1
|
Monge C, Pehrsson EC, Xie C, Duffy AG, Mabry D, Wood BJ, Kleiner DE, Steinberg SM, Figg WD, Redd B, Budhu A, Wang S, Tandon M, Ma L, Wei Wang X, Greten TF. A Phase II Study of Pembrolizumab in Combination with Capecitabine and Oxaliplatin with Molecular Profiling in Patients with Advanced Biliary Tract Carcinoma. Oncologist 2022; 27:e273-e285. [PMID: 35274717 PMCID: PMC8914487 DOI: 10.1093/oncolo/oyab073] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 12/07/2021] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND We conducted a phase II study of the combination of pembrolizumab with capecitabine and oxaliplatin (CAPOX) in patients with advanced biliary tract carcinoma (BTC) to assess response rate and clinical efficacy. Exploratory objectives included correlative studies of immune marker expression, tumor evolution, and immune infiltration in response to treatment. PATIENTS AND METHODS Adult patients with histologically confirmed BTC were enrolled and received oxaliplatin and pembrolizumab on day 1 of cycles 1-6. Capecitabine was administered orally twice daily as intermittent treatment, with the first dose on day 1 and the last dose on day 14 of cycles 1-6. Starting on cycle 7, pembrolizumab monotherapy was continued until disease progression. The primary endpoint was progression-free survival (PFS). Secondary endpoints were safety, tolerability, feasibility, and response rate. Immunohistochemistry (IHC) for PD-L1 and immune infiltrates was analyzed in paired tumor biopsies, as well as bulk transcriptome and exome profiling for five patients and single-cell RNA sequencing for one partial responder. RESULTS Eleven patients enrolled, three of whom had received no prior systemic therapy. Treatment was well tolerated, and the most common treatment-related grade 3 or 4 adverse events were lymphocytopenia, anemia, and decreased platelet count. Three patients (27.3%) achieved a partial response, and six (54%) had stable disease. The disease control rate was 81.8%. The median PFS was 4.1 months with a 6-month PFS rate of 45.5%. Molecular profiling suggests qualitative differences in immune infiltration and clonal evolution based on response. CONCLUSION Capecitabine and oxaliplatin in combination with pembrolizumab is tolerable and a potentially effective treatment for refractory advanced BTC. This study highlights a design framework for the precise characterization of individual BTC tumors. TRIAL REGISTRATION This study was registered in ClinicalTrials.gov (NCT03111732).
Collapse
Affiliation(s)
- Cecilia Monge
- Gastrointestinal Malignancies Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Erica C Pehrsson
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- CCR Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Changqing Xie
- Gastrointestinal Malignancies Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Austin G Duffy
- Gastrointestinal Malignancies Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Donna Mabry
- Gastrointestinal Malignancies Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bradford J Wood
- Center for Interventional Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - David E Kleiner
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - William D Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bernadette Redd
- Radiology and Imaging Sciences, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anuradha Budhu
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sophie Wang
- Gastrointestinal Malignancies Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mayank Tandon
- CCR Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Lichun Ma
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xin Wei Wang
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tim F Greten
- Gastrointestinal Malignancies Section, Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Massa A, Varamo C, Vita F, Tavolari S, Peraldo-Neia C, Brandi G, Rizzo A, Cavalloni G, Aglietta M. Evolution of the Experimental Models of Cholangiocarcinoma. Cancers (Basel) 2020; 12:cancers12082308. [PMID: 32824407 PMCID: PMC7463907 DOI: 10.3390/cancers12082308] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/14/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a rare, aggressive disease with poor overall survival. In advanced cases, surgery is often not possible or fails; in addition, there is a lack of effective and specific therapies. Multidisciplinary approaches and advanced technologies have improved the knowledge of CCA molecular pathogenesis, highlighting its extreme heterogeneity and high frequency of genetic and molecular aberrations. Effective preclinical models, therefore, should be based on a comparable level of complexity. In the past years, there has been a consistent increase in the number of available CCA models. The exploitation of even more complex CCA models is rising. Examples are the use of CRISPR/Cas9 or stabilized organoids for in vitro studies, as well as patient-derived xenografts or transgenic mouse models for in vivo applications. Here, we examine the available preclinical CCA models exploited to investigate: (i) carcinogenesis processes from initiation to progression; and (ii) tools for personalized therapy and innovative therapeutic approaches, including chemotherapy and immune/targeted therapies. For each model, we describe the potential applications, highlighting both its advantages and limits.
Collapse
Affiliation(s)
- Annamaria Massa
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Torino, Italy; (A.M.); (G.C.)
| | - Chiara Varamo
- Department of Oncology, University of Turin, 10126 Torino, Italy; (C.V.); (F.V.)
- Department of Oncology, Laboratory of Tumor Inflammation and Angiogenesis, B3000 KU Leuven, Belgium
| | - Francesca Vita
- Department of Oncology, University of Turin, 10126 Torino, Italy; (C.V.); (F.V.)
| | - Simona Tavolari
- Center for Applied Biomedical Research, S. Orsola-Malpighi University Hospital, 40138 Bologna, Italy;
| | | | - Giovanni Brandi
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, 40138 Bologna, Italy; (G.B.); (A.R.)
| | - Alessandro Rizzo
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, 40138 Bologna, Italy; (G.B.); (A.R.)
| | - Giuliana Cavalloni
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Torino, Italy; (A.M.); (G.C.)
| | - Massimo Aglietta
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Torino, Italy; (A.M.); (G.C.)
- Department of Oncology, University of Turin, 10126 Torino, Italy; (C.V.); (F.V.)
- Correspondence:
| |
Collapse
|
3
|
Jansen H, Pape UF, Utku N. A review of systemic therapy in biliary tract carcinoma. J Gastrointest Oncol 2020; 11:770-789. [PMID: 32953160 PMCID: PMC7475338 DOI: 10.21037/jgo-20-203] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022] Open
Abstract
Biliary tract carcinoma (BTC) has a poor prognosis and is increasing in incidence. Although surgery, chemotherapy and other treatment modalities have improved, surgery remains the only potential curative treatment and is appropriate for only those few patients who present with localized, resectable disease. However, for the majority of patients, unresectable disease is evident at diagnosis and about 95% of patients die within 10 years, despite the majority receiving chemotherapy. Long-term survival is significantly greater for patients with resected BTC compared to those with unresectable disease. In unresected disease, life expectancy is limited, with first-line gemcitabine/cisplatin (GEM/CIS) accepted as standard of care. Currently no standard second-line regimen which provides significant improvement of clinical outcomes exists for those who present with refractory disease or who relapse after first-line treatment. Of particular importance is establishing the impact of best supportive care (BSC) as a benchmark for survival outcomes to which the impact of treatment modalities can be compared. Survival outcome often differs significantly for patients with different prognostic factor profiles even when receiving the same therapy so that it can be difficult to predict which patient subgroup might benefit most from which therapy. Therefore, the influence of prognostic factors on survival under different therapies as well as under BSC needs to be further assessed in order to arrive at truly evidence-based, best therapeutic decisions for individual patients. Encouraging new research into the genomic landscape of BTC may help to further subdivide the BTC population into molecular-genetic clusters likely to be sensitive to different targeted therapy approaches leading to further improvements in survival. Consequently, an unmet need exists not only to develop new and more effective therapies for this devastating disease, but also to integrate original research findings into a more complex, dynamic, individualized therapeutic decision model to aid clinicians in making evidence-based, best therapeutic decisions for individual patients.
Collapse
Affiliation(s)
- Holger Jansen
- Campus Virchow & Mitte Charité, Institute f. Med. Immunologie, Berlin, Germany
| | - Ulrich-Frank Pape
- Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Germany
- Internal Medicine and Gastroenterology, Asklepios Klinik St. Georg, Asklepios Tumor Zentrum Hamburg, Germany
| | - Nalân Utku
- Campus Virchow & Mitte Charité, Institute f. Med. Immunologie, Berlin, Germany
- CellAct Pharma GmbH, Dortmund, Germany
| |
Collapse
|
4
|
Xie C, Duffy AG, Mabry-Hrones D, Wood B, Levy E, Krishnasamy V, Khan J, Wei JS, Agdashian D, Tyagi M, Gangalapudi V, Fioravanti S, Walker M, Anderson V, Venzon D, Figg WD, Sandhu M, Kleiner DE, Morelli MP, Floudas CS, Brar G, Steinberg SM, Korangy F, Greten TF. Tremelimumab in Combination With Microwave Ablation in Patients With Refractory Biliary Tract Cancer. Hepatology 2019; 69:2048-2060. [PMID: 30578687 PMCID: PMC6461476 DOI: 10.1002/hep.30482] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 12/11/2018] [Indexed: 12/15/2022]
Abstract
Treatment options for patients with advanced biliary tract cancer are limited. Dysregulation of the immune system plays an important role in the pathogenesis of biliary tract cancer (BTC). This study aimed to investigate whether tremelimumab, an anti-CTLA4 (cytotoxic T-lymphocyte-associated protein 4) inhibitor, could be combined safely with microwave ablation to enhance the effect of anti-CTLA4 treatment in patients with advanced BTC. Patients were enrolled to receive monthly tremelimumab (10 mg/kg, intravenously) for six doses, followed by infusions every 3 months until off-treatment criteria were met. Thirty-six days after the first tremelimumab dose, patients underwent subtotal microwave ablation. Interval imaging studies were performed every 8 weeks. Adverse events (AEs) were noted and managed. Tumor and peripheral blood samples were collected to perform immune monitoring and whole-exome sequencing (WES). Twenty patients with refractory BTC were enrolled (median age, 56.5 years). No dose-limiting toxicities were encountered. The common treatment-related AEs included lymphopenia, diarrhea, and elevated transaminases. Among 16 patients evaluable for efficacy analysis, 2 (12.5%) patients achieved a confirmed partial response (lasting for 8.0 and 18.1 months, respectively) and 5 patients (31.3%) achieved stable disease. Median progression free survival (PFS) and overall survival (OS) were 3.4 months (95% confidence interval [CI], 2.5-5.2) and 6.0 months (95% CI, 3.8-8.8), respectively. Peripheral blood immune cell subset profiling showed increased circulating activated human leukocyte antigen, DR isotype ([HLA-DR] positive) CD8+ T cells. T-cell receptor (TCR)β screening showed tremelimumab expanded TCR repertoire, but not reaching statistical significance (P = 0.057). Conclusion: Tremelimumab in combination with tumor ablation is a potential treatment strategy for patients with advanced BTC. Increased circulating activated CD8+ T cells and TCR repertoire expansion induced by tremelimumab may contribute to treatment benefit.
Collapse
Affiliation(s)
- Changqing Xie
- Hematology/Oncology Fellowship Program, National Heart, Lung, and Blood Institute/National Cancer Institute, National Institutes of Health
| | - Austin G. Duffy
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Donna Mabry-Hrones
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Bradford Wood
- Radiology and Imaging Sciences, Center for Cancer Research, National Institutes of Health
| | - Elliot Levy
- Radiology and Imaging Sciences, Center for Cancer Research, National Institutes of Health
| | - Venkatesh Krishnasamy
- Radiology and Imaging Sciences, Center for Cancer Research, National Institutes of Health
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Jun S. Wei
- Genetics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - David Agdashian
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Manoj Tyagi
- Genetics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | | | - Suzanne Fioravanti
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Melissa Walker
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Victoria Anderson
- Radiology and Imaging Sciences, Center for Cancer Research, National Institutes of Health
| | - David Venzon
- Biostatistics and Data Management Section, NCI, NIH, Bethesda, Maryland
| | - William D. Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Milan Sandhu
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health
| | - David E. Kleiner
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Maria Pia Morelli
- Hematology/Oncology Fellowship Program, National Heart, Lung, and Blood Institute/National Cancer Institute, National Institutes of Health
| | - Charalampos S Floudas
- Hematology/Oncology Fellowship Program, National Heart, Lung, and Blood Institute/National Cancer Institute, National Institutes of Health
| | - Gagandeep Brar
- Hematology/Oncology Fellowship Program, National Heart, Lung, and Blood Institute/National Cancer Institute, National Institutes of Health
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Institutes of Health
| | - Firouzeh Korangy
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Tim F. Greten
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health,NCI CCR Liver Cancer Program
| |
Collapse
|
5
|
Peraldo-Neia C, Ostano P, Cavalloni G, Pignochino Y, Sangiolo D, De Cecco L, Marchesi E, Ribero D, Scarpa A, De Rose AM, Giuliani A, Calise F, Raggi C, Invernizzi P, Aglietta M, Chiorino G, Leone F. Transcriptomic analysis and mutational status of IDH1 in paired primary-recurrent intrahepatic cholangiocarcinoma. BMC Genomics 2018; 19:440. [PMID: 29871612 PMCID: PMC5989353 DOI: 10.1186/s12864-018-4829-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 05/25/2018] [Indexed: 12/14/2022] Open
Abstract
Background Effective target therapies for intrahepatic cholangiocarcinoma (ICC) have not been identified so far. One of the reasons may be the genetic evolution from primary (PR) to recurrent (REC) tumors. We aim to identify peculiar characteristics and to select potential targets specific for recurrent tumors. Eighteen ICC paired PR and REC tumors were collected from 5 Italian Centers. Eleven pairs were analyzed for gene expression profiling and 16 for mutational status of IDH1. For one pair, deep mutational analysis by Next Generation Sequencing was also carried out. An independent cohort of patients was used for validation. Results Two class-paired comparison yielded 315 differentially expressed genes between REC and PR tumors. Up-regulated genes in RECs are involved in RNA/DNA processing, cell cycle, epithelial to mesenchymal transition (EMT), resistance to apoptosis, and cytoskeleton remodeling. Down-regulated genes participate to epithelial cell differentiation, proteolysis, apoptotic, immune response, and inflammatory processes. A 24 gene signature is able to discriminate RECs from PRs in an independent cohort; FANCG is statistically associated with survival in the chol-TCGA dataset. IDH1 was mutated in the RECs of five patients; 4 of them displayed the mutation only in RECs. Deep sequencing performed in one patient confirmed the IDH1 mutation in REC. Conclusions RECs are enriched for genes involved in EMT, resistance to apoptosis, and cytoskeleton remodeling. Key players of these pathways might be considered druggable targets in RECs. IDH1 is mutated in 30% of RECs, becoming both a marker of progression and a target for therapy. Electronic supplementary material The online version of this article (10.1186/s12864-018-4829-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- C Peraldo-Neia
- Medical Oncology Division, Candiolo Cancer Institute - FPO, IRCCS, Str. Prov. 142, km 3.95, 10060, Candiolo, Turin, Italy. .,Cancer Genomics Lab, Fondazione Edo ed Elvo Tempia Valenta, Biella, Italy.
| | - P Ostano
- Cancer Genomics Lab, Fondazione Edo ed Elvo Tempia Valenta, Biella, Italy
| | - G Cavalloni
- Medical Oncology Division, Candiolo Cancer Institute - FPO, IRCCS, Str. Prov. 142, km 3.95, 10060, Candiolo, Turin, Italy
| | - Y Pignochino
- Department of Oncology, University of Turin, Torino, Italy
| | - D Sangiolo
- Medical Oncology Division, Candiolo Cancer Institute - FPO, IRCCS, Str. Prov. 142, km 3.95, 10060, Candiolo, Turin, Italy.,Department of Oncology, University of Turin, Torino, Italy
| | - L De Cecco
- Functional Genomics and Bioinformatics, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - E Marchesi
- Functional Genomics and Bioinformatics, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - D Ribero
- Division of Hepatobilio-Pancreatic and Colorectal Surgery, Candiolo Cancer Institute - FPO, IRCCS, Str. Prov. 142, km 3.95, Candiolo, Italy
| | - A Scarpa
- ARC-Net Research Centre and Department of Diagnostics and Public Health - Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - A M De Rose
- Hepatobiliary Surgery Unit, Gemelli Hospital, Catholic University of the Sacred Heart, Rome, Italy
| | - A Giuliani
- Department of Health's Sciences and Medicine "V. Tiberio", University of Molise, Campobasso, Italy
| | - F Calise
- Hepatobiliary and Liver Transplant Unit, Cardarelli Hospital, Naples, Italy
| | - C Raggi
- Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano, Italy.,Department of Experimental and Clinical Medicine, University of Firenze, Florence, Italy
| | - P Invernizzi
- Center for Autoimmune Liver Diseases, Humanitas Clinical and Research Center, Rozzano, Italy.,UOC di Gastroenterologia, Azienda Ospedaliera San Gerardo, Monza, Italy
| | - M Aglietta
- Medical Oncology Division, Candiolo Cancer Institute - FPO, IRCCS, Str. Prov. 142, km 3.95, 10060, Candiolo, Turin, Italy.,Department of Oncology, University of Turin, Torino, Italy
| | - G Chiorino
- Cancer Genomics Lab, Fondazione Edo ed Elvo Tempia Valenta, Biella, Italy
| | - F Leone
- Medical Oncology Division, Candiolo Cancer Institute - FPO, IRCCS, Str. Prov. 142, km 3.95, 10060, Candiolo, Turin, Italy. .,Department of Oncology, University of Turin, Torino, Italy.
| |
Collapse
|
6
|
Comparison of FOLFIRINOX Chemotherapy with Other Regimens in Patients with Biliary Tract Cancers: a Retrospective Study. J Gastrointest Cancer 2018; 48:170-175. [PMID: 27714651 DOI: 10.1007/s12029-016-9880-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE The aim of this retrospective study was to compare the different treatment options of patients with advanced biliary tract carcinoma (BTC) who were treated with platinum-gemcitabine (CG) or platinum-5-fluorouracil (CF) or 5-Fluorouracil-oxaliplatin-irinotecan (FOLFIRINOX) chemotherapy. METHODS We included the patients with advanced BTC who were registered at the Department of Oncology in Gaziantep University between January 2008 and January 2016. The following data were analyzed: disease control rate (DCR), progression free survival (PFS) of first and second-line of chemotherapy, and overall survival (OS). Kaplan-Meier method and Log-rank test was used to compare two survival curves, and hazard regression model was used to evaluate risk factors for PFS. RESULT Ninety-two patients were recruited. 53 (57.6 %), 27 (29.3 %), and 12 (13 %) patients received CG, CF, and FOLFIRINOX regimen as first-line chemotherapy, respectively. Median PFS and DCR of CG group were 22 weeks and 56.6 %, and these were 12 weeks and 44.4 % for CF group, and 9 weeks and 41.7 % for FOLFIRINOX group. Median OS of CG, CF, and FOLFIRINOX groups was 28, 21,and 23.5 weeks, respectively (p = 0.497). Second-line PFS of fluoropyrimidine-based chemotherapy group and gemcitabine-based chemotherapy group was 12 vs. 14 weeks (p = 0.988). Second-line PFS of FOLFIRINOX was 20 weeks, whereas it was 14 weeks for other fuoropyrimidine-based chemotherapies (p = 0.190). CONCLUSIONS This was the first study evaluating the FOLFIRINOX regimen in BTC. Cisplatin-gemcitabine therapy still provides better survival in BCT. However, FOLFIRINOX can be an option in the second-line treatment of BTC patients who are eligible for chemotherapy.
Collapse
|
7
|
Predictive Value of Early Skin Rash in Cetuximab-Based Therapy of Advanced Biliary Tract Cancer. Pathol Oncol Res 2017; 24:237-244. [DOI: 10.1007/s12253-017-0238-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/25/2017] [Indexed: 02/06/2023]
|
8
|
Yang H, Zhou J, Mi J, Ma K, Fan Y, Ning J, Wang C, Wei X, Zhao H, Li E. HOXD10 acts as a tumor-suppressive factor via inhibition of the RHOC/AKT/MAPK pathway in human cholangiocellular carcinoma. Oncol Rep 2015; 34:1681-91. [PMID: 26260613 PMCID: PMC4564083 DOI: 10.3892/or.2015.4194] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 06/25/2015] [Indexed: 12/20/2022] Open
Abstract
HOXD10, a key regulator of cell-differentiated phenotype maintainence, has been demonstrated to be involved in the tumorigenesis of many human malignacies. However, the status of HOXD10 expression and its biological function in cholangiocellular carcinoma (CCC) remain to be clarified. In the present study, we investigated the clinical significance and biological functions of HOXD10 in CCC and found that the expression of HOXD10 and its downstream effector RHOC was significantly different in well-differentiated CCC tissues compared with poorly-differentiated lesions. We also observed a significant correlation between low HOXD10 and high RHOC expression levels and worse prognosis. The stable overexpression of HOXD10 by lentivirus vector significantly inhibited cell invasion partly by downregulating the expression of MMP2 and MMP9, and significantly increased early apoptosis in CCC cell lines and induced G1 phase cell cycle arrest, contributing to the inhibition of cell proliferation in vitro. Additionally, we demonstrated that the inactivation of the RHOC/AKT/MAPK pathway was involved in the tumor-suppressive functions of HOXD10 in CCC. These results suggested that HOXD10 may be a putative suppressor gene and can act as a prognostic marker and potentially a novel therapeutic target for CCC.
Collapse
Affiliation(s)
- Haixia Yang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710062, P.R. China
| | - Jiupeng Zhou
- Department of Medical Oncology, Xi'an Chest Hospital of Shaanxi, Xi'an, Shaanxi 710061, P.R. China
| | - Jianqiang Mi
- Department of Pathology, The First Affiliated Hospital of Henan Science and Technology University, Luoyang, Henan 471003, P.R. China
| | - Ke Ma
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710062, P.R. China
| | - Yangwei Fan
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710062, P.R. China
| | - Jing Ning
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710062, P.R. China
| | - Chuying Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710062, P.R. China
| | - Xin Wei
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710062, P.R. China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, Xi'an, Shaanxi 710038, P.R. China
| | - Enxiao Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710062, P.R. China
| |
Collapse
|