1
|
Veneziani AC, Gonzalez-Ochoa E, Alqaisi H, Madariaga A, Bhat G, Rouzbahman M, Sneha S, Oza AM. Heterogeneity and treatment landscape of ovarian carcinoma. Nat Rev Clin Oncol 2023; 20:820-842. [PMID: 37783747 DOI: 10.1038/s41571-023-00819-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 10/04/2023]
Abstract
Ovarian carcinoma is characterized by heterogeneity at the molecular, cellular and anatomical levels, both spatially and temporally. This heterogeneity affects response to surgery and/or systemic therapy, and also facilitates inherent and acquired drug resistance. As a consequence, this tumour type is often aggressive and frequently lethal. Ovarian carcinoma is not a single disease entity and comprises various subtypes, each with distinct complex molecular landscapes that change during progression and therapy. The interactions of cancer and stromal cells within the tumour microenvironment further affects disease evolution and response to therapy. In past decades, researchers have characterized the cellular, molecular, microenvironmental and immunological heterogeneity of ovarian carcinoma. Traditional treatment approaches have considered ovarian carcinoma as a single entity. This landscape is slowly changing with the increasing appreciation of heterogeneity and the recognition that delivering ineffective therapies can delay the development of effective personalized approaches as well as potentially change the molecular and cellular characteristics of the tumour, which might lead to additional resistance to subsequent therapy. In this Review we discuss the heterogeneity of ovarian carcinoma, outline the current treatment landscape for this malignancy and highlight potentially effective therapeutic strategies in development.
Collapse
Affiliation(s)
- Ana C Veneziani
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Eduardo Gonzalez-Ochoa
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Husam Alqaisi
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Ainhoa Madariaga
- Medical Oncology Department, 12 De Octubre University Hospital, Madrid, Spain
| | - Gita Bhat
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Marjan Rouzbahman
- Department of Laboratory Medicine and Pathobiology, Toronto General Hospital, Toronto, Ontario, Canada
| | - Suku Sneha
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Amit M Oza
- Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Lim ST, Choi HS, Kim K, Hahn S, Cho IJ, Noh H, Lee JI, Han A. Hounsfield Units Predict Survival of Patients With Estrogen Receptor-Positive and Human Epithelial Growth Factor Receptor 2-Negative Breast Cancer. Clin Breast Cancer 2023; 23:e424-e433.e3. [PMID: 37438195 DOI: 10.1016/j.clbc.2023.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUNDS Tumor vascularity plays a fundamental role in cancer progression, including breast cancer. This study aimed to elucidate tumor vascularity and its impact on patient survival in the context of breast cancer subtypes using Hounsfield units (HU) on contrast-enhanced computed tomography (CT). MATERIALS AND METHODS Patients with early-stage breast cancer who completed planned treatment between 2003 and 2013 were retrospectively assessed. RESULTS The final cohort comprised 440 patients. Of the 440 patients, 262 had estrogen receptor (ER)-positive disease and 119 had human epidermal growth factor receptor 2 (HER2)-overexpressing disease. The tumor-to-aorta ratio of Hounsfield units (TAR) was related to significantly worse recurrence-free interval (RFI) (P < .001) and overall survival (OS) (P < .001) in patients with TAR > 0.33 for RFI and > 0.35 for OS than their counterparts. In the subgroup analysis, the survival disadvantage was limited only to patients with ER-positive and HER2-negative disease (P < .001 for both RFI and OS). CONCLUSION This study showed that TAR, which reflects tumor vascularity, was significantly related to patients' RFI and OS, suggesting its capacity as a feasible biomarker. This study also showed that TAR was associated with the survival in patients with ER-positive and HER2-negative disease.
Collapse
Affiliation(s)
- Seung Taek Lim
- Department of Oncology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Hyang Suk Choi
- Department of Surgery, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Kwangmin Kim
- Department of Surgery, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Seok Hahn
- Department of Radiology, Inje University College of Medicine, Haeundae Paik Hospital, Busan, Korea
| | - In-Jeong Cho
- Department of Surgery, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Hany Noh
- Department of Surgery, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Jong-In Lee
- Department of Oncology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Airi Han
- Department of Surgery, Yonsei University Wonju College of Medicine, Wonju, Korea.
| |
Collapse
|
3
|
van Dijk E, van Werkhoven E, Asher R, Mooi JK, Espinoza D, van Essen HF, van Tinteren H, van Grieken NCT, Punt CJA, Tebbutt NC, Ylstra B. Predictive value of chromosome 18q11.2-q12.1 loss for benefit from bevacizumab in metastatic colorectal cancer; a post-hoc analysis of the randomized phase III-trial AGITG-MAX. Int J Cancer 2022; 151:1166-1174. [PMID: 35489024 PMCID: PMC9545440 DOI: 10.1002/ijc.34061] [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: 10/26/2021] [Revised: 03/07/2022] [Accepted: 04/12/2022] [Indexed: 11/24/2022]
Abstract
The VEGF‐A monoclonal antibody bevacizumab is currently recommended for first‐line treatment of all metastatic colorectal cancer (mCRC) patients. Cost‐benefit ratio and side‐effects however necessitate patient selection. A large retrospective yet nonrandomized study showed that patients with loss of chromosome 18q11.2‐q12.1 in the tumor and treated with bevacizumab have 3 months improved median progression‐free (PFS) and overall survival (OS) benefit compared to patients without this loss and/or treatment modality. Implementation for loss of chromosome 18q11.2‐q12.1 as a marker in clinical practice mandates evidence in a randomized controlled trial for bevacizumab. Of the trials with randomization of chemotherapy vs chemotherapy with bevacizumab, the AGITG‐MAX trial was the only one with tumor materials available. Chromosome 18q11.2‐q12.1 copy number status was measured for 256 AGITG‐MAX trial patients and correlated with PFS according to a predefined analysis plan with marker‐treatment interaction as the primary end‐point. Chromosome 18q11.2‐q12.1 losses were detected in 71% of patients (181/256) characteristic for mCRC. Consistent with the nonrandomized study, significant PFS benefit of bevacizumab was observed in patients with chromosome 18q11.2‐q12.1 loss (P = .009), and not in patients without 18q loss (P = .67). Although significance for marker‐treatment interaction was not reached (Pinteraction = .28), hazard ratio and 95% confidence interval of this randomized cohort (HRinteraction = 0.72; 95% CI = 0.39‐1.32) shows striking overlap with the nonrandomized study cohorts (HRinteraction = 0.41; 95% CI = 0.32‐0.8) supported by a nonsignificant Cochrane χ2 test (P = .11) for heterogeneity. We conclude that post hoc analysis of the AGITG‐MAX RCT provides supportive evidence for chromosome 18q11.2‐q12.1 as a predictive marker for bevacizumab in mCRC patients.
Collapse
Affiliation(s)
- Erik van Dijk
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands
| | - Erik van Werkhoven
- Biometrics Department, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Rebecca Asher
- Department of Biostatistics, NHMRC Clinical Trials Centre, University of Sydney, Camperdown, Australia
| | - Jennifer K Mooi
- Olivia Newton-John Cancer Research Institute, Heidelberg; Department of Medicine, University of Melbourne, Melbourne, Australia.,Peter MacCallum Cancer Institute, Melbourne, Australia
| | - David Espinoza
- Department of Biostatistics, NHMRC Clinical Trials Centre, University of Sydney, Camperdown, Australia
| | - Hendrik F van Essen
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands
| | - Harm van Tinteren
- Trial and Datacenter, Princess Máxima Center for pedeatric oncology, Utrecht, The Netherlands
| | - Nicole C T van Grieken
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands
| | - Cornelis J A Punt
- Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Epidemiology, University Medical Centre Utrecht, Utrecht University, Utrecht, Netherlands
| | - Niall C Tebbutt
- Department of Medical Oncology, Austin Health, Heidelberg, Australia.,Department of Surgery, University of Melbourne
| | - Bauke Ylstra
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
4
|
Nakamoto S, Watanabe J, Ohtani S, Morita S, Ikeda M. Bevacizumab as First-line Treatment for HER2-negative Advanced Breast Cancer: Paclitaxel plus Bevacizumab Versus Other Chemotherapy. In Vivo 2021; 34:1377-1386. [PMID: 32354934 DOI: 10.21873/invivo.11917] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The efficacy of paclitaxel and bevacizumab (PB) compared with other chemotherapies in patients with human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer is unclear. PATIENTS AND METHODS We retrospectively investigated 301 patients with HER2- ABC who received first-line chemotherapy from January 2011 to December 2016. RESULTS We included 114 patients who received PB and 187 patients who received other chemotherapies. After propensity score matching, the PB group showed a significantly superior overall response rate (77.8% vs. 38.9%, p<0.0001) and median time to treatment failure (7.3 vs. 5.9 months, p=0.035). In subgroup analyses, PB improved the median overall survival of patients with pleural lesions or pulmonary lymphangiopathy (not reached vs. 18.9 months, p=0.037), and of patients with three or more metastatic sites without liver metastases, (48.0 vs. 27.3 months, p=0.015). CONCLUSION Compared with conventional chemotherapy, PB improved the overall response rate and time to treatment failure in patients with HER2- advanced breast cancer and improved overall survival in some patient subgroups.
Collapse
Affiliation(s)
- Shogo Nakamoto
- Department of Breast Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | | | - Shoichiro Ohtani
- Department of Breast Surgery, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masahiko Ikeda
- Department of Breast and Thyroid Gland Surgery, Fukuyama City Hospital, Fukuyama, Japan
| |
Collapse
|
5
|
Haugen MH, Lingjærde OC, Hedenfalk I, Garred Ø, Borgen E, Loman N, Hatschek T, Børresen-Dale AL, Naume B, Mills GB, Mælandsmo GM, Engebraaten O. Protein Signature Predicts Response to Neoadjuvant Treatment With Chemotherapy and Bevacizumab in HER2-Negative Breast Cancers. JCO Precis Oncol 2021; 5:PO.20.00086. [PMID: 34036235 PMCID: PMC8140811 DOI: 10.1200/po.20.00086] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 10/21/2020] [Accepted: 12/07/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Antiangiogenic therapy using bevacizumab has proven effective for a number of cancers; however, in breast cancer (BC), there is an unmet need to identify patients who benefit from such treatment. PATIENTS AND METHODS In the NeoAva phase II clinical trial, patients (N = 132) with large (≥ 25 mm) human epidermal growth factor receptor 2 (HER2)-negative primary tumors were randomly assigned 1:1 to treatment with neoadjuvant chemotherapy (CTx) alone or in combination with bevacizumab (Bev plus CTx). The ratio of the tumor size after relative to before treatment was calculated into a continuous response scale. Tumor biopsies taken prior to neoadjuvant treatment were analyzed by reverse-phase protein arrays (RPPA) for expression levels of 210 BC-relevant (phospho-) proteins. Lasso regression was used to derive a predictor of tumor shrinkage from the expression of selected proteins prior to treatment. RESULTS We identified a nine-protein signature score named vascular endothelial growth factor inhibition response predictor (ViRP) for use in the Bev plus CTx treatment arm able to predict with accuracy pathologic complete response (pCR) (area under the curve [AUC] = 0.85; 95% CI, 0.74 to 0.97) and low residual cancer burden (RCB 0/I) (AUC = 0.80; 95% CI, 0.68 to 0.93). The ViRP score was significantly lower in patients with pCR (P < .001) and in patients with low RCB (P < .001). The ViRP score was internally validated on mRNA data and the resultant surrogate mRNA ViRP score significantly separated the pCR patients (P = .016). Similarly, the mRNA ViRP score was validated (P < .001) in an independent phase II clinical trial (PROMIX). CONCLUSION Our ViRP score, integrating the expression of nine proteins and validated on mRNA data both internally and in an independent clinical trial, may be used to increase the likelihood of benefit from treatment with bevacizumab combined with chemotherapy in patients with HER2-negative BC.
Collapse
Affiliation(s)
- Mads H Haugen
- Department of Tumor Biology, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Ole Christian Lingjærde
- Department of Genetics, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Department of Informatics-Biomedical Informatics, University of Oslo, Oslo, Norway.,K.G. Jebsen-Centre for B Cell Malignancies, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingrid Hedenfalk
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Øystein Garred
- Division of Laboratory Medicine-Pathology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Elin Borgen
- Division of Laboratory Medicine-Pathology, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Niklas Loman
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Skåne, Sweden
| | - Thomas Hatschek
- Department of Oncology-Pathology, Karolinska University Hospital, Stockholm, Sweden
| | - Anne-Lise Børresen-Dale
- Department of Genetics, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Bjørn Naume
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | - Gordon B Mills
- Department of Cell, Developmental and Cancer Biology, School of Medicine, Oregon Health Science University, Portland, OR
| | - Gunhild M Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Institute for Medical Biology, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, Tromsø, Norway
| | - Olav Engebraaten
- Department of Tumor Biology, Institute for Cancer Research, Division of Cancer Medicine, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
6
|
Madu CO, Wang S, Madu CO, Lu Y. Angiogenesis in Breast Cancer Progression, Diagnosis, and Treatment. J Cancer 2020; 11:4474-4494. [PMID: 32489466 PMCID: PMC7255381 DOI: 10.7150/jca.44313] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/04/2020] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is a significant event in a wide range of healthy and diseased conditions. This process frequently involves vasodilation and an increase in vascular permeability. Numerous players referred to as angiogenic factors, work in tandem to facilitate the outgrowth of endothelial cells (EC) and the consequent vascularity. Conversely, angiogenic factors could also feature in pathological conditions. Angiogenesis is a critical factor in the development of tumors and metastases in numerous cancers. An increased level of angiogenesis is associated with decreased survival in breast cancer patients. Therefore, a good understanding of the angiogenic mechanism holds a promise of providing effective treatments for breast cancer progression, thereby enhancing patients' survival. Disrupting the initiation and progression of this process by targeting angiogenic factors such as vascular endothelial growth factor (Vegf)-one of the most potent member of the VEGF family- or by targeting transcription factors, such as Hypoxia-Inducible Factors (HIFs) that act as angiogenic regulators, have been considered potential treatment options for several types of cancers. The objective of this review is to highlight the mechanism of angiogenesis in diseases, specifically its role in the progression of malignancy in breast cancer, as well as to highlight the undergoing research in the development of angiogenesis-targeting therapies.
Collapse
Affiliation(s)
- Chikezie O. Madu
- Departments of Biological Sciences, University of Memphis, Memphis, TN 38152. USA
| | - Stephanie Wang
- Departments of Biology and Advanced Placement Biology, White Station High School, Memphis, TN 38117. USA
| | - Chinua O. Madu
- Departments of Biology and Advanced Placement Biology, White Station High School, Memphis, TN 38117. USA
| | - Yi Lu
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN 38163. USA
| |
Collapse
|
7
|
de Miguel Pérez D, Rodriguez Martínez A, Ortigosa Palomo A, Delgado Ureña M, Garcia Puche JL, Robles Remacho A, Exposito Hernandez J, Lorente Acosta JA, Ortega Sánchez FG, Serrano MJ. Extracellular vesicle-miRNAs as liquid biopsy biomarkers for disease identification and prognosis in metastatic colorectal cancer patients. Sci Rep 2020; 10:3974. [PMID: 32132553 PMCID: PMC7055306 DOI: 10.1038/s41598-020-60212-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 11/05/2019] [Indexed: 12/11/2022] Open
Abstract
Disseminated disease is present in ≈50% of colorectal cancer patients upon diagnosis, being responsible for most of cancer deaths. Addition of biological drugs, as Bevacizumab, to chemotherapy, has increased progression free survival and overall survival of metastatic colorectal cancer (mCRC) patients. However, these benefits have been only reported in a small proportion of patients. To date, there are not biomarkers that could explain the heterogeneity of this disease and would help in treatment selection. Recent findings demonstrated that microRNAs (miRNAs) play an important role in cancer and they can be encapsulated with high stability into extracellular vesicles (EVs) that are released in biological fluids. EVs can act as cell-to-cell communicators, transferring genetic information, such as miRNAs. In this context, we aimed to investigate serum EV associated miRNAs (EV-miRNAs) as novel non-invasive biomarkers for the diagnosis and prognosis of Bevacizumab-treated mCRC patients. We observed that baseline miRNA-21 and 92a outperformed carcinoembryonic antigen levels in the diagnosis of our 44 mCRC patients, compared to 17 healthy volunteers. In addition, patients who died presented higher levels of miRNA-92a and 222 at 24 weeks. However, in the multivariate Cox analysis, higher levels of miRNA-222 at 24 weeks were associated with lower overall survival. Altogether, these data indicate that EV-miRNAs have a strong potential as liquid biopsy biomarkers for the identification and prognosis of mCRC.
Collapse
Affiliation(s)
- Diego de Miguel Pérez
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and metastasis research group, PTS Granada, Avenida de la Ilustración 114, 18016, Granada, Spain.,Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine, University of Granada, Granada, Spain
| | - Alba Rodriguez Martínez
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and metastasis research group, PTS Granada, Avenida de la Ilustración 114, 18016, Granada, Spain.,Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine, University of Granada, Granada, Spain
| | - Alba Ortigosa Palomo
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and metastasis research group, PTS Granada, Avenida de la Ilustración 114, 18016, Granada, Spain
| | - Mayte Delgado Ureña
- Integral Oncology Division, University Hospital Virgen de las Nieves, IBS Granada, Instituto de Investigación Biosanitaria de Granada, 18012, Granada, Spain
| | - Jose Luis Garcia Puche
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and metastasis research group, PTS Granada, Avenida de la Ilustración 114, 18016, Granada, Spain.,Integral Oncology Division, University Hospital Virgen de las Nieves, IBS Granada, Instituto de Investigación Biosanitaria de Granada, 18012, Granada, Spain
| | - Agustín Robles Remacho
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and metastasis research group, PTS Granada, Avenida de la Ilustración 114, 18016, Granada, Spain
| | - José Exposito Hernandez
- Integral Oncology Division, University Hospital Virgen de las Nieves, IBS Granada, Instituto de Investigación Biosanitaria de Granada, 18012, Granada, Spain
| | - Jose Antonio Lorente Acosta
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and metastasis research group, PTS Granada, Avenida de la Ilustración 114, 18016, Granada, Spain.,Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine, University of Granada, Granada, Spain
| | - Francisco Gabriel Ortega Sánchez
- Balearic Islands Health Research Institute (IdISBa), 07010, Palma de Mallorca, Spain. .,Laboratory of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Ma Jose Serrano
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and metastasis research group, PTS Granada, Avenida de la Ilustración 114, 18016, Granada, Spain. .,Integral Oncology Division, University Hospital Virgen de las Nieves, IBS Granada, Instituto de Investigación Biosanitaria de Granada, 18012, Granada, Spain.
| |
Collapse
|
8
|
Sundar R, Tan IBH, Chee CE. Negative Predictive Biomarkers in Colorectal Cancer: PRESSING Ahead. J Clin Oncol 2019; 37:3066-3068. [DOI: 10.1200/jco.19.01977] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Raghav Sundar
- National University Health System, Singapore
- Duke-National University of Singapore Medical School, Singapore
- National University of Singapore, Singapore
| | - Iain Bee Huat Tan
- Duke-National University of Singapore Medical School, Singapore
- National Cancer Centre Singapore, Singapore
- Agency for Science, Technology and Research (A*STAR), Genome Institute of Singapore, Singapore
| | - Cheng E. Chee
- National University Health System, Singapore
- National University of Singapore, Singapore
| |
Collapse
|
9
|
Colombo N, Sessa C, Bois AD, Ledermann J, McCluggage WG, McNeish I, Morice P, Pignata S, Ray-Coquard I, Vergote I, Baert T, Belaroussi I, Dashora A, Olbrecht S, Planchamp F, Querleu D. ESMO-ESGO consensus conference recommendations on ovarian cancer: pathology and molecular biology, early and advanced stages, borderline tumours and recurrent disease. Int J Gynecol Cancer 2019; 29:ijgc-2019-000308. [PMID: 31048403 DOI: 10.1136/ijgc-2019-000308] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/25/2019] [Indexed: 12/29/2022] Open
Abstract
The development of guidelines is one of the core activities of the European Society for Medical Oncology (ESMO) and European Society of Gynaecologial Oncology (ESGO), as part of the mission of both societies to improve the quality of care for patients with cancer across Europe. ESMO and ESGO jointly developed clinically relevant and evidence-based recommendations in several selected areas in order to improve the quality of care for women with ovarian cancer. The ESMO-ESGO consensus conference on ovarian cancer was held on April 12-14, 2018 in Milan, Italy, and comprised a multidisciplinary panel of 40 leading experts in the management of ovarian cancer. Before the conference, the expert panel worked on five clinically relevant questions regarding ovarian cancer relating to each of the following four areas: pathology and molecular biology, early-stage and borderline tumours, advanced stage disease and recurrent disease. Relevant scientific literature, as identified using a systematic search, was reviewed in advance. During the consensus conference, the panel developed recommendations for each specific question and a consensus was reached. The recommendations presented here are thus based on the best available evidence and expert agreement. This article presents the recommendations of this ESMO-ESGO consensus conference, together with a summary of evidence supporting each recommendation.
Collapse
Affiliation(s)
- N Colombo
- Division of Medical Gynecologic Oncology, European Institute of Oncology IRCCS, University of Milan-Bicocca, Milan, Italy
| | - C Sessa
- Department of Medical Oncology, Oncology Institute of Southern Switzerland, Ospedale San Giovanni, Bellinzona, Switzerland
| | - A du Bois
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Essen, Germany
| | - J Ledermann
- Department of Oncology and Cancer Trials, UCL Cancer Institute, London, UK
| | - W G McCluggage
- Department of Pathology, Belfast Health and Social Care Trust, Belfast, UK
| | - I McNeish
- Department of Surgery and Cancer, Imperial College, London, UK
| | - P Morice
- Department of Gynecologic Surgery, Gustave Roussy Cancer Campus, Villejuif, France
| | - S Pignata
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale', Naples, Italy
| | - I Ray-Coquard
- Department of Medical and Surgical Oncology, Centre Léon Bérard, Lyon, France
| | - I Vergote
- Department of Gynaecological Oncology, Leuven Cancer Institute, Leuven, Belgium
- Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium
| | - T Baert
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Essen, Germany
| | - I Belaroussi
- Department of Gynecologic Surgery, Gustave Roussy Cancer Campus, Villejuif, France
| | - A Dashora
- Department of Cellular Pathology, Maidstone and Tunbridge Wells NHS Trust, Kent, UK
| | - S Olbrecht
- Department of Gynaecological Oncology, Leuven Cancer Institute, Leuven, Belgium
- Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium
| | - F Planchamp
- Clinical Research Unit, Institut Bergonié, Bordeaux, France
| | - D Querleu
- Department of Surgery, Institut Bergonié, Bordeaux, France
| |
Collapse
|
10
|
Colombo N, Sessa C, du Bois A, Ledermann J, McCluggage WG, McNeish I, Morice P, Pignata S, Ray-Coquard I, Vergote I, Baert T, Belaroussi I, Dashora A, Olbrecht S, Planchamp F, Querleu D. ESMO-ESGO consensus conference recommendations on ovarian cancer: pathology and molecular biology, early and advanced stages, borderline tumours and recurrent disease†. Ann Oncol 2019; 30:672-705. [PMID: 31046081 DOI: 10.1093/annonc/mdz062] [Citation(s) in RCA: 634] [Impact Index Per Article: 126.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023] Open
Abstract
The development of guidelines recommendations is one of the core activities of the European Society for Medical Oncology (ESMO) and European Society of Gynaecologial Oncology (ESGO), as part of the mission of both societies to improve the quality of care for patients with cancer across Europe. ESMO and ESGO jointly developed clinically relevant and evidence-based recommendations in several selected areas in order to improve the quality of care for women with ovarian cancer. The ESMO-ESGO consensus conference on ovarian cancer was held on 12-14 April 2018 in Milan, Italy, and comprised a multidisciplinary panel of 40 leading experts in the management of ovarian cancer. Before the conference, the expert panel worked on five clinically relevant questions regarding ovarian cancer relating to each of the following four areas: pathology and molecular biology, early-stage and borderline tumours, advanced stage disease and recurrent disease. Relevant scientific literature, as identified using a systematic search, was reviewed in advance. During the consensus conference, the panel developed recommendations for each specific question and a consensus was reached. The recommendations presented here are thus based on the best available evidence and expert agreement. This article presents the recommendations of this ESMO-ESGO consensus conference, together with a summary of evidence supporting each recommendation.
Collapse
Affiliation(s)
- N Colombo
- Division of Medical Gynecologic Oncology, European Institute of Oncology IRCCS, University of Milan-Bicocca, Milan, Italy.
| | - C Sessa
- Department of Medical Oncology, Oncology Institute of Southern Switzerland, Ospedale San Giovanni, Bellinzona, Switzerland
| | - A du Bois
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Essen, Germany
| | - J Ledermann
- Department of Oncology and Cancer Trials, UCL Cancer Institute, London
| | - W G McCluggage
- Department of Pathology, Belfast Health and Social Care Trust, Belfast
| | - I McNeish
- Department of Surgery and Cancer, Imperial College, London, UK
| | - P Morice
- Department of Gynecologic Surgery, Gustave Roussy Cancer Campus, Villejuif, France
| | - S Pignata
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori IRCCS 'Fondazione G. Pascale', Naples, Italy
| | - I Ray-Coquard
- Department of Medical and Surgical Oncology, Centre Léon Bérard, Lyon, France
| | - I Vergote
- Department of Gynaecological Oncology, Leuven Cancer Institute, Leuven; Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium
| | - T Baert
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Essen, Germany
| | - I Belaroussi
- Department of Gynecologic Surgery, Gustave Roussy Cancer Campus, Villejuif, France
| | - A Dashora
- Department of Cellular Pathology, Maidstone and Tunbridge Wells NHS Trust, Kent, UK
| | - S Olbrecht
- Department of Gynaecological Oncology, Leuven Cancer Institute, Leuven; Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium
| | | | - D Querleu
- Department of Surgery, Institut Bergonié, Bordeaux, France.
| |
Collapse
|
11
|
Jiang C, Huang YH, Lu JB, Yang YZ, Rao HL, Zhang B, He WZ, Xia LP. Perivascular cell coverage of intratumoral vasculature is a predictor for bevacizumab efficacy in metastatic colorectal cancer. Cancer Manag Res 2018; 10:3589-3597. [PMID: 30271207 PMCID: PMC6149904 DOI: 10.2147/cmar.s172261] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Purpose Tumor vessels supported by perivascular cells have been implicated in the failure of some anti-angiogenic agents. The relationship between perivascular cell coverage (PC) and bevacizumab efficacy in metastatic colorectal cancer (mCRC) was analyzed. Patients and methods A total of 284 consecutive mCRC patients who received first-line chemotherapy with or without bevacizumab from 2007-2014 in Sun Yat-Sen University Cancer Center were analyzed. Immunohistochemical double-stain for the perivascular cell marker alpha-smooth muscle actin and endothelial cell (cluster of differentiation 31) was performed to characterize the intratumoral microvascular density. Multispectral image capturing and computerized image analyses were used to quantify the microvessels supported by the perivascular cells. The patients were divided into high and low PC group according to a median cutoff value of 0.55. Results No significant differences in overall survival (OS) and progression-free survival (PFS) were noted between the high and low PC group. In the low PC group, the patients with bevacizumab treatment had favorable OS (P=0.03), but without PFS benefit. In the high PC group, neither OS nor PFS was significantly different between the B+C and C subgroup. Tumors with perineural invasion had high PC (P=0.03). Conclusion The data showed that a low PC value could be a predictor for bevacizumab benefit.
Collapse
Affiliation(s)
- Chang Jiang
- VIP Region, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, People's Republic of China,
| | - Yu-Hua Huang
- Department of Pathology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, People's Republic of China
| | - Jia-Bin Lu
- Department of Pathology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, People's Republic of China
| | - Yuan-Zhong Yang
- Department of Pathology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, People's Republic of China
| | - Hui-Lan Rao
- Department of Pathology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, People's Republic of China
| | - Bei Zhang
- VIP Region, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, People's Republic of China,
| | - Wen-Zhuo He
- VIP Region, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, People's Republic of China,
| | - Liang-Ping Xia
- VIP Region, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, People's Republic of China,
| |
Collapse
|
12
|
Delgado-Ureña M, Ortega FG, de Miguel-Pérez D, Rodriguez-Martínez A, García-Puche JL, Ilyine H, Lorente JA, Exposito-Hernandez J, Garrido-Navas MC, Delgado-Ramirez M, Serrano MJ. Circulating tumor cells criteria (CyCAR) versus standard RECIST criteria for treatment response assessment in metastatic colorectal cancer patients. J Transl Med 2018; 16:251. [PMID: 30189880 PMCID: PMC6127986 DOI: 10.1186/s12967-018-1624-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/29/2018] [Indexed: 02/06/2023] Open
Abstract
Background The use of circulating tumor cells (CTCs) as indicators of treatment response in metastatic colorectal cancer (mCRC) needs to be clarified. The objective of this study is to compare the Response Evaluation Criteria in Solid Tumors (RECIST) with the Cytologic Criteria Assessing Response (CyCAR), based on the presence and phenotypic characterization of CTCs, as indicators of FOLFOX–bevacizumab treatment response. Methods 77 mCRC blood samples from FOLFOX–bevacizumab treated patients were analyzed to isolate CTCs before and after (12 and 24 weeks) treatment, using an immunomagnetic separation method. VEGFR expression was identified by double immunostaining. Results We observed a decrease of CTCs (42.8 vs. 18.2%) and VEGFR positivity (69.7% vs. 41.7%) after treatment. According to RECIST, 6.45% of the patients did not show any clinical benefit, whereas 93.55% patients showed a favorable response at 12 weeks. According to CyCAR, 29% had a non-favorable response and 71% patients did not. No significant differences were found between the response assessment by RECIST and CyCAR at 12 or 24 weeks. However, in the multivariate analysis, RECIST at 12 weeks and CyCAR at 24 weeks were independent prognostic factors for OS (HR: 0.1, 95% CI 0.02–0.58 and HR: 0.35, 95% CI 0.12–0.99 respectively). Conclusions CyCAR results were comparable to RECIST in evaluating the response in mCRC and can be used as an alternative when the limitation of RECIST requires additional response analysis techniques.
Collapse
Affiliation(s)
- Mayte Delgado-Ureña
- Integral Oncology Division, Clinical University Hospital, Av. Dr. Olóriz 16, 18012, Granada, Spain
| | - Francisco G Ortega
- Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS, Granada, Avenida de la Ilustración, 114, 18016, Granada, Spain
| | - Diego de Miguel-Pérez
- Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS, Granada, Avenida de la Ilustración, 114, 18016, Granada, Spain.,Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine, University of Granada, Avenida de la Investigación, 11, 18071, Granada, Spain
| | - Alba Rodriguez-Martínez
- Integral Oncology Division, Clinical University Hospital, Av. Dr. Olóriz 16, 18012, Granada, Spain.,Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS, Granada, Avenida de la Ilustración, 114, 18016, Granada, Spain.,Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine, University of Granada, Avenida de la Investigación, 11, 18071, Granada, Spain.,DestiNA Genomics Ltd, 7-11 Melville St, Edinburgh, EH3 7PE, UK.,Division of Preventive Medicine and Public Health, CIBERESP, University of Jaen, Campus de las Lagunillas, 23072, Jaén, Spain
| | - Jose L García-Puche
- Integral Oncology Division, Clinical University Hospital, Av. Dr. Olóriz 16, 18012, Granada, Spain.,Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS, Granada, Avenida de la Ilustración, 114, 18016, Granada, Spain
| | - Hugh Ilyine
- DestiNA Genomics Ltd, 7-11 Melville St, Edinburgh, EH3 7PE, UK
| | - Jose A Lorente
- Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS, Granada, Avenida de la Ilustración, 114, 18016, Granada, Spain.,Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine, University of Granada, Avenida de la Investigación, 11, 18071, Granada, Spain
| | - Jose Exposito-Hernandez
- Integral Oncology Division, Clinical University Hospital, Av. Dr. Olóriz 16, 18012, Granada, Spain
| | - M Carmen Garrido-Navas
- Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS, Granada, Avenida de la Ilustración, 114, 18016, Granada, Spain
| | - Miguel Delgado-Ramirez
- Division of Preventive Medicine and Public Health, CIBERESP, University of Jaen, Campus de las Lagunillas, 23072, Jaén, Spain
| | - M José Serrano
- Integral Oncology Division, Clinical University Hospital, Av. Dr. Olóriz 16, 18012, Granada, Spain. .,Liquid Biopsy and Metastasis Research Group, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS, Granada, Avenida de la Ilustración, 114, 18016, Granada, Spain.
| |
Collapse
|
13
|
van Dijk E, Biesma HD, Cordes M, Smeets D, Neerincx M, Das S, Eijk PP, Murphy V, Barat A, Bacon O, Prehn JHM, Betge J, Gaiser T, Fender B, Meijer GA, McNamara DA, Klinger R, Koopman M, Ebert MPA, Kay EW, Hennessey BT, Verheul HMW, Gallagher WM, O'Connor DP, Punt CJA, Loupakis F, Lambrechts D, Byrne AT, van Grieken NCT, Ylstra B. Loss of Chromosome 18q11.2-q12.1 Is Predictive for Survival in Patients With Metastatic Colorectal Cancer Treated With Bevacizumab. J Clin Oncol 2018; 36:2052-2060. [PMID: 29792754 DOI: 10.1200/jco.2017.77.1782] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Patients with metastatic colorectal cancer (mCRC) have limited benefit from the addition of bevacizumab to standard chemotherapy. However, a subset probably benefits substantially, highlighting an unmet clinical need for a biomarker of response to bevacizumab. Previously, we demonstrated that losses of chromosomes 5q34, 17q12, and 18q11.2-q12.1 had a significant correlation with progression-free survival (PFS) in patients with mCRC treated with bevacizumab in the CAIRO2 clinical trial but not in patients who did not receive bevacizumab in the CAIRO trial. This study was designed to validate these findings. Materials and Methods Primary mCRC samples were analyzed from two cohorts of patients who received bevacizumab as first-line treatment; 96 samples from the European multicenter study Angiopredict (APD) and 81 samples from the Italian multicenter study, MOMA. A third cohort of 90 samples from patients with mCRC who did not receive bevacizumab was analyzed. Copy number aberrations of tumor biopsy specimens were measured by shallow whole-genome sequencing and were correlated with PFS, overall survival (OS), and response. Results Loss of chromosome 18q11.2-q12.1 was associated with prolonged PFS most significantly in both the cohorts that received bevacizumab (APD: hazard ratio, 0.54; P = .01; PFS difference, 65 days; MOMA: hazard ratio, 0.55; P = .019; PFS difference, 49 days). A similar association was found for OS and overall response rate in these two cohorts, which became significant when combined with the CAIRO2 cohort. Median PFS in the cohort of patients with mCRC who did not receive bevacizumab and in the CAIRO cohort was similar to that of the APD, MOMA, and CAIRO2 patients without an 18q11.2-q12.1 loss. Conclusion We conclude that the loss of chromosome 18q11.2-q12.1 is consistently predictive for prolonged PFS in patients receiving bevacizumab. The predictive value of this loss is substantiated by a significant gain in OS and overall response rate.
Collapse
Affiliation(s)
- Erik van Dijk
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Hedde D Biesma
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Martijn Cordes
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Dominiek Smeets
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Maarten Neerincx
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Sudipto Das
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Paul P Eijk
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Verena Murphy
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Anna Barat
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Orna Bacon
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Jochen H M Prehn
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Johannes Betge
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Timo Gaiser
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Bozena Fender
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Gerrit A Meijer
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Deborah A McNamara
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Rut Klinger
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Miriam Koopman
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Matthias P A Ebert
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Elaine W Kay
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Bryan T Hennessey
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Henk M W Verheul
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - William M Gallagher
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Darran P O'Connor
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Cornelis J A Punt
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Fotios Loupakis
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Diether Lambrechts
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Annette T Byrne
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Nicole C T van Grieken
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| | - Bauke Ylstra
- Erik van Dijk, Hedde D. Biesma, Martijn Cordes, Maarten Neerincx, Paul P. Eijk, Henk M.W. Verheul, Nicole C.T. van Grieken, and Bauke Ylstra, Vrije Universiteit Medical Center; Gerrit A. Meijer, Netherlands Cancer Institute; Cornelis J.A. Punt, Academic Medical Center, Amsterdam; Miriam Koopman, University Medical Center Utrecht, Utrecht, the Netherlands; Dominiek Smeets and Diether Lambrechts, KU Leuven, Leuven, Belgium; Sudipto Das, Orna Bacon, Jochen H.M. Prehn, Bryan T. Hennessey, Darran P. O'Connor, and Annette T. Byrne, Royal College of Surgeons in Ireland; Verena Murphy, Cancer Trials Ireland; Anna Barat, Deborah A. McNamara, and Elaine W. Kay, Beaumont Hospital; Bozena Fender and William M. Gallagher, OncoMark Ltd; Rut Klinger and William M. Gallagher, University College Dublin, Dublin, United Kingdom; Johannes Betge, Timo Gaiser, and Matthias P.A. Ebert, University Hospital Mannheim, Mannheim, Germany; and Fotios Loupakis, Veneto Institute of Oncology IOV-IRCCS Padua, Italy
| |
Collapse
|
14
|
Abstract
Vascular endothelial growth factor (VEGF) has been identified as the most potent cytokine involved in tumor angiogenesis and metastasis formation. Clinical results of anti-angiogenic therapies targeting VEGF and its receptors are very modest, resulting in a moderate improvement of overall survival. The clinical outcome is associated with the development of resistance and the increased risk of invasion and metastasis. In this article, I have analyzed the principal mechanisms of resistance to VEGF pathway inhibitors, including normalization of tumor blood vessels, hypoxia, recruitment of inflammatory cells and immature myeloid cells, alternative mechanisms of tumor vessel formation, genomic instability of tumor endothelial cells. In this context, the concept and strategies of anti-angiogenic therapies should be extensively re-considered and re-evaluated. In particular, rational combinations of anti-angiogenic agents based on pharmacokinetic and pharmacodynamics data are needed to overcome resistance and it is extremely important to determine the optimal duration and scheduling of anti-VEGF agents.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.,National Cancer Institute "Giovanni Paolo II", Bari, Italy
| |
Collapse
|
15
|
Prediction of novel target genes and pathways involved in bevacizumab-resistant colorectal cancer. PLoS One 2018; 13:e0189582. [PMID: 29342159 PMCID: PMC5771567 DOI: 10.1371/journal.pone.0189582] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 11/29/2017] [Indexed: 12/22/2022] Open
Abstract
Bevacizumab combined with cytotoxic chemotherapy is the backbone of metastatic colorectal cancer (mCRC) therapy; however, its treatment efficacy is hampered by therapeutic resistance. Therefore, understanding the mechanisms underlying bevacizumab resistance is crucial to increasing the therapeutic efficacy of bevacizumab. The Gene Expression Omnibus (GEO) database (dataset, GSE86525) was used to identify the key genes and pathways involved in bevacizumab-resistant mCRC. The GEO2R web tool was used to identify differentially expressed genes (DEGs). Functional and pathway enrichment analyses of the DEGs were performed using the Database for Annotation, Visualization, and Integrated Discovery(DAVID). Protein–protein interaction (PPI) networks were established using the Search Tool for the Retrieval of Interacting Genes/Proteins database(STRING) and visualized using Cytoscape software. A total of 124 DEGs were obtained, 57 of which upregulated and 67 were downregulated. PPI network analysis showed that seven upregulated genes and nine downregulated genes exhibited high PPI degrees. In the functional enrichment, the DEGs were mainly enriched in negative regulation of phosphate metabolic process and positive regulation of cell cycle process gene ontologies (GOs); the enriched pathways were the phosphoinositide 3-kinase-serine/threonine kinase signaling pathway, bladder cancer, and microRNAs in cancer. Cyclin-dependent kinase inhibitor 1A(CDKN1A), toll-like receptor 4 (TLR4), CD19 molecule (CD19), breast cancer 1, early onset (BRCA1), platelet-derived growth factor subunit A (PDGFA), and matrix metallopeptidase 1 (MMP1) were the DEGs involved in the pathways and the PPIs. The clinical validation of the DEGs in mCRC (TNM clinical stages 3 and 4) revealed that high PDGFA expression levels were associated with poor overall survival, whereas high BRCA1 and MMP1 expression levels were associated with favorable progress free survival(PFS). The identified genes and pathways can be potential targets and predictors of therapeutic resistance and prognosis in bevacizumab-treated patients with mCRC.
Collapse
|
16
|
English WR, Lunt SJ, Fisher M, Lefley DV, Dhingra M, Lee YC, Bingham K, Hurrell JE, Lyons SK, Kanthou C, Tozer GM. Differential Expression of VEGFA Isoforms Regulates Metastasis and Response to Anti-VEGFA Therapy in Sarcoma. Cancer Res 2017; 77:2633-2646. [PMID: 28377452 DOI: 10.1158/0008-5472.can-16-0255] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/13/2016] [Accepted: 03/09/2017] [Indexed: 11/16/2022]
Abstract
Elevated plasma concentrations of soluble VEGFA isoforms are associated with poor prognosis in parallel with improved response to treatment with the anti-VEGFA antibody bevacizumab. To uncover the underlying mechanism to these observations, we administered anti-VEGFA therapy to mice bearing luminescent mouse fibrosarcomas expressing single VEGFA isoforms or their wild-type counterparts expressing all isoforms (fs120, fs164, fs188, or fsWT). Expression of the more soluble isoforms conferred an advantage for lung metastasis from subcutaneous tumors (fs120/164 vs. fs188/WT); fs120 cells also produced more lung colonies than fs188 cells when injected intravenously. Metastasis from subcutaneous fs120 tumors was more sensitive than fs188 to treatment with the anti-VEGFA antibody B20-4.1.1. Despite elevated plasma levels of VEGFA in fs120 tumor-bearing mice and a dependence on VEGF receptor 1 activity for metastasis to the lung, B20-4.1.1 did not affect survival in the lung on intravenous injection. B20-4.1.1 inhibited subcutaneous tumor growth and decreased vascular density in both fs120 and fs188 tumors. However, migration of fs120, but not fs188 cells, in vitro was inhibited by B20-4.1.1. The greater survival of fs120 cells in the lung was associated with VEGFR1-dependent accumulation of CD11b-positive myeloid cells and higher expression of the VEGFR1 ligand, PlGF2, by the fs120 cells in vitro and in the plasma and lungs of fs120 tumor-bearing mice. We conclude that soluble VEGFA isoform expression increases fibrosarcoma metastasis through multiple mechanisms that vary in their sensitivity to anti-VEGF/VEGFR inhibition, with VEGFA-targeted therapy suppressing metastasis through effects on the primary tumor rather than the metastatic site. Cancer Res; 77(10); 2633-46. ©2017 AACR.
Collapse
Affiliation(s)
- William R English
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom.
| | - Sarah Jane Lunt
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Matthew Fisher
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Diane V Lefley
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Mohit Dhingra
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Yu-Chin Lee
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Karina Bingham
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Jack E Hurrell
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Scott K Lyons
- CR-UK Cambridge Institute, The Li Ka Shing Building, Cambridge, United Kingdom
| | - Chryso Kanthou
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Gillian M Tozer
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| |
Collapse
|
17
|
Abstract
Colorectal cancer is commonly diagnosed throughout the world, and treatment options have greatly expanded over the last 2 decades. Targeting angiogenesis has been a major focus of study in a variety of malignancy types. Targeting angiogenesis has been achieved by several mechanisms in colorectal cancer, including use of antiangiogenic small molecule tyrosine kinase inhibitors (TKIs). There have been many attempts and failures to prove efficacy of TKIs in the treatment of colorectal cancer including sorafenib, sunitinib, vatalanib, and tivozanib. Regorafenib was the first TKI to demonstrate efficacy and is an orally active inhibitor of angiogenic (including the vascular endothelial growth factor receptors 1, 2, and 3), stromal, and oncogenic receptor tyrosine kinases. There are ongoing investigations of both regorafenib and ninetanib; however, there remains a critical need to better understand novel combinations with TKIs that could prove more efficacious than available options.
Collapse
|
18
|
Anti-angiogenic treatment in breast cancer: Facts, successes, failures and future perspectives. Cancer Treat Rev 2017; 53:98-110. [PMID: 28088074 DOI: 10.1016/j.ctrv.2016.12.009] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/20/2016] [Accepted: 12/21/2016] [Indexed: 01/06/2023]
Abstract
Angiogenesis is one of the hallmarks of cancer and a crucial requisite in the development of tumors. Interrupting this process by blocking the vascular endothelial growth factor (VEGF) with the monoclonal antibody bevacizumab has been considered a possible breakthrough in the treatment of various types of cancer, especially for advanced disease. However in breast cancer, studies have shown ambivalent results causing debate about the value of this drug. In this article, we review the evidence for anti-angiogenic treatment options for breast cancer, as well as discuss the possible factors limiting the effectiveness of anti-angiogenic agents and offer a recommendation regarding the future research on these therapies for the treatment of breast cancer.
Collapse
|
19
|
Bevacizumab plus paclitaxel versus placebo plus paclitaxel as first-line therapy for HER2-negative metastatic breast cancer (MERiDiAN): A double-blind placebo-controlled randomised phase III trial with prospective biomarker evaluation. Eur J Cancer 2016; 70:146-155. [PMID: 27817944 DOI: 10.1016/j.ejca.2016.09.024] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/26/2016] [Accepted: 09/28/2016] [Indexed: 11/24/2022]
Abstract
AIM MERiDiAN evaluated plasma vascular endothelial growth factor-A (pVEGF-A) prospectively as a predictive biomarker for bevacizumab efficacy in metastatic breast cancer (mBC). METHODS In this double-blind placebo-controlled randomised phase III trial, eligible patients had HER2-negative mBC previously untreated with chemotherapy. pVEGF-A was measured before randomisation to paclitaxel 90 mg/m2 on days 1, 8 and 15 with either placebo or bevacizumab 10 mg/kg on days 1 and 15, repeated every 4 weeks until disease progression, unacceptable toxicity or consent withdrawal. Stratification factors were baseline pVEGF-A, prior adjuvant chemotherapy, hormone receptor status and geographic region. Co-primary end-points were investigator-assessed progression-free survival (PFS) in the intent-to-treat and pVEGF-Ahigh populations. RESULTS Of 481 patients randomised (242 placebo-paclitaxel; 239 bevacizumab-paclitaxel), 471 received study treatment. The stratified PFS hazard ratio was 0.68 (99% confidence interval, 0.51-0.91; log-rank p = 0.0007) in the intent-to-treat population (median 8.8 months with placebo-paclitaxel versus 11.0 months with bevacizumab-paclitaxel) and 0.64 (96% confidence interval, 0.47-0.88; log-rank p = 0.0038) in the pVEGF-Ahigh subgroup. The PFS treatment-by-VEGF-A interaction p value (secondary end-point) was 0.4619. Bevacizumab was associated with increased incidences of bleeding (all grades: 45% versus 27% with placebo), neutropenia (all grades: 39% versus 29%; grade ≥3: 25% versus 13%) and hypertension (all grades: 31% versus 13%; grade ≥3: 11% versus 4%). CONCLUSION The significant PFS improvement with bevacizumab is consistent with previous placebo-controlled first-line trials in mBC. Results do not support using baseline pVEGF-A to identify patients benefitting most from bevacizumab. CLINICAL TRIALS REGISTRATION ClinicalTrials.gov NCT01663727.
Collapse
|
20
|
Sonnenblick A, Pondé N, Piccart M. Metastatic breast cancer: The Odyssey of personalization. Mol Oncol 2016; 10:1147-59. [PMID: 27430154 PMCID: PMC5423195 DOI: 10.1016/j.molonc.2016.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/27/2016] [Accepted: 07/05/2016] [Indexed: 12/31/2022] Open
Abstract
Metastatic breast cancer is the most frequent cause of cancer death for women worldwide. In the last 15 years, a large number of new agents have entered clinical use, a result of the dramatic increase in our understanding of the molecular underpinnings of metastatic breast cancer. However, while these agents have led to better outcomes, they are also at the root cause of increasing financial pressure on healthcare systems. Moreover, decision making in an era where every year new agents are added to the therapeutic armamentarium has also become a significant challenge for medical oncologists. In the present article, we will provide an ample review on the most recent developments in the field of treatment of the different subtypes of metastatic breast cancer with a critical discussion on the slow progress made in identifying response biomarkers. New hopes in the form of ctDNA monitoring and functional imaging will be presented.
Collapse
Affiliation(s)
- A Sonnenblick
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo 125, B 1000 Brussels, Belgium; Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - N Pondé
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo 125, B 1000 Brussels, Belgium
| | - M Piccart
- Department of Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Boulevard de Waterloo 125, B 1000 Brussels, Belgium.
| |
Collapse
|
21
|
Incorporation of Antiangiogenic Therapy Into the Non-Small-Cell Lung Cancer Paradigm. Clin Lung Cancer 2016; 17:493-506. [PMID: 27381269 DOI: 10.1016/j.cllc.2016.05.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/29/2016] [Accepted: 05/31/2016] [Indexed: 12/18/2022]
Abstract
Although molecular targeted agents have improved the treatment of lung cancer, their use has largely been restricted to limited subsets of the overall population that carry specific mutations. Angiogenesis, the formation of new blood vessels from existing networks, is an attractive, more general process for the development of targeted anticancer therapies, because it is critical for the growth of solid tumors, including non-small-cell lung cancer. Growing tissues require a vascular supply within a few millimeters. Therefore, solid tumors create a proangiogenic microenvironment to facilitate the development of new tumor-associated blood vessels, thus providing an adequate vascular supply for continued tumor growth. Antiangiogenic agents can specifically target the vascular endothelial growth factor (VEGF) signaling pathways, broadly inhibit multiple tyrosine kinases, or interfere with other angiogenic processes, such as disruption of existing tumor vasculature. The present report provides an overview of antiangiogenic therapy for non-small-cell lung cancer, including both currently approved antiangiogenic therapies (bevacizumab [anti-VEGF] and ramucirumab [anti-VEGF receptor 2] monoclonal antibodies), and a variety of promising novel agents in development. Although recent data have demonstrated promising efficacy for some novel agents, the overall development of antiangiogenic therapy has been hampered by redundancy in signaling pathways and the highly heterogeneous nature of tumors. An improved understanding of the molecular basis of angiogenesis will guide the development of new antiangiogenic therapies and the identification of biomarkers to predict which patients with lung cancer are most likely to benefit from antiangiogenic therapy.
Collapse
|
22
|
Schuster C, Akslen LA, Straume O. Expression of Heat Shock Protein 27 in Melanoma Metastases Is Associated with Overall Response to Bevacizumab Monotherapy: Analyses of Predictive Markers in a Clinical Phase II Study. PLoS One 2016; 11:e0155242. [PMID: 27166673 PMCID: PMC4864228 DOI: 10.1371/journal.pone.0155242] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 04/25/2016] [Indexed: 11/27/2022] Open
Abstract
The aim of this study was to identify potential predictive biomarkers in 35 patients with metastatic melanoma treated with anti-angiogenic bevacizumab monotherapy in a clinical phase II study. The immunohistochemical expression of various angiogenic factors in tissues from primary melanomas and metastases as well as their concentration in blood samples were examined. Strong expression of Heat Shock Protein 27 (HSP27) in metastases correlated significantly with complete or partial response to bevacizumab (p = 0.044). Furthermore, clinical benefit, i.e., complete or partial response or stable disease for at least 6 months, was more frequent in patients with strong expression of HSP27 in primary tumors (p = 0.046). Tissue expression of vascular endothelial growth factor (VEGF-A), its splicing variant VEGF165b or basic fibroblast growth factor (bFGF) did not correlate with response, and the concentration of HSP27, VEGF-A or bFGF measured in blood samples before treatment did not show predictive value. Further, microvessel density, proliferating microvessel density and presence of glomeruloid microvascular proliferations were assessed in sections of primary tumors and metastases. Microvessel density in primary melanomas was significantly higher in patients with clinical benefit than in non-responders (p = 0.042). In conclusion, our findings suggest that strong HSP27 expression in melanoma metastases predicts response to bevacizumab treatment.
Collapse
Affiliation(s)
- Cornelia Schuster
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Lars A. Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
- * E-mail: (OS); (LAA)
| | - Oddbjørn Straume
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- * E-mail: (OS); (LAA)
| |
Collapse
|
23
|
D'Abramo F, Goerling U, Guastadisegni C. Targeted drugs and Psycho-oncological intervention for breast cancer patients. J Negat Results Biomed 2016; 15:6. [PMID: 27036549 PMCID: PMC4818528 DOI: 10.1186/s12952-016-0049-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/09/2016] [Indexed: 01/06/2023] Open
Abstract
Personalized medicine is a new field based on molecular biology and genomics in which targeted tumor therapies are administered to patients. Psycho-oncology is a complementary approach that considers social and psychological aspects of patients as part of the treatments for cancer patients. The aim of this mini-review is to weigh clinical benefits for breast cancer patients of both treatments and possibily enhance benefits by modulating the use of both interventions. We have compared and evaluated on the one hand the use of anti Vascular Endothelial Growth Factor and, on the other hand, psycho-oncological interventions in metastatic and non-metastatic breast cancer patients. Both treatments did not increase survival of metastatic breast cancer patients, while in a selected study psycho-oncological interventions extended lifespan of non-metastatic breast cancer patients and ameliorate psychological and social factors of metastatic breast cancer patients. Because the two approaches address completely different aspects of cancer patients, if the comparison is limited to the extension of survival, the value of these two treatments cannot be assessed and compared. It is likely that by comparing patients reported outcomes, possibly by using standardized Quality of Life questionnaires, both patients and health care providers can weigh the benefits of the two treatments. It is therefore important to evaluate the use of cancer patients’ quality of life measures as a mean to improve their experiences about life and treatment, and possibly to extend their survival.
Collapse
Affiliation(s)
- Flavio D'Abramo
- Department of Psychology, Freie Universität Berlin, Hittorfstr. 16, 14195, Berlin, Deutschland.
| | - Ute Goerling
- Charité Comprehensive Cancer Center, Charitéplatz 1, 10117, Berlin, Deutschland
| | - Cecilia Guastadisegni
- Department of Environment and Primary Prevention, Italian National Institute of Health, ISS, V.le Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
24
|
Cao Y. Future options of anti-angiogenic cancer therapy. CHINESE JOURNAL OF CANCER 2016; 35:21. [PMID: 26879126 PMCID: PMC4753668 DOI: 10.1186/s40880-016-0084-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 01/18/2016] [Indexed: 12/30/2022]
Abstract
In human patients, drugs that block tumor vessel growth are widely used to treat a variety of cancer types. Many rigorous phase 3 clinical trials have demonstrated significant survival benefits; however, the addition of an anti-angiogenic component to conventional therapeutic modalities has generally produced modest survival benefits for cancer patients. Currently, it is unclear why these clinically available drugs targeting the same angiogenic pathways produce dissimilar effects in preclinical models and human patients. In this article, we discuss possible mechanisms of various anti-angiogenic drugs and the future development of optimized treatment regimens.
Collapse
Affiliation(s)
- Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77, Stockholm, Sweden. .,Department of Medical and Health Sciences, Linköping University, 581 83, Linköping, Sweden. .,Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, LE3 9QP, UK.
| |
Collapse
|
25
|
Büning H, Hacker UT. Inhibitors of Angiogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 917:261-85. [DOI: 10.1007/978-3-319-32805-8_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
26
|
Tobin NP, Foukakis T, De Petris L, Bergh J. The importance of molecular markers for diagnosis and selection of targeted treatments in patients with cancer. J Intern Med 2015; 278:545-70. [PMID: 26373821 DOI: 10.1111/joim.12429] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The past 30 years have seen the introduction of a number of cancer therapies with the aim of restricting the growth and spread of primary and metastatic tumours. A shared commonality among these therapies is their targeting of various aspects of the cancer hallmarks, that is traits that are essential to successful tumour propagation and dissemination. The evolution of molecular-scale technology has been central to the identification of new cancer targets, and it is not a coincidence that improved therapies have emerged at the same time as gene expression arrays and DNA sequencing have enhanced our understanding of cancer genetics. Modern tumour pathology is now viewed at the molecular level ranging from IHC biomarkers, to gene signature classifiers and gene mutations, all of which provide crucial information about which patients will respond to targeted therapy regimens. In this review, we briefly discuss the general types of targeted therapies used in a clinical setting and provide a short background on immunohistochemical, gene expression and DNA sequencing technologies, before focusing on three tumour types: breast, lung and colorectal cancers. For each of these cancer types, we provide a background to the disease along with an overview of the current standard therapies and then focus on the relevant targeted therapies and the pathways they inhibit. Finally, we highlight several strategies that are pivotal to the successful development of targeted anti-cancer drugs.
Collapse
Affiliation(s)
- N P Tobin
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - T Foukakis
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - L De Petris
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - J Bergh
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| |
Collapse
|
27
|
O'Donnell PH, Karovic S, Karrison TG, Janisch L, Levine MR, Harris PJ, Polite BN, Cohen EEW, Fleming GF, Ratain MJ, Maitland ML. Serum C-Telopeptide Collagen Crosslinks and Plasma Soluble VEGFR2 as Pharmacodynamic Biomarkers in a Trial of Sequentially Administered Sunitinib and Cilengitide. Clin Cancer Res 2015. [PMID: 26199386 DOI: 10.1158/1078-0432.ccr-15-0427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE Fit-for-purpose pharmacodynamic biomarkers could expedite development of combination antiangiogenic regimens. Plasma sVEGFR2 concentrations ([sVEGFR2]) mark sunitinib effects on the systemic vasculature. We hypothesized that cilengitide would impair microvasculature recovery during sunitinib withdrawal and could be detected through changes in [sVEGFR2]. EXPERIMENTAL DESIGN Advanced solid tumor patients received 50 mg sunitinib daily for 14 days. For the next 14 days, patients were randomized to arm A (cilengitide 2,000 mg administered intravenously twice weekly) or arm B (no treatment). The primary endpoint was change in [sVEGFR2] between days 14 and 28. A candidate pharmacodynamic biomarker of cilengitide inhibition of integrin αvβ3, serum c-telopeptide collagen crosslinks (CTx), was also measured. RESULTS Of 21 patients, 14 (7 per arm) received all treatments without interruption and had all blood samples available for analysis. The mean change and SD of [sVEGFR2] for all sunitinib-treated patients was consistent with previous data. There was no significant difference in the mean change in [sVEGFR2] from days 14 to 28 between the arms [arm A: 2.8 ng/mL; 95% confidence interval (CI), 2.1-3.6 vs. arm B: 2.0 ng/mL; 95% CI, 0.72-3.4; P = 0.22, 2-sample t test]. Additional analyses suggested (i) prior bevacizumab therapy to be associated with unusually low baseline [sVEGFR2] and (ii) sunitinib causes measurable changes in CTx. CONCLUSIONS Cilengitide had no measurable effects on any circulating biomarkers. Sunitinib caused measurable declines in serum CTx. The properties of [sVEGFR2] and CTx observed in this study inform the design of future combination antiangiogenic therapy trials.
Collapse
Affiliation(s)
- Peter H O'Donnell
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois. Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois. Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois
| | - Sanja Karovic
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Theodore G Karrison
- Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois. Department of Health Studies, The University of Chicago, Chicago, Illinois
| | - Linda Janisch
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Matthew R Levine
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Pamela J Harris
- Cancer Therapy Evaluation Program of the National Cancer Institute, Bethesda, Maryland
| | - Blase N Polite
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois. Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois
| | - Ezra E W Cohen
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois. Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois
| | - Gini F Fleming
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois. Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois. Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois
| | - Mark J Ratain
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois. Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois. Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois
| | - Michael L Maitland
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois. Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois. Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
28
|
Vascular endothelial growth factor D expression is a potential biomarker of bevacizumab benefit in colorectal cancer. Br J Cancer 2015; 113:37-45. [PMID: 26125443 PMCID: PMC4647541 DOI: 10.1038/bjc.2015.209] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/12/2015] [Accepted: 05/14/2015] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Bevacizumab prolongs progression-free survival (PFS) in patients with metastatic colorectal cancer. We analysed the protein expression levels of vascular endothelial growth factor (VEGF) ligands and receptors to determine their prognostic and predictive effects. METHODS We graded expression of VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-R1, and VEGF-R2 to assess whether overexpression predicted bevacizumab resistance in samples from 268 of 471 patients randomised to capecitabine (C), capecitabine and bevacizumab (CB), or CB and mitomycin (CBM) in the MAX trial and extended the analysis to the CAIRO-2 population. RESULTS Patients with low expression of VEGF-D (0, 1þ) benefited from bevacizumab treatment (PFS hazard ratio (HR) (C vs CBþCBM), 0.21; 95% CI, 0.08–0.55; overall survival (OS) HR, 0.35; 95% CI, 0.13–0.90). Patients with higher VEGF-D expression received less benefit (VEGF-D 2þ PFS HR, 0.67; 95% CI, 0.45–1.00; OS HR, 0.82; 95% CI, 0.52–1.30; VEGF-D 3þ PFS HR, 0.77; 95% CI, 0.50–1.17; OS HR, 1.28; 95% CI, 0.79–2.09) (P interaction o0.05). In CAIRO-2, there was no difference in PFS or OS according to VEGF-D expression. CONCLUSIONS The predictive value of VEGF-D expression for bevacizumab may depend on the chemotherapy backbone used. Further evaluation is required before clinical utilisation.
Collapse
|
29
|
Abstract
The humanized monoclonal antibody bevacizumab (Avastin(®)) has been available in the EU since 2005. Results of phase III trials demonstrate that adding intravenous bevacizumab to antineoplastic agents improves progression-free survival and/or overall survival in patients with advanced cancer, including when used as first- or second-line therapy in metastatic colorectal cancer, as first-line therapy in advanced nonsquamous non-small cell lung cancer, as first-line therapy in metastatic renal cell carcinoma, as first-line therapy in metastatic breast cancer, and as first-line therapy in epithelial ovarian, fallopian tube or primary peritoneal cancer or in recurrent, platinum-sensitive or platinum-resistant disease. Results of these studies are supported by the findings of routine oncology practice studies conducted in real-world settings. The tolerability profile of bevacizumab is well defined and adverse events associated with its use (e.g. hypertension, proteinuria, haemorrhage, wound healing complications, arterial thromboembolism, gastrointestinal perforation) are generally manageable. In conclusion, bevacizumab remains an important option for use in patients with advanced cancer.
Collapse
Affiliation(s)
- Gillian M Keating
- Springer, Private Bag 65901, Mairangi Bay, 0754, Auckland, New Zealand.
| |
Collapse
|
30
|
Cramarossa G, Lee EK, Sivanathan L, Georgsdottir S, Lien K, Santos KD, Chan K, Emmenegger U. A systematic literature analysis of correlative studies in low-dose metronomic chemotherapy trials. Biomark Med 2015; 8:893-911. [PMID: 25224945 DOI: 10.2217/bmm.14.14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Low-dose metronomic (LDM) chemotherapy is a beneficial and very well-tolerated form of chemotherapy utilization characterized by the frequent and uninterrupted administration of low doses of conventional chemotherapeutic agents over prolonged periods of time. While patients resistant to standard maximum tolerated dose (MTD) chemotherapy may still benefit from LDM chemotherapy, there is a lack of predictive markers of response to LDM chemotherapy. We searched the MEDLINE, EMBASE, CENTRAL and PubMed databases for correlative studies conducted as part of LDM chemotherapy trials in order to identify the most promising biomarker candidates. Given the antiangiogenic properties of LDM chemotherapy, angiogenesis-related biomarkers were most commonly studied. However, significant correlations between angiogenesis-related biomarkers and study end points were rare and variable, even so far as biomarkers correlating positively with an end point in some studies and negatively with the same end point in other studies. Pursuing biomarkers outside the angiogenesis field may be more promising.
Collapse
Affiliation(s)
- Gemma Cramarossa
- Division of Medical Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Di Salvatore M, Lo Giudice L, Rossi E, Santonocito C, Nazzicone G, Rodriquenz MG, Cappuccio S, Inno A, Fuso P, Orlandi A, Strippoli A, Capoluongo E, Astone A, Cassano A, Barone C. Association of IL-8 and eNOS polymorphisms with clinical outcomes in bevacizumab-treated breast cancer patients: an exploratory analysis. Clin Transl Oncol 2015; 18:40-6. [PMID: 26141413 DOI: 10.1007/s12094-015-1334-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 06/20/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND The role of bevacizumab in metastatic breast cancer is controversial. Identification of predictive biomarkers could help to select patients who really benefit from it. We evaluated the association of angiogenesis-related gene polymorphisms with the treatment outcome of bevacizumab in metastatic breast cancer patients. PATIENTS AND METHODS eNOS-786T/C and -894G/T, IL-8-251T/A genomic polymorphisms were assessed in 31 metastatic breast cancer patients treated with bevacizumab plus chemotherapy in the first-line setting. Testing for association between each polymorphism and treatment outcome was performed. RESULTS Patients with IL-8 251 AA genotype showed a significantly lower progression-free survival in each combination comparison: "TT" vs "AA" (13 vs 8 months; p = 0.008); TT vs TA vs AA (13 vs 11 vs 8 months; p = 0.02); TT vs TA +AA (13 vs 11 months; p = 0.01); TT + TA vs AA (12 vs 8 months; p = 0.01) and a lower overall survival when compared with TT +TA genotype (26 vs 51 months, p = 0.04). Patients carrying eNOS 894 TT genotype showed a statistically significant lower progression-free survival than patients with GG genotype (11.5 vs 26.5 months; p = 0.04) with no differences in the overall survival. No association with response rate was found with any of the polymorphisms analyzed. CONCLUSION These findings suggest that IL-8 251T/A and eNOS-894 G/T polymorphisms might have a role in predicting treatment outcome of bevacizumab in metastatic breast cancer. Our results are hypothesis generating and need to be confirmed in larger clinical trials.
Collapse
Affiliation(s)
- M Di Salvatore
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy.
| | - L Lo Giudice
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - E Rossi
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - C Santonocito
- Laboratory of Clinical Molecular Biology, Institute of Biochemistry and Clinical Biochemistry, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - G Nazzicone
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - M G Rodriquenz
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - S Cappuccio
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - A Inno
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy.,Medical Oncology, Sacro Cuore Don Calabria Hospital, Via Don A. Sempreboni 5, 37024, Negrar, VR, Italy
| | - P Fuso
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - A Orlandi
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - A Strippoli
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - E Capoluongo
- Laboratory of Clinical Molecular Biology, Institute of Biochemistry and Clinical Biochemistry, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - A Astone
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - A Cassano
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| | - C Barone
- Unit of Clinical Oncology, Catholic University of the Sacred Heart, L.go F. Vito 1, 00168, Rome, Italy
| |
Collapse
|
32
|
McIntyre A, Harris AL. Metabolic and hypoxic adaptation to anti-angiogenic therapy: a target for induced essentiality. EMBO Mol Med 2015; 7:368-79. [PMID: 25700172 PMCID: PMC4403040 DOI: 10.15252/emmm.201404271] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 01/12/2015] [Accepted: 01/27/2015] [Indexed: 12/20/2022] Open
Abstract
Anti-angiogenic therapy has increased the progression-free survival of many cancer patients but has had little effect on overall survival, even in colon cancer (average 6-8 weeks) due to resistance. The current licensed targeted therapies all inhibit VEGF signalling (Table 1). Many mechanisms of resistance to anti-VEGF therapy have been identified that enable cancers to bypass the angiogenic blockade. In addition, over the last decade, there has been increasing evidence for the role that the hypoxic and metabolic responses play in tumour adaptation to anti-angiogenic therapy. The hypoxic tumour response, through the transcription factor hypoxia-inducible factors (HIFs), induces major gene expression, metabolic and phenotypic changes, including increased invasion and metastasis. Pre-clinical studies combining anti-angiogenics with inhibitors of tumour hypoxic and metabolic adaptation have shown great promise, and combination clinical trials have been instigated. Understanding individual patient response and the response timing, given the opposing effects of vascular normalisation versus reduced perfusion seen with anti-angiogenics, provides a further hurdle in the paradigm of personalised therapeutic intervention. Additional approaches for targeting the hypoxic tumour microenvironment are being investigated in pre-clinical and clinical studies that have potential for producing synthetic lethality in combination with anti-angiogenic therapy as a future therapeutic strategy.
Collapse
Affiliation(s)
- Alan McIntyre
- Hypoxia and angiogenesis Group, Department of Oncology Weatherall Institute of Molecular Medicine University of Oxford, Oxford, UK
| | - Adrian L Harris
- Hypoxia and angiogenesis Group, Department of Oncology Weatherall Institute of Molecular Medicine University of Oxford, Oxford, UK
| |
Collapse
|
33
|
An exploratory biomarker study in metastatic tumors from colorectal cancer patients treated with bevacizumab. Int J Biol Markers 2015; 30:e73-80. [PMID: 24980447 DOI: 10.5301/jbm.5000097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2014] [Indexed: 01/26/2023]
Abstract
AIMS Inhibition of angiogenesis is an effective treatment option for metastatic colorectal cancer. Predictive biomarkers to select patients who are most likely to benefit from this therapeutic strategy are lacking. We conducted a pilot, retrospective biomarker study in a cohort of metastatic colorectal cancer patients treated with bevacizumab. The objectives of this study were to evaluate the prognostic value of biomarker expression in metastases and to compare their expression in paired tumor specimens. MATERIALS AND METHODS Eligible patients were treated with a bevacizumab-containing therapy; from these patients, tumor tissue from metastases was available. PTEN, PI3K p110a, c-MET, and CAIX were analyzed by immunohistochemistry. RESULTS Forty-two patients received bevacizumab, 13 (31%) with first-line and 29 (69%) with second-line chemotherapy. Expression of CAIX, PI3K p110a, and c-MET in metastases did not predict objective response. PTEN loss was associated with response to treatment (p=0.02) and this association remained significant after adjusting for prognostic variables (p=0.006). However, no association with survival outcomes was found. In 32 patients (76%) with available paired specimens, we observed an equal expression between primary tumors and corresponding metastases in 75% of cases for CAIX in epithelial tumor cells, 56% for CAIX in stromal cells, 63% for PTEN, and 87% for c-MET. CONCLUSION PTEN loss in metastases appears to be associated with response to bevacizumab-based therapy. However, larger studies are necessary to confirm the potential role of the PI3K/AKT/mTOR pathway in modulating the therapeutic effect of bevacizumab. Tumor heterogeneity should be taken into consideration when analyzing tumor tissues for biomarker studies.
Collapse
|
34
|
Schmainda KM, Zhang Z, Prah M, Snyder BS, Gilbert MR, Sorensen AG, Barboriak DP, Boxerman JL. Dynamic susceptibility contrast MRI measures of relative cerebral blood volume as a prognostic marker for overall survival in recurrent glioblastoma: results from the ACRIN 6677/RTOG 0625 multicenter trial. Neuro Oncol 2015; 17:1148-56. [PMID: 25646027 DOI: 10.1093/neuonc/nou364] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/24/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The study goal was to determine whether changes in relative cerebral blood volume (rCBV) derived from dynamic susceptibility contrast (DSC) MRI are predictive of overall survival (OS) in patients with recurrent glioblastoma multiforme (GBM) when measured 2, 8, and 16 weeks after treatment initiation. METHODS Patients with recurrent GBM (37/123) enrolled in ACRIN 6677/RTOG 0625, a multicenter, randomized, phase II trial of bevacizumab with irinotecan or temozolomide, consented to DSC-MRI plus conventional MRI, 21 with DSC-MRI at baseline and at least 1 postbaseline scan. Contrast-enhancing regions of interest were determined semi-automatically using pre- and postcontrast T1-weighted images. Mean tumor rCBV normalized to white matter (nRCBV) and standardized rCBV (sRCBV) were determined for these regions of interest. The OS rates for patients with positive versus negative changes from baseline in nRCBV and sRCBV were compared using Wilcoxon rank-sum and Kaplan-Meier survival estimates with log-rank tests. RESULTS Patients surviving at least 1 year (OS-1) had significantly larger decreases in nRCBV at week 2 (P = .0451) and sRCBV at week 16 (P = .014). Receiver operating characteristic analysis found the percent changes of nRCBV and sRCBV at week 2 and sRCBV at week 16, but not rCBV data at week 8, to be good prognostic markers for OS-1. Patients with positive change from baseline rCBV had significantly shorter OS than those with negative change at both week 2 and week 16 (P = .0015 and P = .0067 for nRCBV and P = .0251 and P = .0004 for sRCBV, respectively). CONCLUSIONS Early decreases in rCBV are predictive of improved survival in patients with recurrent GBM treated with bevacizumab.
Collapse
Affiliation(s)
- Kathleen M Schmainda
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - Zheng Zhang
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - Melissa Prah
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - Bradley S Snyder
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - Mark R Gilbert
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - A Gregory Sorensen
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - Daniel P Barboriak
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| | - Jerrold L Boxerman
- Department of Radiology, Medical College of Wisconsin, Milwaukee, Wisconsin (K.M.S., M.P.); Department of Biostatistics and Center for Statistical Sciences, Brown University, Providence, Rhode Island (Z.Z., B.S.S.); Department of Neuro-Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas (M.R.G.); Department of Radiology, Massachusetts General Hospital, Charlestown, Massachusetts (A.G.S.); Department of Radiology, Duke University Medical Center, Durham, North Carolina (D.P.B.); Department of Diagnostic Imaging, Rhode Island Hospital, Providence, Rhode Island (J.L.B.); Alpert Medical School of Brown University, Providence, Rhode Island (J.L.B.)
| |
Collapse
|
35
|
|
36
|
Fontana E, Sclafani F, Cunningham D. Anti-angiogenic therapies for advanced esophago-gastric cancer. Indian J Med Paediatr Oncol 2014; 35:253-62. [PMID: 25538401 PMCID: PMC4264270 DOI: 10.4103/0971-5851.144985] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Neo-vascularization is a vital process for tumor growth and development which involves the interaction between tumor cells and stromal endothelial cells through several growth factors and membranous receptors which ultimately activate pro-angiogenic intracellular signaling pathways. Inhibition of angiogenesis has become a standard treatment option for several tumor types including colorectal cancer, glioblastoma and ovarian cancer. In gastric cancer, the therapeutic role of anti-angiogenic agents is more controversial. Bevacizumab and ramucirumab, two monoclonal antibodies, which target vascular endothelial growth factor-A and vascular endothelial growth factor receptor-2, respectively, have been demonstrated antitumor activity in patients with tumors of the stomach or esophagogastric junction. However, especially for bevacizumab, this antitumor activity has not consistently translated into a survival advantage over standard treatment in randomized trials. In this article, we provide an overview of the role of angiogenesis in gastric cancer and discuss the results of clinical trials that investigated safety and effectiveness of antiangiogenic therapies in this disease. A review of the literature has been done using PubMed, ClinicalTrials.gov website and the ASCO Annual Meeting Library.
Collapse
Affiliation(s)
- Elisa Fontana
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Francesco Sclafani
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - David Cunningham
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| |
Collapse
|
37
|
Klemm F, Joyce JA. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol 2014; 25:198-213. [PMID: 25540894 DOI: 10.1016/j.tcb.2014.11.006] [Citation(s) in RCA: 522] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 11/20/2014] [Accepted: 11/21/2014] [Indexed: 02/08/2023]
Abstract
The tumor microenvironment (TME) not only plays a pivotal role during cancer progression and metastasis but also has profound effects on therapeutic efficacy. In the case of microenvironment-mediated resistance this can involve an intrinsic response, including the co-option of pre-existing structural elements and signaling networks, or an acquired response of the tumor stroma following the therapeutic insult. Alternatively, in other contexts, the TME has a multifaceted ability to enhance therapeutic efficacy. This review examines recent advances in our understanding of the contribution of the TME during cancer therapy and discusses key concepts that may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Florian Klemm
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Johanna A Joyce
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
38
|
Ganesan P, Moulder S, Lee JJ, Janku F, Valero V, Zinner RG, Naing A, Fu S, Tsimberidou AM, Hong D, Stephen B, Stephens P, Yelensky R, Meric-Bernstam F, Kurzrock R, Wheler JJ. Triple-negative breast cancer patients treated at MD Anderson Cancer Center in phase I trials: improved outcomes with combination chemotherapy and targeted agents. Mol Cancer Ther 2014; 13:3175-84. [PMID: 25253784 PMCID: PMC4258414 DOI: 10.1158/1535-7163.mct-14-0358] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Patients with metastatic triple-negative breast cancer (TNBC) have poor treatment outcomes. We reviewed the electronic records of consecutive patients with metastatic TNBC treated in phase I clinic at MD Anderson Cancer Center (Houston, TX) between Augu st 2005 and May 2012. One hundred and six patients received at least 1 phase I trial. Twelve of 98 evaluable patients (12%) had either complete response (CR; n = 1), partial response (PR; n = 7), or stable disease ≥ 6 months (SD; n = 4). Patients treated on matched therapy (n = 16) compared with those on nonmatched therapy (n = 90) had improved SD ≥ 6 months/PR/CR (33% vs. 8%; P = 0.018) and longer progression-free survival (PFS; median, 6.4 vs. 1.9 months; P = 0.001). Eleven of 57 evaluable patients (19%) treated with combination chemotherapy and targeted therapy had SD ≥ 6 months/PR/CR versus 1 of 41 evaluable patients (2%) treated on other phase I trials (P = 0.013), and longer PFS (3.0 vs. 1.6 months; P < 0.0001). Patients with molecular alterations in the PI3K/AKT/mTOR pathway treated on matched therapy (n = 16) had improved PFS compared with those with and without molecular alterations treated on nonmatched therapy (n = 27; 6.4 vs. 3.2 months; P = 0.036). On multivariate analysis, improved PFS was associated with treatment with combined chemotherapy and targeted agents (P = 0.0002), ≤ 2 metastatic sites (P = 0.003), therapy with PI3K/AKT/mTOR inhibitors for those with cognate pathway abnormalities (P = 0.018), and treatment with antiangiogenic agents (P = 0.023). In summary, combinations of chemotherapy and angiogenesis and/or PI3K/AKT/mTOR inhibitors demonstrated improved outcomes in patients with metastatic TNBC.
Collapse
Affiliation(s)
- Prasanth Ganesan
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stacy Moulder
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Filip Janku
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ralph G Zinner
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aung Naing
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Hong
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bettzy Stephen
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, Moores Cancer Center, The University of California San Diego, La Jolla, California
| | - Jennifer J Wheler
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
39
|
Allegrini G, Coltelli L, Orlandi P, Fontana A, Camerini A, Ferro A, Cazzaniga M, Casadei V, Lucchesi S, Bona E, Di Lieto M, Pazzagli I, Villa F, Amoroso D, Scalese M, Arrighi G, Molinaro S, Fioravanti A, Finale C, Triolo R, Di Desidero T, Donati S, Marcucci L, Goletti O, Del Re M, Salvadori B, Ferrarini I, Danesi R, Falcone A, Bocci G. Pharmacogenetic interaction analysis of VEGFR-2 and IL-8 polymorphisms in advanced breast cancer patients treated with paclitaxel and bevacizumab. Pharmacogenomics 2014; 15:1985-99. [DOI: 10.2217/pgs.14.140] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aim: To investigate pharmacogenetic interactions among VEGF-A, VEGFR-2, IL-8, HIF-1α, EPAS-1 and TSP-1 SNPs and their role on progression-free survival in a population of metastatic breast cancer patients treated with bevacizumab in combination with first-line paclitaxel. Patients & methods: Analyses were performed on germline DNA obtained from blood samples and SNPs were investigated by real-time polymerase chain reaction technique. The multifactor dimensionality reduction methodology was applied to investigate the interaction between SNPs. Results: One hundred and thirteen patients were enrolled from eight Italian Oncology Units ( clinicaltrial.gov : NCT01935102). The multifactor dimensionality reduction software provided two pharmacogenetic interaction profiles consisting of the combination between specific VEGFR-2 rs11133360 and IL-8 rs4073 genotypes. The median progression-free survival was 14.1 months (95% CI: 11.4–16.8) and 10.2 months (95% CI: 8.8–11.5) for the favorable and the unfavorable genetic profile, respectively (HR: 0.44, 95% CI: 0.29–0.66, p < 0.0001). Conclusion: The pharmacogenetic statistical interaction between VEGFR-2 rs11133360 and IL-8 rs4073 genotypes may identify a population of patients with a better outcome.
Collapse
Affiliation(s)
| | - Luigi Coltelli
- Division of Medical Oncology, Pontedera Hospital, Pisa, Italy
| | - Paola Orlandi
- Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea Fontana
- Division of Medical Oncology II, Azienda Ospedaliero-Universitaria Pisana, S. Chiara Hospital, Pisa, Italy
| | - Andrea Camerini
- Division of Medical Oncology, Versilia Hospital, Lucca, Italy
| | - Antonella Ferro
- Division of Medical Oncology, S. Chiara Hospital, Trento, Italy
| | | | - Virginia Casadei
- Division of Medical Oncology, S. Salvatore Hospital, Pesaro, Italy
| | - Sara Lucchesi
- Division of Medical Oncology, Pontedera Hospital, Pisa, Italy
| | - Eleonora Bona
- Division of Medical Oncology II, Azienda Ospedaliero-Universitaria Pisana, S. Chiara Hospital, Pisa, Italy
| | - Marco Di Lieto
- Division of Medical Oncology, Azienda USL 3, Pistoia, Italy
| | - Ilaria Pazzagli
- Division of Medical Oncology, S. Cosma & Damiano Hospital, Pescia, Pistoia, Italy
| | - Federica Villa
- Division of Medical Oncology, AO S. Gerardo, Monza, Italy
| | | | - Marco Scalese
- Institute of Clinical Physiology, Italian National Research Council – CNR, Pisa Italy
| | - Giada Arrighi
- Division of Medical Oncology, Pontedera Hospital, Pisa, Italy
| | - Sabrina Molinaro
- Institute of Clinical Physiology, Italian National Research Council – CNR, Pisa Italy
| | - Anna Fioravanti
- Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy
| | - Chiara Finale
- Division of Medical Oncology, Pontedera Hospital, Pisa, Italy
| | - Renza Triolo
- Division of Medical Oncology, S. Chiara Hospital, Trento, Italy
| | - Teresa Di Desidero
- Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy
| | - Sara Donati
- Division of Medical Oncology, Versilia Hospital, Lucca, Italy
| | | | - Orlando Goletti
- Department of Translational Research & New Technology in Medicine & Surgery, University of Pisa, Italy
| | - Marzia Del Re
- Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy
| | - Barbara Salvadori
- Division of Medical Oncology II, Azienda Ospedaliero-Universitaria Pisana, S. Chiara Hospital, Pisa, Italy
| | - Ilaria Ferrarini
- Division of Medical Oncology II, Azienda Ospedaliero-Universitaria Pisana, S. Chiara Hospital, Pisa, Italy
| | - Romano Danesi
- Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alfredo Falcone
- Division of Medical Oncology II, Azienda Ospedaliero-Universitaria Pisana, S. Chiara Hospital, Pisa, Italy
- Division of Medical Oncology, Department of Translational Research & New Technology in Medicine & Surgery, University of Pisa, Italy
| | - Guido Bocci
- Department of Clinical & Experimental Medicine, University of Pisa, Pisa, Italy
- Istituto Toscano Tumori, Firenze, Italy
| |
Collapse
|
40
|
Bahce I, Huisman MC, Verwer EE, Ooijevaar R, Boutkourt F, Vugts DJ, van Dongen GA, Boellaard R, Smit EF. Pilot study of (89)Zr-bevacizumab positron emission tomography in patients with advanced non-small cell lung cancer. EJNMMI Res 2014; 4:35. [PMID: 26055936 PMCID: PMC4884046 DOI: 10.1186/s13550-014-0035-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 06/26/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The aim of this pilot study was to evaluate whether the uptake of (89)Zr-bevacizumab in non-small cell lung cancer (NSCLC) tumors could be visualized and quantified. The correlation between tumor (89)Zr-bevacizumab uptake and tumor response to antitumor therapy with a bevacizumab-based regimen was explored. METHODS Seven NSCLC patients underwent static PET scans at days 4 and 7 after injection of 36.4 ± 0.9 MBq (mean ± SD) (89)Zr-bevacizumab, prior to commencing carboplatin-paclitaxel-bevacizumab chemotherapy (CPB). Overall survival (OS) and progression-free survival (PFS) to CPB followed by bevacizumab maintenance therapy was correlated to tumor tracer uptake, quantified using peak standardized uptake values (SUVpeak). RESULTS Zr-bevacizumab uptake (SUVpeak) was approximately four times higher in tumor tissues (primary tumor and metastases) than in non-tumor tissues (healthy muscle, lung, and fat) on days 4 and 7. A positive trend but no significant correlation could be found between SUVpeak and OS or PFS. CONCLUSIONS This pilot study shows that (89)Zr-bevacizumab PET imaging in NSCLC is feasible. Further investigation to validate this technique as a predictive biomarker for selecting patients for bevacizumab treatment is warranted.
Collapse
Affiliation(s)
- Idris Bahce
- Department of Pulmonary Diseases, VU University Medical Center, De Boelelaan 1117, Amsterdam, 1081HV, The Netherlands,
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Smith MA. Lessons learned from adult clinical experience to inform evaluations of VEGF pathway inhibitors in children with cancer. Pediatr Blood Cancer 2014; 61:1497-505. [PMID: 24760743 DOI: 10.1002/pbc.25036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 02/27/2014] [Indexed: 01/07/2023]
Abstract
Agents targeting the vascular endothelial growth factor (VEGF) pathway have been studied in adults with cancer for nearly two decades. It is important to assess the lessons learned from this adult experience and to see how these lessons can help inform pediatric development of agents in this class. The benefit achieved from the use of VEGF pathway targeted agents for adult cancers has primarily been to delay for several months disease progression and less commonly time to death for conditions in which cure is not a reasonable expectation. VEGF pathway targeted agents have shown no efficacy when applied in the adjuvant setting. For adults with advanced cancer, prolongation of survival by 2-3 months is considered an important achievement in some settings. However, the primary goal of pediatric oncology clinical research is to identify treatments that allow children to be cured of their cancer and to grow to adulthood without treatment-induced limitations that lower their quality of survival. An important question for the pediatric oncology research community, pharmaceutical companies, and regulatory agencies to address in planning for future clinical trials is whether existing data support a role for VEGF pathway targeted agents in contributing to a therapeutic pathway to cure for children with cancer.
Collapse
|
42
|
de Haas S, Delmar P, Bansal AT, Moisse M, Miles DW, Leighl N, Escudier B, Van Cutsem E, Carmeliet P, Scherer SJ, Pallaud C, Lambrechts D. Genetic variability of VEGF pathway genes in six randomized phase III trials assessing the addition of bevacizumab to standard therapy. Angiogenesis 2014; 17:909-20. [PMID: 25012543 DOI: 10.1007/s10456-014-9438-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 07/05/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND Despite extensive translational research, no validated biomarkers predictive of bevacizumab treatment outcome have been identified. METHODS We performed a meta-analysis of individual patient data from six randomized phase III trials in colorectal, pancreatic, lung, renal, breast, and gastric cancer to explore the potential relationships between 195 common genetic variants in the vascular endothelial growth factor (VEGF) pathway and bevacizumab treatment outcome. RESULTS The analysis included 1,402 patients (716 bevacizumab-treated and 686 placebo-treated). Twenty variants were associated (P < 0.05) with progression-free survival (PFS) in bevacizumab-treated patients. Of these, 4 variants in EPAS1 survived correction for multiple testing (q < 0.05). Genotype-by-treatment interaction tests revealed that, across these 20 variants, 3 variants in VEGF-C (rs12510099), EPAS1 (rs4953344), and IL8RA (rs2234671) were potentially predictive (P < 0.05), but not resistant to multiple testing (q > 0.05). A weak genotype-by-treatment interaction effect was also observed for rs699946 in VEGF-A, whereas Bayesian genewise analysis revealed that genetic variability in VHL was associated with PFS in the bevacizumab arm (q < 0.05). Variants in VEGF-A, EPAS1, and VHL were located in expression quantitative loci derived from lymphoblastoid cell lines, indicating that they affect the expression levels of their respective gene. CONCLUSIONS This large genetic analysis suggests that variants in VEGF-A, EPAS1, IL8RA, VHL, and VEGF-C have potential value in predicting bevacizumab treatment outcome across tumor types. Although these associations did not survive correction for multiple testing in a genotype-by-interaction analysis, they are among the strongest predictive effects reported to date for genetic variants and bevacizumab efficacy.
Collapse
|
43
|
Zirconium-89 labeled antibodies: a new tool for molecular imaging in cancer patients. BIOMED RESEARCH INTERNATIONAL 2014; 2014:203601. [PMID: 24991539 PMCID: PMC4058511 DOI: 10.1155/2014/203601] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 04/23/2014] [Indexed: 01/15/2023]
Abstract
Antibody based positron emission tomography (immuno-PET) imaging is of increasing importance to visualize and characterize tumor lesions. Additionally, it can be used to identify patients who may benefit from a particular therapy and monitor the therapy outcome. In recent years the field is focused on 89Zr, a radiometal with near ideal physical and chemical properties for immuno-PET. In this review we will discuss the production of 89Zr, the bioconjugation strategies, and applications in (pre-)clinical studies of 89Zr-based immuno-PET in oncology. To date, 89Zr-based PET imaging has been investigated in a wide variety of cancer-related targets. Moreover, clinical studies have shown the feasibility for 89Zr-based immuno-PET to predict and monitor treatment, which could be used to tailor treatment for the individual patient. Further research should be directed towards the development of standardized and robust conjugation methods and improved chelators to minimize the amount of released Zr4+ from the antibodies. Additionally, further validation of the imaging method is required. The ongoing development of new 89Zr-labeled antibodies directed against novel tumor targets is expected to expand applications of 89Zr-labeled immuno-PET to a valuable method in the medical imaging.
Collapse
|
44
|
Kümler I, Christiansen OG, Nielsen DL. A systematic review of bevacizumab efficacy in breast cancer. Cancer Treat Rev 2014; 40:960-73. [PMID: 24909311 DOI: 10.1016/j.ctrv.2014.05.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/08/2014] [Accepted: 05/12/2014] [Indexed: 12/21/2022]
Abstract
UNLABELLED Angiogenesis is a key component of cancer growth, invasion and metastasis. Therefore, inhibition of angiogenesis is an attractive strategy for the treatment of cancer. We systematically describe phase II and III clinical trials of bevacizumab for the treatment of breast cancer. METHODS A computer-based literature search was carried out using PUBMED and conference databases. Original phase II and III studies reporting ≥15 patients who received bevacizumab were included. RESULTS 41 phase II trials were identified in the metastatic setting. Most trials found bevacizumab treatment feasible. Response rates (RR) varied from 0% to 76.5%, time to progression (TTP)/progression free survival (PFS) from 2.4 to 25.3 months and overall survival from 11.5 to more than 38 months. 14 phase III trials including more than 4400 patients with MBC unanimously showed increased RR and PFS, however, no trials demonstrated an OS benefit. In the neoadjuvant setting 23 phase II and III trials were identified. All studies found increased pCR/tpCR but no benefit in terms of OS could be demonstrated. The only study conducted in the adjuvant setting failed to show any survival benefit of bevacizumab. CONCLUSION Despite increased response rates in both the metastatic and neoadjuvant setting, bevacizumab has failed to show any OS benefit. Future trials should include identification of robust predictive biomarkers in order to improve our understanding of molecular biomarkers and mechanisms.
Collapse
Affiliation(s)
- Iben Kümler
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, DK-2730 Herlev, Denmark.
| | - Ole Grummedal Christiansen
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, DK-2730 Herlev, Denmark.
| | - Dorte Lisbet Nielsen
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, DK-2730 Herlev, Denmark.
| |
Collapse
|
45
|
Adham SAI, Al Harrasi I, Al Haddabi I, Al Rashdi A, Al Sinawi S, Al Maniri A, Ba-Omar T, Coomber BL. Immunohistological insight into the correlation between neuropilin-1 and epithelial-mesenchymal transition markers in epithelial ovarian cancer. J Histochem Cytochem 2014; 62:619-31. [PMID: 24850663 DOI: 10.1369/0022155414538821] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The mechanism by which neuropilin-1 (NRP-1) induces malignancy in Epithelial Ovarian Cancer (EOC) is still unknown. This study is the first to demonstrate the relationship between NRP-1 expression and EMT markers vimentin, N-cadherin, E-cadherin and Slug. We used tissue microarrays containing the three main subtypes of EOC tumors: serous, mucinous cystadenocarcinoma and endometrioid adenocarcinoma and representative cases retrieved from our pathology archives. Immunohistochemistry was performed to detect the expression levels and location of NRP-1 and the aforementioned EMT proteins. NRP-1 was mainly expressed on cancer cells but not in normal ovarian surface epithelium (OSE). The Immunoreactive Scoring (IRS) values revealed that the expression of NRP-1, Slug and E-cadherin in the malignant subtypes of ovarian tissues was significantly higher (5.18 ± 0.64, 4.84 ± 0.7, 4.98 ± 0.68, respectively) than their expression in the normal and benign tissues (1.04 ± 0.29, 0.84 ± 0.68, 1.71 ± 0.66, respectively), with no significant differences among the studied subtypes. Vimentin was expressed in the cancer cell component of 43% of tumors and it was exclusively localized in the stroma of all mucinous tumors. The Spearman's rho value indicated that NRP-1 is positively related to the EMT markers E-cadherin and Slug. This notion might indicate that NRP-1 is a partner in the EMT process in EOC tumors.
Collapse
Affiliation(s)
- Sirin A I Adham
- Department of Biology, College of Science (SAIA, IAH, TBO) Sultan Qaboos University, Muscat, OmanDepartment of Pathology, College of Medicine (IAH, AAR, SAS) Sultan Qaboos University, Muscat, OmanThe Research Council, Muscat, Oman (AAM)Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada (BLC)
| | - Ibtisam Al Harrasi
- Department of Biology, College of Science (SAIA, IAH, TBO) Sultan Qaboos University, Muscat, OmanDepartment of Pathology, College of Medicine (IAH, AAR, SAS) Sultan Qaboos University, Muscat, OmanThe Research Council, Muscat, Oman (AAM)Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada (BLC)
| | - Ibrahim Al Haddabi
- Department of Biology, College of Science (SAIA, IAH, TBO) Sultan Qaboos University, Muscat, OmanDepartment of Pathology, College of Medicine (IAH, AAR, SAS) Sultan Qaboos University, Muscat, OmanThe Research Council, Muscat, Oman (AAM)Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada (BLC)
| | - Afrah Al Rashdi
- Department of Biology, College of Science (SAIA, IAH, TBO) Sultan Qaboos University, Muscat, OmanDepartment of Pathology, College of Medicine (IAH, AAR, SAS) Sultan Qaboos University, Muscat, OmanThe Research Council, Muscat, Oman (AAM)Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada (BLC)
| | - Shadia Al Sinawi
- Department of Biology, College of Science (SAIA, IAH, TBO) Sultan Qaboos University, Muscat, OmanDepartment of Pathology, College of Medicine (IAH, AAR, SAS) Sultan Qaboos University, Muscat, OmanThe Research Council, Muscat, Oman (AAM)Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada (BLC)
| | - Abdullah Al Maniri
- Department of Biology, College of Science (SAIA, IAH, TBO) Sultan Qaboos University, Muscat, OmanDepartment of Pathology, College of Medicine (IAH, AAR, SAS) Sultan Qaboos University, Muscat, OmanThe Research Council, Muscat, Oman (AAM)Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada (BLC)
| | - Taher Ba-Omar
- Department of Biology, College of Science (SAIA, IAH, TBO) Sultan Qaboos University, Muscat, OmanDepartment of Pathology, College of Medicine (IAH, AAR, SAS) Sultan Qaboos University, Muscat, OmanThe Research Council, Muscat, Oman (AAM)Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada (BLC)
| | - Brenda L Coomber
- Department of Biology, College of Science (SAIA, IAH, TBO) Sultan Qaboos University, Muscat, OmanDepartment of Pathology, College of Medicine (IAH, AAR, SAS) Sultan Qaboos University, Muscat, OmanThe Research Council, Muscat, Oman (AAM)Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada (BLC)
| |
Collapse
|
46
|
Hoshino Y, Hayashida T, Hirata A, Takahashi H, Chiba N, Ohmura M, Wakui M, Jinno H, Hasegawa H, Maheswaran S, Suematsu M, Kitagawa Y. Bevacizumab terminates homeobox B9-induced tumor proliferation by silencing microenvironmental communication. Mol Cancer 2014; 13:102. [PMID: 24885802 PMCID: PMC4023179 DOI: 10.1186/1476-4598-13-102] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 05/01/2014] [Indexed: 12/20/2022] Open
Abstract
Background Homeobox B9 (HOXB9), a transcriptional factor, regulates developmental processes and tumor progression and has recently been recognized as one of important transcriptional factors related to angiogenesis. This study aimed to investigate the role of HOXB9 in tumorigenesis and angiogenesis. Methods We examined the expression of HOXB9 in colorectal cancer using qPCR and in situ hybridization. We also examined the effect of HOXB9 overexpression in colorectal cancer using a proliferation assay, ELISA, a multiplex assay, and xenograft models. The clinical significance of HOXB9 was statistically evaluated in resected specimens. Results HOXB9 was expressed in colorectal cancer specimens. HOXB9 induced angiogenesis and tumor proliferation in vitro, which resulted in high tumorigenicity in vivo and poor overall survival. Bevacizumab, an anti-vascular endothelial growth factor (VEGF) antibody, remarkably suppressed tumor proliferation by inhibiting angiogenesis in HOXB9-overexpressing xenografts, and it improved overall survival and provided prolonged progression-free survival in HOXB9-overexpressing patients. A comprehensive multiplex assay of the supernatant of cancer cells co-cultured with human vascular endothelial cells and fibroblasts indicated significantly higher interleukin-6 (IL6) levels than those in the supernatant of monocultured cells. HOXB9 overexpression in clinical specimens was significantly correlated with increased IL6 expression. An IL6-neutralizing antibody inhibited VEGF secretion and tumor proliferation in the co-culture system. Conclusions HOXB9 promotes the secretion of angiogenic factors, including VEGF, to induce tumor proliferation through microenvironmental production of cytokines including IL6 signaling. Moreover, silencing of VEGF or IL6 terminates cytokine release in tumor microenvironment. Thus, HOXB9 and IL6 may be potential biomarkers for bevacizumab treatment.
Collapse
Affiliation(s)
| | - Tetsu Hayashida
- Department of Surgery, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Parametric response maps of perfusion MRI may identify recurrent glioblastomas responsive to bevacizumab and irinotecan. PLoS One 2014; 9:e90535. [PMID: 24675671 PMCID: PMC3968002 DOI: 10.1371/journal.pone.0090535] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 02/02/2014] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Perfusion weighted imaging (PWI) can be used to measure key aspects of tumor vascularity in vivo and recent studies suggest that perfusion imaging may be useful in the early assessment of response to angiogenesis inhibitors. Aim of this work is to compare Parametric Response Maps (PRMs) with the Region Of Interest (ROI) approach in the analysis of tumor changes induced by bevacizumab and irinotecan in recurrent glioblastomas (rGBM), and to evaluate if changes in tumor blood volume measured by perfusion MRI may predict clinical outcome. METHODS 42 rGBM patients with KPS ≥ 50 were treated until progression, as defined by MRI with RANO criteria. Relative cerebral blood volume (rCBV) variation after 8 weeks of treatment was calculated through semi-automatic ROI placement in the same anatomic region as in baseline. Alternatively, rCBV variations with respect to baseline were calculated into the evolving tumor region using a voxel-by-voxel difference. PRMs were created showing where rCBV significantly increased, decreased or remained unchanged. RESULTS An increased blood volume in PRM (PRMCBV+) higher than 18% (first quartile) after 8 weeks of treatment was associated with increased progression free survival (PFS; 24 versus 13 weeks, p = 0.045) and overall survival (OS; 38 versus 25 weeks, p = 0.016). After 8 weeks of treatment ROI analysis showed that mean rCBV remained elevated in non responsive patients (4.8 ± 0.9 versus 5.1 ± 1.2, p = 0.38), whereas decreased in responsive patients (4.2 ± 1.3 versus 3.8 ± 1.6 p = 0.04), and re-increased progressively when patients approached tumor progression. CONCLUSIONS Our data suggest that PRMs can provide an early marker of response to antiangiogenic treatment and warrant further confirmation in a larger cohort of GBM patients.
Collapse
|
48
|
Genetic markers of bevacizumab-induced hypertension. Angiogenesis 2014; 17:685-94. [PMID: 24558090 DOI: 10.1007/s10456-014-9424-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 02/17/2014] [Indexed: 12/19/2022]
Abstract
PURPOSE There are currently no validated biomarkers predicting bevacizumab treatment outcome or toxicity. We combined biomarker data from six phase III trials of bevacizumab to assess whether genetic variation in vascular endothelial growth factor-A (VEGF-A) pathway or hypertension-related genes are associated with bevacizumab-induced hypertension. EXPERIMENTAL DESIGN Germline DNA was available from 1,631 patients receiving bevacizumab-containing therapy for advanced solid tumors. Overall, 194 white patients had grade 1-4 bevacizumab-induced hypertension. In total, 236 single nucleotide polymorphisms (SNPs) located in VEGF-A, VEGF-A receptors (FLT1 and KDR), and other genes were selected using a SNP tagging approach and genotyped. A logistic regression on individual patient data was performed after adjustment for cancer type and five other covariates. RESULTS Ten SNPs were associated with bevacizumab-induced hypertension (P ≤ 0.05), but none surpassed the threshold adjusted for multiple testing (P < 0.0002). The most significant VEGF-A pathway SNP was rs1680695 in EGLN3 [allelic odds ratio (OR) 1.50 [95 % confidence interval (Cl) 1.09-2.07], P = 0.012]. Two additional SNPs, rs4444903 in EGF and rs2305949 in KDR, were associated with hypertension (allelic OR 1.57 [95 % CI 1.17-2.11], P = 0.0025; allelic OR 0.62 [95 % CI 0.42-0.93], P = 0.020, respectively) and closely linked to nearby functional variants. Consistent with previous reports, rs11064560 in WNK1 was also associated with bevacizumab-induced hypertension (OR 1.41 [95 % CI 1.04-1.92], P = 0.028). CONCLUSIONS The genes described in this large genetic analysis using pooled datasets warrant further functional investigation regarding their role in mediating bevacizumab-induced hypertension.
Collapse
|
49
|
Kim HK, Green JE. Predictive biomarker candidates for the response of gastric cancer to targeted and cytotoxic agents. Pharmacogenomics 2014; 15:375-84. [PMID: 24533716 PMCID: PMC7670597 DOI: 10.2217/pgs.13.250] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gastric cancer is the second most common cause of cancer death worldwide. Recent development of targeted agents provides clinicians with additional systemic treatment options to conventional cytotoxic agents. Predictive markers are undoubtedly important for guiding the appropriate use of targeted and cytotoxic agents. Currently, however, HER2 is the only predictive biomarker validated for gastric cancer. In this review, candidate predictive markers for response to other targeted agents and cytotoxic chemotherapeutic agents are discussed.
Collapse
Affiliation(s)
- Hark Kyun Kim
- Center for Gastric Cancer, National Cancer Center, Goyang, 410-769, Republic of Korea.
| | | |
Collapse
|
50
|
Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis 2014; 17:471-94. [PMID: 24482243 PMCID: PMC4061466 DOI: 10.1007/s10456-014-9420-y] [Citation(s) in RCA: 518] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 01/15/2014] [Indexed: 12/17/2022]
Abstract
Tumours require a vascular supply to grow and can achieve this via the expression of pro-angiogenic growth factors, including members of the vascular endothelial growth factor (VEGF) family of ligands. Since one or more of the VEGF ligand family is overexpressed in most solid cancers, there was great optimism that inhibition of the VEGF pathway would represent an effective anti-angiogenic therapy for most tumour types. Encouragingly, VEGF pathway targeted drugs such as bevacizumab, sunitinib and aflibercept have shown activity in certain settings. However, inhibition of VEGF signalling is not effective in all cancers, prompting the need to further understand how the vasculature can be effectively targeted in tumours. Here we present a succinct review of the progress with VEGF-targeted therapy and the unresolved questions that exist in the field: including its use in different disease stages (metastatic, adjuvant, neoadjuvant), interactions with chemotherapy, duration and scheduling of therapy, potential predictive biomarkers and proposed mechanisms of resistance, including paradoxical effects such as enhanced tumour aggressiveness. In terms of future directions, we discuss the need to delineate further the complexities of tumour vascularisation if we are to develop more effective and personalised anti-angiogenic therapies.
Collapse
|