1
|
Erythroblastic oncogene B-2 status and intracranial metastatic disease in patients with gastrointestinal cancer: a systematic review. J Neurooncol 2022; 160:735-742. [PMID: 36372832 DOI: 10.1007/s11060-022-04195-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/07/2022] [Indexed: 11/15/2022]
Abstract
PURPOSE The incidence of intracranial metastatic disease (IMD) in patients with gastrointestinal (GI) cancers is rising. Expression of the erythroblastic oncogene B-2 (ERBB2) is associated with an in increased risk of IMD in patients with breast cancer. The implications of ERBB2 expression for IMD risk in patients with GI cancers is less clear. The objective of this systematic review was to determine the incidence of IMD and OS in patients with ERBB2+ gastrointestinal cancers. METHODS A literature search of MEDLINE, EMBASE, CENTRAL, and grey literature sources was conducted from date of database inception to July 2021. Included studies reported outcomes on patients with IMD secondary to ERBB2 GI cancers. RESULTS Fourteen cohort studies met inclusion criteria, of which thirteen were retrospective. Eleven studies reported on gastric, esophageal, or gastroesophageal junction cancers. Three studies directly compared incidence of IMD based on ERBB2 status and among these, ERBB2+ patients had a higher incidence of IMD. One study indicated that ERBB2+ patients had significantly longer OS from the times of primary cancer (P = .015) and IMD diagnosis (P = .01), compared with patients with ERBB2- disease. CONCLUSIONS In this systematic review, patients with ERBB2+ GI cancer were more likely to develop IMD. Future study is required on the prognostic and predictive value of ERBB2 status in patients with GI cancers.
Collapse
|
2
|
Guimond E, Tsai CJ, Hosni A, O'Kane G, Yang J, Barry A. Safety and Tolerability of Metastasis Directed Radiotherapy in the Era of Evolving Systemic, Immune and Targeted Therapies. Adv Radiat Oncol 2022; 7:101022. [PMID: 36177487 PMCID: PMC9513086 DOI: 10.1016/j.adro.2022.101022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/02/2022] [Indexed: 11/17/2022] Open
Abstract
AbstractPurpose Systemic, immune, and target therapies are growing in use in the management of metastatic cancers. The aim of this review was to describe up-to-date published data on the safety and tolerability of metastasis-directed hypofractionated radiation therapy (RT) when combined with newer systemic, immune, and targeted therapies and to provide suggested strategies to mitigate potential toxicities in the clinical setting. Methods and Materials A comprehensive search was performed for the time period between 1946 and August 2021 using predetermined keywords describing the use of noncentral nervous system palliative RT with commonly used targeted systemic therapies on PubMed and Medline databases. A total of 1022 articles were screened, and 130 met prespecified criteria to be included in this review. Results BRAF and MEK inhibitors are reported to be toxic when given concurrently with RT; suspension 3 days and 1 to 2 days, respectively, prior and post-RT is suggested. Cetuximab, erlotinib/gefitinib, and osimertinib were generally safe to use concomitantly with conventional radiation. But in a palliative/hypofractionated RT setting, suspending cetuximab during radiation week, erlotinib/gefitinib 1 to 2 days, and osimertinib ≥2 days pre- and post-RT is suggested. Vascular endothelial growth factor inhibitors such as bevacizumab reported substantial toxicities, and the suggestion is to suspend 4 weeks before and after radiation. Less data exist on sorafenib and sunitinib; 5 to 10 days suspension before and after RT should be considered. As a precaution, until further data are available, for cyclin-dependent kinase 4-6 inhibitors, consideration of suspending treatment 1 to 2 days before and after RT should be given. Ipilimumab should be suspended 2 days before and after RT, and insufficient data exist for other immunotherapy agents. Trastuzumab and pertuzumab are generally safe to use in combination with RT, but insufficient data exist for other HER2 target therapy. Conclusions Suggested approaches are described, using up-to-date literature, to aid clinicians in navigating the integration of newer targeted agents with hypofractionated palliative and/or ablative metastatic RT. Further prospective studies are required.
Collapse
Affiliation(s)
- Elizabeth Guimond
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada
- University of Toronto, Toronto, Ontario
- Corresponding author: Elizabeth Guimond, MD, FRCPC
| | - Chiaojung Jillian Tsai
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ali Hosni
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada
- University of Toronto, Toronto, Ontario
| | - Grainne O'Kane
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada
- University of Toronto, Toronto, Ontario
| | - Jonathan Yang
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Aisling Barry
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada
- University of Toronto, Toronto, Ontario
| |
Collapse
|
3
|
Safran HP, Winter K, Ilson DH, Wigle D, DiPetrillo T, Haddock MG, Hong TS, Leichman LP, Rajdev L, Resnick M, Kachnic LA, Seaward S, Mamon H, Diaz Pardo DA, Anderson CM, Shen X, Sharma AK, Katz AW, Salo J, Leonard KL, Moughan J, Crane CH. Trastuzumab with trimodality treatment for oesophageal adenocarcinoma with HER2 overexpression (NRG Oncology/RTOG 1010): a multicentre, randomised, phase 3 trial. Lancet Oncol 2022; 23:259-269. [PMID: 35038433 PMCID: PMC8903071 DOI: 10.1016/s1470-2045(21)00718-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/06/2021] [Accepted: 12/06/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Trastuzumab is a monoclonal antibody against HER2 (also known as ERBB2). The primary objective of the NRG Oncology/RTOG-1010 trial was to establish whether trastuzumab improves disease-free survival when combined with trimodality treatment (paclitaxel plus carboplatin and radiotherapy, followed by surgery) for patients with untreated HER2-overexpressing oesophageal adenocarcinoma. METHODS NRG Oncology/RTOG-1010 was an open label, randomised, phase 3 trial for which patients were accrued from 111 NRG-affiliated institutions in the USA. Eligible patients were adults (aged ≥18 years) with newly diagnosed pathologically confirmed oesophageal adenocarcinoma, American Joint Committee on Cancer 7th edition T1N1-2 or T2-3N0-2 stage disease, and a Zubrod performance status of 0-2. Patients were stratified by adenopathy (no vs yes [coeliac absent] vs yes [coeliac present ≤2 cm]) and randomly assigned (1:1) to receive weekly intravenous paclitaxel (50 mg/m2 intravenously over 1 h) and carboplatin (area under the curve 2, intravenously over 30-60 min) for 6 weeks with radiotherapy 50·4 Gy in 28 fractions (chemoradiotherapy) followed by surgery, with or without intravenous trastuzumab (4 mg/kg in week one, 2 mg/kg per week for 5 weeks during chemoradiotherapy, 6 mg/kg once presurgery, and 6 mg/kg every 3 weeks for 13 treatments starting 21-56 days after surgery). The primary endpoint, disease-free survival, was defined as the time from randomisation to death or first of locoregional disease persistence or recurrence, distant metastases, or second primary malignancy. Analyses were done by modified intention to treat. This study is registered with Clinicaltrials.gov, NCT01196390; it is now closed and in follow-up. FINDINGS 606 patients were entered for HER2 assessment from Dec 30, 2010 to Nov 10, 2015, and 203 eligible patients who were HER2-positive were enrolled and randomly assigned to chemoradiotherapy plus trastuzumab (n=102) or chemoradiotherapy alone (n=101). Median duration of follow-up was 2·8 years (IQR 1·4-5·7). Median disease-free survival was 19·6 months (95% CI 13·5-26·2) with chemoradiotherapy plus trastuzumab compared with 14·2 months (10·5-23·0) for chemoradiotherapy alone (hazard ratio 0·99 [95% CI 0·71-1·39], log-rank p=0·97). Grade 3 treatment-related adverse events occurred in 41 (43%) of 95 patients in the chemoradiotherapy plus trastuzumab group versus 52 (54%) of 96 in the chemoradiotherapy group and grade 4 events occurred in 20 (21%) versus 21 (22%). The most common grade 3 or worse treatment-related adverse events for both groups were haematological (53 [56%] of 95 patients in the chemoradiotherapy plus trastuzumab group vs 55 [57%] of 96 patients in the chemotherapy group) or gastrointestinal disorders (28 [29%] vs 20 [21 %]). 34 (36%) of 95 patients in the chemoradiotherapy plus trastuzumab group and 27 (28%) of 96 patients in the chemoradiotherapy only group had treatment-related serious adverse events. There were eight treatment-related deaths: five (5%) of 95 patients in the chemoradiotherapy plus trastuzumab group (bronchopleural fistula, oesophageal anastomotic leak, lung infection, sudden death, and death not otherwise specified), and three (3%) of 96 in the chemoradiotherapy group (two multiorgan failure and one sepsis). INTERPRETATION The addition of trastuzumab to neoadjuvant chemoradiotherapy for HER2-overexpressing oesophageal cancer was not effective. Trastuzumab did not lead to increased toxicities, suggesting that future studies combining it with or using other agents targeting HER2 in oesophageal cancer are warranted. FUNDING National Cancer Institute and Genentech.
Collapse
|
4
|
Stroes CI, van den Ende T, Derks S, van Laarhoven HWM. A systematic review of HER2 blockade for the curative treatment of gastroesophageal adenocarcinoma: Successes achieved and opportunities ahead. Cancer Treat Rev 2021; 99:102249. [PMID: 34171733 DOI: 10.1016/j.ctrv.2021.102249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Despite multimodality treatment for curatively-treated gastroesophageal adenocarcinoma (GEA), prognosis remains dismal. The benefit of adding trastuzumab to chemotherapy for advanced Human Epidermal Growth Factor 2 (HER2) positive GEA has been established in the ToGA trial. However, it remains unclear if HER2 inhibition might also be beneficial in the curative setting. Therefore, we conducted a systematic review to investigate the role of HER2 inhibitors for the curative treatment of GEA. METHODS A systematic literature search was performed in PubMed, EMBASE, CENTRAL, and clinicaltrials.gov to identify clinical trials investigating HER2 inhibition for the curative treatment of GEA. Study quality was assessed using the GRADE methodology. RESULTS From the 1825 studies retrieved, 17 were included (seven published articles; three published conference abstracts; seven ongoing studies). From the published studies, eight studies investigated single-agent HER2 inhibition. Four studies had a nonrandomized design, and two were randomized controlled trials. Two published studies were assessed as high-quality. The addition of single-agent HER2 inhibition to chemo(radio)therapy showed a pathological complete response rate (pCR) of 22.2% (range, 9.6-25%) and dual HER2 inhibition of 34.5% (34-35%). Two-year disease-free survival (DFS) was 51.0% (40-71%) with single-agent and 70.0% (70-70%) with dual HER2 therapy. DISCUSSION Dual-agent HER2 inhibition showed promising pCR rates and DFS. Given the limited additional toxicity of the addition of HER2 targeting agents and the potential benefit of dual-targeting, further investigation is required in a phase III randomized clinical trial. Next steps include combining checkpoint inhibitors and HER2 blockade given the suggested synergism, as well as investigating new anti-HER2 agents.
Collapse
Affiliation(s)
- Charlotte I Stroes
- Amsterdam UMC, University of Amsterdam, Department of Medical Oncology, Cancer Center Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| | - Tom van den Ende
- Amsterdam UMC, University of Amsterdam, Department of Medical Oncology, Cancer Center Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Sarah Derks
- Amsterdam UMC, VU University, Department of Medical Oncology, Cancer Center Amsterdam, De Boelelaan 1117, 1118, 1081HV Amsterdam, the Netherlands; Oncode Institute, Jaarbeursplein 6, 3521AL Utrecht, the Netherlands
| | - Hanneke W M van Laarhoven
- Amsterdam UMC, University of Amsterdam, Department of Medical Oncology, Cancer Center Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Cancer Center Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| |
Collapse
|
5
|
Faut-il moduler les contraintes de dose dans les organes à risque lors d’une irradiation en association avec un traitement anticancéreux systémique ? Cancer Radiother 2020; 24:594-601. [DOI: 10.1016/j.canrad.2020.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 05/27/2020] [Accepted: 05/30/2020] [Indexed: 11/23/2022]
|
6
|
Stroes CI, Schokker S, Creemers A, Molenaar RJ, Hulshof MC, van der Woude SO, Bennink RJ, Mathôt RA, Krishnadath KK, Punt CJ, Verhoeven RH, van Oijen MG, Creemers GJ, Nieuwenhuijzen GA, van der Sangen MJ, Beerepoot LV, Heisterkamp J, Los M, Slingerland M, Cats A, Hospers GA, Bijlsma MF, van Berge Henegouwen MI, Meijer SL, van Laarhoven HW. Phase II Feasibility and Biomarker Study of Neoadjuvant Trastuzumab and Pertuzumab With Chemoradiotherapy for Resectable Human Epidermal Growth Factor Receptor 2-Positive Esophageal Adenocarcinoma: TRAP Study. J Clin Oncol 2020; 38:462-471. [PMID: 31809243 PMCID: PMC7007286 DOI: 10.1200/jco.19.01814] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2019] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Approximately 15% to 43% of esophageal adenocarcinomas (EACs) are human epidermal growth factor receptor 2 (HER2) positive. Because dual-agent HER2 blockade demonstrated a survival benefit in breast cancer, we conducted a phase II feasibility study of trastuzumab and pertuzumab added to neoadjuvant chemoradiotherapy (nCRT) in patients with EAC. PATIENTS AND METHODS Patients with resectable HER2-positive EAC received standard nCRT with carboplatin and paclitaxel and 41.4 Gy of radiotherapy, with 4 mg/kg of trastuzumab on day 1, 2 mg/kg per week during weeks 2 to 6, and 6 mg/kg per week during weeks 7, 10, and 13 and 840 mg of pertuzumab every 3 weeks. The primary end point was feasibility, defined as ≥ 80% completion of treatment with both trastuzumab and pertuzumab. An exploratory comparison of survival with a propensity score-matched cohort receiving standard nCRT was performed, as were exploratory pharmacokinetic and biomarker analyses. RESULTS Of the 40 enrolled patients (78% men; median age, 63 years), 33 (83%) completed treatment with trastuzumab and pertuzumab. No unexpected safety events were observed. R0 resection was achieved in all patients undergoing surgery, with pathologic complete response in 13 patients (34%). Three-year progression-free and overall survival (OS) were 57% and 71%, respectively (median follow-up, 32.1 months). Compared with the propensity score-matched cohort, a significantly longer OS was observed with HER2 blockade (hazard ratio, 0.58; 95% CI, 0.34 to 0.97). Results of pharmacokinetic analysis and activity on [18F]fluorodeoxyglucose positron emission tomography scans did not correlate with survival or pathologic response. Patients with HER2 3+ overexpression or growth factor receptor-bound protein 7 (Grb7) -positive tumors at baseline demonstrated significantly better survival (P = .007) or treatment response (P = .016), respectively. CONCLUSION Addition of trastuzumab and pertuzumab to nCRT in patients with HER2-positive EAC is feasible and demonstrates potentially promising activity compared with historical controls. HER2 3+ overexpression and Grb7 positivity are potentially predictive for survival and treatment response, respectively.
Collapse
Affiliation(s)
- Charlotte I. Stroes
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Sandor Schokker
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Aafke Creemers
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Remco J. Molenaar
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Maarten C.C.M. Hulshof
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Stephanie O. van der Woude
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Roel J. Bennink
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Ron A.A. Mathôt
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Kausilia K. Krishnadath
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Cornelis J.A. Punt
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | | | - Martijn G.H. van Oijen
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | | - Maartje Los
- Sint Antonius Hospital, Nieuwegein, the Netherlands
| | | | - Annemieke Cats
- Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Maarten F. Bijlsma
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
- Oncode Institute, Amsterdam, the Netherlands
| | - Mark I. van Berge Henegouwen
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Sybren L. Meijer
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Hanneke W.M. van Laarhoven
- Amsterdam University Medical Center, University of Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
7
|
Belgioia L, Desideri I, Errico A, Franzese C, Daidone A, Marino L, Fiore M, Borghetti P, Greto D, Fiorentino A. Safety and efficacy of combined radiotherapy, immunotherapy and targeted agents in elderly patients: A literature review. Crit Rev Oncol Hematol 2018; 133:163-170. [PMID: 30661652 DOI: 10.1016/j.critrevonc.2018.11.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/11/2018] [Accepted: 11/23/2018] [Indexed: 01/31/2023] Open
Abstract
PURPOSE Aim of the present review is to assess present data about the use of the association of Radiotherapy (RT) and targeted therapy/immunotherapy (TT/IT) in elderly people. DESIGN PubMed database was searched for English literature published up to December 2017 using the keywords "radiotherapy" combined with "bevacizumab", "cetuximab", "trastuzumab", "erlotinib", "gefitinib", "sorafenib", "sunitinib", "vismodegib", "sonidegib", "ipilimumab", "pembrolizumab", "nivolumab". Studies performing RT and TT/IT in people aged >65-years were evaluated focusing on safety, toxicity and efficacy. Studies eligible for inclusion were: case reports, retrospective/prospective studies in which RT and new drugs were used concomitantly or sequentially, focusing on elderly sub-group. RESULTS The systematic search identified 626 records. After exclusion of duplicates, full-text review, cross-referencing and paper that did not respect the inclusion criteria, 81 studies were included in this review. In elderly patients the combination of RT with cetuximab or bevacizumab seems feasible but with higher reported side effects. Patients' age should not limit the association of trastuzumab and RT in HER2 positive breast cancer. The concurrent administration of TKIs and RT appears to be feasible and effective. Regarding the Immune Check Point inhibitors and RT, tolerance seems similar among older and younger people but definitive data are lacking. Instead, the association of RT and vismodegib/sonidegib remains investigational. CONCLUSION TT/IT in association of RT seems to be safe, but in elderly patients data concerning safety and toxicity are limited. Specific clinical trials on this population are encouraged.
Collapse
Affiliation(s)
- Liliana Belgioia
- Health Science Department (DISSAL) - University of Genoa and Radiation Oncology Department, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Isacco Desideri
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Italy
| | - Angelo Errico
- Radiation Oncology Department, Mons. R. Dimiccoli Hospital - ASL BT, Barletta, Italy
| | - Ciro Franzese
- Department of Radiotherapy and Radiosurgery Istituto Clinico Humanitas "Humanitas Cancer Center", Rozzano, Italy
| | - Antonino Daidone
- U.O Radioterapia Oncologica, Ospedale A. Ajello, Mazara del Vallo, Trapani - Radioterapia Oncologica, Centro di Medicina Nucleare, San Gaetano, Bagheria, Palermo, Italy
| | | | - Michele Fiore
- Radiation Oncology, Campus Bio-Medico University, Rome, Italy
| | - Paolo Borghetti
- Radiation Oncology Department University and Spedali Civili, Brescia, Italy
| | - Daniela Greto
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", Section of Radiation Oncology, University of Florence, Italy
| | - Alba Fiorentino
- Radiation Oncology Department, General Regional Hospital "F. Miulli", Acquaviva delle fonti, BA, Italy.
| | | |
Collapse
|
8
|
Arcangeli S, Jereczek-Fossa BA, Alongi F, Aristei C, Becherini C, Belgioia L, Buglione M, Caravatta L, D'Angelillo RM, Filippi AR, Fiore M, Genovesi D, Greco C, Livi L, Magrini SM, Marvaso G, Mazzola R, Meattini I, Merlotti A, Palumbo I, Pergolizzi S, Ramella S, Ricardi U, Russi E, Trovò M, Sindoni A, Valentini V, Corvò R. Combination of novel systemic agents and radiotherapy for solid tumors - Part II: An AIRO (Italian association of radiotherapy and clinical oncology) overview focused on treatment toxicity. Crit Rev Oncol Hematol 2018; 134:104-119. [PMID: 30658887 DOI: 10.1016/j.critrevonc.2018.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 11/13/2018] [Accepted: 11/20/2018] [Indexed: 12/31/2022] Open
Abstract
Clinical development and use of novel systemic agents in combination with radiotherapy (RT) is at nowadays most advanced in the field of treatment of solid tumors. Although for many of these substances preclinical studies provide sufficient evidences on their principal capability to enhance radiation effects, the majority of them have not been investigated in even phase I clinical trials for safety in the context of RT. In clinical practice, unexpected acute and late side effects may emerge especially in combination with RT. As a matter of fact, despite combined modality treatment holds potential for enhancing the therapeutic ratio, some concerns are raised from the lack of high-quality clinical data to guide the care of patients who are treated with novel compounds in conjunction with RT. The aim of this review is to provide, from a radio-oncological point of view, an overview of the most advanced combined treatment concepts for solid tumors focusing on treatment toxicity.
Collapse
Affiliation(s)
- Stefano Arcangeli
- Department of Radiation Oncology, Policlinico S. Gerardo and University of Milan "Bicocca", Milan, Italy.
| | | | - Filippo Alongi
- Department of Radiation Oncology, Sacro Cuore Don Calabria Cancer Care Center, Negrar-Verona, University of Brescia, Brescia, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, Department of Surgical and Biomedical Science, University of Perugia, Perugia General Hospital, Perugia, Italy
| | - Carlotta Becherini
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | - Liliana Belgioia
- Department of Radiation Oncology, Ospedale Policlinico San Martino and University of Genoa, Genoa, Italy
| | - Michela Buglione
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Luciana Caravatta
- Department of Radiation Oncology, SS. Annunziata Hospital, G. D'Annunzio University of Chieti, Chieti, Italy
| | | | | | - Michele Fiore
- Radiotherapy Unit, Campus Bio-Medico University, Rome, Italy
| | - Domenico Genovesi
- Department of Radiation Oncology, SS. Annunziata Hospital, G. D'Annunzio University of Chieti, Chieti, Italy
| | - Carlo Greco
- Radiotherapy Unit, Campus Bio-Medico University, Rome, Italy
| | - Lorenzo Livi
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | - Stefano Maria Magrini
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Giulia Marvaso
- Deparment of Radiation Oncology of IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Rosario Mazzola
- Department of Radiation Oncology, Sacro Cuore Don Calabria Cancer Care Center, Negrar-Verona, University of Brescia, Brescia, Italy
| | - Icro Meattini
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | - Anna Merlotti
- Department of Radiation Oncology, S. Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Isabella Palumbo
- Radiation Oncology Section, Department of Surgical and Biomedical Science, University of Perugia, Perugia General Hospital, Perugia, Italy
| | - Stefano Pergolizzi
- Department of Biomedical Sciences and Morphological and Functional Images, University of Messina, Italy
| | - Sara Ramella
- Radiotherapy Unit, Campus Bio-Medico University, Rome, Italy
| | | | - Elvio Russi
- Department of Radiation Oncology, S. Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Marco Trovò
- Department of Radiation Oncology, Azienda Sanitaria Universitaria Integrata of Udine, Udine, Italy
| | - Alessandro Sindoni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Vincenzo Valentini
- Gemelli Advanced Radiation Therapy Center, Fondazione Policlinico Universitario "A. Gemelli", Catholic University of Sacred Heart, Rome, Italy
| | - Renzo Corvò
- Department of Radiation Oncology, Ospedale Policlinico San Martino and University of Genoa, Genoa, Italy
| |
Collapse
|
9
|
Wu SG, Zhang WW, Sun JY, Li FY, Lin Q, He ZY. Patterns of Distant Metastasis Between Histological Types in Esophageal Cancer. Front Oncol 2018; 8:302. [PMID: 30135855 PMCID: PMC6092597 DOI: 10.3389/fonc.2018.00302] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/18/2018] [Indexed: 12/25/2022] Open
Abstract
Introduction: Distant metastasis remains the major cause of treatment failure in esophageal cancer, though there have been few large-scale studies of the patterns of distant metastasis in different histological types. We investigated the patterns of distant metastasis in esophageal adenocarcinoma (AC) and squamous cell carcinoma (SCC) using a population-based approach. Methods: Patients with de novo stage IV esophageal cancer at diagnosis were identified using the Surveillance, Epidemiology, and End Results database. Multivariable logistic regression was performed to identify potential risk factors for site-specific distant metastasis to the distant lymph nodes, bone, liver, brain, and lung at diagnosis. Results: We identified 1,470 patients with complete data for analysis including 1,096 (74.6%) patients with AC and 374 (25.4%) patients with SCC. A total of 2,243 sites of distant metastasis were observed, the liver was the most common site of distant metastasis (727, 32.4%), followed by the distant lymph nodes (637, 28.4%), lung (459, 20.5%), bone (344, 15.3%), and brain (76, 3.4%). Multivariable logistic regression showed that compared to patients with SCC, patients with AC were more likely to have metastasis to the brain (odds ratio [OR] 3.026, 95% confidence interval [CI] 1.441-6.357, p = 0.003) and liver (OR 1.848, 95% CI 1.394–2.451, p < 0.001), and less likely to have metastasis to the lung (OR 0.404, 95% CI 0.316–0.516, p < 0.001). Histological type had no effect on metastasis to the distant lymph nodes or bone. Conclusions: Patients with esophageal AC are more likely to present with liver and brain metastases, and less likely to present with lung metastasis than patients with esophageal SCC.
Collapse
Affiliation(s)
- San-Gang Wu
- Department of Radiation Oncology, Xiamen Cancer Hospital, First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Wen-Wen Zhang
- State Key Laboratory of Oncology in South China, Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Jia-Yuan Sun
- State Key Laboratory of Oncology in South China, Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Feng-Yan Li
- State Key Laboratory of Oncology in South China, Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Qin Lin
- Department of Radiation Oncology, Xiamen Cancer Hospital, First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Zhen-Yu He
- State Key Laboratory of Oncology in South China, Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| |
Collapse
|
10
|
Somatic DNA Copy-Number Alterations Detection for Esophageal Adenocarcinoma Using Digital Polymerase Chain Reaction. Methods Mol Biol 2018. [PMID: 29600372 DOI: 10.1007/978-1-4939-7734-5_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Somatic copy-number alterations are commonly found in cancer and play key roles in activating oncogenes and deactivating tumor suppressor genes. Digital polymerase chain reaction is an effective way to detect the changes in copy number. In esophageal adenocarcinoma, detection of somatic copy-number alterations could predict the prognosis of patients as well as the response to therapy. This chapter will review the methods involved in digital polymerase chain reaction for the research or potential clinical applications in esophageal adenocarcinoma.
Collapse
|
11
|
Shim JW, Madsen JR. VEGF Signaling in Neurological Disorders. Int J Mol Sci 2018; 19:ijms19010275. [PMID: 29342116 PMCID: PMC5796221 DOI: 10.3390/ijms19010275] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/06/2018] [Accepted: 01/10/2018] [Indexed: 12/19/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is a potent growth factor playing diverse roles in vasculogenesis and angiogenesis. In the brain, VEGF mediates angiogenesis, neural migration and neuroprotection. As a permeability factor, excessive VEGF disrupts intracellular barriers, increases leakage of the choroid plexus endothelia, evokes edema, and activates the inflammatory pathway. Recently, we discovered that a heparin binding epidermal growth factor like growth factor (HB-EGF)—a class of EGF receptor (EGFR) family ligands—contributes to the development of hydrocephalus with subarachnoid hemorrhage through activation of VEGF signaling. The objective of this review is to entail a recent update on causes of death due to neurological disorders involving cerebrovascular and age-related neurological conditions and to understand the mechanism by which angiogenesis-dependent pathological events can be treated with VEGF antagonisms. The Global Burden of Disease study indicates that cancer and cardiovascular disease including ischemic and hemorrhagic stroke are two leading causes of death worldwide. The literature suggests that VEGF signaling in ischemic brains highlights the importance of concentration, timing, and alternate route of modulating VEGF signaling pathway. Molecular targets distinguishing two distinct pathways of VEGF signaling may provide novel therapies for the treatment of neurological disorders and for maintaining lower mortality due to these conditions.
Collapse
Affiliation(s)
- Joon W Shim
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Joseph R Madsen
- Department of Neurosurgery, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
12
|
Long-term survival after multidisciplinary therapy for brain metastases from asymptomatic esophageal adenocarcinoma. Clin J Gastroenterol 2017; 11:113-117. [PMID: 29181738 DOI: 10.1007/s12328-017-0803-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 11/20/2017] [Indexed: 01/30/2023]
Abstract
Asymptomatic T1 (invaded submucosa) esophageal carcinoma rarely metastasizes to the brain. A 53-year-old Japanese man complaining of right hemiparesis and convulsion was admitted to our hospital. Brain imaging demonstrated a ring-like, enhanced brain tumor in the left parietal lobe. The pathological findings of the resected tumor were consistent with a metastatic adenocarcinoma from the gastrointestinal tract. Additional examinations revealed an elevated-type tumor in the lower third of the thoracic esophagus. The patient underwent thoracoscopic esophagectomy with lymph node dissection followed by reconstruction with gastric tube substitution. The immunohistochemical findings of the resected specimen were similar to those of the metastatic brain tumor. Although the patient received adjuvant chemotherapy (5-fluorouracil, docetaxel plus cisplatin), a solitary small brain metastasis was detected 4 months after esophagectomy. Excision of the sequential metastases with whole-brain radiation therapy and gamma-knife therapy were performed. The patient survived for 50 months after beginning the initial treatment. This report describes a rare case of brain metastases from T1 esophageal adenocarcinoma in a patient without gastrointestinal symptoms.
Collapse
|
13
|
Mignot F, Ajgal Z, Xu H, Geraud A, Chen JY, Mégnin-Chanet F, Kirova Y. Concurrent administration of anti-HER2 therapy and radiotherapy: Systematic review. Radiother Oncol 2017; 124:190-199. [DOI: 10.1016/j.radonc.2017.07.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 06/22/2017] [Accepted: 07/04/2017] [Indexed: 12/18/2022]
|
14
|
So B, Marcu LG, Olver I, Gowda R, Bezak E. Cocktail without hangover: in search for the optimal chemotherapy in the combined management of non-operable esophageal carcinomas. Acta Oncol 2017; 56:899-908. [PMID: 28375694 DOI: 10.1080/0284186x.2017.1307518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND The worldwide incidence of esophageal cancer has greatly increased over the past few decades making it the sixth deadliest cancer. The disease is often detected in advanced stages when surgery is no longer an option. The standard treatment in these situations is combined chemoradiotherapy, by employing drug cocktails that lead to optimal treatment outcomes both from the perspective of tumor control and normal tissue toxicity. METHODS The aim of this work was to collate the existing trials and clinical studies reported on non-operable esophageal cancer and to analyze the results based on treatment outcomes after various drug combinations. RESULTS Of all drug combinations, cisplatin/5-FU is the most well established chemotherapy regimen for esophageal cancer as both neoadjuvant therapy, an alternative option to surgery, and for palliative purposes. Although this regimen is associated with the most toxicity, it also appears to have the best survival benefit and relief of symptoms. CONCLUSIONS More research is warranted to further increase the therapeutic ratio in non-operable esophageal cancers.
Collapse
Affiliation(s)
- Bianca So
- Faculty of Medicine, University of New South Wales, Sydney, Australia
- School of Health Sciences, University of South Australia, Adelaide, Australia
| | - Loredana G. Marcu
- Department of Physics, Faculty of Science, University of Oradea, Oradea, Romania
- School of Physical Sciences, University of Adelaide, Adelaide, Australia
| | - Ian Olver
- Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
| | - Raghu Gowda
- Department of Radiation Oncology, Royal Adelaide Hospital, Adelaide, Australia
| | - Eva Bezak
- School of Health Sciences, University of South Australia, Adelaide, Australia
- Department of Physics, Faculty of Science, University of Oradea, Oradea, Romania
- Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
| |
Collapse
|
15
|
Abstract
Gastroesophageal junction tumors have been increasing in incidence over time, with most tumors presenting at a locally advanced stage. The treatment plan depends on the stage at diagnosis. PET-CT and endoscopic ultrasound are used to determine clinical stage. Depending on the location of the tumor in the esophagus and stomach, treatment can include chemotherapy with or without radiation, followed by surgery if there is no disease progression. Prognosis is related to stage at diagnosis and response to preoperative treatment. Most surgery for gastroesophageal junction tumors can be performed minimally invasively, which helps decrease postoperative length of stay and morbidity from surgery.
Collapse
Affiliation(s)
- Ikenna C Okereke
- Thoracic Surgery, Division of Cardiothoracic Surgery, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555, USA.
| |
Collapse
|
16
|
Jamorabo DS, Lin SH, Jabbour SK. Successes and Failures of Combined Modalities in Upper Gastrointestinal Malignancies: New Directions. Semin Radiat Oncol 2016; 26:307-19. [PMID: 27619252 PMCID: PMC10794083 DOI: 10.1016/j.semradonc.2016.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Upper gastrointestinal malignancies generally have moderate to poor cure rates, even in the earliest stages, thereby implying that both local and systemic treatments have room for improvement. Therapeutic options are broadening, however, with the development of new immunotherapies and targeted agents, which can have synergistic effects with radiotherapy. Here we discuss the current state of combined modality therapy for upper gastrointestinal malignancies, specifically recent successes and setbacks in trials of radiation therapy with targeted therapies, vaccines, immunotherapies, and chemotherapies.
Collapse
Affiliation(s)
- Daniel S Jamorabo
- Department of Internal Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ.
| |
Collapse
|
17
|
Kothari N, Mellon E, Hoffe SE, Frakes J, Shridhar R, Pimiento J, Meredith K, Tran ND, Saeed N, Almhanna K. Outcomes in patients with brain metastasis from esophageal carcinoma. J Gastrointest Oncol 2016; 7:562-9. [PMID: 27563446 DOI: 10.21037/jgo.2016.03.12] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Brain metastases from esophageal carcinoma have historically been rare and associated with poor prognosis. With improvements in systemic disease control, the incidence of brain metastases is expected to rise. To better inform management decisions, we sought to identify factors associated with survival in patients with brain metastasis from esophageal cancer. METHODS We retrospectively identified 49 patients with brain metastasis from stage I-IV primary esophageal cancer treated with surgery, radiation, or a combination of modalities at our tertiary referral center between 1998 and 2015. Medical records were reviewed to collect demographic and clinical information. RESULTS Median age at diagnosis of the primary esophageal cancer was 60 years. Forty-one (84%) patients were male and forty patients (82%) had adenocarcinoma. Median overall survival (MS) following esophageal cancer diagnosis was 24 months (range, 3-71 months), and median survival after the identification of brain metastases was 5 months (range, 1-52 months). On univariate analysis, only patients with poor Karnofsky performance status (KPS <70), recursive partitioning analysis (RPA) classification (III), or 3 or more brain metastases were found to have worsened survival after the diagnosis of brain metastases (all P<0.01). Factors not associated with survival were age, gender, histology (adenocarcinoma vs. other), palliative-intent treatment of the primary tumor, time to diagnosis of brain metastases from initial diagnosis, uncontrolled primary tumor at time of brain metastasis diagnosis, or extracranial metastases. On multivariate analysis (MVA, KPS excluded), patients with RPA class I (MS, 14.6 months) or II (MS, 5.0 months) disease had significantly improved overall survival compared to class III disease (MS, 1.6 months, P<0.01). Also on MVA, patients with 1 (MS, 10.7 months) or 2 (MS, 4.7 months) brain metastases had significantly improved overall survival compared to patients with 3 or more brain metastases (MS, 0.3 months, P<0.01). For the 36 patients with 1-2 brain metastases and KPS ≥70, MS was 11.1 months. CONCLUSIONS While the prognosis for esophageal cancer metastatic to brain remains poor overall, we found that patients with good performance status and limited number of brain lesions have superior survival. Aggressive management may further improve outcomes in these patients.
Collapse
Affiliation(s)
- Nishi Kothari
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Eric Mellon
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Sarah E Hoffe
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Jessica Frakes
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Ravi Shridhar
- Department of Radiation Oncology, Florida Hospital Cancer Institute, Orlando, FL, USA
| | - Jose Pimiento
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Ken Meredith
- Department of Gastrointestinal Oncology, First Physicians Group, Sarasota, FL, USA
| | - Nam D Tran
- Department of Neuro-oncology. H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Nadia Saeed
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Khaldoun Almhanna
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| |
Collapse
|
18
|
Fokas E, Rödel C. Targeted agents in GI radiotherapy: Clinical efficacy and side effects. Best Pract Res Clin Gastroenterol 2016; 30:537-49. [PMID: 27644903 DOI: 10.1016/j.bpg.2016.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/26/2016] [Accepted: 05/08/2016] [Indexed: 01/31/2023]
Abstract
Approximately 50% of all patients with cancer receive radiotherapy (RT) at some point during their treatment. Despite the advent of modern imaging and advances in planning and delivering highly-conformal and precise RT, further dose escalation to improve clinical outcome is often limited by the potential side-effects to adjacent tissues. Addition of chemotherapy to radiotherapy (CRT) has led to significant clinical improvements in many gastrointestinal malignancies but at the expense of increased toxicity as most chemotherapy drugs lack specificity. Targeted agents modulate specific biological pathways and can potentially enhance RT efficacy. However, so far, the majority of clinical studies incorporating targeted agents into RT and CRT have produced disappointing results in gastrointestinal malignancies. Also, we lack validated biomarkers and methods for monitoring and predicting the efficacy of these agents when combined with RT/CRT. In the present article, we will review the most important targeted therapies, and examine the efficacy and toxicity of these agents when combined with RT/CRT in gastrointestinal malignancies. The shortcomings as well as future challenges and perspectives for the successful use of these compounds with RT/CRT in future trials will also be outlined.
Collapse
Affiliation(s)
- Emmanouil Fokas
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK; Department of Radiotherapy and Oncology, University of Frankfurt, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK) partner site: Frankfurt, Germany.
| | - Claus Rödel
- Department of Radiotherapy and Oncology, University of Frankfurt, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany and German Cancer Consortium (DKTK) partner site: Frankfurt, Germany
| |
Collapse
|
19
|
Yoon HH, Lewis MA, Foster NR, Sukov WR, Khan M, Sattler CA, Wiktor AE, Wu TT, Jenkins RB, Sinicrope FA. Central nervous system relapse in patients with untreated HER2-positive esophageal or gastroesophageal junction adenocarcinoma. Int J Cancer 2016; 139:1626-31. [PMID: 27198655 DOI: 10.1002/ijc.30200] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/25/2016] [Accepted: 04/27/2016] [Indexed: 01/20/2023]
Abstract
Although HER2-positive breast cancers demonstrate a propensity for central nervous system (CNS) metastasis, it is unknown whether other HER2-positive tumors, including adenocarcinomas of the esophagus/gastroesophageal junction (EAC), share this characteristic. Insight into this association may inform the development of HER2-targeted therapies that penetrate the blood-brain barrier. We examined HER2 overexpression and gene amplification in 708 patients with EAC who underwent curative-intent surgery during a time period (1980-1997) when no patient received HER2-targeted therapy. We identified patients whose site of first cancer recurrence was CNS and those who had a CNS relapse at any time. After a median follow-up of 61.2 months, 3.4% (24/708) of patients developed CNS relapse (all involved the brain). Patients with HER2-positive (vs -negative) primary tumors showed a higher 5-year cumulative incidence of CNS relapse as first recurrence (5.8% vs. 1.2%; p = 0.0058) and at any time (8.3% vs. 2.4%; p = 0.0062). In a multivariable model that included covariates previously associated with HER2 or with CNS relapse in breast cancer, HER2 positivity was the only variable that was statistically significantly associated with shorter time to CNS relapse as first recurrence (p = 0.0026) or at any time (hazard ratio 4.3 [95% confidence interval 1.8 to 10.3]; p = 0.001). These are the first data in a non-breast cancer to demonstrate an association between HER2 positivity and higher CNS relapse risk after surgery, and suggest that HER2-positive EACs have a predilection for CNS metastases.
Collapse
Affiliation(s)
- Harry H Yoon
- Department of Medical Oncology, Mayo Clinic, Rochester, MN
| | - Mark A Lewis
- Department of Medical Oncology, MD Anderson Cancer Center, Houston, TX
| | - Nathan R Foster
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | | | - Maliha Khan
- Department of Leukemia Department, MD Anderson Cancer Center, Houston, TX
| | | | - Anne E Wiktor
- Mayo Collaborative Services, Mayo Clinic, Rochester, MN
| | | | | | | |
Collapse
|
20
|
Chan E, Duckworth LV, Alkhasawneh A, Toro TZ, Lu X, Ben-David K, Hughes SJ, Rossidis G, Zlotecki R, Lightsey J, Daily KC, Dang L, Allegra CJ, King B, George TJ. Discordant HER2 expression and response to neoadjuvant chemoradiotherapy in esophagogastric adenocarcinoma. J Gastrointest Oncol 2016; 7:173-80. [PMID: 27034783 PMCID: PMC4783750 DOI: 10.3978/j.issn.2078-6891.2015.071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 06/05/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Targeting human epidermal growth factor receptor 2 (HER2) with trastuzumab in metastatic esophagogastric adenocarcinoma (EGA) improves survival. The impact of HER2 inhibition in combination with chemoradiotherapy (CRT) in early stage EGA is under investigation. This study analyzed the pattern of HER2 overexpression in matched-pair tumor samples of patients who underwent neoadjuvant CRT followed by surgery. METHODS All patients with EGA who underwent standard neoadjuvant CRT followed by esophagectomy at the University of Florida were included. Demographics, risk factors, tumor features, and outcome data were analyzed. Descriptive statistics, Chi-square exact test, uni- and multivariate analyses, and Kaplan Meier method were used. HER2 expression determined by immunohistochemical (IHC) was scored as negative (0, 1+), indeterminate (2+) or positive (3+). RESULTS Among 49 sequential patients (41 M/8 F) with matched-pair tumor samples, 9/49 patients (18%) had pathologic complete response (pCR), 10/49 had near pCR or not enough tumor (NET) to examine in the post- treatment samples. Patients with initial HER2 negativity demonstrated conversion to HER2 positivity after neoadjuvant CRT (7/30 cases; 23%). Baseline HER2 overexpression was more common in lower stage/node negative patients (67% in stages I, IIA vs. 33% in stages IIB, III) and did not correlate with treatment response or survival. CONCLUSIONS Although limited by a relatively small sample size, our study failed to demonstrate that baseline HER2 protein over-expression in EGA predicts response to standard CRT. However, our data suggested that HER2 was up regulated by CRT resulting in unreliable concordance between pre-treatment (pre-tx) and post-treatment (post-tx) samples. Pre-therapy HER2 expression may not reliably reflect the HER2 status of persistent or recurrent disease.
Collapse
|
21
|
Abstract
INTRODUCTION Human epidermal growth factor receptor 2 (HER2) and HER3 are altered in multiple tumor types, including gastrointestinal cancer. The HER2/HER3 dimer is crucial for HER2-mediated signaling in HER2-positive tumors. HER2-targeting agents, including trastuzumab, lapatinib, trastuzumab emtansine, and pertuzumab, have been approved for the treatment of HER2-positive breast cancer, with trastuzumab also approved for the treatment of HER2-positive gastric cancer. Pertuzumab, a recombinant humanized immunoglobulin (Ig) G1 monoclonal antibody targeting HER-2, binds to the dimerization domain (extracellular domain II) of HER2, which leads to blocking of ligand-induced HER2 heterodimerization. It is under investigation in gastrointestinal cancers, including HER2-positive gastric cancer. AREA COVERED In this review, the authors summarize the biology of HER2/HER3 and its alterations in gastrointestinal cancers. The authors focus specifically on the current status of development of pertuzumab in gastrointestinal cancers. EXPERT OPINION The HER2/HER3 alteration in gastrointestinal cancers is quite interesting. In HER2-positive gastric cancer, the dual blockade of HER2 and HER3 using trastuzumab and pertuzumab is being tested in an international phase III trial, the JACOB study. This strategy may benefit HER2-positive gastric cancer patients more as in the case of HER2-positive breast cancer. In other gastrointestinal cancers, including biliary tract cancer, esophageal cancer, pancreatic cancer, and colorectal cancer, there is huge room for the development of pertuzumab.
Collapse
Affiliation(s)
- Do-Youn Oh
- a Department of Internal Medicine , Seoul National University Hospital , Seoul , Korea.,b Cancer Research Institute , Seoul National University College of Medicine , Seoul , Korea
| | - Yung-Jue Bang
- a Department of Internal Medicine , Seoul National University Hospital , Seoul , Korea.,b Cancer Research Institute , Seoul National University College of Medicine , Seoul , Korea
| |
Collapse
|
22
|
Woo J, Cohen SA, Grim JE. Targeted therapy in gastroesophageal cancers: past, present and future. Gastroenterol Rep (Oxf) 2015; 3:316-29. [PMID: 26510453 PMCID: PMC4650980 DOI: 10.1093/gastro/gov052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 09/09/2015] [Indexed: 12/12/2022] Open
Abstract
Gastroesophageal cancer is a significant global problem that frequently presents at an incurable stage and has very poor survival with standard chemotherapy approaches. This review will examine the epidemiology and molecular biology of gastroesophageal cancer and will focus on the key deregulated signaling pathways that have been targeted in the clinic. A comprehensive overview of clinical data highlighting successes and failures with targeted agents will be presented. Most notably, HER2-targeted therapy with the monoclonal antibody trastuzumab has proven beneficial in first-line therapy and has been incorporated into standard practice. Targeting the VEGF pathway has also proven beneficial, and the VEGFR-targeted monoclonal antibody ramucirumab is now approved for second-line therapy. In contrast to these positive results, agents targeting the EGFR and MET pathways have been evaluated extensively in gastroesophageal cancer but have repeatedly failed to show benefit. An increased understanding of the molecular predictors of response to targeted therapies is sorely needed. In the future, improved molecular pathology approaches should subdivide this heterogeneous disease entity to allow individualization of cancer therapy based on integrated and global identification of deregulated signaling pathways. Better patient selection, rational combinations of targeted therapies and incorporation of emerging immunotherapeutic approaches should further improve the treatment of this deadly disease.
Collapse
Affiliation(s)
- Janghee Woo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA, Division of Medical Oncology, University of Washington, Seattle, WA, USA and
| | - Stacey A Cohen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA, Division of Medical Oncology, University of Washington, Seattle, WA, USA and
| | - Jonathan E Grim
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA, Division of Medical Oncology, University of Washington, Seattle, WA, USA and Hospital and Specialty Medicine, VA Puget Sound Health Care System, Seattle, WA, USA
| |
Collapse
|
23
|
Nagaraja V, Shaw N, Morey AL, Cox MR, Eslick GD. HER2 expression in oesophageal carcinoma and Barrett's oesophagus associated adenocarcinoma: An Australian study. Eur J Surg Oncol 2015; 42:140-8. [PMID: 26422587 DOI: 10.1016/j.ejso.2015.08.159] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/05/2015] [Accepted: 08/06/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Several studies have evaluated the prognostic value of HER2 in oesophageal cancer, but the prognostic influence of HER2 overexpression in oesophageal cancer remains uncertain. The aim of this study was to assess the incidence of HER2 positivity and relationship with clinicopathological features in patients with oesophageal cancer. DESIGN The study cohort consisted of 269 patients diagnosed with oesophageal carcinoma in a single institution. HER2 expression was analysed by immunohistochemistry (IHC) and silver in situ hybridization (SISH) in 152 archival oesophageal cancer specimens. Survival analysis was assessed using Hazard models. RESULTS HER2 expression was IHC3+ in 14 (9.2%), IHC2+ in 14 (9.2%), IHC1+ in 57 (37.5%), and IHC0 in 67 (44.1%) cases. SISH results confirmed that 15 specimens (9.9%) were HER2 gene amplified. Among 27 squamous cell carcinomas (SCCs) only 3.7% were HER2 positive whereas 11.2% of 125 adenocarcinomas were HER2 positive. The HER2 positive tumours were more likely to occur in men (OR: 5.00, 95% CI: 1.69-14.29), smokers (OR: 10.00, 95% CI: 4.17-25) and in patients with Barrett's oesophagus (OR: 8.33, 95% CI: 3.71-20.00). There was no significant difference in survival between the (HER2 +ve, 14.3 months vs HER2 -ve, 24.6 months, p = 0.42) CONCLUSION: A HER2 prevalence rate of 9.9% was found among patients with oesophageal cancer and no correlation with survival was detected overall.
Collapse
Affiliation(s)
- V Nagaraja
- The Whiteley-Martin Research Centre, Discipline of Surgery, The University of Sydney, Nepean Hospital, Penrith, New South Wales, Australia
| | - N Shaw
- Department of Pathology, St. Vincent's Hospital, Darlinghurst, New South Wales, Australia
| | - A L Morey
- Department of Pathology, St. Vincent's Hospital, Darlinghurst, New South Wales, Australia
| | - M R Cox
- The Whiteley-Martin Research Centre, Discipline of Surgery, The University of Sydney, Nepean Hospital, Penrith, New South Wales, Australia
| | - G D Eslick
- The Whiteley-Martin Research Centre, Discipline of Surgery, The University of Sydney, Nepean Hospital, Penrith, New South Wales, Australia.
| |
Collapse
|
24
|
Ruffier-Loubière A, Janoray G, Chapet S, de Calan L, Dumont P, Dorval É, Orain I, Calais G. [Long-term outcome of neoadjuvant radiochemotherapy followed by surgery for esophageal cancer: a single institution retrospective study of 102 patients]. Cancer Radiother 2015. [PMID: 26215366 DOI: 10.1016/j.canrad.2015.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE AND OBJECTIVES To report survival and morbidity of a large homogeneous cohort of patients with a locally advanced esophageal or cardia carcinoma and put in evidence predictive factors of locoregional control and survival. PATIENTS AND METHODS Hundred and two patients were treated at the university hospital of Tours between 1990 and 2010 and received neo-adjuvant chemoradiation therapy with external irradiation (40Gy-44Gy) and two courses of chemotherapy (5-fluoro-uracile and cisplatine). Esophagectomy associated with lymph node dissection was performed about ten weeks after the end of chemoradiation therapy. RESULTS The median follow-up was 22.4 months [6-185 months]. The overall survival rates at 2 and 5years were 53% and 27%, respectively. The median overall survival was estimated at 27months. The overall 2-year survival between patients "responders" and patients "non-responders" was 67% vs 26%, respectively (P<0.0001). In case of histological response, there was a benefit in terms of overall survival (P<0.0001), locoregional control (P<0.0036) and disease-free survival (P<0.001). Overall survival at 2years was 64% for ypN0 group vs 32% for ypN1 group (P<0.0001). The median survival was estimated at 37months against 15months in the absence of lymph node involvement (P<0.0001). CONCLUSION Our results in terms of survival, tolerance and morbidity and mortality were comparable to those in the literature. Complete histological response of lymph node was associated with an improvement of local control, disease-free survival and overall survival.
Collapse
Affiliation(s)
- A Ruffier-Loubière
- Clinique d'oncologie-radiothérapie, hôpital Bretonneau, CHRU de Tours, 2, boulevard Tonnellé, 37044 Tours cedex 9, France
| | - G Janoray
- Clinique d'oncologie-radiothérapie, hôpital Bretonneau, CHRU de Tours, 2, boulevard Tonnellé, 37044 Tours cedex 9, France.
| | - S Chapet
- Clinique d'oncologie-radiothérapie, hôpital Bretonneau, CHRU de Tours, 2, boulevard Tonnellé, 37044 Tours cedex 9, France
| | - L de Calan
- Service de chirurgie digestive, hôpital Trousseau, CHRU de Tours, avenue de la République, 37170 Chambray-les-Tours, France
| | - P Dumont
- Service de chirurgie thoracique, hôpital Trousseau, CHRU de Tours, avenue de la République, 37170 Chambray-les-Tours, France
| | - É Dorval
- Service de gastroentérologie, hôpital Trousseau, CHRU de Tours, avenue de la République, 37170 Chambray-les-Tours, France
| | - I Orain
- Service d'anatomopathologie, hôpital Trousseau, CHRU de Tours, avenue de la République, 37170 Chambray-les-Tours, France
| | - G Calais
- Clinique d'oncologie-radiothérapie, hôpital Bretonneau, CHRU de Tours, 2, boulevard Tonnellé, 37044 Tours cedex 9, France
| |
Collapse
|
25
|
Tian GY, Miu M, Huang XE. Systematic analysis of pemetrexed-based chemoradiotherapy for patients with locally advanced or metastatic esophageal cancer. Asian Pac J Cancer Prev 2015; 15:8475-8. [PMID: 25339049 DOI: 10.7314/apjcp.2014.15.19.8475] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
PURPOSE This systematic analysis was conducted to evaluate the efficacy and safety of pemetrexed-based chemoradiotherapy in treating patients with locally advanced or metastatic esophageal cancer. METHODS Clinical studies evaluating the efficacy and safety of pemetrexed based regimens on response and safety for relevant patients were identified using a predefined search strategy. Pooled response rates (RRs) were calculated. RESULTS For pemetrexed-based regimens, 4 clinical studies including 47 patients with locally advanced or metastatic esophageal cancer were considered eligible for inclusion. Systematic analysis showed that, in all patients, the pooled RR was 51% (24/47) . Major adverse effects of grade III/IV were esophagitis, neutropenia, thrombocytopenia, anemia anorexia, fatigue, diarrhea, dysphagia and vomiting. No treatment related death occurred with pemetrexed-based treatment. CONCLUSION This systematic analysis suggests that pemetrexed based radiotherapy is associated with reasonable activity and good tolerability in treating patients with locally advanced or metastatic esophageal cancer.
Collapse
Affiliation(s)
- Guang-Yu Tian
- Department of Chemotherapy, the Affiliated Jiangsu Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, China E-mail :
| | | | | |
Collapse
|
26
|
Niccolai E, Taddei A, Prisco D, Amedei A. Gastric cancer and the epoch of immunotherapy approaches. World J Gastroenterol 2015; 21:5778-5793. [PMID: 26019442 PMCID: PMC4438012 DOI: 10.3748/wjg.v21.i19.5778] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 12/05/2014] [Accepted: 04/17/2015] [Indexed: 02/06/2023] Open
Abstract
The incidence of gastric cancer (GC) fell dramatically over the last 50 years, but according to IARC-Globocan 2008, it is the third most frequent cause of cancer-related deaths with a case fatality GC ratio higher than other common malignancies. Surgical resection is the primary curative treatment for GC though the overall 5-year survival rate remains poor (approximately 20%-25%). To improve the outcome of resectable gastric cancer, different treatment strategies have been evaluated such as adjuvant or perioperative chemotherapy. In resected gastric cancer, the addition of radiotherapy to chemotherapy does not appear to provide any additional benefit. Moreover, in metastatic patients, chemotherapy is the mainstay of palliative therapy with a median overall survival of 8-10 mo and objective response rates of merely 20%-40%. Therefore, the potential for making key beneficial progress is to investigate the GC molecular biology to realize innovative therapeutic strategies, such as specific immunotherapy. In this review, we provide a panoramic view of the different immune-based strategies used for gastric cancer treatment and the results obtained in the most significant clinical trials. In detail, firstly we describe the therapeutic approaches that utilize the monoclonal antibodies while in the second part we analyze the cell-based immunotherapies.
Collapse
|
27
|
Chemoradiation in oesophageal cancer. Best Pract Res Clin Gastroenterol 2015; 29:193-209. [PMID: 25743466 DOI: 10.1016/j.bpg.2014.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 11/07/2014] [Accepted: 11/16/2014] [Indexed: 01/31/2023]
Abstract
Oesophageal cancer is the 8th most common cancer worldwide, and has significant mortality and morbidity rates. The two most common histological types, squamous cell carcinoma and adenocarcinoma, have different localizations, distinctive risk factors, and molecular mechanisms. Survival for patients with locoregional oesophageal cancer is poor when treated with surgery only, with 5-year survival less than 10-15%. Radiation therapy has limited efficacy when given alone. Concurrent chemoradiation improves local-regional control and facilitates margin-free resection when delivered preoperatively. Chemoradiation prolongs survival when given as definitive treatment or combined with surgery. Neoadjuvant chemoradiation also reduces risk of distant recurrence. To date, there is no data supporting the addition of targeted therapy to concurrent chemoradiation. Understanding molecular pathways regulating both radiosensitivity and tumorigenesis/invasion may lead to the discovery of new targeted agents, improving outcome of chemoradiation in terms of both locoregional and systemic control, ultimately resulting in prolonged survival.
Collapse
|
28
|
Boonstra JJ, Tilanus HW, Dinjens WNM. Translational research on esophageal adenocarcinoma: from cell line to clinic. Dis Esophagus 2015; 28:90-6. [PMID: 23795680 DOI: 10.1111/dote.12095] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human esophageal adenocarcinoma (EAC) cell lines have made a substantial contribution to elucidating mechanisms of carcinogenesis and drug discovery. Model research on EAC relies almost entirely on a relatively small set of established tumor cell lines because appropriate animal models are lacking. Nowadays, more than 20% of all fundamental translational research studies regarding EAC are partially or entirely based on these cell lines. The ready availability of these cell lines to investigators worldwide have resulted in more than 250 publications, including many examples of important biomedical discoveries. The high genomic similarities (but certainly not completely identical) between the EAC cell lines and their original tumors provide rational for their use. Recently, in a collaborative effort all available EAC cell lines have been verified resulting in the establishment of a reliable panel of 10 EAC cell lines. It could be expected that the value of these cell lines increases as unlimited source of tumor material because new biomedical techniques require more tumor cells and the supply of viable tumor cells is diminishing because of neoadjuvant chemo(radio)therapy of patients with EAC. Here, we review the history of the EAC cell lines and their utility in translational research and biomedical discovery.
Collapse
Affiliation(s)
- J J Boonstra
- Department of Pathology, Josephine Nefkens Institute, University Medical Center, Rotterdam, The Netherlands; Department of Surgery, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|
29
|
Shen K, Huang XE, Lu YY, Wu XY, Liu J, Xiang J. Phase II study of docetaxel (Aisu®) combined with three- dimensional conformal external beam radiotherapy in treating patients with inoperable esophageal cancer. Asian Pac J Cancer Prev 2014; 13:6523-6. [PMID: 23464486 DOI: 10.7314/apjcp.2012.13.12.6523] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE This study was designed to investigate treatment efficacy and side effects of concomitant Aisu® (docetaxel) with three-dimensional conformal external beam radiotherapy for the treatment of inoperable patients with esophageal cancer. METHODS Inoperable patients were treated with three-dimensional conformal external beam radiotherapy (5/week, 2 GY/day, and total dose 60GY) plus docetaxel (30-45 mg/m2, iv, d1, 8). RESULTS Twenty eight patients met the study eligibility criteria and the response rate was evaluated according to RICIST guidelines. Among 28 patients, 2 achieved CR, 22 PR, 3 SD and 1 patient was documented PD. Mild gastrointestinal reaction and bone marrow suppression were also documented. All treatment related side effects were tolerable. CONCLUSION Three-dimensional conformal external beam radiotherapy combined with docetaxel is an active and safe regimen for inoperable patients with esophageal cancer.
Collapse
Affiliation(s)
- Kang Shen
- Department of Chemotherapy, Affiliated Jiangsu Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, China
| | | | | | | | | | | |
Collapse
|
30
|
Uzunoglu FG, Koenig AM, Izbicki JR. The potential for targeting HER2 therapeutically in esophageal cancer – a grasp at straws? Expert Opin Ther Targets 2014; 18:1421-6. [DOI: 10.1517/14728222.2014.956086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
31
|
Rackley T, Leong T, Foo M, Crosby T. Definitive Chemoradiotherapy for Oesophageal Cancer — A Promising Start on an Exciting Journey. Clin Oncol (R Coll Radiol) 2014; 26:533-40. [DOI: 10.1016/j.clon.2014.06.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 06/02/2014] [Indexed: 10/25/2022]
|
32
|
Zhang J, Jiang D, Li X, Lv J, Xie L, Zheng L, Gavine PR, Hu Q, Shi Y, Tan L, Ge D, Xu S, Li L, Zhu L, Hou Y, Wang Q. Establishment and characterization of esophageal squamous cell carcinoma patient-derived xenograft mouse models for preclinical drug discovery. J Transl Med 2014; 94:917-26. [PMID: 24999713 DOI: 10.1038/labinvest.2014.77] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 03/27/2014] [Accepted: 04/08/2014] [Indexed: 12/22/2022] Open
Abstract
The purpose of this study was to establish and characterize patient-derived esophageal squamous cell carcinoma xenograft (PDECX) mice for utilization in antitumor drug discovery. A total of 96 esophageal squamous cell carcinoma (ESCC) tissues from Chinese patients were transplanted subcutaneously into immunodeficient mice. Histology, EGFR, K-ras, B-raf, and PIK3CA mutations, and HER2 gene amplifications were analyzed in both patient tumors and mouse xenograft tissues using immunohistochemistry, mutant-enriched liquid chip sequencing and fluorescence in situ hybridization assays, respectively. Furthermore, in vivo efficacy studies using five PDECX mice harboring a variety of genetic aberrations were performed using the chemotherapy agents 5-fluorouracil (5-FU) and cisplatin. Thirty-seven PDECX mouse models were successfully established in immunodeficient mice. Pathological analysis revealed similar histological architecture and degrees of differentiation between patient ESCC and xenografted tumors. No mutations were identified in EGFR, K-ras, and B-raf genes in either xenograft models or patient ESCC tissues. In contrast, PIK3CA gene mutations were detected in 12.5% (12/96) ESCC patients and 18.9% (7/37) PDECX models. Interestingly, patient ESCC tissues exhibiting HER2 overexpression or gene amplification were unable to survive in immunodeficient mice. Further analysis showed that PDECX models carrying HER2 2+ expression had no response to 5-FU/cisplatin, compared with HER2-negative models. In conclusion, a panel of PDECX mouse models, which include PIK3CA mutant and HER2-positive models, was established and characterized thus mimicking the current clinical genetic setting of esophageal carcinoma. The sensitivity of HER2-negative ESCC models to chemotherapy supports stratification approaches in the treatment of esophageal carcinoma patients and warrants further investigation of the impact of PI3KCA on treatment response.
Collapse
Affiliation(s)
- Jingchuan Zhang
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xiaojing Li
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jing Lv
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Liang Xie
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Li Zheng
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Paul R Gavine
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Qin Hu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yuan Shi
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Lijie Tan
- Department of Thorax Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Di Ge
- Department of Thorax Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Songtao Xu
- Department of Thorax Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Leon Li
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Lifang Zhu
- Innovation Center China, AstraZeneca Global R&D, Zhangjiang Hi-Tech Park, Shanghai, People's Republic of China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Qun Wang
- Department of Thorax Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
33
|
Moorcraft SY, Chau I. Investigational therapies targeting the ErbB family in oesophagogastric cancer. Expert Opin Investig Drugs 2014; 23:1349-63. [PMID: 24949530 DOI: 10.1517/13543784.2014.930126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The prognosis for patients with oesophagogastric (OG) cancer remains poor, with a median survival of approximately 9 - 11 months for patients with metastatic disease. However, a more personalised approach to treatment, using drugs tailored to the molecular characteristics of patients' tumours, has the potential to improve patient outcomes. Drugs targeting the ErbB family of receptors have been developed, but these have had varying degrees of success in clinical practice. AREAS COVERED The authors provide an overview of the ErbB receptor family with regard to OG cancers. Furthermore, they evaluate the evidence from preclinical and clinical trials of therapeutics targeting this family, including monoclonal antibodies, tyrosine kinase inhibitors and novel agents. EXPERT OPINION Drugs targeting the ErbB family have been evaluated in OG cancer, with a notable success story in the case of trastuzumab, although there have been disappointing failures with anti-EGFR therapy. The response to targeted treatment remains variable and further biomarker research is essential to identify patients most likely to benefit from these therapies. The treatment of OG cancer remains challenging, but new anti-HER2 therapies and combination therapies hold promise for the future.
Collapse
Affiliation(s)
- Sing Yu Moorcraft
- The Royal Marsden NHS Foundation Trust, Gastrointestinal Unit, Department of Medicine , Sutton SM2 5PT , UK +44 020 8642 6011 ; +44 020 8643 9414 ;
| | | |
Collapse
|
34
|
Guo XF, Zhu XF, Yang WC, Zhang SH, Zhen YS. An EGFR/HER2-Bispecific and enediyne-energized fusion protein shows high efficacy against esophageal cancer. PLoS One 2014; 9:e92986. [PMID: 24664246 PMCID: PMC3963964 DOI: 10.1371/journal.pone.0092986] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 02/27/2014] [Indexed: 01/27/2023] Open
Abstract
Esophageal cancer is one of the most common cancers, and the 5-year survival rate is less than 10% due to lack of effective therapeutic agents. This study was to evaluate antitumor activity of Ec-LDP-Hr-AE, a recently developed bispecific enediyne-energized fusion protein targeting both epidermal growth factor receptor (EGFR) and epidermal growth factor receptor 2 (HER2), on esophageal cancer. The fusion protein Ec-LDP-Hr-AE consists of two oligopeptide ligands and an enediyne antibiotic lidamycin (LDM) for receptor binding and cell killing, respectively. The current study demonstrated that Ec-LDP-Hr had high affinity to bind to esophageal squamous cell carcinoma (ESCC) cells, and enediyne-energized fusion protein Ec-LDP-Hr-AE showed potent cytotoxicity to ESCC cells with differential expression of EGFR and HER2. Ec-LDP-Hr-AE could cause significant G2-M arrest in EC9706 and KYSE150 cells, and it also induced apoptosis in ESCC cells in a dosage-dependent manner. Western blot assays showed that Ec-LDP-Hr-AE promoted caspase-3 and caspase-7 activities as well as PARP cleavage. Moreover, Ec-LDP-Hr-AE inhibited cell proliferation via decreasing phosphorylation of EGFR and HER2, and further exerted inhibition of the activation of their downstream signaling molecules. In vivo, at a tolerated dose, Ec-LDP-Hr-AE inhibited tumor growth by 88% when it was administered to nude mice bearing human ESCC cell KYSE150 xenografts. These results indicated that Ec-LDP-Hr-AE exhibited potent anti-caner efficacy on ESCC, suggesting it could be a promising candidate for targeted therapy of esophageal cancer.
Collapse
Affiliation(s)
- Xiao-Fang Guo
- Department of Microbiology, Xinxiang Medical University, Xinxiang, China
| | - Xiao-Fei Zhu
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Wan-Cai Yang
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
- Department of Pathology, University of Illinois at Chicago, Chicago, United States of America
| | - Sheng-Hua Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Perking Union Medical College, Beijing, China
| | - Yong-Su Zhen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Perking Union Medical College, Beijing, China
- * E-mail:
| |
Collapse
|
35
|
van Hagen P, Biermann K, Boers JE, Stoss O, Sleddens HF, van Lanschot JJB, Dinjens WNM, Rueschoff J, Wijnhoven BPL. Human epidermal growth factor receptor 2 overexpression and amplification in endoscopic biopsies and resection specimens in esophageal and junctional adenocarcinoma. Dis Esophagus 2014; 28:380-5. [PMID: 24611982 DOI: 10.1111/dote.12204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) is overexpressed in a subset of esophageal adenocarcinomas. Frequently, biopsy material is used for evaluation of HER2 status. The aim of the study was to determine if HER2 expression in preoperative endoscopic biopsies is representative for the entire tumor. Preoperative endoscopic biopsies and matched resection specimens were collected from 75 patients who underwent esophagectomy for esophageal adenocarcinoma. Immunohistochemical staining (IHC) on HER2 and dual-color in situ hybridization (ISH) were performed. HER2 status was determined by following a clinical algorithm, first determining HER2 overexpression on immunohistochemistry and, when equivocal (2+), determining HER2 amplification on ISH. Seventy-one of 75 (95%) biopsies and 69/75 (92%) resection specimens could be analyzed due to technical failure. HER2 positivity was seen in 18/71 (25%) biopsies and in 15/69 (22%) resection specimens. Overall, HER2 status in the biopsy was concordant with HER2 status in the resection specimen in 94% of cases. Interobserver agreement on IHC scoring for all three observers was 83% in biopsies and 85% in resection specimens. HER2 positivity was detected in 22% of esophageal adenocarcinomas. Although interobserver agreement was moderate, HER2 status of a primary tumor can be reliably determined based on the endoscopically obtained pretreatment biopsy.
Collapse
Affiliation(s)
- P van Hagen
- Department of Surgery, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Liang J, E M, Wu G, Zhao L, Li X, Xiu X, Li N, Chen B, Hui Z, Lv J, Fang H, Tang Y, Bi N, Wang W, Zhai Y, Li T, Chen D, Zou S, Lu N, Perez-Rodríguez R, Zheng J, Wang L. Nimotuzumab combined with radiotherapy for esophageal cancer: preliminary study of a Phase II clinical trial. Onco Targets Ther 2013; 6:1589-96. [PMID: 24235844 PMCID: PMC3825695 DOI: 10.2147/ott.s50945] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE To determine the safety and therapeutic effects of nimotuzumab (h-R3) combined with radiotherapy in esophageal cancer. METHODS This Phase II clinical trial involved 42 patients with stage II (inoperable or refused surgery) to stage IV (supraclavicular lymph node metastasis only) esophageal cancers treated between November 2008 and July 2010. All patients had squamous cell carcinomas, and all received three-dimensional conformal radiotherapy and 200 mg nimotuzumab per week during radiotherapy. RESULTS There were 9, 25, and 8 patients with stage II, III and IV disease, respectively. All except two patients received 50-70 Gy radiation; 37 patients (88.1%) received more than five nimotuzumab doses. Grade III toxicities (21.4% of all adverse events) included esophagitis and gastrointestinal, dermatological and hematological toxicities. Complete response, partial response, stable disease, and progressive disease were observed in 0, 22 (52.4%), 17 (40.5%) and 3 (7.1%) patients at 1 month after the treatment. The epidermal growth factor receptor (EGFR) overexpression rate was 95.2%. After a median follow-up of 37 months, the median survival time (MST) was 14 months. The 2 year and 3 year overall survival (OS) rates were 33.3% and 26.2%, respectively. The median progression-free survival (PFS) time was 10 months. The 2 year and 3 year PFS rates were 24.5% and 22.1%, respectively. The MST in the 13 patients with (+++) EGFR expression (group A) and 7 patients with (++) EGFR expression (group B) was 15 and 11 months, respectively. The 2 year and 3 year OS rates were 46.2% and 38.5% in group A and 28.6% and 28.6% in group B, respectively (P = 0.405). CONCLUSION Although concurrent chemoradiotherapy was the standard care for locally advanced esophageal cancer, radiotherapy was the choice for those who were refused or could not tolerate chemoradiotherapy. Our study shows that nimotuzumab combined with radiotherapy was well tolerated in patients with esophageal cancer. EGFR overexpression was more common than previously reported. OS was higher after combined therapy than after historical control radiotherapy alone. Further studies are required to confirm the therapeutic efficacy of nimotuzumab in esophageal cancer.
Collapse
Affiliation(s)
- Jun Liang
- Department of Radiotherapy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Mingyan E
- Department of Radiotherapy, Cancer Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Gang Wu
- Department of Radiotherapy, Tongji Cancer Center Hospital, Wuhan, People’s Republic of China
| | - Lujun Zhao
- Department of Radiotherapy, Cancer Hospital of Tianjin Medical University, Tianjin, People’s Republic of China
| | - Xia Li
- Department of Radiotherapy, Liaoning Province Cancer Hospital, Shenyang, People’s Republic of China
| | - Xia Xiu
- Department of Radiotherapy, Beijing Hospital, Beijing, People’s Republic of China
| | - Ning Li
- Department of Radiotherapy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Bo Chen
- Department of Radiotherapy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Zhouguang Hui
- Department of Radiotherapy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Jima Lv
- Department of Radiotherapy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Hui Fang
- Department of Radiotherapy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Yu Tang
- Department of Radiotherapy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Nan Bi
- Department of Radiotherapy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Wenqing Wang
- Department of Radiotherapy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Yirui Zhai
- Department of Radiotherapy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Tao Li
- Department of Radiotherapy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Dongfu Chen
- Department of Radiotherapy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Shuangmei Zou
- Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | - Ning Lu
- Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| | | | - Junqi Zheng
- School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Luhua Wang
- Department of Radiotherapy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, People’s Republic of China
| |
Collapse
|
37
|
Wijnhoven BPL, Toxopeus ELA, Vallböhmer D, Knoefel WT, Krasna MJ, Perez K, van Rossum PSN, Ruurda JP, van Hillegersberg R, Schiesser M, Schneider P, Felix VN. New therapeutic strategies for squamous cell cancer and adenocarcinoma. Ann N Y Acad Sci 2013; 1300:213-225. [PMID: 24117644 DOI: 10.1111/nyas.12247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This paper presents commentaries on neoadjuvant treatment esophagectomy; the prognostic and predictive effects of single nucleotide polymorphisms (SNP) in the multimodality therapy of esophageal cancer; optimal preoperative treatment prior to surgery for esophageal cancer; a possible role for trastuzumab in treating esophageal adenocarcinoma or any esophageal dysplasia/intra-epithelial neoplasia; surgery after chemoradiation in resectable esophageal cancer; whether para-aortic lymph node dissection should be performed in esophagogastric junction (EGJ) tumors; and transhiatal esophagectomy in treatment of the esophageal cancer.
Collapse
Affiliation(s)
- Bas P L Wijnhoven
- Department of Surgery, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Eelke L A Toxopeus
- Department of Surgery, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Daniel Vallböhmer
- Department of General, Visceral and Pediatric Surgery, University of Düsseldorf, Düsseldorf, Germany
| | - Wolfram T Knoefel
- Department of General, Visceral and Pediatric Surgery, University of Düsseldorf, Düsseldorf, Germany
| | - Mark J Krasna
- Jersey Shore University Medical Center, Neptune, New Jersey
| | - Kimberly Perez
- Division of Hematology - Oncology, Warren Alpert School of Medicine, Brown University, Providence, Rhode Island
| | - Peter S N van Rossum
- Departments of Surgery and Radiotherapy, University Medical Center, Utrecht, The Netherlands
| | - Jelle P Ruurda
- Departments of Surgery and Radiotherapy, University Medical Center, Utrecht, The Netherlands
| | | | - Marc Schiesser
- Department of Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Paul Schneider
- Department of Surgery, University Hospital Zurich, Zurich, Switzerland
| | | |
Collapse
|
38
|
Meng X, Wang J, Sun X, Wang L, Ye M, Feng P, Zhu G, Lu Y, Han C, Zhu S, Liao Z, Yu J. Cetuximab in combination with chemoradiotherapy in Chinese patients with non-resectable, locally advanced esophageal squamous cell carcinoma: a prospective, multicenter phase II trail. Radiother Oncol 2013; 109:275-80. [PMID: 24128808 DOI: 10.1016/j.radonc.2013.09.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 08/29/2013] [Accepted: 09/01/2013] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND PURPOSE This multicenter phase II trial investigated cetuximab combined with chemoradiotherapy in patients with esophageal squamous cell carcinoma (ESCC). MATERIAL AND METHODS Eligible patients with non-resectable, locally-advanced ESCC received cetuximab 400mg/m(2) loading dose on day 1; and on day 1 of the 2nd-7th weeks: cetuximab 250mg/m(2), paclitaxel 45mg/m(2), and cisplatin 20mg/m(2), concurrent with 59.4Gy/33 fractions of radiation therapy. Primary endpoint was clinical response rate. Secondary endpoints included overall survival (OS), progression-free survival (PFS), safety, and KRAS status. RESULTS Of 55 patients enrolled, 45 completed therapy. Forty-four patients had a clinical response: 29 complete response and 15 partial response. One-year PFS and OS of 45 evaluable patients were 84.23% and 93.33%, respectively, and 2-year PFS and OS were 74.87% and 80.00%, respectively. Non-hematologic adverse events were generally grade 1 or 2; primarily rash (92.7%), mucositis (45.5%), fatigue (41.8%), and nausea (38.2%). Grade 3 hematologic adverse events included neutropenia (32.7%) and anemia (1.8%). No KRAS mutations were identified in 50 evaluated samples. CONCLUSIONS Cetuximab can be safely administered with chemoradiotherapy to patients with locally-advanced ESCC and may improve clinical response rate.
Collapse
Affiliation(s)
- Xue Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Nagaraja V, Eslick GD. Advances in biomarkers for esophageal cancer. Expert Rev Anticancer Ther 2013; 13:1169-1180. [PMID: 24134419 DOI: 10.1586/14737140.2013.844953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
|
40
|
Ree AH, Hollywood D. Design and conduct of early-phase radiotherapy trials with targeted therapeutics: lessons from the PRAVO experience. Radiother Oncol 2013; 108:3-16. [PMID: 23830196 DOI: 10.1016/j.radonc.2013.06.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 05/28/2013] [Accepted: 06/10/2013] [Indexed: 12/28/2022]
Abstract
New strategies to facilitate the improvement of physical and integrated biological optimization of high-precision treatment protocols are an important priority for modern radiation oncology. From a clinical perspective, as knowledge accumulates from molecular radiobiology, there is a complex and exciting opportunity to investigate novel approaches to rational patient treatment stratification based on actionable tumor targets, together with the appropriate design of next-generation early-phase radiotherapy trials utilizing targeted therapeutics, to formally evaluate relevant clinical and biomarker endpoints. A unique aspect in the development pathway of systemic agents with presumed radiosensitizing activity will also be the need for special attention on patient eligibility and the rigorous definition of radiation dose-volume relationships and potential dose-limiting toxicities. Based on recent experience from systematically investigating histone deacetylase inhibitors as radiosensitizing agents, from initial studies in preclinical tumor models through the conduct of a phase I clinical study to evaluate tumor activity of the targeted agent as well as patient safety and tumor response to the combined treatment modality, this communication will summarize principles relating to early clinical evaluation of combining radiotherapy and targeted therapeutics.
Collapse
Affiliation(s)
- Anne Hansen Ree
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway.
| | | |
Collapse
|
41
|
Guo XF, Zhu XF, Zhong GS, Deng BG. Lapatinib, a dual inhibitor of epidermal growth factor receptor and human epidermal growth factor receptor 2, potentiates the antitumor effects of cisplatin on esophageal carcinoma. Dis Esophagus 2013; 26:487-95. [PMID: 22458639 DOI: 10.1111/j.1442-2050.2012.01332.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2) overexpression occurs in over 30% of esophageal carcinomas. Combination therapies of EGFR- and HER2-targeting agents with cytotoxic agents are considered a potential therapeutic strategy for esophageal cancer. The antitumor effects of lapatinib, a dual tyrosine kinase inhibitor of EGFR and HER2, cisplatin alone, and the combination of the two drugs on esophageal cancer cells were evaluated. The growth inhibition activity of lapatinib, cisplatin, and lapatinib plus cisplatin was measured by 3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide (MTT) assays, and the combination index values were calculated. Additionally, cell cycle distribution and cell apoptosis treated with lapatinib or cisplatin alone and the combination of the two drugs were detected by flow cytometry analysis. The activation of EGFR and HER2 signaling pathways was monitored by Western blot analysis. These experimental data showed that the combination of lapatinib and cisplatin synergistically inhibited cell proliferation and exhibited an enhanced pro-apoptotic effect on esophageal cancer cells. The underlying mechanisms of potentiated effects of combined treatment were associated with reduced phosphorylation of EGFR and HER2, and the downstream signaling molecules AKT and extracellular regulated protein kinases (ERK). Our findings indicated that the combination of lapatinib and cisplatin is one of the promising treatment strategies for esophageal carcinomas with EGFR and HER2 overexpression.
Collapse
Affiliation(s)
- X F Guo
- Xinxiang Medical University, Xinxiang The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, China.
| | | | | | | |
Collapse
|
42
|
Toomey PG, Vohra NA, Ghansah T, Sarnaik AA, Pilon-Thomas SA. Immunotherapy for gastrointestinal malignancies. Cancer Control 2013; 20:32-42. [PMID: 23302905 DOI: 10.1177/107327481302000106] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Gastrointestinal (GI) cancers are the most common human tumors encountered worldwide. The majority of GI cancers are unresectable at the time of diagnosis, and in the subset of patients undergoing resection, few are cured. There is only a modest improvement in survival with the addition of modalities such as chemotherapy and radiation therapy. Due to an increasing global cancer burden, it is imperative to integrate alternative strategies to improve outcomes. It is well known that cancers possess diverse strategies to evade immune detection and destruction. This has led to the incorporation of various immunotherapeutic strategies, which enable reprogramming of the immune system to allow effective recognition and killing of GI tumors. METHODS A review was conducted of the results of published clinical trials employing immunotherapy for esophageal, gastroesophageal, gastric, hepatocellular, pancreatic, and colorectal cancers. RESULTS Monoclonal antibody therapy has come to the forefront in the past decade for the treatment of colorectal cancer. Immunotherapeutic successes in solid cancers such as melanoma and prostate cancer have led to the active investigation of immunotherapy for GI malignancies, with some promising results. CONCLUSIONS To date, monoclonal antibody therapy is the only immunotherapy approved by the US Food and Drug Administration for GI cancers. Initial trials validating new immunotherapeutic approaches, including vaccination-based and adoptive cell therapy strategies, for GI malignancies have demonstrated safety and the induction of antitumor immune responses. Therefore, immunotherapy is at the forefront of neoadjuvant as well as adjuvant therapies for the treatment and eradication of GI malignancies.
Collapse
Affiliation(s)
- Paul G Toomey
- Department of Surgery, USF Health Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | | | | | | |
Collapse
|
43
|
Hong TS, Wo JY, Kwak EL. Targeted therapies with chemoradiation in esophageal cancer: development and future directions. Semin Radiat Oncol 2013. [PMID: 23207045 DOI: 10.1016/j.semradonc.2012.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Targeted therapies have been studied in combination with chemoradiation therapy for esophageal and gastroesophageal junction cancers. To date, the Human Epidermal Growth Factor Receptor 2 (HER2) HER2, epidermal growth factor receptor, and vascular endothelial growth factor pathways have been most extensively studied in esophagogastric cancers. However, despite strong preclinical rationale and evidence of activity in other diseases, the efficacy of these agents has been mixed when studied in the metastatic setting or in combination with chemoradiation. In this review, the history of the development of targeted therapies in esophagogastric cancers is reviewed, with a specific focus on the incorporation of these agents with chemoradiation. Additionally, more recently identified targets of interest in esophagogastric cancers, including c-MET, heat shock protein 90, and Sonic hedgehog, are discussed in the context of chemoradiation.
Collapse
Affiliation(s)
- Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | | | | |
Collapse
|
44
|
Prins M, Ruurda J, van Diest P, van Hillegersberg R, ten Kate F. The significance of the HER-2 status in esophageal adenocarcinoma for survival: an immunohistochemical and an in situ hybridization study. Ann Oncol 2013; 24:1290-7. [DOI: 10.1093/annonc/mds640] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
45
|
Pretz JL, Wo JY, Mamon HJ, Kachnic LA, Hong TS. Chemoradiation therapy: localized esophageal, gastric, and pancreatic cancer. Surg Oncol Clin N Am 2013; 22:511-24. [PMID: 23622077 DOI: 10.1016/j.soc.2013.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chemoradiation plays an important role in management of locally advanced gastrointestinal tumors. This article reviews data regarding chemoradiation for tumors of the upper gastrointestinal tract. For esophageal and gastroesophageal junction cancers, chemoradiation is standard of care in the preoperative setting. In gastric cancer, 2 standards have emerged: definitively treating with perioperative chemotherapy alone and using chemoradiation postoperatively. For pancreatic cancer, the benefit of radiation is less well defined. The future of treatment sites lies in trials evaluating new chemotherapy regimens, alternative systemic therapies, and different radiation fractionation schema. Because care of these patients is complex, multimodality team evaluation before treatment is encouraged.
Collapse
Affiliation(s)
- Jennifer L Pretz
- Harvard Radiation Oncology Program, Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
46
|
Cidon EU, Ellis SG, Inam Y, Adeleke S, Zarif S, Geldart T. Molecular targeted agents for gastric cancer: a step forward towards personalized therapy. Cancers (Basel) 2013; 5:64-91. [PMID: 24216699 PMCID: PMC3730303 DOI: 10.3390/cancers5010064] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/01/2013] [Accepted: 01/14/2013] [Indexed: 12/19/2022] Open
Abstract
Gastric cancer (GC) represents a major cancer burden worldwide, and remains the second leading cause of cancer-related death. Due to its insidious nature, presentation is usually late and often carries a poor prognosis. Despite having improved treatment modalities over the last decade, for most patients only modest improvements have been seen in overall survival. Recent progress in understanding the molecular biology of GC and its signaling pathways, offers the hope of clinically significant promising advances for selected groups of patients. Patients with Her-2 overexpression or amplification have experienced benefit from the integration of monoclonal antibodies such as trastuzumab to the standard chemotherapy. Additionally, drugs targeting angiogenesis (bevacizumab, sorafenib, sunitinib) are under investigation and other targeted agents such as mTOR inhibitors, anti c-MET, polo-like kinase 1 inhibitors are in preclinical or early clinical development. Patient selection and the development of reliable biomarkers to accurately select patients most likely to benefit from these tailored therapies is now key. Future trials should focus on these advances to optimize the treatment for GC patients. This article will review recent progress and current status of targeted agents in GC.
Collapse
Affiliation(s)
- Esther Una Cidon
- Medical Oncology Department, The Royal Bournemouth and Christchurch Hospitals NHS Foundation Trust, Castle Lane East, BH7 7DW Bournemouth, Dorset, UK.
| | | | | | | | | | | |
Collapse
|
47
|
Yin YM, Zhou Y, Shao YF. Targeted therapies for esophageal cancer. Shijie Huaren Xiaohua Zazhi 2012; 20:3499-3504. [DOI: 10.11569/wcjd.v20.i35.3499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Esophageal cancer is a highly frequent malignancy, being the fourth leading cause of cancer-related deaths in China. Most patients are initially diagnosed with advanced disease. Despite recent advances in surgical techniques and adjuvant/neoadjuvant radiotherapy and chemotherapy, the prognosis of esophageal cancer is still poor. An emerging understanding of molecular pathways that characterizes cell growth, cell cycle, apoptosis, angiogenesis, and invasion has provided novel targets for cancer therapy. This review focuses on novel targeted treatments for esophageal cancer.
Collapse
|
48
|
Villaflor VM, Allaix ME, Minsky B, Herbella FA, Patti MG. Multidisciplinary approach for patients with esophageal cancer. World J Gastroenterol 2012; 18:6737-46. [PMID: 23239911 PMCID: PMC3520162 DOI: 10.3748/wjg.v18.i46.6737] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 11/19/2012] [Accepted: 11/24/2012] [Indexed: 02/06/2023] Open
Abstract
Patients with esophageal cancer have a poor prognosis because they often have no symptoms until their disease is advanced. There are no screening recommendations for patients unless they have Barrett's esophagitis or a significant family history of this disease. Often, esophageal cancer is not diagnosed until patients present with dysphagia, odynophagia, anemia or weight loss. When symptoms occur, the stage is often stage III or greater. Treatment of patients with very early stage disease is fairly straight forward using only local treatment with surgical resection or endoscopic mucosal resection. The treatment of patients who have locally advanced esophageal cancer is more complex and controversial. Despite multiple trials, treatment recommendations are still unclear due to conflicting data. Sadly, much of our data is difficult to interpret due to many of the trials done have included very heterogeneous groups of patients both histologically as well as anatomically. Additionally, studies have been underpowered or stopped early due to poor accrual. In the United States, concurrent chemoradiotherapy prior to surgical resection has been accepted by many as standard of care in the locally advanced patient. Patients who have metastatic disease are treated palliatively. The aim of this article is to describe the multidisciplinary approach used by an established team at a single high volume center for esophageal cancer, and to review the literature which guides our treatment recommendations.
Collapse
|
49
|
Cooper SL, Russo JK, Chin S. Definitive chemoradiotherapy for esophageal carcinoma. Surg Clin North Am 2012; 92:1213-48. [PMID: 23026279 DOI: 10.1016/j.suc.2012.07.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Radiation therapy plays an important role in the treatment of esophageal cancer. Radiation therapy may be combined with chemotherapy, used as a component of induction therapy, used in the adjuvant setting, or used for palliation of advanced disease. Chemotherapy is also occasionally used as a solitary treatment modality for patients with esophageal cancer. Current treatment protocols include multiple agents, and agents directed against specific molecular targets have been investigated in clinical trials. This article discusses future directions related to the selection of radiation treatment protocols, novel targeted chemotherapeutic agents, and the selection of patients for surgery.
Collapse
Affiliation(s)
- S Lewis Cooper
- Department of Radiation Oncology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | |
Collapse
|
50
|
Al-Momani H, Barnes R, El-Hadi A, Shah R, Lewis WG, Edwards P. Human epidermal growth factor receptor-2 in oesophageal cancers: An observational study. World J Gastroenterol 2012; 18:6447-6451. [PMID: 23197890 PMCID: PMC3508639 DOI: 10.3748/wjg.v18.i44.6447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine the incidence of human epidermal growth factor receptor 2 (HER2) over expression in oesophageal cancers.
METHODS: A retrospective study, of one hundred consecutive cases of endoscopic histological samples of oesophageal cancers from a single British cancer network were included. Cancer cases were diagnosed between April 2007 and June 2010. HER2 over expression was assessed using immunohistochemistry, those that scored “0” and “+1” were considered “negative” for HER2; those that scored “+3” were considered “Positive”. Cases that were scored “+2” on immunohistochemistry further went on to have HER2 gene analysis using the Ventana HER brightfield dual-colour in situ hybridisations (HER B DISH) assay and either came back to be positive or negative for HER2 over expression. Overall survival was measured from date of histological diagnosis until date of death. 93% of the cases were followed up till five years or death, and all were followed up till two years. Cases of gastro-oesophageal junctional tumours were excluded.
RESULTS: The median age of our sample was 66 years (range: 38-91 years). Eighty one were male and 19 female. Ninety-one of the cases were adenocarcinoma of the oesophagus and the rest were cases of squamous cell carcinoma. The anatomical distribution of the tumours was; upper oesophagus 2, middle oesophagus 11, and 87 were in the lower oesophagus. Operative resection was completed in 15 cases; seven cases had attempted surgical resections, i.e., open and close, 33 patients received definitive chemo-radiation and 52 had palliative treatment. Twenty-five of the cancers showed evidence of HER2 over expression, all were adenocarcinomas. Of the 25 cases that showed evidence of HER2 over expression, 21 (84%) were located in the lower third of the oesophagus. On staging, 24 out of the 25 HER2 positive cases were at stage 3 or more (13 at stage 3 and 11 at stage 4), For HER2 negative cases 37 were at stage 3 and 32 were staged as stage 4. Seventeen out of twenty five cases (68%) with HER2 over expression received palliative therapy, in comparison to thirty five out of seventy five (46.7%) in tumours not expressing HER2. No significant difference in overall survival was demonstrated between patients whose cancers showed evidence of HER2 over expression and those who did not; median overall survival for HER2 positive tumours was 15 mo (95%CI, 11-19 mo) compared to 13 mo (95%CI, 9-17 mo) for HER2 negative ones. Two years cumulative survival for cases with HER2 over expression was 33.7% compared to 31.6% in cases without HER2 over expression (P = 0.576). Only cancer’s stage significantly affected overall survival on both univariant and multivariable analysis (P = 0.034 and P = 0.009 respectively). None of the patients included in this study received Trastuzumab.
CONCLUSION: Twenty-seven point five percent of oesophageal adenocarcinomas showed evidence of HER2 over expression. Routine testing for human HER2 in oesophageal adenocarcinomas can have significant implication on treatments offered to patients that may potentially affect their prognosis.
Collapse
|