlled, phase 3 study included patients from 49 hospitals in six countries aged at least 18 years with histologically confirmed, advanced, IDH1-mutant cholangiocarcinoma who had progressed on previous therapy, and had up to two previous treatment regimens for advanced disease, an Eastern Cooperative Oncology Group performance status score of 0 or 1, and a measurable lesion as defined by Response Evaluation Criteria in Solid Tumors version 1.1. Patients were randomly assigned (2:1) with a block size of 6 and stratified by number of previous systemic treatment regimens for advanced disease to oral ivosidenib 500 mg or matched placebo once daily in continuous 28-day cycles, by means of an interactive web-based response system. Placebo to ivosidenib crossover was permitted on radiological progression per investigator assessment. The primary endpoint was progression-free survival by independent central review. The intention-to-treat population was used for the primary efficacy analyses. Safety was assessed in all patients who had received at least one dose of ivosidenib or placebo. Enrolment is complete; this study is registered with ClinicalTrials.gov, NCT02989857. FINDINGS Between Feb 20, 2017, and Jan 31, 2019, 230 patients were assessed for eligibility, and as of the Jan 31, 2019 data cutoff date, 185 patients were randomly assigned to ivosidenib (n=124) or placebo (n=61). Median follow-up for progression-free survival was 6·9 months (IQR 2·8-10·9). Progression-free survival was significantly improved with ivosidenib compared with placebo (median 2·7 months [95% CI 1·6-4·2] vs 1·4 months [1·4-1·6]; hazard ratio 0·37; 95% CI 0·25-0·54; one-sided p<0·0001). The most common grade 3 or worse adverse event in both treatment groups was ascites (four [7%] of 59 patients receiving placebo and nine [7%] of 121 patients receiving ivosidenib). Serious adverse events were reported in 36 (30%) of 121 patients receiving ivosidenib and 13 (22%) of 59 patients receiving placebo. There were no treatment-related deaths. INTERPRETATION Progression-free survival was significantly improved with ivosidenib compared with placebo, and ivosidenib was well tolerated. This study shows the clinical benefit of targeting IDH1 mutations in advanced, IDH1-mutant cholangiocarcinoma. FUNDING Agios Pharmaceuticals.
Collapse
Affiliation(s)
- Ghassan K Abou-Alfa
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Medical College at Cornell University, New York, NY, USA
| | - Teresa Macarulla
- Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Milind M Javle
- Department of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Robin K Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Sam J Lubner
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Jorge Adeva
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - James M Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Daniel V Catenacci
- Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Mitesh J Borad
- Department of Hematology-Oncology, Mayo Clinic Cancer Center, Phoenix, AZ, USA
| | - John Bridgewater
- Department of Medical Oncology, UCL Cancer Institute, London, UK
| | - William P Harris
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Adrian G Murphy
- Department of Oncology-Gastrointestinal Cancer, Johns Hopkins University, Baltimore, MD, USA
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jonathan Whisenant
- Medical Oncology and Hematology, Utah Cancer Specialists, Salt Lake City, UT, USA
| | - Maeve A Lowery
- Trinity St James Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Lipika Goyal
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Rachna T Shroff
- Department of Medicine, University of Arizona Cancer Center, Tucson, AZ, USA
| | - Anthony B El-Khoueiry
- Department of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Bin Fan
- Agios Pharmaceuticals, Cambridge, MA, USA
| | - Bin Wu
- Agios Pharmaceuticals, Cambridge, MA, USA
| | | | | | | | | | - Juan W Valle
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Andrew X Zhu
- Department of Medicine, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA; Jiahui International Cancer Center, Jiahui Health, Shanghai, China.
| |
Collapse
|
17
|
Chakrabarti S, Zemla TJ, Ahn DH, Ou F, Fruth B, Borad MJ, Hartgers ML, Wessling J, Walkes RL, Alberts SR, McWilliams RR, Liu MC, Durgin LM, Bekaii‐Saab TS, Mahipal A. Phase II Trial of Trifluridine/Tipiracil in Patients with Advanced, Refractory Biliary Tract Carcinoma. Oncologist 2020; 25:380-e763. [PMID: 31826977 PMCID: PMC7216452 DOI: 10.1634/theoncologist.2019-0874] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/04/2019] [Indexed: 01/04/2023] Open
Abstract
LESSONS LEARNED Trifluridine/tipiracil (FTD/TPI) shows promising antitumor activity in heavily pretreated patients with advanced biliary tract carcinoma, including patients with 5-fluorouracil refractory tumors. FTD/TPI has an acceptable safety profile and should be studied further in patients with advanced biliary tract carcinoma after progression on standard first-line therapy. BACKGROUND Patients with advanced biliary tract carcinoma (BTC) refractory to first-line therapy lack an established second-line option. Trifluridine/tipiracil (FTD/TPI) has activity in both fluoropyrimidine-sensitive and -resistant tumors, which led us to conduct a single arm phase II trial to evaluate the safety and efficacy of FTD/TPI for patients previously treated for advanced BTC. METHODS Patients with advanced BTC previously treated with at least one line of chemotherapy were enrolled and treated with FTD/TPI until disease progression or unacceptable toxicity. The primary endpoint target was to have at least 6 patients who were progression free and alive at 16 weeks among 25 evaluable patients. Secondary endpoints included overall survival (OS), overall response rate (ORR), progression-free survival (PFS), and toxicity. RESULTS Of 27 evaluable patients, 59.3% received at least three prior lines of therapy, and 81.5% had previous exposure to fluoropyrimidine. Eight (32%, 95% confidence interval [CI], 14.9%-53.5%) patients were progression free at 16 weeks in the primary analysis population (n = 25), which met the predefined efficacy criteria. Median PFS and OS were 3.8 (95% CI, 2-5.8 months) and 6.1 (95% CI, 4.4-11.4 months) months, respectively. No objective responses were seen. There were no unexpected safety signals noted. CONCLUSION FTD/TPI demonstrated promising antitumor activity, with acceptable toxicity, in heavily pretreated patients with advanced BTC.
Collapse
Affiliation(s)
| | - Tyler J. Zemla
- Department of Biostatistics, Mayo ClinicRochesterMinnesotaUSA
| | | | - Fang‐Shu Ou
- Department of Biostatistics, Mayo ClinicRochesterMinnesotaUSA
| | - Briant Fruth
- Department of Biostatistics, Mayo ClinicRochesterMinnesotaUSA
| | | | | | | | | | | | | | | | | | | | - Amit Mahipal
- Medical Oncology, Mayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
18
|
Bekric D, Neureiter D, Ritter M, Jakab M, Gaisberger M, Pichler M, Kiesslich T, Mayr C. Long Non-Coding RNAs in Biliary Tract Cancer-An Up-to-Date Review. J Clin Med 2020; 9:jcm9041200. [PMID: 32331331 PMCID: PMC7231154 DOI: 10.3390/jcm9041200] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/31/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
The term long non-coding RNA (lncRNA) describes non protein-coding transcripts with a length greater than 200 base pairs. The ongoing discovery, characterization and functional categorization of lncRNAs has led to a better understanding of the involvement of lncRNAs in diverse biological and pathological processes including cancer. Aberrant expression of specific lncRNA species was demonstrated in various cancer types and associated with unfavorable clinical characteristics. Recent studies suggest that lncRNAs are also involved in the development and progression of biliary tract cancer, a rare disease with high mortality and limited therapeutic options. In this review, we summarize current findings regarding the manifold roles of lncRNAs in biliary tract cancer and give an overview of the clinical and molecular consequences of aberrant lncRNA expression as well as of underlying regulatory functions of selected lncRNA species in the context of biliary tract cancer.
Collapse
Affiliation(s)
- Dino Bekric
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria;
- Cancer Cluster Salzburg, 5020 Salzburg, Austria
| | - Markus Ritter
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Paracelsus Medical University, 5020 Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Martin Jakab
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
| | - Martin Gaisberger
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Paracelsus Medical University, 5020 Salzburg, Austria
- Gastein Research Institute, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Martin Pichler
- Research Unit of Non-Coding RNAs and Genome Editing, Division of Clinical Oncology, Department of Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria;
| | - Tobias Kiesslich
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria
| | - Christian Mayr
- Institute of Physiology and Pathophysiology, Paracelsus Medical University, 5020 Salzburg, Austria; (D.B.); (M.R.); (M.J.); (M.G.); (T.K.)
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK), 5020 Salzburg, Austria
- Correspondence:
| |
Collapse
|
19
|
Filippi R, Lombardi P, Quarà V, Fenocchio E, Aimar G, Milanesio M, Leone F, Aglietta M. Pharmacotherapeutic options for biliary tract cancer: current standard of care and new perspectives. Expert Opin Pharmacother 2019; 20:2121-2137. [PMID: 31550186 DOI: 10.1080/14656566.2019.1667335] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Biliary tract cancer (BTC), which comprises gallbladder cancer, ampullary cancer, and cholangiocarcinoma, is a rare and heterogeneous entity, with limited approved therapeutic options. However, interest in this disease has grown exponentially in recent years, as a mounting body of evidence has shed light on the complex molecular and microenvironmental background of BTC, and clinical investigations have explored a variety of new agents and combinations, with promising results.Areas covered: This review describes the standard of care in advanced BTC and summarizes the most recent evidence available on the pharmacological treatment of resected and advanced disease, focusing on chemotherapy, targeted therapy, and immunotherapy.Expert opinion: The therapeutic armamentarium of BTC has made radical progress after almost a decade of very few positive results. Phase-III evidence now supports the use of adjuvant capecitabine after resection of localized disease, while investigations into improved regimens in the advanced setting are underway, exploring alternative options to the standard gemcitabine-cisplatin doublet. The first positive phase-III trial supports the use of the mFOLFOX6 regimen as a second-line chemotherapy. Targeted therapy against specific genomic alterations can combine with chemotherapy in specific subsets of patients. Despite recent advancements, conducting clinical trials for BTC is still a real challenge.
Collapse
Affiliation(s)
- Roberto Filippi
- Department of Oncology, University of Turin, Candiolo, Italy.,Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Pasquale Lombardi
- Department of Oncology, University of Turin, Candiolo, Italy.,Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Virginia Quarà
- Department of Oncology, University of Turin, Candiolo, Italy.,Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Elisabetta Fenocchio
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Giacomo Aimar
- Department of Oncology, University of Turin, Candiolo, Italy.,Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Michela Milanesio
- Department of Oncology, University of Turin, Candiolo, Italy.,Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Francesco Leone
- Medical Oncology, Ospedale degli Infermi, Azienda Sanitaria Locale di Biella, Biella, Italy
| | - Massimo Aglietta
- Department of Oncology, University of Turin, Candiolo, Italy.,Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| |
Collapse
|