1
|
Qureshi Z, Jamil A, Fatima E, Altaf F, Siddique R, Shah S. Pembrolizumab in combination with trastuzumab for treatment of HER2-positive advanced gastric or gastro-esophageal junction cancer. Ann Med Surg (Lond) 2024; 86:4647-4656. [PMID: 39118760 PMCID: PMC11305801 DOI: 10.1097/ms9.0000000000002305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/14/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Gastric cancer remains a challenging malignancy with a high global mortality rate. Recent advances in targeted therapy and immunotherapy have shown promise in improving patient outcomes. This paper reviews the impact of incorporating targeted agents such as trastuzumab and immunotherapeutic agents like pembrolizumab into standard chemotherapy regimens for gastric cancer treatment. Methods A comprehensive analysis was conducted on pivotal clinical trials, including KEYNOTE-590, KEYNOTE-811, and ToGA, focusing on their methodologies, patient populations, treatment regimens, and outcome measures. The review also explored emerging research avenues in precision medicine, particularly genomic sequencing and biomarker identification. Aim To assess the efficacy and survival benefits of adding trastuzumab and pembrolizumab to standard chemotherapy in the treatment of gastric cancer and to outline future directions in gastric cancer research. Results Including trastuzumab and pembrolizumab in treatment regimens for human epidermal growth factor receptor 2 (HER2)-positive and PD-L1-expressing gastric cancers significantly improved progression-free and overall survival rates compared to chemotherapy alone. These findings highlight the potential of personalized therapy in enhancing treatment outcomes. Furthermore, ongoing research into the gastric cancer microenvironment and the role of the microbiome suggests novel targets for future therapeutic interventions. Conclusion The integration of targeted and immunotherapeutic agents with traditional chemotherapy represents a pivotal shift in gastric cancer treatment, moving towards more personalized and effective regimens.
Collapse
Affiliation(s)
- Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, CT
| | - Abdur Jamil
- Department of Medicine, Samaritan Medical Centre, Watertown, NY
| | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY, USA
| | | | - Shivendra Shah
- Department of Medicine, Nepalgunj Medical College, Chisapani, Nepal
| |
Collapse
|
2
|
Wang T, Chen S, Wang Z, Li S, Fei X, Wang T, Zhang M. KIRREL promotes the proliferation of gastric cancer cells and angiogenesis through the PI3K/AKT/mTOR pathway. J Cell Mol Med 2024; 28:e18020. [PMID: 37909722 PMCID: PMC10805501 DOI: 10.1111/jcmm.18020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/14/2023] [Accepted: 10/19/2023] [Indexed: 11/03/2023] Open
Abstract
Anti-angiogenesis is a promising therapeutic strategy for delaying tumour progression that offers, new hope for gastric cancer targeted therapy. The purpose of this study was to investigate the precise mechanism by which Kin of IRRE-like protein 1 (KIRREL) contributes to the development of gastric cancer, particularly in terms of tumour angiogenesis. Differential expression of KIRREL in tissues and cells was detected using quantitative real-time polymerase chain reaction, western blotting and immunohistochemistry. A bioinformatics analysis was conducted to screen for the function and pathway enrichment of KIRREL in gastric cancer. Lentivirus-induced KIRREL silencing in SNU-5 cells and lentivirus-induced KIRREL overexpression in AGS cells were used to study the effect of KIRREL on the proliferation, cell cycle and angiogenesis of gastric cancer cells. Moreover, the expressions of PI3K, P-PI3K, AKT, P-AKT, mTOR, P-mTOR, HIF-1α and VEGF were also detected. Gastric cancer tissues and cells had high levels of KIRREL expression, which is associated with the proliferation, cell cycle and angiogenesis of gastric cancer cells. After silencing and overexpressing KIRREL in SNU-5 and AGS cells, respectively, the proliferation and angiogenesis of SNU-5 cells were inhibited, while the proliferation and angiogenesis of AGS cells were promoted. According to a bioinformatics analysis of the KIRREL gene, angiogenesis regulation and the PI3K/AKT pathway were highly connected. The PI3K/AKT/mTOR pathway was repressed and stimulated by KIRREL silencing and overexpression, respectively. IGF-1, an AKT agonist, and LY294002, an inhibitor, reversed the effects of KIRREL silencing and overexpression on the PI3K/AKT/mTOR pathway and on gastric cancer cell proliferation and angiogenesis. KIRREL may mediate the proliferation and angiogenesis of gastric cancer cells through the PI3K/AKT/mTOR signalling pathway. These findings could help in the further development of potential anti-angiogenesis targets.
Collapse
Affiliation(s)
- Tao Wang
- Department of OncologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Shuo Chen
- Department of OncologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Ziliang Wang
- Department of OncologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Siyu Li
- Department of OncologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Xichang Fei
- Department of OncologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Tong Wang
- Department of General PracticeThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Mingjun Zhang
- Department of OncologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| |
Collapse
|
3
|
Xu D, Luo Y, Wang P, Li J, Ma L, Huang J, Zhang H, Yang X, Li L, Zheng Y, Fang G, Yan P. Clinical progress of anti-angiogenic targeted therapy and combination therapy for gastric cancer. Front Oncol 2023; 13:1148131. [PMID: 37384288 PMCID: PMC10295723 DOI: 10.3389/fonc.2023.1148131] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/12/2023] [Indexed: 06/30/2023] Open
Abstract
The incidence of gastric cancer is increasing year by year. Most gastric cancers are already in the advanced stage with poor prognosis when diagnosed, which means the current treatment is not satisfactory. Angiogenesis is an important link in the occurrence and development of tumors, and there are multiple anti-angiogenesis targeted therapies. To comprehensively evaluate the efficacy and safety of anti-angiogenic targeted drugs alone and in combination against gastric cancer, we systematically searched and sorted out relevant literature. In this review, we summarized the efficacy and safety of Ramucirumab, Bevacizumab, Apatinib, Fruquintinib, Sorafenib, Sunitinib, Pazopanib on gastric cancer when used alone or in combination based on prospective clinical trials reported in the literature, and sorted response biomarkers. We also summarized the challenges faced by anti-angiogenesis therapy for gastric cancer and available solutions. Finally, the characteristics of the current clinical research are summarized and suggestions and prospects are raised. This review will serve as a good reference for the clinical research of anti-angiogenic targeted drugs in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Donghan Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Yehao Luo
- School of Second Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Wang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Jiaxin Li
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Linrui Ma
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Jie Huang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Hao Zhang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Xiaoman Yang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Liqi Li
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Yuhong Zheng
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Gang Fang
- Guangxi Key Laboratory of Applied Fundamental Research of Zhuang Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Peiyu Yan
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, Macau University of Science and Technology, Macao, Macao SAR, China
| |
Collapse
|
4
|
Haque E, Esmail A, Muhsen I, Salah H, Abdelrahim M. Recent Trends and Advancements in the Diagnosis and Management of Gastric Cancer. Cancers (Basel) 2022; 14:5615. [PMID: 36428707 PMCID: PMC9688354 DOI: 10.3390/cancers14225615] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022] Open
Abstract
Gastric cancer is an enigmatic malignancy that has recently been shown to be increasing in incidence globally. There has been recent progress in emerging technologies for the diagnosis and treatment of the disease. Improvements in non-invasive diagnostic techniques with serological tests and biomarkers have led to decreased use of invasive procedures such as endoscopy. A multidisciplinary approach is used to treat gastric cancer, with recent significant advancements in systemic therapies used in combination with cytotoxic chemotherapies. New therapeutic targets have been identified and clinical trials are taking place to assess their efficacy and safety. In this review, we provide an overview of the current and emerging treatment strategies and diagnostic techniques for gastric cancer.
Collapse
Affiliation(s)
- Emaan Haque
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Abdullah Esmail
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston, TX 77030, USA
| | - Ibrahim Muhsen
- Section of Hematology and Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haneen Salah
- Department of Pathology, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Maen Abdelrahim
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston, TX 77030, USA
- Cockrell Center for Advanced Therapeutic Phase I Program, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| |
Collapse
|
5
|
Salati M, Caputo F, Bocconi A, Cerri S, Baldessari C, Piacentini F, Dominici M, Gelsomino F. Successes and failures of angiogenesis blockade in gastric and gastro-esophageal junction adenocarcinoma. Front Oncol 2022; 12:993573. [PMID: 36212393 PMCID: PMC9540203 DOI: 10.3389/fonc.2022.993573] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Gastric and gastro-esophageal junction adenocarcinoma (GEA) remains a considerable major public health problem worldwide, being the fifth most common cancer with a fatality-to-case ratio that stands still at 70%. Angiogenesis, which is a well-established cancer hallmark, exerts a fundamental role in cancer initiation and progression and its targeting has been actively pursued as a promising therapeutic strategy in GEA. A wealth of clinical trials has been conducted, investigating anti-angiogenic agents including VEGF-directed monoclonal antibodies, small molecules tyrosine kinase inhibitors and VEGF-Trap agents both in the resectable and advanced setting, reporting controversial results. While phase III randomized trials testing the anti-VEGFR-2 antibody Ramucirumab and the selective VEGFR-2 tyrosine kinase inhibitor Apatinib demonstrated a significant survival benefit in later lines, the shift of angiogenesis inhibitors in the perioperative and first-line setting failed to improve patients' outcome in GEAs. The molecular landscape of disease, together with novel combinatorial strategies and biomarker-selected approaches are under investigation as key elements to the success of angiogenesis blockade in GEA. In this article, we critically review the existing literature on the biological rationale and clinical development of antiangiogenic agents in GEA, discussing major achievements, limitations and future developments, aiming at fully realizing the potential of this therapeutic approach.
Collapse
Affiliation(s)
- Massimiliano Salati
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
- PhD Program Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Caputo
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Alessandro Bocconi
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Sara Cerri
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Cinzia Baldessari
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Federico Piacentini
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Fabio Gelsomino
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| |
Collapse
|
6
|
Novel Biomarkers of Gastric Adenocarcinoma: Current Research and Future Perspectives. Cancers (Basel) 2021; 13:cancers13225660. [PMID: 34830815 PMCID: PMC8616337 DOI: 10.3390/cancers13225660] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Gastric cancer is characterized by poor survival rates despite surgery and chemotherapy. Current research focuses on biomarkers to improve diagnosis and prognosis, and to enable targeted treatment strategies. The aim of our review was to give an overview over the wide range of novel biomarkers in gastric cancer. These biomarkers are targets of a specific treatment, such as antibodies against human epidermal growth factor receptor 2. Other promising biomarkers for targeted therapies that have shown relevance in clinical trials are vascular endothelial growth factor, programmed cell death protein 1, and Claudin 18.2. There is a vast number of biomarkers based on DNA, RNA, and protein expression, as well as detection of circulating tumor cells and the immune tumor microenvironment. Abstract Overall survival of gastric cancer remains low, as patients are often diagnosed with advanced stage disease. In this review, we give an overview of current research on biomarkers in gastric cancer and their implementation in treatment strategies. The HER2-targeting trastuzumab is the first molecular targeted agent approved for gastric cancer treatment. Other promising biomarkers for targeted therapies that have shown relevance in clinical trials are VEGF and Claudin 18.2. Expression of MET has been shown to be a negative prognostic factor in gastric cancer. Targeting the PD-1/PD-L1 pathway with immune checkpoint inhibitors has proven efficacy in advanced gastric cancer. Recent technology advances allow the detection of circulating tumor cells that may be used as diagnostic and prognostic indicators and for therapy monitoring in gastric cancer patients. Prognostic molecular subtypes of gastric cancer have been identified using genomic data. In addition, transcriptome profiling has allowed a comprehensive characterization of the immune and stromal microenvironment in gastric cancer and development of novel risk scores. These prognostic and predictive markers highlight the rapidly evolving field of research in gastric cancer, promising improved treatment stratification and identification of molecular targets for individualized treatment in gastric cancer.
Collapse
|
7
|
Högner A, Al-Batran SE, Siveke JT, Lorenz M, Bartels P, Breithaupt K, Malfertheiner P, Homann N, Stein A, Gläser D, Tamm I, Hinke A, Vogel A, Thuss-Patience P. Pazopanib with 5-FU and oxaliplatin as first line therapy in advanced gastric cancer: A randomized phase-II study-The PaFLO trial. A study of the Arbeitsgemeinschaft Internistische Onkologie AIO-STO-0510. Int J Cancer 2021; 150:1007-1017. [PMID: 34741530 DOI: 10.1002/ijc.33864] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/24/2021] [Accepted: 10/08/2021] [Indexed: 12/30/2022]
Abstract
VEGF inhibition in gastric cancer has a proven benefit in the second line setting. Pazopanib, an oral tyrosine kinase inhibitor, selectively inhibits VEGFR-1, -2 and -3, c-kit and PDGF-R resulting in inhibition of angiogenesis. This open-label randomized phase II trial (2:1) investigated the efficacy of combining pazopanib with FLO (5-fluorouracil, oxaliplatin) vs FLO alone (internal control arm) as first-line treatment in patients with advanced adenocarcinoma of the stomach and gastroesophageal junction (GEJ). Eighty-seven patients were randomized and 78 patients were eligible and evaluable (PaFLO arm 51 patients, FLO arm 27 patients). The PFS rate at 6 months (primary endpoint) was 34% in the PaFLO arm vs 30% in the FLO arm. Comparing PaFLO with FLO median PFS was 4.66 months (95% confidence interval [CI] 2.87-6.46) vs 4.47 months (95% CI 1.79-7.14) (95% CI, hazard ratio [HR] 0.96 (0.60-1.55), P = .882 [exploratory]); median OS was 10.19 months (95% CI 5.46-14.92) vs 7.33 months (95% CI 4.93-9.73), (95% CI HR 1.01 [0.62-1.65], P = .953, exploratory), disease control rate was 72% vs 59%. PaFLO was well tolerable, toxicities were slightly higher in the PaFLO arm. Major adverse events were loss of appetite, nausea, fatigue, diarrhea, neutropenia and thrombocytopenia. Adding pazopanib to chemotherapy shows signs of efficacy but no major improvement in this randomized phase 2 trial. The PFS at 6 months in both arms was lower than expected from the literature. Biomarkers identifying subgroups who benefit and novel combinations are needed. ClinicalTrials.gov: NCT01503372.
Collapse
Affiliation(s)
- Anica Högner
- Medizinische Klinik m.S. Hämatologie, Onkologie und Tumorimmunologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Salah-Eddin Al-Batran
- Institut für Klinische Krebsforschung IKF GmbH am Krankenhaus Nordwest, Frankfurt, Germany
| | - Jens T Siveke
- II. Medizinische Klinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Brückeninstitut für Experimentelle Tumortherapie, Westdeutsches Tumorzentrum, Universitätsklinikum Essen, Essen, Germany.,Abteilung für Translationale Onkologie Solider Tumore, Deutsches Konsortium für Translationale Krebsforschung (DKTK), Parnterstandort Essen, Germany.,Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Mario Lorenz
- Medizinische Klinik m.S. Hämatologie, Onkologie und Tumorimmunologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Prisca Bartels
- Medizinische Klinik m.S. Hämatologie, Onkologie und Tumorimmunologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Kirstin Breithaupt
- Medizinische Klinik m.S. Hämatologie, Onkologie und Tumorimmunologie, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Palliativnetz Travebogen gGmbH, Lübeck, Germany
| | - Peter Malfertheiner
- Klinik für Gastroenterologie, Hepatologie und Infektiologie, Otto-von-Guericke-Universität, Magdeburg, Germany
| | - Nils Homann
- Medizinische Klinik II, Klinikum Wolfsburg, Wolfsburg, Germany
| | - Alexander Stein
- Medizinische Klinik II, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Dietrich Gläser
- Klinik für Innere Medizin III, Klinikum Südstadt Rostock, Rostock, Germany
| | - Ingo Tamm
- Onkologische Schwerpunktpraxis Kurfürstendamm, Berlin, Germany
| | - Axel Hinke
- CCRC Cancer Clinical Research Consulting, Düsseldorf, Germany
| | - Arndt Vogel
- Medizinische Hochschule Hannover, Klinik für Gastroenterologie, Hepatologie und Endokrinologie, Hannover, Germany
| | - Peter Thuss-Patience
- Medizinische Klinik m.S. Hämatologie, Onkologie und Tumorimmunologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
8
|
Hsu A, Chudasama R, Almhanna K, Raufi A. Targeted therapies for gastroesophageal cancers. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1104. [PMID: 33145323 PMCID: PMC7576008 DOI: 10.21037/atm-20-3265] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022]
Abstract
Gastroesophageal cancers are some of the most common malignancies worldwide. A significant portion of patients are diagnosed with advanced or metastatic disease given the insidious nature of gastroesophageal cancers. In the instance where surgical resection for cure is no longer an option, the prognosis is poor and generally less than a year. Traditionally, standard front-line chemotherapy included two- to three-drug regimens with modest improvements in overall survival. Over the past two decades, with increased understanding of the biology of cancer, targeted therapies have been developed to stop the actions of molecules that are key in the growth and spread of cancer cells and have been successful in a number of cancers. In gastroesophageal cancer, these gains have been more modest with limited approval-trastuzumab being incorporated into front-line use in HER2-positive disease, and ramucirumab alone or in combination with paclitaxel becoming the preferred second-line regimen in progressive disease. However, with increased understanding of the biology of cancer, new and promising targeted therapies have emerged along with novel strategies in combining targeted therapies with traditional chemotherapy and immunotherapy. In this article, we will review the use of targeted therapies in the treatment of gastroesophageal cancer and touch upon future treatment strategies and therapeutics currently under investigation.
Collapse
Affiliation(s)
- Andrew Hsu
- Division of Hematology/Oncology, The Warren Alpert Medical School of Brown University, Lifespan Cancer Institute, Rhode Island Hospital, Providence, RI, USA
| | - Rani Chudasama
- Division of Hematology/Oncology, The Warren Alpert Medical School of Brown University, Lifespan Cancer Institute, Rhode Island Hospital, Providence, RI, USA
| | - Khaldoun Almhanna
- Division of Hematology/Oncology, The Warren Alpert Medical School of Brown University, Lifespan Cancer Institute, Rhode Island Hospital, Providence, RI, USA
| | - Alexander Raufi
- Division of Hematology/Oncology, The Warren Alpert Medical School of Brown University, Lifespan Cancer Institute, Rhode Island Hospital, Providence, RI, USA
| |
Collapse
|
9
|
Nie S, Yang G, Lu H. Current Molecular Targeted Agents for Advanced Gastric Cancer. Onco Targets Ther 2020; 13:4075-4088. [PMID: 32494161 PMCID: PMC7229784 DOI: 10.2147/ott.s246412] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 04/20/2020] [Indexed: 12/26/2022] Open
Abstract
Gastric cancer is the third leading cause of malignant tumor-related mortality worldwide. Traditional cytotoxic agents prolong the overall survival and progression-free survival of patients with advanced gastric cancer (AGC) compared to that with best supportive care. Due to the occurrence of serious adverse drug reactions that result in discontinued treatment, the survival benefit in AGC remains unsatisfactory. Systemic chemotherapy regimens have changed greatly, especially since the introduction of trastuzumab. Nevertheless, HER2 positivity is present in only approximately 20% of tumors. Due to the genetic heterogeneity and complexity of patients, there are many studies in progress that are exploring novel targeted drugs as an alternative to chemotherapy or adjuvant treatment in early-stage, progressive, and advanced gastric cancer. On the basis of the differences in gene expression profiles among patients, searching for specific and sensitive predictive biomarkers is important for identifying patients who will benefit from a specific targeted drug. With the development of targeted therapies and available chemotherapeutic drugs, there is no doubt that, over time, more patients will achieve better survival outcomes. Recently, immune checkpoint blockade has been well developed as a promising anticancer strategy. This review outlines the currently available information on clinically tested molecular targeted drugs and immune checkpoint inhibitors for AGC to provide support for decision-making in clinical practice.
Collapse
Affiliation(s)
- Shanshan Nie
- Center for Clinical Pharmacology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| | - Guoping Yang
- Center for Clinical Pharmacology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| | - Hongwei Lu
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
10
|
Vogl UM, Vormittag L, Winkler T, Kafka A, Weiser-Jasch O, Heinrich B, Roider-Schur S, Andalibi H, Autzinger E, Schima W, Klaus A, Zacherl J, Wimberger GM, Öhler L. Ramucirumab plus paclitaxel or FOLFIRI in platinum-refractory advanced or metastatic gastric or gastroesophageal junction adenocarcinoma-experience at two centres. J Gastrointest Oncol 2020; 11:366-375. [PMID: 32399277 DOI: 10.21037/jgo.2020.03.10] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background Ramucirumab is a VEGFR-2 antibody that has proven to prolong overall survival (OS) in patients with pretreated metastatic gastric/gastrooesophageal junction (GEJ) adenocarcinoma. We present data from patients treated with ramucirumab and paclitaxel or FOLFIRI after failure of at least one platinum- and 5-FU-containing chemotherapy (CHT) regimen. Methods In this retrospective two-center study, 56 patients with metastatic gastric cancer (47%) or adenocarcinoma of the GEJ (53%) were treated with paclitaxel and ramucirumab (n=38) as second-line (75%) or beyond second-line (25%) therapy. FOLFIRI-ramucirumab (FOLFIRI-R) (n=16) was given to patients with a short interval between taxane-based perioperative CHT and occurrence of metastatic disease or to those ineligible for paclitaxel. Results The median progression-free survival (PFS) and OS for patients treated with paclitaxel-ramucirumab (pacl-R) were 2.9 (95% CI: 2.3-3.6) and 4.4 (4.1-4.7) months, respectively, and those for patients treated with FOLFIRI-R were 5.9 (95% CI: 0.35-11.4) and 8.3 (6.6-10) months, respectively (P=0.05). We observed a trend towards prolonged PFS after perioperative taxane-based FLOT CHT (n=12) with FOLFIRI-R compared with pacl-R. Adverse events were manageable, with neutropenia and polyneuropathy (PNP) being the most common events. More than two treatment lines were given to 48.2% of patients. Conclusions The use of ramucirumab in combination with FOLFIRI showed favourable PFS and OS in patients with prior treatments with platinum and/or taxane-based agents and allows further treatment lines after progression. In patients with taxane pretreatment or persistent high-grade PNP, the combination of FOLFIRI-R might be a promising combination.
Collapse
Affiliation(s)
- Ursula M Vogl
- Department of Medicine I, Oncology, Barmherzige Schwestern Krankenhaus Wien, Vienna, Austria
| | - Laurenz Vormittag
- Department of Medicine I, Oncology, St. Josef Krankenhaus, Vienna, Austria
| | - Thomas Winkler
- Department of Medicine I, Oncology, St. Josef Krankenhaus, Vienna, Austria
| | - Alice Kafka
- Department of Medicine I, Oncology, St. Josef Krankenhaus, Vienna, Austria
| | | | - Bettina Heinrich
- Department of Medicine I, Oncology, St. Josef Krankenhaus, Vienna, Austria
| | | | - Haleh Andalibi
- Department of Medicine I, Oncology, Barmherzige Schwestern Krankenhaus Wien, Vienna, Austria
| | - Eva Autzinger
- Department of Medicine I, Oncology, Barmherzige Schwestern Krankenhaus Wien, Vienna, Austria
| | - Wolfgang Schima
- Department of Diagnostic and Interventional Radiology, Barmherzige Schwestern Krankenhaus Wien, St. Josef Krankenhaus and Krankenhaus Göttlicher Heiland, Vienna, Austria
| | - Alexander Klaus
- Department of Surgery, Barmherzige Schwestern Krankenhaus Wien, Vienna, Austria
| | | | | | - Leopold Öhler
- Department of Medicine I, Oncology, Barmherzige Schwestern Krankenhaus Wien, Vienna, Austria.,Department of Medicine I, Oncology, St. Josef Krankenhaus, Vienna, Austria
| |
Collapse
|
11
|
Hironaka S. Anti-angiogenic therapies for gastric cancer. Asia Pac J Clin Oncol 2019; 15:208-217. [PMID: 31111678 DOI: 10.1111/ajco.13174] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 05/07/2019] [Indexed: 12/12/2022]
Abstract
Tumor angiogenesis plays an important role in cancer cell proliferation and metastasis. In gastric cancer, among the numerous clinical trials investigating various anti-angiogenic therapies, such as antivascular endothelial growth factor (VEGF) or anti-VEGF receptor (VEGFR)-2 monoclonal antibodies, VEGF-Trap and VEGFR tyrosine kinase inhibitors, the anti-VEGFR-2 antibody ramucirumab was shown to prolong overall survival not only as a single agent but also in combination with paclitaxel as a second-line chemotherapy. Additionally, apatinib, a selective VEGFR-2 tyrosine kinase inhibitor, prolonged survival as a third-line or later treatment option in patients with advanced gastric cancer. Preliminary results of studies investigating ramucirumab plus immune checkpoint inhibitors in gastric cancer were encouraging, and further investigations are ongoing. In China, apatinib in combination with cytotoxic agents is being investigated for systemic chemotherapy or maintenance therapy as an earlier treatment option. The clinical activity in gastric cancer of the multikinase inhibitor regorafenib was suggested in a randomized phase II study. A global phase III trial comparing regorafenib with placebo is currently ongoing. Further studies of anti-angiogenic therapy combined with not only chemotherapy but also immune checkpoint inhibitors are also being pursued, providing hope for improved survival in patients with gastric cancer.
Collapse
Affiliation(s)
- Shuichi Hironaka
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, Oita, Japan
| |
Collapse
|
12
|
Amerizadeh F, Khazaei M, Maftouh M, Mardani R, Bahrami A. miRNA Targeting Angiogenesis as a Potential Therapeutic Approach in the Treatment of Colorectal Cancers. Curr Pharm Des 2019; 24:4668-4674. [DOI: 10.2174/1381612825666190110161843] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/27/2018] [Accepted: 01/01/2019] [Indexed: 12/11/2022]
Abstract
Angiogenesis refers to the formation of recent blood vessels, which is one of the characteristics of
cancer progression and it has been deliberated as a putative target to the treatment of many kinds of cancers. The
VEGF signaling substrate is very important for angiogenesis and is commonly high-regulated in tumors. As a
result, this molecule has attracted the attention of most of the researchers to develop antiangiogenic therapies. We
have presented that VEGF blockage in neoadjuvant setting via bevacizumab, aflibercept and sunitinib not only
has revealed some promising benefits but also has shown a large negative outcome in the adjuvant trials. However,
at an advanced stage of tumors, suppression of VEGF alone is inadequate to stop advancement, encouraging
drug resistance, and probably enhancing metastasis and invasion in the tumor microenvironment, thereby suggesting
the therapeutic potential of targeting angiogenic pathways in gastrointestinal cancers.
Collapse
Affiliation(s)
- Forouzan Amerizadeh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Maftouh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramin Mardani
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Afsane Bahrami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
13
|
Tan AC, Chan DL, Faisal W, Pavlakis N. New drug developments in metastatic gastric cancer. Therap Adv Gastroenterol 2018; 11:1756284818808072. [PMID: 30455742 PMCID: PMC6236851 DOI: 10.1177/1756284818808072] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 09/27/2018] [Indexed: 02/04/2023] Open
Abstract
Metastatic gastric cancer is associated with a poor prognosis and novel treatment options are desperately needed. The development of targeted therapies heralded a new era for the management of metastatic gastric cancer, however results from clinical trials of numerous targeted agents have been mixed. The advent of immune checkpoint inhibitors has yielded similar promise and results from early trials are encouraging. This review provides an overview of the systemic treatment options evaluated in metastatic gastric cancer, with a focus on recent evidence from clinical trials for targeted therapies and immune checkpoint inhibitors. The failure to identify appropriate predictive biomarkers has hampered the success of many targeted therapies in gastric cancer, and a deeper understanding of specific molecular subtypes and genomic alterations may allow for more precision in the application of novel therapies. Identifying appropriate biomarkers for patient selection is essential for future clinical trials, for the most effective use of novel agents and in combination approaches to account for growing complexity of treatment options.
Collapse
Affiliation(s)
- Aaron C. Tan
- Northern Sydney Cancer Centre, Royal North Shore
Hospital, Sydney, NSW, Australia/National Cancer Centre Singapore,
Singapore
| | - David L. Chan
- Northern Sydney Cancer Centre, Royal North Shore
Hospital, Sydney, NSW, Australia/Northern Clinical School, University of
Sydney, NSW, Australia
| | - Wasek Faisal
- Ballarat Regional Integrated Cancer Centre,
Ballarat, VIC, Australia
| | - Nick Pavlakis
- Northern Sydney Cancer Centre, Royal North Shore
Hospital, Reserve Road, St. Leonards, NSW 2065, Australia
| |
Collapse
|
14
|
Bai ZG, Zhang ZT. A systematic review and meta-analysis on the effect of angiogenesis blockade for the treatment of gastric cancer. Onco Targets Ther 2018; 11:7077-7087. [PMID: 30410364 PMCID: PMC6200090 DOI: 10.2147/ott.s169484] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Introduction To date, anti-vascular endothelial growth factor (VEGF) monoclonal antibody (mAb, bevacizumab), anti-VEGF receptor mAb (ramucirumab) and selective vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (sunitinib, sorafenib and apatinib) have been tested in the clinical trials. Materials and methods In the current study, results of 32 clinical trials (24 Phase I or II, 8 Phase III) were systematically reviewed and meta-analysis was performed in 8 Phase III trial results. Results It was found that median overall survival (OS) time and progression-free survival (PFS) time were significantly longer in the patients treated with antiangiogenic reagents compared to that in the patients with placebo when all of 8 Phase III clinical trials were analyzed together (OS: odds ratio = 0.805, 95% CI: 0.719–0.901, P < 0.001; PFS: odds ratio = 0.719, 95% CI: 0.533–969, P = 0.030). Conclusion Meta-analysis on bevacizumab (4 out 8 Phase III trials) indicated that neither OS nor PFS was significantly different between the groups treated with bevacizumab or placebo with or without combination of other chemotherapeutic reagents (OS: odds ratio = 0.909, 95% CI: 0.780–1.059, P = 0.221; PFS: odds ratio = 0.985, 95% CI: 0.865–1.122, P = 0.826). By contrast, meta-analysis on ramucirumab (3 out of 8 Phase III trials) revealed that ramucirumab was significantly favored in the treatment of gastric cancer with significant different OS between the two groups (odds ratio = 0.720, 95% CI: 0.604–0.858, P < 0.001). In addition, patients treated with VEGF or VEGFR blockers had higher morbidity of hypertension and neutropenia, but lower risk of side effects of vomiting and anemia. These findings suggest that addition of antiangiogenesis reagents, especially anti-VEGFR-mAb, to the first- or second-line chemotherapy could prolong patient’s OS and PFS time in the advanced or metastatic gastric cancer.
Collapse
Affiliation(s)
- Zhi-Gang Bai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Cancer Invasion and Metastasis Research and National Clinical Research Center for Digestive Diseases, Beijing 100050, People's Republic of China,
| | - Zhong-Tao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Cancer Invasion and Metastasis Research and National Clinical Research Center for Digestive Diseases, Beijing 100050, People's Republic of China,
| |
Collapse
|
15
|
The Hippo pathway as a drug target in gastric cancer. Cancer Lett 2018; 420:14-25. [PMID: 29408652 DOI: 10.1016/j.canlet.2018.01.062] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 02/08/2023]
Abstract
The Hippo tumor suppressor pathway is critical for balancing cellular differentiation and proliferation in response to cell-cell contact, mechanical signals and diffusible signals such as lysophosphatidic acid. Hippo pathway signaling is frequently dysregulated in gastric cancer (GC), as well as many other kinds of solid tumors, contributing to multiple aspects of malignant progression including unchecked cell division and metastasis. Considering the importance of this Hippo pathway in cancer, its pharmacological disruption may be of huge benefit in the fight against this disease. In this review, we summarize the components of the Hippo pathway, its crosstalk with other major oncogenic signaling pathways, common mechanisms of its dysregulation, as well as potential therapeutic approaches of targeting this pathway for cancer treatment, specifically in a GC context.
Collapse
|
16
|
Liu D, Ma X, Xiao D, Jia Y, Wang Y. Efficacy and safety of targeting VEGFR drugs in treatment for advanced or metastatic gastric cancer: a systemic review and meta-analysis. Oncotarget 2018; 9:8120-8132. [PMID: 29487720 PMCID: PMC5814287 DOI: 10.18632/oncotarget.23429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 11/29/2017] [Indexed: 12/21/2022] Open
Abstract
The value of targeting VEGFR (vascular endothelial growth factor receptor) drugs has demonstrated encouraging anti-cancer activity in advanced solid tumors within current clinical trials. This study aimed to serve as the first systemic review to assess their safety and efficacy according to biochemical characteristics of targeting VEGFR drugs in gastric cancer. We analyzed eight clinical trials on targeting VEGFR drugs in gastric cancer. Results showed that targeting VEGFR drugs significantly improved overall survival (OS) [Hazard Ratio (HR) 0.69, 95% confidence interval (CI) (0.55, 0.83), P < 0.001], progression free survival (PFS) [HR 0.50, 95% CI (0.34, 0.66), P < 0.001], disease control rate (DCR) [Odds Ratio (OR) 3.83, 95% CI (2.39, 6.15), P < 0.001] and significantly decreased the progressive disease rate(PDR)[OR 0.45, 95% CI (0.34, 0.59), P < 0.001], but not objective response rate (ORR) [OR 1.46, 95% CI (0.93, 2.29), P = 0.098]. Further subgroup revealed that VEGFR antibody (VEGFR-Ab) drugs were superior to VEGFR tyrosine kinase inhibitor (VEGFR-TKI) drugs in terms of the OS, PFS and PDR. To determine the toxic effect of targeting VEGFR drugs, the relative risk of adverse events (grade ≥ 3) of special interest(AESIs) were estimated. Most of these were predictable and manageable. Furthermore, less AESIs were observed in the VEGFR-Ab than the VEGFR-TKI drugs. In conclusion, VEGFR drugs were effective targeted therapy in advanced or metastatic gastric cancer, and its toxicity is within a controllable range. VEGFR-Ab drugs were more effective than VEGFR-TKI drugs in terms of the OS, PFS and PDR of gastric cancer patients with little toxicity.
Collapse
Affiliation(s)
- Duanrui Liu
- Central Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan 250013, People's Republic of China
| | - Xiaoli Ma
- Central Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan 250013, People's Republic of China
| | - Dongjie Xiao
- Central Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan 250013, People's Republic of China
| | - Yanfei Jia
- Central Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan 250013, People's Republic of China
- Shandong Province Key Lab of Tumor Target Molecule, Jinan Central Hospital Affiliated to Shandong University, Jinan 250013, People's Republic of China
| | - Yunshan Wang
- Central Laboratory, Jinan Central Hospital Affiliated to Shandong University, Jinan 250013, People's Republic of China
- Shandong Province Key Lab of Tumor Target Molecule, Jinan Central Hospital Affiliated to Shandong University, Jinan 250013, People's Republic of China
| |
Collapse
|
17
|
Chen LT, Oh DY, Ryu MH, Yeh KH, Yeo W, Carlesi R, Cheng R, Kim J, Orlando M, Kang YK. Anti-angiogenic Therapy in Patients with Advanced Gastric and Gastroesophageal Junction Cancer: A Systematic Review. Cancer Res Treat 2017; 49:851-868. [PMID: 28052652 PMCID: PMC5654167 DOI: 10.4143/crt.2016.176] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 12/20/2016] [Indexed: 02/08/2023] Open
Abstract
Despite advancements in therapy for advanced gastric and gastroesophageal junction cancers, their prognosis remains dismal. Tumor angiogenesis plays a key role in cancer growth and metastasis, and recent studies indicate that pharmacologic blockade of angiogenesis is a promising approach to therapy. In this systematic review, we summarize current literature on the clinical benefit of anti-angiogenic agents in advanced gastric cancer. We conducted a systematic search of PubMed and conference proceedings including the American Society of Clinical Oncology, the European Society for Medical Oncology, and the European Cancer Congress. Included studies aimed to prospectively evaluate the efficacy and safety of anti-angiogenic agents in advanced gastric or gastroesophageal junction cancer. Each trial investigated at least one of the following endpoints: overall survival, progression-free survival/time to progression, and/or objective response rate. Our search yielded 139 publications. Forty-two met the predefined inclusion criteria. Included studies reported outcomes with apatinib, axitinib, bevacizumab, orantinib, pazopanib, ramucirumab, regorafenib, sorafenib, sunitinib, telatinib, and vandetanib. Second-line therapy with ramucirumab and third-line therapy with apatinib are the only anti-angiogenic agents so far shown to significantly improve survival of patients with advanced gastric cancer. Overall, agents that specifically target the vascular endothelial growth factor ligand or receptor have better safety profile compared to multi-target tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes and National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Do-Youn Oh
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kun-Huei Yeh
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Winnie Yeo
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | - Yoon-Koo Kang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Song Z, Wu Y, Yang J, Yang D, Fang X. Progress in the treatment of advanced gastric cancer. Tumour Biol 2017; 39:1010428317714626. [PMID: 28671042 DOI: 10.1177/1010428317714626] [Citation(s) in RCA: 603] [Impact Index Per Article: 86.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer is one of the most common malignant tumors in the digestive system. Surgery is currently considered to be the only radical treatment. As surgical techniques improve and progress is made in traditional radiotherapy, chemotherapy, and the implementation of neoadjuvant therapy, the 5-year survival rate of early gastric cancer can reach >95%. However, the low rate of early diagnosis means that most patients have advanced-stage disease at diagnosis and so the best surgical window is missed. Therefore, the main treatment for advanced gastric cancer is the combination of neoadjuvant chemoradiotherapy, molecular-targeted therapy, and immunotherapy. In this article, we summarize several common methods used to treat advanced gastric cancer and discuss the progress made in the treatment of gastric cancer in detail. Only clinical practice and clinical research will allow us to prolong the survival time of patients and allow the patients to truly benefit by paying attention to the individual patient characteristics, drug choice, and developing a reasonable and comprehensive treatment plan.
Collapse
Affiliation(s)
- Zheyu Song
- 1 Department of General Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Yuanyu Wu
- 1 Department of General Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Jiebing Yang
- 2 Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, People's Republic of China
| | - Dingquan Yang
- 1 Department of General Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Xuedong Fang
- 1 Department of General Surgery, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| |
Collapse
|
19
|
Narita Y, Muro K. Challenges in molecular targeted therapy for gastric cancer: considerations for efficacy and safety. Expert Opin Drug Saf 2017; 16:319-327. [PMID: 27976952 DOI: 10.1080/14740338.2017.1273348] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION The Cancer Genome Atlas Research Network recently proposed a molecular classification for gastric cancer (GC) into four subtypes based on comprehensive evaluation. While the mechanisms of molecular targeted therapies in GC were confirmed by multiple clinical studies, only a limited number of therapeutics for GC have been approved to date. Areas covered: In this systematic review of the available literature, we discuss the completed and ongoing clinical trials of molecular targeted therapies in patients with GC, with a focus on their efficacy and safety profiles. Expert opinion: Results of recent studies clearly demonstrated that trastuzumab and ramucirumab, monoclonal antibodies (mAbs) against human epidermal growth factor receptor 2 (HER2) and vascular endothelial growth factor (VEGF), respectively, improved overall survival (OS) in GC with manageable safety profiles. Careful surveillance of ongoing clinical trials and timely profiling and monitoring of genetic signatures are imperative to establish a strong foundation for precision medicine in GC.
Collapse
Affiliation(s)
- Yukiya Narita
- a Department of Clinical Oncology , Aichi Cancer Center Hospital , Nagoya , Japan
| | - Kei Muro
- a Department of Clinical Oncology , Aichi Cancer Center Hospital , Nagoya , Japan
| |
Collapse
|
20
|
Pasini F, Fraccon AP, Modena Y, Bencivenga M, Giacopuzzi S, La Russa F, Gusella M, de Manzoni G. Targeted therapies for advanced and metastatic adenocarcinoma of the gastroesophageal junction: is there something new? Gastric Cancer 2017; 20:31-42. [PMID: 27568322 DOI: 10.1007/s10120-016-0626-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 08/02/2016] [Indexed: 02/07/2023]
Abstract
Despite improvements in systemic chemotherapy (CT), the prognosis of metastatic adenocarcinoma of the gastroesophageal junction remains poor. Over the years, new targeting agents have become available and were tested, with or without CT, in first or subsequent lines of therapy. The epidermal growth factor receptor family was targeted with monoclonal antibodies (MoAbs) (trastuzumab, cetuximab, panitumumab) and tyrosin kinase inhibitors (TKIs) (lapatinib, erlotinib, gefitinib). Only trastuzumab, in combination with cisplatin and fluoropyrimidines, significantly improved overall survival (OS) in first-line therapy (13.8 vs. 11.1 months). Angiogenesis also was targeted with MoAbs (bevacizumab and ramucirumab); ramucirumab, a vascular endothelial growth factor-receptor 2 antagonist, enhanced OS in two phase III studies in the first (9.6 vs. 7.4 months) and subsequent lines of treatment (5.2 vs. 3.8 months), while the bevacizumab study was negative. TKIs (sunitinib, sorafenib, regorafenib, apatinib) were tested in this setting in phase II studies in the second/third line, only showing modest antitumor activity. The hepatocyte growth factor receptor (MET) was targeted in untreated patients in a phase III trial with MoAb rilotumumab, with or without CT, but the study was stopped because of mortality excess in the rilotumumab arm. Mammalian target of rapamycin (MTOR) pathway inhibition with everolimus was tested in pretreated patients in a placebo-controlled phase III trial who failed to improve OS (5.4 vs. 4.3 months). In conclusion, considering the modest survival gain obtained overall, the high cost of these therapies and the quality of life issue must be primarily considered in treating these patients.
Collapse
Affiliation(s)
- Felice Pasini
- Department of Medical Oncology, Ospedale S. Maria della Misericordia, Viale Tre Martiri, 140-45100, Rovigo, Italy.
| | - Anna Paola Fraccon
- Medical Oncology Unit, Casa di Cura Pederzoli, Peschiera del Garda, Verona, Italy
| | - Yasmina Modena
- Department of Medical Oncology, Ospedale S. Maria della Misericordia, Viale Tre Martiri, 140-45100, Rovigo, Italy
| | - Maria Bencivenga
- General and Upper GI Surgery Division, University of Verona, Verona, Italy
| | - Simone Giacopuzzi
- General and Upper GI Surgery Division, University of Verona, Verona, Italy
| | - Francesca La Russa
- Department of Medical Oncology, Ospedale S. Maria della Misericordia, Viale Tre Martiri, 140-45100, Rovigo, Italy
| | - Milena Gusella
- Department of Medical Oncology, Ospedale S. Maria della Misericordia, Viale Tre Martiri, 140-45100, Rovigo, Italy
| | | |
Collapse
|
21
|
Horgan AM, Darling G, Wong R, Guindi M, Liu G, Jonker DJ, Lister J, Xu W, MacKay HM, Dinniwell R, Kim J, Pierre A, Shargall Y, Asmis TR, Agboola O, Seely AJ, Ringash J, Wells J, Marginean EC, Haider M, Knox JJ. Adjuvant sunitinib following chemoradiotherapy and surgery for locally advanced esophageal cancer: a phase II trial. Dis Esophagus 2016; 29:1152-1158. [PMID: 26663741 DOI: 10.1111/dote.12444] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The prognosis for locally advanced esophageal cancer is poor despite the use of trimodality therapy. In this phase II study, we report the feasibility, tolerability and efficacy of adjuvant sunitinib. Included were patients with stage IIa, IIB or III cancer of the thoracic esophagus or gastroesophageal junction. Neoadjuvant therapy involved Irinotecan (65 mg/m2 ) + Cisplatin (30 mg/m2 ) on weeks 1 and 2, 4 and 5, 7 and 8 with concurrent radiation (50Gy/25 fractions) on weeks 4-8. Sunitinib was commenced 4-13 weeks after surgery and continued for one year. Sixty-one patients were included in the final analysis, 36 patients commenced adjuvant sunitinib. Fourteen patients discontinued sunitinib due to disease recurrence (39%) within the 12-month period, 12 (33%) discontinued due to toxicity, and 3 (8%) requested cessation of therapy. In the overall population, median survival was 26 months with a 2 and 3-year survival rate of 52% and 35%, respectively. The median survival for the 36 patients treated with sunitinib was 35 months and 2-year survival probability of 68%. In a historical control, a prior phase II study with the same trimodality therapy (n = 43), median survival was 36 months, with a 2-year survival of 67%. Initiation of adjuvant sunitinib is feasible, but poorly tolerated, with no signal of additional benefit over trimodality therapy for locally advanced esophageal cancer.
Collapse
Affiliation(s)
- A M Horgan
- Department of Medical Oncology, Princess Margaret and Toronto General Hospitals, University Health Network, Toronto, Ontario, Canada
| | - G Darling
- Department of Surgical Oncology, Division of Thoracic Surgery, Princess Margaret and Toronto General Hospitals, University Health Network, Toronto, Ontario, Canada
| | - R Wong
- Department of Radiation Oncology, Princess Margaret and Toronto General Hospitals, University Health Network, Toronto, Ontario, Canada
| | - M Guindi
- Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - G Liu
- Department of Medical Oncology, Princess Margaret and Toronto General Hospitals, University Health Network, Toronto, Ontario, Canada
| | - D J Jonker
- Department of Medical Oncology, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - J Lister
- Department of Surgical Oncology, Division of Thoracic Surgery, Princess Margaret and Toronto General Hospitals, University Health Network, Toronto, Ontario, Canada
| | - W Xu
- Department of Biostatistics, Princess Margaret and Toronto General Hospitals, University Health Network, Toronto, Ontario, Canada
| | - H M MacKay
- Department of Medical Oncology, Princess Margaret and Toronto General Hospitals, University Health Network, Toronto, Ontario, Canada
| | - R Dinniwell
- Department of Radiation Oncology, Princess Margaret and Toronto General Hospitals, University Health Network, Toronto, Ontario, Canada
| | - J Kim
- Department of Radiation Oncology, Princess Margaret and Toronto General Hospitals, University Health Network, Toronto, Ontario, Canada
| | - A Pierre
- Department of Surgical Oncology, Division of Thoracic Surgery, Princess Margaret and Toronto General Hospitals, University Health Network, Toronto, Ontario, Canada
| | - Y Shargall
- Division of Thoracic Surgery, McMaster University, Hamilton, Ontario, Canada
| | - T R Asmis
- Department of Medical Oncology, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - O Agboola
- Department of Thoracic Surgery, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - A J Seely
- Department of Pathology, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - J Ringash
- Department of Radiation Oncology, Princess Margaret and Toronto General Hospitals, University Health Network, Toronto, Ontario, Canada
| | - J Wells
- Department of Radiation Oncology, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - E C Marginean
- Department of Pathology, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - M Haider
- Department of Medical Oncology, Princess Margaret and Toronto General Hospitals, University Health Network, Toronto, Ontario, Canada
| | - J J Knox
- Department of Medical Oncology, Princess Margaret and Toronto General Hospitals, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Yu J, Zhang Y, Leung LH, Liu L, Yang F, Yao X. Efficacy and safety of angiogenesis inhibitors in advanced gastric cancer: a systematic review and meta-analysis. J Hematol Oncol 2016; 9:111. [PMID: 27756337 PMCID: PMC5070169 DOI: 10.1186/s13045-016-0340-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 10/08/2016] [Indexed: 01/05/2023] Open
Abstract
Monoclonal antibodies and small molecule tyrosine kinase inhibitors (TKIs) directed against the vascular endothelial growth factor (VEGF) or its receptors have been investigated in several studies for the treatment of advanced gastric cancer (GC). In the present study, we aimed to evaluate the efficacy and safety of angiogenesis inhibitors in advanced GC. We searched published randomized controlled trials (RCTs) comparing angiogenesis inhibitors with non-angiogenesis inhibitors for the treatment of GC. MEDLINE, EMBASE, and the Cochrane Controlled Trials Register were searched. The extracted data on progression-free survival (PFS) and overall survival (OS) were measured in terms of hazard ratios (HR) and corresponding 95 % confidence intervals (CIs). In addition, risk ratios (RR) and corresponding 95 % CIs were pooled for objective response rate (ORR), disease control rate (DCR), and risk of adverse events (AEs). Ten RCTs involving 2786 patients were included. Compared with non-angiogenesis inhibitor-containing regimens, angiogenesis inhibitor-containing regimens resulted in a significant improvement in OS (HR 0.80, 95 % CI 0.69-0.93, P = 0.004), prolonged PFS (HR 0.66, 95 % CI 0.51-0.86, P = 0.002), and superior ORR (RR 1.34, 95 % CI 1.09-1.65, P = 0.005) and DCR (RR 1.37, 95 % CI 1.17-1.61, P = 0.0001). Angiogenesis inhibitors were associated with a greater number of AEs, but most of these were predictable and manageable. However, hand-foot syndrome, diarrhea, and gastrointestinal (GI) perforation were significantly increased in patients treated with angiogenesis inhibitors. In summary, angiogenesis inhibitor-containing regimens were superior to non-angiogenesis inhibitor-containing regimens in terms of OS, PFS, RR, and DCR in patients with advanced GC.
Collapse
Affiliation(s)
- Jing Yu
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng District, Beijing, 100050 China
| | - Yue Zhang
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng District, Beijing, 100050 China
| | - Lai-Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau 999078 China
| | - Lian Liu
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng District, Beijing, 100050 China
| | - Fan Yang
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng District, Beijing, 100050 China
| | - Xiaojun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau 999078 China
| |
Collapse
|
23
|
Hwang J. Beyond HER2: recent advances and future directions in targeted therapies in esophagogastric cancers. J Gastrointest Oncol 2016; 7:763-770. [PMID: 27747090 PMCID: PMC5056259 DOI: 10.21037/jgo.2016.08.13] [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: 04/20/2016] [Accepted: 06/06/2016] [Indexed: 12/19/2022] Open
Abstract
Esophagogastric cancers (EGCa) are a leading cause of cancer related mortality worldwide. It has been recognized that they represent heterogenous diseases based on histology and anatomy. However, it is also increasingly evident that these are diverse malignancies based on genetic alterations, and this is increasingly making these diseases amenable to targeted therapies. While epidermal growth factor receptor (EGFR) and mTOR inhibitors have failed to prove effective in the treatment of advanced EGCa, vascular endothelial growth factor (VEGF) inihibitor have now been demonstrated to improve survival, at least in the 2nd line setting of adenocarcinomas. Other promising approaches are being investigated, including targeted therapies such as MET and FGFR inhibitors, as well as immunotherapy and agents that may affect synthetic lethality.
Collapse
Affiliation(s)
- Jimmy Hwang
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC 28204, USA
| |
Collapse
|
24
|
Moehler M, Gepfner-Tuma I, Maderer A, Thuss-Patience PC, Ruessel J, Hegewisch-Becker S, Wilke H, Al-Batran SE, Rafiyan MR, Weißinger F, Schmoll HJ, Kullmann F, von Weikersthal LF, Siveke JT, Weusmann J, Kanzler S, Schimanski CC, Otte M, Schollenberger L, Koenig J, Galle PR. Sunitinib added to FOLFIRI versus FOLFIRI in patients with chemorefractory advanced adenocarcinoma of the stomach or lower esophagus: a randomized, placebo-controlled phase II AIO trial with serum biomarker program. BMC Cancer 2016; 16:699. [PMID: 27582078 PMCID: PMC5006426 DOI: 10.1186/s12885-016-2736-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 08/20/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND As a multi-targeted anti-angiogenic receptor tyrosine kinase (RTK) inhibitor sunitinib (SUN) has been established for renal cancer and gastrointestinal stromal tumors. In advanced refractory esophagogastric cancer patients, monotherapy with SUN was associated with good tolerability but limited tumor response. METHODS This double-blind, placebo-controlled, multicenter, phase II clinical trial was conducted to evaluate the efficacy, safety and tolerability of SUN as an adjunct to second and third-line FOLFIRI (NCT01020630). Patients were randomized to receive 6-week cycles including FOLFIRI plus sodium folinate (Na-FOLFIRI) once every two weeks and SUN or placebo (PL) continuously for four weeks followed by a 2-week rest period. The primary study endpoint was progression-free survival (PFS). Preplanned serum analyses of VEGF-A, VEGF-D, VEGFR2 and SDF-1α were performed retrospectively. RESULTS Overall, 91 patients were randomized, 45 in each group (one patient withdrew). The main grade ≥3 AEs were neutropenia and leucopenia, observed in 56 %/20 % and 27 %/16 % for FOLFIRI + SUN/FOLFIRI + PL, respectively. Median PFS was similar, 3.5 vs. 3.3 months (hazard ratio (HR) 1.11, 95 % CI 0.70-1.74, P = 0.66) for FOLFIRI + SUN vs. FOLFIRI + PL, respectively. For FOLFIRI + SUN, a trend towards longer median overall survival (OS) compared with placebo was observed (10.4 vs. 8.9 months, HR 0.82, 95 % CI 0.50-1.34, one-sided P = 0.21). In subgroup serum analyses, significant changes in VEGF-A (P = 0.017), VEGFR2 (P = 0.012) and VEGF-D (P < 0.001) serum levels were observed. CONCLUSIONS Although sunitinib combined with FOLFIRI did not improve PFS and response in chemotherapy-resistant gastric cancer, a trend towards better OS was observed. Further biomarker-driven studies with other anti-angiogenic RTK inhibitors are warranted. TRIAL REGISTRATION This study was registered prospectively in the NCT Clinical Trials Registry (ClinicalTrials.gov) under NCT01020630 on November 23, 2009 after approval by the leading ethics committee of the Medical Association of Rhineland-Palatinate, Mainz, in coordination with the participating ethics committees (see Additional file 2) on September 16, 2009.
Collapse
Affiliation(s)
- Markus Moehler
- University Medical Center, Johannes Gutenberg-University Mainz, I. Medizinische Klinik und Poliklinik, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Irina Gepfner-Tuma
- University Medical Center, Johannes Gutenberg-University Mainz, I. Medizinische Klinik und Poliklinik, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Annett Maderer
- University Medical Center, Johannes Gutenberg-University Mainz, I. Medizinische Klinik und Poliklinik, Langenbeckstraße 1, 55131 Mainz, Germany
| | | | - Joern Ruessel
- University Hospital Halle (Saale), Halle (Saale), Germany
| | | | | | | | | | | | | | - Frank Kullmann
- Kliniken Nordoberpfalz - Klinikum Weiden, Weiden, Germany
| | | | - Jens T. Siveke
- Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Jens Weusmann
- University Medical Center, Johannes Gutenberg-University Mainz, I. Medizinische Klinik und Poliklinik, Langenbeckstraße 1, 55131 Mainz, Germany
| | | | | | - Melanie Otte
- Praxisgemeinschaft für Onkologie und Urologie, Wilhelmshaven, Germany
| | | | - Jochem Koenig
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI) of the University Medical Center Mainz, Mainz, Germany
| | - Peter R. Galle
- University Medical Center, Johannes Gutenberg-University Mainz, I. Medizinische Klinik und Poliklinik, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
25
|
Roberto M, Romiti A, Onesti CE, Zullo A, Falcone R, Marchetti P. Evolving treatments for advanced gastric cancer: appraisal of the survival trend. Expert Rev Anticancer Ther 2016; 16:717-29. [PMID: 27137418 DOI: 10.1080/14737140.2016.1184979] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction and areas covered: We analysed the results of the main clinical studies looking at patients with advanced gastric or esophagogastric junction cancer, in order to differentiate between what is already clinical evidence and what is a promise for the cure of such patients. Thus, achievements from key studies, which had been purposely directed at chemotherapy, molecular target therapies and immunotherapy in both first and second-line setting were analysed. Metronomic chemotherapy, which consists of the administration of continuative low-dose anticancer drugs, was considered also. Expert commentary: It was found that patients included in experimental arms of randomized trials compared with controls have often benefited from a statistically significant extension of overall survival. However, further studies are awaited to bring new drugs into clinical practice and to validate candidate biomarkers predictive of response.
Collapse
Affiliation(s)
- Michela Roberto
- a Clinical and Molecular Medicine Department , Sapienza University, Sant'Andrea Hospital , Rome , Italy
| | - Adriana Romiti
- a Clinical and Molecular Medicine Department , Sapienza University, Sant'Andrea Hospital , Rome , Italy
| | - Concetta Elisa Onesti
- a Clinical and Molecular Medicine Department , Sapienza University, Sant'Andrea Hospital , Rome , Italy
| | - Angelo Zullo
- b Gastroenterology and Digestive Endoscopy , Nuovo Regina Margherita Hospital , Rome , Italy
| | - Rosa Falcone
- a Clinical and Molecular Medicine Department , Sapienza University, Sant'Andrea Hospital , Rome , Italy
| | - Paolo Marchetti
- a Clinical and Molecular Medicine Department , Sapienza University, Sant'Andrea Hospital , Rome , Italy
| |
Collapse
|
26
|
Abstract
Background Gastric cancer is the fourth most common cancer worldwide. Surgery in combination with multimodal therapy provides the only curative therapy until now. The importance of targeted therapy became clear over the last few years. Due to the implication of HER2 and angiogenesis-directed targeted therapies major advances in the treatment of gastric cancer could be reached. Nevertheless, benefits in survival remain unsatisfactory and the development of resistance to monoclonal antibodies is arising. Methods A comprehensive and comparative literature research was performed to evaluate the status of HER2 and angiogenesis-directed targeted therapy in gastric cancer. Results Up to now, trastuzumab and ramucirumab are the only agents showing remarkable benefits in the therapy for the patients suffering from gastric cancer. The limitations of targeted therapies in gastric cancer are mainly associated with the development of secondary resistance. Conclusion Addition of targeted therapy in second-line treatment is beneficial when compared with chemotherapy alone. Nevertheless, results in first-line treatment remain modest. Therefore, new therapeutic agents and combinations in the first-line treatment of gastric cancer are urgently needed and remain to be validated in clinical trials.
Collapse
Affiliation(s)
- G Jomrich
- Department of Surgery, Gastroesophageal Tumor Unit, Comprehensive Cancer Center (CCC), Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| | - S F Schoppmann
- Department of Surgery, Gastroesophageal Tumor Unit, Comprehensive Cancer Center (CCC), Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
| |
Collapse
|
27
|
Tomasello G, Ghidini M, Liguigli W, Ratti M, Toppo L, Passalacqua R. Targeted therapies in gastric cancer treatment: where we are and where we are going. Invest New Drugs 2016; 34:378-93. [PMID: 26873643 DOI: 10.1007/s10637-016-0330-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/09/2016] [Indexed: 12/12/2022]
Abstract
Gastric cancer (GC) is one of the most common malignancies and a major cause of cancer-related deaths worldwide. Its incidence has significantly declined over the last few decades, probably due to the identification of specific etiologic agents such as Helicobacter pylori and other dietary and environmental risk factors. Nevertheless, most of the cases are unfortunately diagnosed at an advanced stage justifying median overall survival rates frequently not exceeding one year. Palliative combination chemotherapy usually represented by a platinum-based doublet is the mainstay of treatment in the metastatic setting. Adding a third drug such as an anthracycline or a taxane has been shown to improve response rate and provide limited survival benefits in fit selected patients. Unlike other tumors, the introduction of molecularly targeted drugs in the medical armamentarium for GC is relatively recent with trastuzumab and ultimately ramucirumab constituting the only agents approved to date. Recent advances in the understanding of GC biology have led to the development of novel targeted therapies holding the promise to further improve treatment outcomes. The aim of this paper is to review the main available data coming from clinical trials of targeted drugs and to describe some of the most interesting molecules in clinical development in GC. These include drugs targeting EGFR, angiogenesis, c-MET, FGFR2, mTOR and immune checkpoints.
Collapse
Affiliation(s)
- Gianluca Tomasello
- Oncology Division, Azienda Socio Sanitaria Territoriale di Cremona, Ospedale di Cremona, Viale Concordia 1, 26100, Cremona, Italy.
| | - Michele Ghidini
- Oncology Division, Azienda Socio Sanitaria Territoriale di Cremona, Ospedale di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Wanda Liguigli
- Oncology Division, Azienda Socio Sanitaria Territoriale di Cremona, Ospedale di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Margherita Ratti
- Oncology Division, Azienda Socio Sanitaria Territoriale di Cremona, Ospedale di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Laura Toppo
- Oncology Division, Azienda Socio Sanitaria Territoriale di Cremona, Ospedale di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| | - Rodolfo Passalacqua
- Oncology Division, Azienda Socio Sanitaria Territoriale di Cremona, Ospedale di Cremona, Viale Concordia 1, 26100, Cremona, Italy
| |
Collapse
|
28
|
Aguiar PN, Muniz TP, Miranda RR, Tadokoro H, Forones NM, Monteiro IDP, Castelo-Branco P, Janjigian YY, De Mello RA. Current advances in targeted therapies for metastatic gastric cancer: improving patient care. Future Oncol 2016; 12:839-54. [PMID: 26838766 DOI: 10.2217/fon.15.348] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this article, we review the literature on the current advances in targeted therapies for metastatic gastric cancer aimed at improving patient care. We conclude that the key to guiding targeted therapy is individual biomarkers, which are not completely elucidated. HER2 overexpression is the only predictive biomarker currently in use. Furthermore, it is necessary to understand that gastric tumors are heterogeneous; therefore, is impossible to evaluate a novel biological compound without evaluating personal biomarkers. The selection of patients who are able to receive each treatment is paramount for improving advanced gastric cancer survival and reducing unnecessary costs.
Collapse
Affiliation(s)
| | | | | | - Hakaru Tadokoro
- Division of Medical Oncology, Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Pedro Castelo-Branco
- Department of Biomedical Sciences & Medicine, Division of Oncology, University of Algarve, Faro, Portugal
| | - Yelena Y Janjigian
- Department of Medicine, Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ramon Andrade De Mello
- Department of Medicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Biomedical Sciences & Medicine, Division of Oncology, University of Algarve, Faro, Portugal.,Clinical Research Center & Department of Medical Oncology, Centro Oncológico São Mateus, Ceará Cancer Institute, Rua Papi Junior 1222, Rodolfo Teófilo, CEP 60430-235, Fortaleza-CE, Brazil
| |
Collapse
|
29
|
Roviello G, Petrioli R, Marano L, Polom K, Marrelli D, Perrella A, Roviello F. Angiogenesis inhibitors in gastric and gastroesophageal junction cancer. Gastric Cancer 2016; 19:31-41. [PMID: 26329368 DOI: 10.1007/s10120-015-0537-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 08/15/2015] [Indexed: 02/07/2023]
Abstract
Despite significant improvements in systemic chemotherapy during the past two decades, the prognosis of patients with advanced gastric and gastroesophageal junction adenocarcinoma remains poor. Because of molecular heterogeneity, it is essential to classify tumors based on the underlying oncogenic pathways and to develop targeted therapies acting on individual tumors. Unfortunately, although a number of molecular targets have been studied, very few of these agents can be used in a clinical setting. In this review, we summarize the available data on anti-angiogenic agents in advanced/metastatic gastric cancer.
Collapse
Affiliation(s)
| | - Roberto Petrioli
- Medical Oncology Unit, University of Siena, Viale Bracci 11, 53100, Siena, Italy
| | - Luigi Marano
- Unit of General and Minimally Invasive Surgery, Department of Medical, Surgical and Neuroscience, University of Siena, Viale Bracci 11, Siena, 53100, Italy
| | - Karol Polom
- Unit of General and Minimally Invasive Surgery, Department of Medical, Surgical and Neuroscience, University of Siena, Viale Bracci 11, Siena, 53100, Italy
| | - Daniele Marrelli
- Section of Advanced Surgical Oncology, Department of Medical, Surgical and Neurosciences, University of Siena, Viale Bracci 11, Siena, 53100, Italy
| | - Armando Perrella
- Medical Oncology Unit, University of Siena, Viale Bracci 11, 53100, Siena, Italy
| | - Franco Roviello
- Unit of General and Minimally Invasive Surgery, Department of Medical, Surgical and Neuroscience, University of Siena, Viale Bracci 11, Siena, 53100, Italy
| |
Collapse
|
30
|
Carlisle B, Demko N, Freeman G, Hakala A, MacKinnon N, Ramsay T, Hey S, London AJ, Kimmelman J. Benefit, Risk, and Outcomes in Drug Development: A Systematic Review of Sunitinib. J Natl Cancer Inst 2016; 108:djv292. [PMID: 26547927 PMCID: PMC5943825 DOI: 10.1093/jnci/djv292] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 05/19/2015] [Accepted: 09/22/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Little is known about the total patient burden associated with clinical development and where burdens fall most heavily during a drug development program. Our goal was to quantify the total patient burden/benefit in developing a new drug. METHODS We measured risk using drug-related adverse events that were grade 3 or higher, benefit by objective response rate, and trial outcomes by whether studies met their primary endpoint with acceptable safety. The differences in risk (death rate) and benefit (overall response rate) between industry and nonindustry trials were analyzed with an inverse-variance weighted fixed effects meta-analysis implemented as a weighted regression analysis. All statistical tests were two-sided. RESULTS We identified 103 primary publications of sunitinib monotherapy, representing 9092 patients and 3991 patient-years of involvement over 10 years and 32 different malignancies. In total, 1052 patients receiving sunitinib monotherapy experienced objective tumor response (15.7% of intent-to-treat population, 95% confidence interval [CI] = 15.3% to 16.0%), 98 died from drug-related toxicities (1.08%, 95% CI = 1.02% to 1.14%), and at least 1245 experienced grade 3-4 drug-related toxicities (13.7%, 95% CI = 13.3% to 14.1%). Risk/benefit worsened as the development program matured, with several instances of replicated negative studies and almost no positive trials after the first responding malignancies were discovered. CONCLUSIONS Even for a successful drug, the risk/benefit balance of trials was similar to phase I cancer trials in general. Sunitinib monotherapy development showed worsening risk/benefit, and the testing of new indications responded slowly to evidence that sunitinib monotherapy would not extend to new malignancies. Research decision-making should draw on evidence from whole research programs rather than a narrow band of studies in the same indication.
Collapse
Affiliation(s)
- Benjamin Carlisle
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Nadine Demko
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Georgina Freeman
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Amanda Hakala
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Nathalie MacKinnon
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Tim Ramsay
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Spencer Hey
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Alex John London
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL)
| | - Jonathan Kimmelman
- Studies of Translation, Ethics and Medicine (STREAM), Biomedical Ethics Unit, McGill University, Montréal, QC, Canada (BC, ND, GF, AH, NM, SH, JK); University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada (TR); Program On Regulation, Therapeutics, And Law (PORTAL), Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (SH); Department of Philosophy and Center for Ethics and Policy, Carnegie Mellon University, Pittsburgh, PA (AJL).
| |
Collapse
|
31
|
Shan F, Miao R, Xue K, Li Z, Li Z, Bu Z, Wu A, Zhang L, Wu X, Zong X, Wang X, Li S, Ji X, Jia Z, Li Z, Ji J. Controlling angiogenesis in gastric cancer: A systematic review of anti-angiogenic trials. Cancer Lett 2015; 380:598-607. [PMID: 26724681 DOI: 10.1016/j.canlet.2015.12.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 12/31/2022]
Abstract
PURPOSE Angiogenesis is a promising therapeutic target to inhibit tumor growth. This review summarizes data from clinical trials of anti-angiogenic agents in gastric cancer. DESIGN A systematic search of PubMed, Embase and conference databases is performed to identify clinical trials with specific anti-angiogenic agents in gastric cancer treatment RESULTS The risk of disease progression (37-52%) and death (19-22%) with ramucirumab as second-line treatment decreases in phase III trials in advanced gastric cancer. No significant improvement in overall survival (OS) with the addition of bevacizumab to chemotherapy is shown. Bevacizumab or ramucirumab combined with traditional chemotherapy is associated with higher adverse event rate compared to chemotherapy alone. Except for apatinib, phase II trials of other tyrosine kinase inhibitors (TKIs) may improve overall response rate, but there are no significant improvements in OS and progression-free survival (PFS) when combined with chemotherapy. CONCLUSION Phase III trials in advanced gastric cancer have demonstrated improved outcome with ramucirumab as second-line treatment. Most of the other studies on anti-angiogenic agents in gastric cancer have reported improvement in response rate but not in OS compared to chemotherapy alone. Future research is expected in optimizing the anti-angiogenic therapy combined with traditional treatment.
Collapse
Affiliation(s)
- Fei Shan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Rulin Miao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Kan Xue
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhemin Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ziyu Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhaode Bu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Aiwen Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lianhai Zhang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaojiang Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xianglong Zong
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaohong Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shuangxi Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xin Ji
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ziyu Jia
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ziran Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jiafu Ji
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| |
Collapse
|
32
|
Abstract
Gastric cancer is one of the most commonly diagnosed and the second leading cause of cancer death worldwide. Surgery combined with multimodal therapy remains the only curative therapy. However, local relapse or distant metastases occur in more than 50% of radically resected patients. Due to molecular therapies, targeting HER2 and angiogenesis, major advances in the treatment of gastric cancer could be achieved. Nevertheless, development of resistance to monoclonal antibodies, such as trastuzumab, is arising. Currently a number of promising new therapeutic are under investigation, combining chemotherapy with newly developed agents to overcome cancer resistance. In this review we report current clinical applications of targeted therapies and overview ongoing trials, investigating the use of monoclonal antibodies in (HER2 positive) gastric cancer.
Collapse
Affiliation(s)
- G Jomrich
- a Department of Surgery, Gastroesophageal Tumor Unit, Comprehensive Cancer Center (CCC) , Medical University of Vienna , Vienna , Austria
| | - S F Schoppmann
- a Department of Surgery, Gastroesophageal Tumor Unit, Comprehensive Cancer Center (CCC) , Medical University of Vienna , Vienna , Austria
| |
Collapse
|
33
|
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: 1.7] [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
|
34
|
Dahle-Smith A, Petty RD. Biomarkers and novel agents in esophago-gastric cancer: are we making progress? Expert Rev Anticancer Ther 2015; 15:1103-19. [DOI: 10.1586/14737140.2015.1071669] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
35
|
Aprile G, Ongaro E, Del Re M, Lutrino SE, Bonotto M, Ferrari L, Rihawi K, Cardellino GG, Pella N, Danesi R, Fasola G. Angiogenic inhibitors in gastric cancers and gastroesophageal junction carcinomas: A critical insight. Crit Rev Oncol Hematol 2015; 95:165-78. [PMID: 25800976 DOI: 10.1016/j.critrevonc.2015.02.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 12/28/2014] [Accepted: 02/24/2015] [Indexed: 12/11/2022] Open
Abstract
Advanced gastric cancer ranks second as the global leading cause of cancer-related death and improvements in systemic chemotherapy have reached a plateau. Advanced molecular sequencing techniques help identifying patients more likely to respond to targeted agents; nevertheless we are still far from major breakthroughs. Although antiangiogenic drugs have produced notable advances, redundant pathways or mechanisms of resistance may limit their efficacy. Novel compounds have been recently developed to specifically target VEGF receptors, PlGF, FGF, MET, and angiopoietin. Ramucirumab, a monoclonal antibody specifically directed against the VEGFR-2, has emerged as a novel therapeutic opportunity. REGARD and RAINBOW were the first phase III studies to report the value of this strategy in gastric cancer patients, and other ongoing trials are testing novel antiangiogenic compounds. The aim of our review is to present the state-of-the-art of novel antiangiogenic compounds in advanced gastric cancer, underlying the biology, their mechanism of action, and their clinical results.
Collapse
Affiliation(s)
- Giuseppe Aprile
- Department of Medical Oncology, University and General Hospital, Udine, Italy.
| | - Elena Ongaro
- Department of Medical Oncology, University and General Hospital, Udine, Italy
| | - Marzia Del Re
- Clinical Pharmacology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | | | - Marta Bonotto
- Department of Medical Oncology, University and General Hospital, Udine, Italy
| | - Laura Ferrari
- Department of Medical Oncology, University and General Hospital, Udine, Italy
| | - Karim Rihawi
- Department of Medical Oncology, University and General Hospital, Udine, Italy
| | | | - Nicoletta Pella
- Department of Medical Oncology, University and General Hospital, Udine, Italy
| | - Romano Danesi
- Clinical Pharmacology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Gianpiero Fasola
- Department of Medical Oncology, University and General Hospital, Udine, Italy
| |
Collapse
|
36
|
ElHalawani H, Abdel-Rahman O. Critical evaluation of ramucirumab in the treatment of advanced gastric and gastroesophageal cancers. Ther Clin Risk Manag 2015; 11:1123-32. [PMID: 26251608 PMCID: PMC4524527 DOI: 10.2147/tcrm.s71045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Gastric (GC) and gastroesophageal junction (GEJ) cancers are two global health problems with a relatively high mortality, particularly in the advanced stage. Inhibition of angiogenesis is now contemplated as a classic treatment preference for myriad tumor types encompassing renal cell carcinoma, non-small cell lung cancer, colorectal cancer, glioblastoma, and ovarian cancer, among others. Bevacizumab and ramucirumab have been widely investigated in GC and GEJ cancer, with some controversy about their therapeutic role. Ramucirumab is a monoclonal antibody for vascular endothelial growth factor receptor-2, with demonstrated activity both as a monotherapy and as a part of combination strategy in the management of advanced GC/GEJ cancer. In this review article, we present a critical evaluation of the preclinical and clinical data underlying the use of this drug in this indication. Moreover, we provide a spotlight on the future perspectives in systemic therapy for advanced GC/GEJ cancer.
Collapse
Affiliation(s)
- Hesham ElHalawani
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
37
|
Vincenzi B, Imperatori M, Silletta M, Marrucci E, Santini D, Tonini G. Emerging kinase inhibitors of the treatment of gastric cancer. Expert Opin Emerg Drugs 2015; 20:479-93. [PMID: 26021342 DOI: 10.1517/14728214.2015.1051467] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Gastric cancer (GC) is the fifth most common malignancy in the world. In the last years, for the first time in literature, the addition of a targeted therapy to standard chemotherapy has proved to prolong median overall survival. In this scenario, kinase inhibitors (KIs), smaller intracellular agents, could be an interesting and novel type of targeted treatment of metastatic GC both in first and further lines of therapy. AREAS COVERED Several KI have been evaluated in the preclinical setting. This review will underline the most relevant targeted pathways involved in GC tumorigenesis and disease progression including EGFR, VEGFR, c-MET, mTOR, fibroblast growth factor receptor, Src and Aurora kinases. EXPERT OPINION Despite the good results of TOGA, RAINBOW and REGARD trials about the addition of monoclonal antibodies to standard of care in GC, the addition of KI seems not to achieve comparable interesting results in management of GC. However, an improved patient selection before and during treatment according to molecular characteristics, as well as combination studies evaluating the synergistic effect of combination schedules of different KIs and standard chemotherapy, or KI plus KI or KI plus antibodies-based therapy may reveal interesting results and lead to understand mechanisms of multi-drug resistance.
Collapse
Affiliation(s)
- Bruno Vincenzi
- a University Campus Biomedico - Medical Oncology , Via Alvaro del Portillo, 200, Rome 00128, Italy
| | | | | | | | | | | |
Collapse
|
38
|
Oh DY, Doi T, Shirao K, Lee KW, Park SR, Chen Y, Yang L, Valota O, Bang YJ. Phase I Study of Axitinib in Combination with Cisplatin and Capecitabine in Patients with Previously Untreated Advanced Gastric Cancer. Cancer Res Treat 2015; 47:687-96. [PMID: 25687867 PMCID: PMC4614203 DOI: 10.4143/crt.2014.225] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 10/15/2014] [Indexed: 12/14/2022] Open
Abstract
PURPOSE This phase I trial evaluated the question of whether the standard starting dose of axitinib could be administered in combination with therapeutic doses of cisplatin/capecitabine in patients with previously untreated advanced gastric cancer, and assessed overall safety, pharmacokinetics, and preliminary antitumor activity of this combination. MATERIALS AND METHODS Patients in dose level (DL) 1 received axitinib 5 mg twice a day (days 1 to 21) with cisplatin 80 mg/m(2) (day 1) and capecitabine 1,000 mg/m(2) twice a day (days 1 to 14) in 21-day cycles. Maximum tolerated dose (MTD) was the highest dose at which ≤ 30% of the first 12 patients experienced a dose-limiting toxicity (DLT) during cycle 1. Ten additional patients were enrolled and treated at the MTD in order to obtain additional safety and pharmacokinetic data. RESULTS Three DLTs occurred during cycle 1 in three (25%) of the first 12 patients: ruptured abdominal aortic aneurysm, acute renal failure, and > 5 consecutive days of missed axitinib due to thrombocytopenia. DL1 was established as the MTD, since higher DL cohorts were not planned. Common grade 3/4 non-hematologic adverse events in 22 patients treated at DL1 included hypertension (36.4%) and decreased appetite and stomatitis (18.2% each). Cisplatin/capecitabine slightly increased axitinib exposure; axitinib decreased capecitabine and 5-fluorouracil exposure. Eight patients (36.4%) each had partial response or stable disease. Median response duration was 9.1 months; median progression-free survival was 3.8 months. CONCLUSION In patients with advanced gastric cancer, standard doses of axitinib plus therapeutic doses of cisplatin and capecitabine could be administered in combination. Adverse events were manageable.
Collapse
Affiliation(s)
- Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Toshihiko Doi
- Division of Gastroenterology, National Cancer Center Hospital East, Chiba, Japan
| | - Kuniaki Shirao
- Department of Medical Oncology, Oita University Faculty of Medicine, Yufu, Japan
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Sook Ryun Park
- Department of Internal Medicine, Research Institute and Hospital, National Cancer Center, Goyang, Korea
| | | | | | | | - Yung-Jue Bang
- Department of Internal Medicine, Seoul National University Hospital and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
39
|
Sorafenib reverses resistance of gastric cancer to treatment by cisplatin through down-regulating MDR1 expression. Med Oncol 2015; 32:470. [DOI: 10.1007/s12032-014-0470-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 12/17/2014] [Indexed: 12/13/2022]
|
40
|
Abdel-Rahman O. Targeting vascular endothelial growth factor (VEGF) pathway in gastric cancer: preclinical and clinical aspects. Crit Rev Oncol Hematol 2015; 93:18-27. [PMID: 24970311 DOI: 10.1016/j.critrevonc.2014.05.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/16/2014] [Accepted: 05/30/2014] [Indexed: 12/16/2022] Open
Abstract
The prognosis of advanced gastric cancer has been dreadful with the majority of patients dying of their disease within 1 year of the diagnosis. In the advanced stage several therapeutic options can be discussed, including molecular targeted agents, but biological predicting factors are lacking. A number of molecular targets have been studied over the last decade bringing to several phase II studies; however very few agents moved into phase III clinical trials. The VEGFR-2 inhibitor monoclonal antibody ramucirumab has been recently approved in advanced progressing gastric cancer. This article reviews the basic science as well as clinical data of VEGF signaling in advanced gastric cancer with special emphasis on the different VEGF targeting agents tested previously in this disease.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
41
|
Hultman B, Mahteme H, Sundbom M, Ljungman M, Larsson R, Nygren P. Benchmarking of gastric cancer sensitivity to anti-cancer drugs ex vivo as a basis for drug selection in systemic and intraperitoneal therapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:110. [PMID: 25528067 PMCID: PMC4304126 DOI: 10.1186/s13046-014-0110-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 12/11/2014] [Indexed: 12/13/2022]
Abstract
Background The choice of drugs for treatment of advanced gastric cancer (GC) is empirical. The purpose of the current study was to benchmark ex vivo the sensitivity of GC tumor cells from patients to standard cytotoxic and some newly introduced targeted drugs (TDs), as a basis for drug selection in the treatment of GC. Methods Tumor cell samples from patients with GC were analyzed for sensitivity to 5-fluorouracil, cisplatin, oxaliplatin, irinotecan, mitomycin C, doxorubicin and docetaxel as well as for the targeted drugs bortezomib, sorafenib, sunitinib and rapamycin using a short-term in vitro assay based on retention of viable tumor cells of fluorescent fluorescein. Samples of normal mononuclear cells, chronic lymphocytic leukemia, ovarian cancer and colorectal cancer were included for comparison. Results The GC samples were essentially as sensitive to the standard drugs and the TDs as those from colorectal cancer whereas the ovarian cancer samples were more sensitive. The individual GC samples varied considerably in sensitivity to increasing concentrations of the clinically used standard drugs. In GC, cisplatin was cross-resistant to oxaliplatin and 5-fluorouracil which, on the other hand, was not cross-resistant to the other cytotoxic drugs. The activity of sunitinib did not obviously correlate to that of the standard drugs. Conclusion Ex vivo assessment of drug sensitivity of tumor cells from patients with GC is feasible and may provide information that could be useful for selection of drugs for treatment. Drug sensitivity varies considerably between and within individual samples arguing for individualized selection of drugs for chemotherapy.
Collapse
Affiliation(s)
- Bo Hultman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| | - Haile Mahteme
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| | - Magnus Sundbom
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| | - Martin Ljungman
- Surgery Department, Västmanlands Hospital, SE-721 89, Västerås, Sweden.
| | - Rolf Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| | - Peter Nygren
- Department of Radiology, Oncology and Radiation Sciences, Uppsala University, SE-751 85, Uppsala, Sweden.
| |
Collapse
|
42
|
Abdel-Rahman O. Targeting platelet-derived growth factor (PDGF) signaling in gastrointestinal cancers: preclinical and clinical considerations. Tumour Biol 2014; 36:21-31. [DOI: 10.1007/s13277-014-2797-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 10/30/2014] [Indexed: 12/16/2022] Open
|
43
|
Vita FD, Martino ND, Fabozzi A, Laterza MM, Ventriglia J, Savastano B, Petrillo A, Gambardella V, Sforza V, Marano L, Auricchio A, Galizia G, Ciardiello F, Orditura M. Clinical management of advanced gastric cancer: the role of new molecular drugs. World J Gastroenterol 2014; 20:14537-58. [PMID: 25356019 PMCID: PMC4209522 DOI: 10.3748/wjg.v20.i40.14537] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/19/2014] [Accepted: 06/02/2014] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is the fourth most common malignant neoplasm and the second leading cause of death for cancer in Western countries with more than 20000 new cases yearly diagnosed in the United States. Surgery represents the main approach for this disease but, notwithstanding the advances in surgical techniques, we observed a minimal improvement in terms of overall survival with a significant increasing of relapsing disease rates. Despite the development of new drugs has significantly improved the effectiveness of chemotherapy, the prognosis of patients with unresectable or metastatic gastric adenocarcinoma remains poor. Recently, several molecular target agents have been investigated; in particular, trastuzumab represents the first target molecule showing improvements in overall survival in human epithelial growth factor 2-positive gastric cancer patients. New molecules targeting vascular epithelial growth factor, mammalian target of rapamycin, and anti hepatocyte growth factor-c-Met pathway are also under investigation, with interesting results. Anyway, it seems necessary to select more accurately the population to treat with new agents by the identification of new biomarkers in order to optimize the results. In this paper we review the actual "scenario" of targeted treatments, also focusing on the new agents in development for gastric cancer and gastro-esophageal carcinoma, discussing their efficacy and potential applications in clinical practice.
Collapse
|
44
|
Moehler M, Maderer A, Schimanski C, Kanzler S, Denzer U, Kolligs FT, Ebert MP, Distelrath A, Geissler M, Trojan J, Schütz M, Berie L, Sauvigny C, Lammert F, Lohse A, Dollinger MM, Lindig U, Duerr EM, Lubomierski N, Zimmermann S, Wachtlin D, Kaiser AK, Schadmand-Fischer S, Galle PR, Woerns M. Gemcitabine plus sorafenib versus gemcitabine alone in advanced biliary tract cancer: a double-blind placebo-controlled multicentre phase II AIO study with biomarker and serum programme. Eur J Cancer 2014; 50:3125-35. [PMID: 25446376 DOI: 10.1016/j.ejca.2014.09.013] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/13/2014] [Accepted: 09/22/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND Since sorafenib has shown activity in different tumour types and gemcitabine regimens improved the outcome for biliary tract cancer (BTC) patients, we evaluated first-line gemcitabine plus sorafenib in a double-blind phase II study. PATIENTS AND METHODS 102 unresectable or metastatic BTC patients with histologically proven adenocarcinoma of gallbladder or intrahepatic bile ducts, Eastern Cooperative Oncology Group (ECOG) 0-2 were randomised to gemcitabine (1000 mg/m2 once weekly, first 7-weeks+1-week rest followed by once 3-weeks+1-week rest) plus sorafenib (400 mg twice daily) or placebo. Treatment continued until progression or unacceptable toxicity. Tumour samples were prospectively stained for sorafenib targets and potential biomarkers. Serum samples (first two cycles) were measured for vascular endothelial growth factors (VEGFs), vascular endothelial growth factor receptor 2 (VEGFR-2) and stromal cell-derived factor 1 (SDF1)α by enzyme-linked immunosorbent assay (ELISA). RESULTS Gemcitabine plus sorafenib was generally well tolerated. Four and three patients achieved partial responses in the sorafenib and placebo groups, respectively. There was no difference in the primary end-point, median progression-free survival (PFS) for gemcitabine plus sorafenib versus gemcitabine plus placebo (3.0 versus 4.9 months, P=0.859), and no difference for median overall survival (OS) (8.4 versus 11.2 months, P=0.775). Patients with liver metastasis after resection of primary BTC survived longer with sorafenib (P=0.019) compared to placebo. Patients who developed hand-foot syndrome (HFS) showed longer PFS and OS than patients without HFS. Two sorafenib targets, VEGFR-2 and c-kit, were not expressed in BTC samples. VEGFR-3 and Hif1α were associated with lymph node metastases and T stage. Absence of PDGFRβ expression correlated with longer PFS. CONCLUSION The addition of sorafenib to gemcitabine did not demonstrate improved efficacy in advanced BTC patients. Biomarker subgroup analysis suggested that some patients might benefit from combined treatment.
Collapse
Affiliation(s)
- M Moehler
- Department of Internal Medicine, Johannes Gutenberg-University of Mainz, Mainz, Germany.
| | - A Maderer
- Department of Internal Medicine, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - C Schimanski
- Department of Internal Medicine, Marienhospital Darmstadt, Darmstadt, Germany
| | - S Kanzler
- 2nd Department of Medicine, Leopoldina Hospital, Schweinfurt, Germany
| | - U Denzer
- 1st Department of Medicine, University Hospital Hamburg, Hamburg, Germany
| | - F T Kolligs
- Department of Medicine II, University Hospital Munich, Munich, Germany
| | - M P Ebert
- 2nd Department of Medicine, University Hospital Mannheim, Mannheim, Germany
| | - A Distelrath
- Tumor Department, Hospital Fulda, Fulda, Germany
| | - M Geissler
- Department of Internal Medicine, Hospital Esslingen, Esslingen, Germany
| | - J Trojan
- Department of Internal Medicine I, University Hospital Frankfurt, Frankfurt, Germany
| | - M Schütz
- Department of Internal Medicine, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - L Berie
- Department of Internal Medicine, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - C Sauvigny
- Department of Internal Medicine, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - F Lammert
- Department of Internal Medicine II, University Hospital Homburg, Homburg, Germany
| | - A Lohse
- 1st Department of Medicine, University Hospital Hamburg, Hamburg, Germany
| | - M M Dollinger
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - U Lindig
- Department of Internal Medicine II, University Hospital Jena, Jena, Germany
| | - E M Duerr
- Department of Medicine II, University Hospital Munich, Munich, Germany
| | - N Lubomierski
- Department of Internal Medicine I, University Hospital Frankfurt, Frankfurt, Germany
| | - S Zimmermann
- Department of Internal Medicine II, University Hospital Homburg, Homburg, Germany
| | - D Wachtlin
- Interdisciplinary Center for Clinical Trials of the University Medical Center Mainz, Germany
| | - A-K Kaiser
- Interdisciplinary Center for Clinical Trials of the University Medical Center Mainz, Germany
| | - S Schadmand-Fischer
- Department of Radiology, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - P R Galle
- Department of Internal Medicine, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - M Woerns
- Department of Internal Medicine, Johannes Gutenberg-University of Mainz, Mainz, Germany
| | | |
Collapse
|
45
|
Aoyagi K, Kouhuji K, Kizaki J, Isobe T, Hashimoto K, Shirouzu K. Molecular targeting to treat gastric cancer. World J Gastroenterol 2014; 20:13741-55. [PMID: 25320512 PMCID: PMC4194558 DOI: 10.3748/wjg.v20.i38.13741] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 01/13/2014] [Accepted: 05/23/2014] [Indexed: 02/06/2023] Open
Abstract
Trastuzumab that targets human epidermal growth factor receptor 2 (HER2) protein is the only approved molecular targeting agent for treating gastric cancer in Japan and the outcomes have been favorable. However, trastuzumab is effective for only 10% to 20% of the population with gastric cancer that expresses HER2 protein. Molecular targeting therapy with bevacizumab against vascular endothelial growth factors (VEGF) and with cetuximab and panitumumab against the epidermal growth factors pathway that have been approved for treating colorectal cancer are not considered effective for treating gastric cancer according to several clinical trials. However, ramucirumab that targets VEGF receptor-2 prolonged overall survival in a large phase III clinical trial and it might be an effective molecular targeting therapy for gastric cancer. The significance of molecular targeting therapy for gastric cancer remains controversial. A large-scale randomized clinical trial of novel molecular targeting agents with which to treat gastric cancer is needed.
Collapse
|
46
|
Al-Batran SE, Werner D. Recent advances and future trends in the targeted therapy of metastatic gastric cancer. Expert Rev Gastroenterol Hepatol 2014; 8:555-69. [PMID: 24665840 DOI: 10.1586/17474124.2014.902304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The better understanding of the molecular mechanisms behind gastric cancer has led to the development of new therapeutic strategies that are likely to improve patient outcomes in the near future. Recently, targeting the HER2 and the VEGF pathways with trastuzumab and ramucirumab, respectively, have been found to improve survival, while directed therapies against a number of other pathways are under clinical evaluation. These include the hepatocyte growth factor and its receptor c-MET, the insulin-like growth factor 1, the fibroblast growth factor, the mammalian target of rapamycin (mTOR), the epidermal growth factor receptor, and other pathways, as well as relevant immunotherapeutic strategies. This article reviews recent advances and future trends of these concepts for gastric cancer and adenocarcinoma of the gastroesophageal junction.
Collapse
Affiliation(s)
- Salah-Eddin Al-Batran
- Krankenhaus Nordwest, UCT-University Cancer Center Frankfurt, Frankfurt am Main, Germany
| | | |
Collapse
|
47
|
Thomaidis T, Maderer A, Al-Batran SE, Kany J, Pauligk C, Steinmetz K, Schad A, Hofheinz R, Schmalenberg H, Homann N, Galle PR, Moehler M. VEGFR-3 and CXCR4 as predictive markers for treatment with fluorouracil, leucovorin plus either oxaliplatin or cisplatin in patients with advanced esophagogastric cancer: a comparative study of the Arbeitsgemeinschaft Internistische Onkologie (AIO). BMC Cancer 2014; 14:476. [PMID: 24981311 PMCID: PMC4094395 DOI: 10.1186/1471-2407-14-476] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 06/18/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Combination of fluoropyrimidines and a platinum derivative are currently standards for systemic chemotherapy in advanced adenocarcinoma of the stomach and gastroesophageal junction (GEJ). Nevertheless, individual likelihood for response to these therapeutic regimes remains uncertain. Even more, no predictive markers are available to determine which patients may benefit more from oxaliplatin versus cisplatin or vice versa. The new invasion and stem cell markers VEGFR-3 and CXCR4 have been linked prognostically with more aggressive esophagogastric cancer types. Thus, we aimed to assess correlations of VEGFR-3 and CXCR4 expression levels with clinical outcome in a randomized phase III study of patients with oxaliplatin/leucovorin/5-FU (FLO) versus cisplatin/leucovorin/5-FU (FLP). METHODS The patients data examined in this study (n = 72) were from the collective of the FLO vs. FLP phase III AIO trial. Tumour tissues were stained via immunohistochemistry for VEGFR-3 and CXCR4 expression and results were evaluated by two independent, blinded investigators.Outcome parameter: Survival analysis was calculated for patients receiving FLO vs. FLP in relation to VEGFR-3 and CXCR4 expression. RESULTS 54% and 36% of the examined tumour tissues showed strong positive expression of VEGFR-3 and CXCR4 respectively. No superiority of each regime was detected in terms of overall survival (OS) in the whole population. Patients with strong expression of CXCR4 on their tumour tissues profited more in terms of OS under the treatment of FLP (mOS: 28 vs 15 months, p = 0.05 respectively). Patients with negative VEGFR-3 and CXCR4 expression had a trend to live longer when FLO regime was applied (mOS: 22 vs. 9 months, p = 0.099 and 20 vs. 10 months, p = 0.073 respectively). In an exploratory analysis of patients older than 60 years at diagnosis, we observed a significant benefit in overall survival for VEGFR-3 and CXCR4-positive patients when treated with FLP (p = 0.002, p = 0.021 respectively). CONCLUSIONS CXCR4 positive patients profited in terms of OS from FLP, whereas FLO proved to be more effective in CXCR4 and VEGFR-3 negative patients. Our results suggest, despite the limited size of the study, a predictive value of these biomarkers concerning chemotherapy with FLP or FLO in advanced esophagogastric cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Markus Moehler
- I, Medical Department, Johannes-Gutenberg University of Mainz, Langenbeckstr,1, 55131 Mainz, Germany.
| |
Collapse
|
48
|
Prasad V, Massey PR, Fojo T. Oral anticancer drugs: how limited dosing options and dose reductions may affect outcomes in comparative trials and efficacy in patients. J Clin Oncol 2014; 32:1620-9. [PMID: 24711558 PMCID: PMC4026582 DOI: 10.1200/jco.2013.53.0204] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Historically, cancer medicine has avoided the problem of unequal dosing by comparing maximum-tolerated doses of intravenous regimens with proportionate dose reductions for toxicity. However, in recent years, with the development of numerous oral anticancer agents, dosing options are arbitrarily and increasingly limited by the size of pills. We contend that an underappreciated consequence of pill size is unequal dosing in comparative clinical trials and that this can have an impact on outcomes. We discuss how comparative effectiveness trials can be unbalanced and how the use of doses that are not sustainable might affect outcomes, especially marginal ones. We further argue that because of their poor tolerability and their limited dosing options, which often result in large dose adjustments in response to toxicity, the real-world clinical effectiveness of oral anticancer agents may be diminished and may not emulate results achieved in registration trials.
Collapse
Affiliation(s)
- Vinay Prasad
- Vinay Prasad and Tito Fojo, National Cancer Institute, National Institutes of Health, Bethesda, MD; and Paul R. Massey, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA.
| | - Paul R Massey
- Vinay Prasad and Tito Fojo, National Cancer Institute, National Institutes of Health, Bethesda, MD; and Paul R. Massey, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Tito Fojo
- Vinay Prasad and Tito Fojo, National Cancer Institute, National Institutes of Health, Bethesda, MD; and Paul R. Massey, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
49
|
Aprile G, Giampieri R, Bonotto M, Bittoni A, Ongaro E, Cardellino GG, Graziano F, Giuliani F, Fasola G, Cascinu S, Scartozzi M. The challenge of targeted therapies for gastric cancer patients: the beginning of a long journey. Expert Opin Investig Drugs 2014; 23:925-42. [PMID: 24806575 DOI: 10.1517/13543784.2014.912631] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Despite significant improvements in systemic chemotherapy over the last two decades, the prognosis of patients with advanced disease remains dismal. Collaborative, high-quality research and advances in high-throughput technologies have contributed to elucidate molecular pathways underpinning disease progression and have stimulated many clinical studies testing target therapies in the advanced disease setting. Although progress has been made thanks to trastuzumab in HER2 positive tumours, antiangiogenic drugs have produced conflicting results and EGFR-inhibitors have failed to show major improvements. AREAS COVERED While commenting on the results of many key Phase III randomized trials, the Authors discuss the most promising classes of novel targeted agents and present the current challenges toward a customized treatment. EXPERT OPINION Palliative chemotherapy became the worldwide standard of care for patients with advanced gastric cancers, producing significant life prolongation and improvement of life quality. Nevertheless, long-term outcomes of those patients remain poor. Because of the encouraging advancement in novel targeted therapies, such a disappointing scenario is now evolving. While results serve as a springboard for future research, more comprehensive efforts are needed to clarify the biological mechanisms underpinning cancer progression and help clinicians to develop new effective treatments.
Collapse
Affiliation(s)
- Giuseppe Aprile
- University and General Hospital, Department of Medical Oncology , Piazzale S Maria della Misericordia 1, 33100, Udine , Italy +39 432 559308 ; +39 432 559305 ;
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
BACKGROUND Gastric cancer is the second most common cause of cancer-related deaths worldwide. There are large geographic variations in the incidence of these tumors, with 60% occurring in East Asia. For patients with resectable disease, surgery and perioperative treatment can be effective. For patients with advanced gastric cancer, chemotherapy regimens result in a median survival of 9-11 months. In general, the prognosis for advanced disease is poor and 5-year overall survival rates are around 15%. Combination therapies yield better survival rates, albeit with increased toxicity. Therefore, more effective and less toxic treatment regimens are needed. SUMMARY The molecular aberrations that characterize the different subgroups of gastric cancer have been used as therapeutic targets. However, the heterogeneity and complexity of gastric cancers is a major challenge for the development of effective targeted therapies. This review examines the main molecular targets in the treatment of gastric cancer, namely the vascular endothelial growth factor (VEGF), human epidermal growth factor receptor 2 (HER2), hepatocyte growth factor (HGF)/c-Met, epidermal growth factor receptor (EGFR) and phosphoinositide 3-kinase (PI3K)/Akt pathways. KEY MESSAGE The molecular aberrations characteristic of gastric cancer are being explored for the development of targeted therapies, including the VEGF, HER2, HGF/c-Met, EGFR and PI3K/Akt signaling pathways. PRACTICAL IMPLICATIONS Trastuzumab, an antibody which targets HER2, is the first approved targeted therapy for the treatment of gastric cancer. However, trastuzumab is only effective in HER2-positive tumors (about 10-20% of all gastric cancers). Ramucirumab, which targets the VEGF receptor 2, has yielded benefits with respect to overall survival in a phase III trial and is an effective treatment for advanced gastric cancer with approval in second-line treatment. Apatinib and rilotumumab are another two promising new agents currently under development.
Collapse
Affiliation(s)
- Nadine Schulte
- Department of Medicine II, University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | | | | |
Collapse
|