1
|
Volk A, von Bülow C, Stern A, Simon R, Burandt E, Sauter G, Schmalfeldt B, Oliveira-Ferrer L. Stromal PDGFR-beta expression is a prognostic factor in high-grade serous ovarian cancer patients but is it also predictive for response to antiangiogenic treatment? J Cancer Res Clin Oncol 2025; 151:44. [PMID: 39849231 PMCID: PMC11759463 DOI: 10.1007/s00432-025-06090-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/09/2025] [Indexed: 01/25/2025]
Abstract
OBJECTIVE In advanced ovarian cancer, the majority of patients receive anti-angiogenic treatment with bevacizumab. However, its use is often associated with severe side effects, and not all patients benefit from the therapy. Currently, there are no reliable biomarkers to predict response to treatment. Given their role as key regulators of angiogenesis, platelet-derived growth factor receptor-beta (PDGFR-beta) and vascular endothelial growth factor receptor-2 (VEGFR-2) are promising candidates for predictive biomarkers. This study evaluates their potential. METHODS PDGFR-beta and VEGFR-2 expression was evaluated using immunohistochemistry in a tissue microarray assay including 391 ovarian tissue samples. Correlation analyses with clinical and histopathological parameters were performed in a homogeneous cohort of 199 high grade serous ovarian cancer samples (HGSOC). RESULTS In HGSOC, strong stromal PDGFR-beta expression was associated with significantly shorter overall survival compared to weak/moderate expression. The impact of stromal PDGFR-beta expression on patient survival was however not restricted to the subgroup of patients receiving therapy with bevacizumab, and therefore cannot be considered as a predictive factor. For VEGFR-2, no or weak protein expression was found in the majority of the tumor samples. Survival analyses showed a more favorable prognosis with no or weak VEGFR-2 expression. CONCLUSIONS High stromal expression levels of PDGFR-beta correlate with shorter overall survival in HGSOC. Thus, stromal PDGFR-beta might serve as a prognostic biomarker. No predictive effect in response to bevacizumab therapy could be attributed.
Collapse
Affiliation(s)
- Annabelle Volk
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
| | - Charlotte von Bülow
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Aysche Stern
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Department of Internal Medicine, Southwest Healthcare, Temecula, USA
| | - Ronald Simon
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eike Burandt
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Barbara Schmalfeldt
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Leticia Oliveira-Ferrer
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| |
Collapse
|
2
|
Mehta K, Hegde M, Girisa S, Vishwa R, Alqahtani MS, Abbas M, Shakibaei M, Sethi G, Kunnumakkara AB. Targeting RTKs/nRTKs as promising therapeutic strategies for the treatment of triple-negative breast cancer: evidence from clinical trials. Mil Med Res 2024; 11:76. [PMID: 39668367 PMCID: PMC11636053 DOI: 10.1186/s40779-024-00582-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 11/08/2024] [Indexed: 12/14/2024] Open
Abstract
The extensive heterogeneity and the limited availability of effective targeted therapies contribute to the challenging prognosis and restricted survival observed in triple-negative breast cancer (TNBC). Recent research indicates the aberrant expression of diverse tyrosine kinases (TKs) within this cancer, contributing significantly to tumor cell proliferation, survival, invasion, and migration. The contemporary paradigm shift towards precision medicine has highlighted TKs and their receptors as promising targets for pharmacotherapy against a range of malignancies, given their pivotal roles in tumor initiation, progression, and advancement. Intensive investigations have focused on various monoclonal antibodies (mAbs) and small molecule inhibitors that specifically target proteins such as epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor (VEGFR), cellular mesenchymal-epithelial transition factor (c-MET), human epidermal growth factor receptor 2 (HER2), among others, for combating TNBC. These agents have been studied both in monotherapy and in combination with other chemotherapeutic agents. Despite these advances, a substantial terrain of unexplored potential lies within the realm of TK targeted therapeutics, which hold promise in reshaping the therapeutic landscape. This review summarizes the various TK targeted therapeutics that have undergone scrutiny as potential therapeutic interventions for TNBC, dissecting the outcomes and revelations stemming from diverse clinical investigations. A key conclusion from the umbrella clinical trials evidences the necessity for in-depth molecular characterization of TNBCs for the maximum efficiency of TK targeted therapeutics, either as standalone treatments or a combination. Moreover, our observation highlights that the outcomes of TK targeted therapeutics in TNBC are substantially influenced by the diversity of the patient cohort, emphasizing the prioritization of individual patient genetic/molecular profiles for precise TNBC patient stratification for clinical studies.
Collapse
Affiliation(s)
- Kasshish Mehta
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Ravichandran Vishwa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, 61421, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, 61421, Abha, Saudi Arabia
| | - Mehdi Shakibaei
- Department of Human-Anatomy, Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Ludwig-Maximilian-University, 80336, Munich, Germany
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117699, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati (IITG), Guwahati, Assam, 781039, India.
| |
Collapse
|
3
|
Chen X, Wei X, Yao P, Liu Y, Guan H, Kang H, Liu D, Diao Y, Ma X, Min W, Shan C, Zhao Y, Zhao F, Chen Y, Xiao D, She Q, Liu Y, Zhang Y, Zhang S. The Efficiency and Toxicity Of Anlotinib in Combination With Docetaxel Followed by Epirubicin and Cyclophosphamide Regimen as Neoadjuvant Treatment in IIB to IIIA Triple Negative Breast Cancer: A Single-Arm, Multicenter, Open-Label, Phase II Study. Clin Breast Cancer 2024; 24:e195-e202. [PMID: 38670862 DOI: 10.1016/j.clbc.2024.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 12/03/2023] [Accepted: 01/25/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND The combination of neoadjuvant chemotherapy and anti-angiogenesis therapy for patients with triple-negative breast cancer (TNBC) remains inadequately supported by evidence. We conducted a single-arm, open-label, multicenter, phase II trial to evaluate the efficacy and toxicity of anlotinib plus epirubicin and cyclophosphamide followed by paclitaxel in patients with IIB to IIIA stage TNBC. METHODS Newly diagnosed patients received epirubicin at 90 mg/m2 and cyclophosphamide at 600 mg/m2 followed by docetaxel at 100 mg/m2 (21 days per cycle; total of 4 cycles), along with oral anlotinib (12 mg qd, d1-14; 21 days per cycle; total of 4 cycles). Subsequently, patients underwent surgery. The primary endpoint of this study was pathologic complete response (pCR). RESULTS Among the 34 included patients, the median age was 46.5 years (range: 27-72); all were female. Pathological assessment revealed that 17 patients achieved RCB 0 response, which is currently defined as pathologic complete response; 3 patients achieved RCB 1; 12 patients achieved RCB 2; and 1 patient achieved RCB 3. The probability of a grade 3 adverse reaction was 17.6%, and no grade 4 adverse reactions occurred. The most common hematological adverse reaction was leukopenia (13/34, 38.2%), of which 5.9% (2/34) were grade 3. The most common non-hematological adverse reactions were oral mucositis (16/34, 58.8%), fatigue (50.0%), hand-foot syndrome (50.0%), hypertension (44.1%), bleeding (44.1%), and alopecia (32.4%). CONCLUSION The combination of anlotinib and EC-T chemotherapy demonstrated tolerable side effects in the neoadjuvant treatment of early TNBC. pCR was higher than what has been reported in previous clinical studies of chemotherapy alone. This study provides additional rationale for using anlotinib plus docetaxel-epirubicin-based chemotherapy regimen in patients with early-stage TNBCs.
Collapse
Affiliation(s)
- Xi Chen
- Xi'an Jiaotong University, Shaanxi Province, China
| | - Xinyu Wei
- Xi'an Jiaotong University, Shaanxi Province, China
| | - Peizhuo Yao
- Xi'an Jiaotong University, Shaanxi Province, China
| | - Yanbin Liu
- Xi'an Jiaotong University, Shaanxi Province, China
| | - Haitao Guan
- The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Province, China
| | - Huafeng Kang
- The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Province, China
| | - Di Liu
- The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Province, China
| | - Yan Diao
- The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Province, China
| | - Xiaobin Ma
- The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Province, China
| | - Weili Min
- The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Province, China
| | - Changyou Shan
- The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Province, China
| | - Yang Zhao
- The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Province, China
| | - Fang Zhao
- The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Province, China
| | - Yuanyuan Chen
- Baoji Central Hospital of Shaanxi Province, Shaanxi Province, China
| | - Dong Xiao
- 3201 Hospital of Hanzhong City, Shaanxi Province, China
| | - Qing She
- Baoji Central Hospital of Shaanxi Province, Shaanxi Province, China
| | - Youhuai Liu
- Baoji Central Hospital of Shaanxi Province, Shaanxi Province, China
| | - Yinbin Zhang
- The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Province, China
| | - Shuqun Zhang
- The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi Province, China.
| |
Collapse
|
4
|
Schirizzi A, Arshadi A, Tolomeo D, Schirosi L, Valentini AM, De Leonardis G, Refolo MG, Donghia R, Storlazzi CT, Zito A, Ricci AD, Vallarelli S, Ostuni C, Bencivenga M, De Manzoni G, Messa C, Armentano R, Giannelli G, Lotesoriere C, D’Alessandro R. VEGFA Status as a Predictive Marker of Therapy Outcome in Metastatic Gastric Cancer Patients Following Ramucirumab-Based Treatment. Biomedicines 2023; 11:2721. [PMID: 37893095 PMCID: PMC10603940 DOI: 10.3390/biomedicines11102721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Metastatic gastric cancer (mGC) often has a poor prognosis and may benefit from a few targeted therapies. Ramucirumab-based anti-angiogenic therapy targeting the VEGFR2 represents a milestone in the second-line treatment of mGC. Several studies on different cancers are focusing on the major VEGFR2 ligand status, meaning VEGFA gene copy number and protein overexpression, as a prognostic marker and predictor of response to anti-angiogenic therapy. Following this insight, our study aims to examine the role of VEGFA status as a predictive biomarker for the outcome of second-line therapy with Ramucirumab and paclitaxel in mGC patients. To this purpose, the copy number of the VEGFA gene, by fluorescence in situ hybridization experiments, and its expression in tumor tissue as well as the density of micro-vessels, by immunohistochemistry experiments, were assessed in samples derived from mGC patients. This analysis found that amplification of VEGFA concomitantly with VEGFA overexpression and overexpression of VEGFA with micro-vessels density are more represented in patients showing disease control during treatment with Ramucirumab. In addition, in the analyzed series, it was found that amplification was not always associated with overexpression of VEGFA, but overexpression of VEGFA correlates with high micro-vessel density. In conclusion, overexpression of VEGFA could emerge as a potential biomarker to predict the response to anti-angiogenic therapy.
Collapse
Affiliation(s)
- Annalisa Schirizzi
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.); (C.M.)
| | - Aram Arshadi
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.A.); (D.T.); (C.T.S.)
| | - Doron Tolomeo
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.A.); (D.T.); (C.T.S.)
| | - Laura Schirosi
- Pathology Department, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (L.S.); (A.Z.)
| | - Anna Maria Valentini
- Histopathology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.M.V.); (R.A.)
| | - Giampiero De Leonardis
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.); (C.M.)
| | - Maria Grazia Refolo
- Clinical Pathology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy;
| | - Rossella Donghia
- Data Science Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy;
| | - Clelia Tiziana Storlazzi
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.A.); (D.T.); (C.T.S.)
| | - Alfredo Zito
- Pathology Department, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (L.S.); (A.Z.)
| | - Angela Dalia Ricci
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.D.R.); (S.V.); (C.O.)
| | - Simona Vallarelli
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.D.R.); (S.V.); (C.O.)
| | - Carmela Ostuni
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.D.R.); (S.V.); (C.O.)
| | - Maria Bencivenga
- General and Upper GI Surgery Division, University of Verona, 37126 Verona, Italy; (M.B.); (G.D.M.)
| | - Giovanni De Manzoni
- General and Upper GI Surgery Division, University of Verona, 37126 Verona, Italy; (M.B.); (G.D.M.)
| | - Caterina Messa
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.); (C.M.)
| | - Raffaele Armentano
- Histopathology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.M.V.); (R.A.)
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy;
| | - Claudio Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.D.R.); (S.V.); (C.O.)
| | - Rosalba D’Alessandro
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.); (C.M.)
| |
Collapse
|
5
|
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
|
6
|
Apatinib plus vinorelbine versus vinorelbine for metastatic triple-negative breast cancer who failed first/second-line treatment: the NAN trial. NPJ Breast Cancer 2022; 8:110. [PMID: 36127351 PMCID: PMC9489776 DOI: 10.1038/s41523-022-00462-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 07/05/2022] [Indexed: 11/20/2022] Open
Abstract
While therapies such as chemotherapy combined with immunotherapy, sacituzumab govitecan, and PARP inhibitors are available for metastatic TNBC, on disease progression after these therapies, the mainstay of therapy is chemotherapy. Apatinib is a small-molecule tyrosine kinase inhibitor that has promising anti-angiogenesis and antitumor activity for TNBC. We aimed to evaluate the safety and efficacy of adding apatinib to chemotherapy in patients with advanced TNBC with failed first/second-line treatment. A total of 66 patients were randomly assigned, in a 1:1 ratio, to receive vinorelbine or vinorelbine with apatinib in 28-day cycles. The primary endpoint was progression-free survival (PFS). Secondary endpoints included overall survival (OS), overall response rate (ORR) and safety. 33 received apatinib plus vinorelbine and 32 received vinorelbine (1 was withdrawal). Median PFS was significantly longer in the apatinib plus vinorelbine group than in the vinorelbine group (3.9 months vs. 2.0 months; hazard ratio, 1.82; 95% confidence interval [CI], 1.06 to 3.11; P = 0.026). Median OS was 11.5 months with apatinib plus vinorelbine and 9.9 months with vinorelbine (HR,1.01; 95% CI, 0.51 to 1.97; P = 0.985). The ORR was 9.1% in the apatinib plus vinorelbine group and 6.3% in the vinorelbine group (P = 0.667). The most common treatment-related hematologic grade 3–4 adverse events in apatinib plus vinorelbine group, were leukopenia, granulocytopenia, anemia, and thrombocytopenia. no treatment-related nonhematologic grade 4 adverse events or treatment-related deaths were observed. Collectively, adding apatinib to vinorelbine shows a promising benefit in PFS compared to vinorelbine monotherapy, with an excellent toxicity profile, warranting further exploration.
Collapse
|
7
|
Triple-negative breast cancer brain metastasis: an update on druggable targets, current clinical trials, and future treatment options. Drug Discov Today 2022; 27:1298-1314. [DOI: 10.1016/j.drudis.2022.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/20/2021] [Accepted: 01/22/2022] [Indexed: 12/12/2022]
|
8
|
Abstract
ABSTRACT Triple-negative breast cancer (TNBC) is pathologically defined by lack of expression of the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 amplification and portends an aggressive clinical course with worse outcomes compared with other breast cancers. Until recently, standard treatment options consisted of sequential cytotoxic chemotherapies for both early and metastatic disease. Advances in sequencing technology have led to the identification of 4 main subtypes of TNBC based on recurrent genetic alterations, transcriptional patterns, and molecular features: basal-like 1 (BL1), basal-like 2 (BL2), mesenchymal (M), and luminal androgen receptor (LAR). Frequent alterations found in DNA damage response pathways, germline and somatic BRCA1/2 genes, PI3K signaling pathways, and the presence of androgen receptors and infiltrating immune cells could serve as actionable targets to optimize treatments and improve outcomes for patients with TNBC. Recent approvals for immune checkpoint inhibitors and the antibody-drug conjugate, sacituzumab govitecan-hziy, for advanced TNBC illustrate the advances in treatment that can result from these molecular discoveries. This review will explore the molecular subtypes of TNBC and their distinct characteristics, as well as highlight the molecular features and potential "drivers" that have been identified as promising targets for new treatment strategies.
Collapse
|
9
|
Liao M, Zhang J, Wang G, Wang L, Liu J, Ouyang L, Liu B. Small-Molecule Drug Discovery in Triple Negative Breast Cancer: Current Situation and Future Directions. J Med Chem 2021; 64:2382-2418. [PMID: 33650861 DOI: 10.1021/acs.jmedchem.0c01180] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Triple negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, but an effective targeted therapy has not been well-established so far. Considering the lack of effective targets, where do we go next in the current TNBC drug development? A promising intervention for TNBC might lie in de novo small-molecule drugs that precisely target different molecular characteristics of TNBC. However, an ideal single-target drug discovery still faces a huge challenge. Alternatively, other new emerging strategies, such as dual-target drug, drug repurposing, and combination strategies, may provide new insight into the improvement of TNBC therapeutics. In this review, we focus on summarizing the current situation of a series of candidate small-molecule drugs in TNBC therapy, including single-target drugs, dual-target drugs, as well as drug repurposing and combination strategies that will together shed new light on the future directions targeting TNBC vulnerabilities with small-molecule drugs for future therapeutic purposes.
Collapse
Affiliation(s)
- Minru Liao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Leiming Wang
- The Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Jie Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
10
|
Huang L, Lang GT, Liu Q, Shi JX, Shao ZM, Cao AY. A predictor of pathological complete response to neoadjuvant chemotherapy in triple-negative breast cancer patients with the DNA repair genes. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:301. [PMID: 33708928 PMCID: PMC7944335 DOI: 10.21037/atm-20-4852] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background We conducted this study to investigate the prevalence of potential chemo-response-related gene mutations in triple-negative breast cancer (TNBC) patients and to evaluate the potential relationship between these gene mutations and neoadjuvant chemotherapy response in TNBC patients. Methods One hundred sixty-two TNBC patients in Fudan University Shanghai Cancer Center who received NAC with 4 cycles of paclitaxel and carboplatin were enrolled in this study. Fifty-six pathological complete response (pCR) patients and 56 non-pCR patients were enrolled in this retrospective study for the training set. Clinical assessments of postoperative residual tumors were performed according to Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 criteria. Forty chemo-response-related genes were screened in each tumor specimen by second-generation sequencing analysis. Fifty TNBC patients who received neoadjuvant chemotherapy with paclitaxel and carboplatin were enrolled in the validation group. Results Fifty-seven of 112 (50.9%) TNBCs contained at least one detected somatic mutation. As expected, TP53 mutation was the most common alteration, which was observed in 21.4% of patients. BRCA1, BRCA2, RET, PI3KCA, and PTEN mutations were each observed in 11.6%, 4.5%, 5.4%, 2.7% and 3.6% of all cases, respectively. No significant differences in any gene mutation frequency between pCR and non-pCR groups were identified. We found that the mutation status of 10 DNA repair genes involved in homologous recombination (HR) pathway successfully discriminated between responding and nonresponding tumors in the training group. Up to 18 patients who were mutation-positive experienced pCR compared to only 6 in the non-pCR group (P=0.006), and 75% the HR related gene mutation patients achieved pCR. In the validation group, TNBC patients with DNA repair gene mutations achieved 77.8% pCR. Conclusions A subset of TNBC patients carry deleterious somatic mutations in 10 HR-related genes. The mutation status of this expanded gene panel is likely to effectively predict respond rate to neoadjuvant chemotherapy based on paclitaxel and carboplatin. Our findings need to be validated through follow-up studies in this and additional cohorts.
Collapse
Affiliation(s)
- Liang Huang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guan-Tian Lang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qi Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Radiation Oncology, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, China
| | - Jin-Xiu Shi
- Department of Genetics, Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center and Shanghai Industrial Technology Institute (SITI), Shanghai, China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - A-Yong Cao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center/Cancer Institute, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
11
|
O'Melia MJ, Manspeaker MP, Thomas SN. Tumor-draining lymph nodes are survival niches that support T cell priming against lymphatic transported tumor antigen and effects of immune checkpoint blockade in TNBC. Cancer Immunol Immunother 2021; 70:2179-2195. [PMID: 33459842 DOI: 10.1007/s00262-020-02792-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/07/2020] [Indexed: 12/21/2022]
Abstract
Triple negative breast cancer (TNBC) is a significant clinical problem to which immunotherapeutic strategies have been applied with limited success. Using the syngeneic E0771 TNBC mouse model, this work explores the potential for antitumor CD8+ T cell immunity to be primed extratumorally in lymphoid tissues and therapeutically leveraged. CD8+ T cell viability and responses within the tumor microenvironment (TME) were found to be severely impaired, effects coincident with local immunosuppression that is recapitulated in lymphoid tissues in late stage disease. Prior to onset of a locally suppressed immune microenvironment, however, CD8+ T cell priming within lymph nodes (LN) that depended on tumor lymphatic drainage remained intact. These results demonstrate tumor-draining LNs (TdLN) to be lymphoid tissue niches that support the survival and antigenic priming of CD8+ T lymphocytes against lymph-draining antigen. The therapeutic effects of and CD8+ T cells response to immune checkpoint blockade were furthermore improved when directed to LNs within the tumor-draining lymphatic basin. Therefore, TdLNs represent a unique potential tumor immunity reservoir in TNBC for which strategies may be developed to improve the effects of ICB immunotherapy.
Collapse
Affiliation(s)
- Meghan J O'Melia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, IBB 2310, 315 Ferst Drive NW, Atlanta, GA, 30332, USA
| | - Margaret P Manspeaker
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA.,School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Susan N Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, IBB 2310, 315 Ferst Drive NW, Atlanta, GA, 30332, USA. .,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA. .,George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA. .,Winship Cancer Institute, Emory University, Atlanta, GA, 30332, USA.
| |
Collapse
|
12
|
Zarrilli G, Businello G, Dieci MV, Paccagnella S, Carraro V, Cappellesso R, Miglietta F, Griguolo G, Guarneri V, Lo Mele M, Fassan M. The Tumor Microenvironment of Primitive and Metastatic Breast Cancer: Implications for Novel Therapeutic Strategies. Int J Mol Sci 2020; 21:8102. [PMID: 33143050 PMCID: PMC7662409 DOI: 10.3390/ijms21218102] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/24/2022] Open
Abstract
Breast cancer evolves thanks to a dense and close interaction with the surrounding tumor microenvironment (TME). Fibroblasts, leukocytes, blood and lymphatic endothelial cells and extracellular matrix are the constituents of this entity, and they synergistically play a pivotal role in all of the stages of breast cancer development, from its onset to its metastatic spread. Moreover, it has been widely demonstrated that variations to the TME can correspond to prognosis variations. Breast cancer not only modulates the transformation of the environment within the mammary gland, but the same process is observed in metastases as well. In this minireview, we describe the features of TME within the primitive breast cancer, throughout its evolution and spread into the main metastatic sites.
Collapse
Affiliation(s)
- Giovanni Zarrilli
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy; (G.Z.); (G.B.); (S.P.); (V.C.); (R.C.)
| | - Gianluca Businello
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy; (G.Z.); (G.B.); (S.P.); (V.C.); (R.C.)
| | - Maria Vittoria Dieci
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128 Padua, Italy; (M.V.D.); (G.G.); (V.G.)
- Department of Surgery, Oncology and Gastroenterology (DISCoG), University of Padua, 35121 Padua, Italy;
| | - Silvia Paccagnella
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy; (G.Z.); (G.B.); (S.P.); (V.C.); (R.C.)
| | - Valentina Carraro
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy; (G.Z.); (G.B.); (S.P.); (V.C.); (R.C.)
| | - Rocco Cappellesso
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy; (G.Z.); (G.B.); (S.P.); (V.C.); (R.C.)
| | - Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology (DISCoG), University of Padua, 35121 Padua, Italy;
| | - Gaia Griguolo
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128 Padua, Italy; (M.V.D.); (G.G.); (V.G.)
- Department of Surgery, Oncology and Gastroenterology (DISCoG), University of Padua, 35121 Padua, Italy;
| | - Valentina Guarneri
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, 35128 Padua, Italy; (M.V.D.); (G.G.); (V.G.)
- Department of Surgery, Oncology and Gastroenterology (DISCoG), University of Padua, 35121 Padua, Italy;
| | - Marcello Lo Mele
- Surgical Pathology Unit, University Hospital of Padua, 35121 Padua, Italy;
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy; (G.Z.); (G.B.); (S.P.); (V.C.); (R.C.)
| |
Collapse
|
13
|
Vaklavas C, Roberts BS, Varley KE, Lin NU, Liu MC, Rugo HS, Puhalla S, Nanda R, Storniolo AM, Carey LA, Saleh MN, Li Y, Delossantos JF, Grizzle WE, LoBuglio AF, Myers RM, Forero-Torres A. TBCRC 002: a phase II, randomized, open-label trial of preoperative letrozole with or without bevacizumab in postmenopausal women with newly diagnosed stage 2/3 hormone receptor-positive and HER2-negative breast cancer. Breast Cancer Res 2020; 22:22. [PMID: 32070401 PMCID: PMC7027068 DOI: 10.1186/s13058-020-01258-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 02/06/2020] [Indexed: 12/19/2022] Open
Abstract
Background In preclinical studies, the expression of vascular endothelial growth factor (VEGF) in hormone receptor-positive breast cancer is associated with estrogen-independent tumor growth and resistance to endocrine therapies. This study investigated whether the addition of bevacizumab, a monoclonal antibody against VEGF, to letrozole enhanced the antitumor activity of the letrozole in the preoperative setting. Methods Postmenopausal women with newly diagnosed stage 2 or 3 estrogen and/or progesterone receptor-positive, HER2-negative breast cancer were randomly assigned (2:1) between letrozole 2.5 mg PO daily plus bevacizumab 15 mg/kg IV every 3 weeks (Let/Bev) and letrozole 2.5 mg PO daily (Let) for 24 weeks prior to definitive surgery. Primary objective was within-arm pathologic complete remission (pCR) rate. Secondary objectives were safety, objective response, and downstaging rate. Results Seventy-five patients were randomized (Let/Bev n = 50, Let n = 25). Of the 45 patients evaluable for pathological response in the Let/Bev arm, 5 (11%; 95% CI, 3.7–24.1%) achieved pCR and 4 (9%; 95% CI, 2.5–21.2%) had microscopic residual disease; no pCRs or microscopic residual disease was seen in the Let arm (0%; 95% CI, 0–14.2%). The rates of downstaging were 44.4% (95% CI, 29.6–60.0%) and 37.5% (95% CI, 18.8–59.4%) in the Let/Bev and Let arms, respectively. Adverse events typically associated with letrozole (hot flashes, arthralgias, fatigue, myalgias) occurred in similar frequencies in the two arms. Hypertension, headache, and proteinuria were seen exclusively in the Let/Bev arm. The rates of grade 3 and 4 adverse events and discontinuation due to adverse events were 18% vs 8% and 16% vs none in the Let/Bev and Let arms, respectively. A small RNA-based classifier predictive of response to preoperative Let/Bev was developed and confirmed on an independent cohort. Conclusion In the preoperative setting, the addition of bevacizumab to letrozole was associated with a pCR rate of 11%; no pCR was seen with letrozole alone. There was additive toxicity with the incorporation of bevacizumab. Responses to Let/Bev can be predicted from the levels of 5 small RNAs in a pretreatment biopsy. Trial registration This trial is registered with ClinicalTrials.gov (Identifier: NCT00161291), first posted on September 12, 2005, and is completed.
Collapse
Affiliation(s)
- Christos Vaklavas
- University of Alabama at Birmingham, Birmingham, AL, USA.,Present Address: Huntsman Cancer Institute, University of Utah, Salt Lake City, USA
| | - Brian S Roberts
- HudsonAlpha, Institute for Biotechnology, Huntsville, AL, USA
| | | | - Nancy U Lin
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Minetta C Liu
- Lombardi Cancer Center, Georgetown University Hospital, Washington, DC, USA
| | - Hope S Rugo
- University of California, San Francisco, USA.,Hellen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Shannon Puhalla
- University of Pittsburgh Medical Center, Magee Women's Cancer Program, Pittsburgh, PA, USA
| | | | - Anna Maria Storniolo
- Melvin and Bren Simon Cancer Center at Indiana University, Indianapolis, IN, USA
| | - Lisa A Carey
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | | | - Yufeng Li
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | - Richard M Myers
- HudsonAlpha, Institute for Biotechnology, Huntsville, AL, USA
| | | | | |
Collapse
|
14
|
Tiainen L, Hämäläinen M, Luukkaala T, Tanner M, Lahdenperä O, Vihinen P, Jukkola A, Karihtala P, Moilanen E, Kellokumpu-Lehtinen PL. Low Plasma IL-8 Levels During Chemotherapy Are Predictive of Excellent Long-Term Survival in Metastatic Breast Cancer. Clin Breast Cancer 2019; 19:e522-e533. [DOI: 10.1016/j.clbc.2019.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/14/2019] [Accepted: 03/16/2019] [Indexed: 10/27/2022]
|
15
|
Hwang SY, Park S, Kwon Y. Recent therapeutic trends and promising targets in triple negative breast cancer. Pharmacol Ther 2019; 199:30-57. [PMID: 30825473 DOI: 10.1016/j.pharmthera.2019.02.006] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/04/2019] [Indexed: 12/14/2022]
Abstract
Breast cancer accounts for 25% of all types of cancer in women, and triple negative breast cancer (TNBC) comprises around 15~20% of breast cancers. Conventional chemotherapy and radiation are the primary systemic therapeutic strategies; no other FDA-approved targeted therapies are yet available as for TNBC. TNBC is generally characterized by a poor prognosis and high rates of proliferation and metastases. Due to these aggressive features and lack of targeted therapies, numerous attempts have been made to discover viable molecular targets for TNBC. Massive cohort studies, clinical trials, and in-depth analyses have revealed diverse molecular alterations in TNBC; however, controversy exists as to whether many of these changes are beneficial or detrimental in caner progression. Here we review the complicated tumorigenic processes and discuss critical findings and therapeutic trends in TNBC with a focus on promising therapeutic approaches, the clinical trials currently underway, and potent experimental compounds under preclinical and evaluation.
Collapse
Affiliation(s)
- Soo-Yeon Hwang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seojeong Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Youngjoo Kwon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
16
|
Liang X, Li H, Coussy F, Callens C, Lerebours F. An update on biomarkers of potential benefit with bevacizumab for breast cancer treatment: Do we make progress? Chin J Cancer Res 2019; 31:586-600. [PMID: 31564802 PMCID: PMC6736652 DOI: 10.21147/j.issn.1000-9604.2019.04.03] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
As the first monoclonal antibody against vascular endothelial growth factor (VEGF), bevacizumab (BEV) is a definitely controversial antiangiogenic therapy in breast cancer. The initial excitement over improvements in progression-free survival (PFS) with BEV was tempered by an absence of overall survival (OS) benefit and serious adverse effects. Missing targeted population urged us to identify the predictive biomarkers for BEV efficacy. In this review we focus on the research in breast cancer and provide recent investigations on clinical, radiological, molecular and gene profiling markers of BEV efficacy, including the new results from randomized phase III clinical trials evaluating the efficacy of BEV in combination with comprehensive biomarker analyses. Current evidences indicate some predictive values for genetic variants, molecular imaging, VEGF pathway factors or associated factors in peripheral blood and gene profiling. The current challenge is to validate those potential biomarkers and implement them into clinical practice.
Collapse
Affiliation(s)
- Xu Liang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China.,Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris 75005, France
| | - Huiping Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Florence Coussy
- Department of Medical Oncology, Institut Curie, PSL Research University, Paris 75005, France
| | - Celine Callens
- Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris 75005, France
| | - Florence Lerebours
- Department of Medical Oncology, Institut Curie, René Huguenin Hospital, Saint-Cloud 92210, France
| |
Collapse
|
17
|
Höglander EK, Nord S, Wedge DC, Lingjærde OC, Silwal-Pandit L, Gythfeldt HV, Vollan HKM, Fleischer T, Krohn M, Schlitchting E, Borgen E, Garred Ø, Holmen MM, Wist E, Naume B, Van Loo P, Børresen-Dale AL, Engebraaten O, Kristensen V. Time series analysis of neoadjuvant chemotherapy and bevacizumab-treated breast carcinomas reveals a systemic shift in genomic aberrations. Genome Med 2018; 10:92. [PMID: 30497530 PMCID: PMC6262977 DOI: 10.1186/s13073-018-0601-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/13/2018] [Indexed: 01/23/2023] Open
Abstract
Background Chemotherapeutic agents such as anthracyclines and taxanes are commonly used in the neoadjuvant setting. Bevacizumab is an antibody which binds to vascular endothelial growth factor A (VEGFA) and inhibits its receptor interaction, thus obstructing the formation of new blood vessels. Methods A phase II randomized clinical trial of 123 patients with Her2-negative breast cancer was conducted, with patients treated with neoadjuvant chemotherapy (fluorouracil (5FU)/epirubicin/cyclophosphamide (FEC) and taxane), with or without bevacizumab. Serial biopsies were obtained at time of diagnosis, after 12 weeks of treatment with FEC ± bevacizumab, and after 25 weeks of treatment with taxane ± bevacizumab. A time course study was designed to investigate the genomic landscape at the three time points when tumor DNA alterations, tumor percentage, genomic instability, and tumor clonality were assessed. Substantial differences were observed with some tumors changing mainly between diagnosis and at 12 weeks, others between 12 and 25 weeks, and still others changing in both time periods. Results In both treatment arms, good responders (GR) and non-responders (NR) displayed significant difference in genomic instability index (GII) at time of diagnosis. In the combination arm, copy number alterations at 25 loci at the time of diagnosis were significantly different between the GR and NR. An inverse aberration pattern was also observed between the two extreme response groups at 6p22-p12 for patients in the combination arm. Signs of subclonal reduction were observed, with some aberrations disappearing and others being retained during treatment. Increase in subclonal amplification was observed at 6p21.1, a locus which contains the VEGFA gene for the protein which are targeted by the study drug bevacizumab. Of the 13 pre-treatment samples that had a gain at VEGFA, 12 were responders. Significant decrease of frequency of subclones carrying gains at 17q21.32-q22 was observed at 12 weeks, with the peak occurring at TMEM100, an ALK1 receptor signaling-dependent gene essential for vasculogenesis. This implies that cells bearing amplifications of VEGFA and TMEM100 are particularly sensitive to this treatment regime. Conclusions Taken together, these results suggest that heterogeneity and subclonal architecture influence the response to targeted treatment in combination with chemotherapy, with possible implications for clinical decision-making and monitoring of treatment efficacy. Trial registration NCT00773695. Registered 15 October 2008 Electronic supplementary material The online version of this article (10.1186/s13073-018-0601-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elen Kristine Höglander
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Postboks 4953 Nydalen, 0424, Oslo, Norway.,KG Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Silje Nord
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Postboks 4953 Nydalen, 0424, Oslo, Norway.,KG Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - David C Wedge
- Big Data Institute, University of Oxford, Oxford, UK
| | - Ole Christian Lingjærde
- KG Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway.,Biomedical Informatics, Department of Informatics and Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
| | - Laxmi Silwal-Pandit
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Postboks 4953 Nydalen, 0424, Oslo, Norway.,KG Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hedda vdL Gythfeldt
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Postboks 4953 Nydalen, 0424, Oslo, Norway.,Department of Oncology, Oslo University Hospital, 0407, Oslo, Norway
| | - Hans Kristian Moen Vollan
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Postboks 4953 Nydalen, 0424, Oslo, Norway.,KG Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Oncology, Oslo University Hospital, 0407, Oslo, Norway
| | - Thomas Fleischer
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Postboks 4953 Nydalen, 0424, Oslo, Norway.,KG Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Marit Krohn
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Postboks 4953 Nydalen, 0424, Oslo, Norway.,KG Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ellen Schlitchting
- Section for Breast and Endocrine Surgery, Oslo University Hospital, Oslo, Norway
| | - Elin Borgen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Marit M Holmen
- Department of Radiology, Oslo University Hospital, Oslo, Norway
| | - Erik Wist
- Department of Oncology, Oslo University Hospital, 0407, Oslo, Norway
| | - Bjørn Naume
- KG Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Oncology, Oslo University Hospital, 0407, Oslo, Norway
| | - Peter Van Loo
- Cancer Research UK London Research Institute, London, UK
| | - Anne-Lise Børresen-Dale
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Postboks 4953 Nydalen, 0424, Oslo, Norway.,KG Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Olav Engebraaten
- KG Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway. .,Department of Oncology, Oslo University Hospital, 0407, Oslo, Norway.
| | - Vessela Kristensen
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Postboks 4953 Nydalen, 0424, Oslo, Norway. .,KG Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, University of Oslo, Oslo, Norway. .,Department of Clinical Molecular Biology (EpiGen), Divison of Medicine, Akershus University Hospital, Lørenskog, Norway. .,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
18
|
Miller KD, O’Neill A, Gradishar W, Hobday TJ, Goldstein LJ, Mayer IA, Bloom S, Brufsky AM, Tevaarwerk AJ, Sparano JA, Le-Lindqwister NA, Hendricks CB, Northfelt DW, Dang CT, Sledge GW. Double-Blind Phase III Trial of Adjuvant Chemotherapy With and Without Bevacizumab in Patients With Lymph Node-Positive and High-Risk Lymph Node-Negative Breast Cancer (E5103). J Clin Oncol 2018; 36:2621-2629. [PMID: 30040523 PMCID: PMC6118403 DOI: 10.1200/jco.2018.79.2028] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Purpose Bevacizumab improves progression-free survival but not overall survival in patients with metastatic breast cancer. E5103 tested the effect of bevacizumab in the adjuvant setting in patients with human epidermal growth factor receptor 2-negative disease. Patients and Methods Patients were assigned 1:2:2 to receive placebo with doxorubicin and cyclophosphamide (AC) followed by weekly paclitaxel (arm A), bevacizumab only during AC and paclitaxel (arm B), or bevacizumab during AC and paclitaxel followed by bevacizumab monotherapy for 10 cycles (arm C). Random assignment was stratified and bevacizumab dose adjusted for choice of AC schedule. Radiation and hormonal therapy were administered concurrently with bevacizumab in arm C. The primary end point was invasive disease-free survival (IDFS). Results Four thousand nine hundred ninety-four patients were enrolled. Median age was 52 years; 64% of patients were estrogen receptor positive, 27% were lymph node negative, and 78% received dose-dense AC. Chemotherapy-associated adverse events including myelosuppression and neuropathy were similar across all arms. Grade ≥ 3 hypertension was more common in bevacizumab-treated patients, but thrombosis, proteinuria, and hemorrhage were not. The cumulative incidence of clinical congestive heart failure at 15 months was 1.0%, 1.9%, and 3.0% in arms A, B, and C, respectively. Bevacizumab exposure was less than anticipated, with approximately 24% of patients in arm B and approximately 55% of patients in arm C discontinuing bevacizumab before completing planned therapy. Five-year IDFS was 77% (95% CI, 71% to 81%) in arm A, 76% (95% CI, 72% to 80%) in arm B, and 80% (95% CI, 77% to 83%) in arm C. Conclusion Incorporation of bevacizumab into sequential anthracycline- and taxane-containing adjuvant therapy does not improve IDFS or overall survival in patients with high-risk human epidermal growth factor receptor 2-negative breast cancer. Longer duration bevacizumab therapy is unlikely to be feasible given the high rate of early discontinuation.
Collapse
Affiliation(s)
- Kathy D. Miller
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - Anne O’Neill
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - William Gradishar
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - Timothy J. Hobday
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - Lori J. Goldstein
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - Ingrid A. Mayer
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - Stuart Bloom
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - Adam M. Brufsky
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - Amye J. Tevaarwerk
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - Joseph A. Sparano
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - Nguyet Anh Le-Lindqwister
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - Carolyn B. Hendricks
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - Donald W. Northfelt
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - Chau T. Dang
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| | - George W. Sledge
- Kathy D. Miller, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; Anne O’Neill, Dana-Farber Cancer Institute, Boston, MA; William Gradishar, Northwestern University, Chicago; Nguyet Anh Le-Lindqwister, Heartland Cancer Research National Cancer Institute Community Oncology Research Program, Peoria, IL; Timothy J. Hobday, Mayo Clinic, Rochester; Stuart Bloom, Abbott Northwestern Hospital, Minneapolis, MN; Lori J. Goldstein, Fox Chase Cancer Center, Philadelphia; Adam M. Brufsky, University of Pittsburgh, Pittsburgh, PA; Ingrid A. Mayer, Vanderbilt University, Nashville, TN; Amye J. Tevaarwerk, University of Wisconsin, Madison, WI; Joseph A. Sparano, Montefiore Hospital and Medical Center, Bronx; Chau T. Dang, Memorial Sloan Kettering Cancer Center, New York, NY; Carolyn B. Hendricks, Association Community Clinical Oncology Program, Bethesda, MD; Donald W. Northfelt, Mayo Clinic, Scottsdale, AZ; and George W. Sledge JR, Stanford University, Stanford, CA
| |
Collapse
|
19
|
Sanford T, Meng MV, Railkar R, Agarwal PK, Porten SP. Integrative analysis of the epigenetic basis of muscle-invasive urothelial carcinoma. Clin Epigenetics 2018; 10:19. [PMID: 29456764 PMCID: PMC5809922 DOI: 10.1186/s13148-018-0451-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 01/30/2018] [Indexed: 12/20/2022] Open
Abstract
Background Elucidation of epigenetic alterations in bladder cancer will lead to further understanding of the biology of the disease and hopefully improved therapies. Our aim was to perform an integrative epigenetic analysis of invasive urothelial carcinoma of the bladder to identify the epigenetic abnormalities involved in the development and progression of this cancer. Methods Pre-processed methylation data and RNA-seq data were downloaded from The Cancer Genome Atlas (TCGA) and processed using the R package TCGA-Assembler. An R package MethylMix was used to perform an analysis incorporating both methylation and gene expression data on all samples, as well as a subset analysis comparing patients surviving less than 2 years and patients surviving more than 2 years. Genes associated with poor prognosis were individually queried. Pathway analysis was performed on statistically significant genes identified by MethylMix criteria using ConsensusPathDB. Validation was performed using flow cytometry on bladder cancer cell lines. Results A total of 408 patients met all inclusion criteria. There were a total of 240 genes differentially methylated by MethylMix criteria. Review of individual genes specific to poor-prognosis patients revealed the majority to be candidate tumor suppressors in other cancer types. Pathway analysis showed increase in methylation of genes involved in antioxidant pathways including glutathione and NRF2. Genes involved in estrogen metabolism were also hypermethylated while genes involved in the EGFR pathway were found to be hypomethylated. EGFR expression was confirmed to be elevated in six bladder cancer cell lines. Conclusions In patients with invasive urothelial carcinoma, we found differential methylation in patients with better and worse prognosis after cystectomy. Differentially methylated genes are involved in many relevant oncologic pathways, including EGFR and antioxidant pathways, that may be a target for therapy or chemoprevention.
Collapse
Affiliation(s)
- Thomas Sanford
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Building 10—Hatfield CRC, Room 2-5952, Bethesda, MD 20892-1210 USA
| | - Maxwell V. Meng
- Department of Urology, University of California, Mail code 1695, 550 16th Street, 6th Floor, San Francisco, CA 94143 USA
| | - Reema Railkar
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Building 10—Hatfield CRC, Room 2-5952, Bethesda, MD 20892-1210 USA
| | - Piyush K. Agarwal
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Building 10—Hatfield CRC, Room 2-5952, Bethesda, MD 20892-1210 USA
| | - Sima P. Porten
- Department of Urology, University of California, Mail code 1695, 550 16th Street, 6th Floor, San Francisco, CA 94143 USA
| |
Collapse
|
20
|
Di Salvatore M, Pietrantonio F, Orlandi A, Del Re M, Berenato R, Rossi E, Caporale M, Guarino D, Martinetti A, Basso M, Mennitto R, Santonocito C, Mennitto A, Schinzari G, Bossi I, Capoluongo E, Danesi R, de Braud F, Barone C. IL-8 and eNOS polymorphisms predict bevacizumab-based first line treatment outcomes in RAS mutant metastatic colorectal cancer patients. Oncotarget 2017; 8:16887-16898. [PMID: 28129643 PMCID: PMC5370008 DOI: 10.18632/oncotarget.14810] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/02/2016] [Indexed: 12/19/2022] Open
Abstract
Background Predictive biomarkers of efficacy and toxicity of bevacizumab have not yet been validated. This study assessed the influence of IL-8, eNOS and VEGF-A polymorphisms in RAS mutated metastatic colorectal cancer patients receiving bevacizumab-based chemotherapy. Methods 120 patients treated with first-line combination FOLFOX6 plus bevacizumab were included. A historical cohort of 112 RAS mutated colorectal cancer patients treated with FOLFOX6 alone served as control group. The following SNPs were analyzed: IL-8 c.-251T>A; eNOS c.-786T>C and c.-894G>T; VEGF-A c.936C>T, c.958T>C, c.1154A>G and c.2578C>A. Correlation of SNPs, baseline IL-8 serum levels and bevacizumab-efficacy was done. Results In the bevacizumab group, carriers of the IL-8 alleles c.-251TA+AA showed a shorter PFS (P=0.002) and OS (P=0.03) compared to TT alleles. Patients with pre-treatment IL-8 < 18.25 pg/ml showed significantly longer median PFS and OS (PFS: 10.9 vs 7.6 months, P=0.005; OS: 30.7 vs 18.2 months, P<0.001) compared to patients with IL-8 higher levels (>18,25 pg/ml). IL-8 c.-251TA+AA carriers had significantly higher IL-8 levels (P<0.0001). Multivariate analysis confirmed association of IL-8 polymorphism with PFS, and of IL-8 baseline levels with both PFS and OS. IL-8 SNP did not affect the outcome in the control group. The eNOS polymorphism c.-894G>T was found associated with higher severe toxicity (P=0.0002) in patients carrying the c.-894TT genotype. Conclusions Although our data need prospective validation, IL-8 and eNOS SNPs may be have a role as predictive biomarkers for bevacizumab efficacy and toxicity.
Collapse
Affiliation(s)
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Armando Orlandi
- Unit of Clinical Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marzia Del Re
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Rosa Berenato
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Ernesto Rossi
- Unit of Clinical Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marta Caporale
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Donatella Guarino
- Laboratory of Clinical Molecular Biology, Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonia Martinetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Michele Basso
- Unit of Clinical Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Roberta Mennitto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Concetta Santonocito
- Laboratory of Clinical Molecular Biology, Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Alessia Mennitto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Giovanni Schinzari
- Unit of Clinical Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Ilaria Bossi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Ettore Capoluongo
- Laboratory of Clinical Molecular Biology, Institute of Biochemistry and Clinical Biochemistry, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetic Unit, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Carlo Barone
- Unit of Clinical Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
21
|
Xue S, He L, Zhang X, Zhou J, Li F, Wang X. Expression of Jagged1/Notch3 Signaling Pathway and their Relationship with the Tumor Angiogenesis in TNBC. Arch Med Res 2017. [PMID: 28625320 DOI: 10.1016/j.arcmed.2017.03.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Jagged1/Notch3 signaling pathway plays a key role in angiogenesis of breast cancer, but little is known in TNBC. This study was designed to investigate the expression of Jagged1/Notch3 mRNA and protein in TNBC, analyze their correlations with clinicopathological characteristics and prognosis. Moreover, the interrelationship among Jagged1/Notch3 and VEGF was initially evaluated. METHODS Jagged1/Notch3 mRNA and protein expression levels were determined by Q-RT-PCR and Western blotting. Additionally, Immunohistochemistry for Jagged1/Notch3 was detected by Ventana platform, VEGF and CD34 was performed using the EnVision/HRP technique. RESULTS mRNA transcriptionof Jagged1/Notch3 was in accord with protein expression. TNBC patients with positive Jagged1 expression had poorer DFS (p = 0.008) and OS (p = 0.004). Jagged1 expression was independent predictors of OS (p = 0.038). The expression of VEGF was positively correlative to MVD (p = 0.018), MVD was significantly associated with Jagged1 (p <0.0001) and Notch3 (p <0.0001). The expression of Jagged1/Notch3 has no correlation with VEGF, only in positive VEGF expression of TNBC patients Jagged1/Notch3 had influence on DFS and OS (p <0.05). CONCLUSION Jagged1/Notch3 was -expressed at both the mRNA and protein levels, Jagged1 served as an independent predictor of poor prognosis. We speculate that there is a cross-talk between Jagged1/Notch3 and VEGF in TNBC angiogenesis. Jagged1/Notch3 is expected to be an important signaling pathway for TNBC progression and a potential target for TNBC neovascularization therapy.
Collapse
Affiliation(s)
- Siliang Xue
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lang He
- Cancer Center, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Xiao Zhang
- Department of Breast Surgery, Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Jin Zhou
- Department of Chemotherapy, Sichuan Cancer Hospital, Chengdu, Sichuan, China
| | - Fanghua Li
- Department of Pathology, Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Xiaoshan Wang
- Cancer Center, Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
22
|
Jitariu AA, Cîmpean AM, Ribatti D, Raica M. Triple negative breast cancer: the kiss of death. Oncotarget 2017; 8:46652-46662. [PMID: 28445140 PMCID: PMC5542300 DOI: 10.18632/oncotarget.16938] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/19/2017] [Indexed: 12/22/2022] Open
Abstract
One of the most controversial women malignancies, triple negative breast cancers (TNBCs) are critically overviewed here, being focused on data useful in clinical practice or to improve the therapy and patients survival. TNBCs "choose" young women and its "kiss" is, unfortunately deadly in most cases. Currently, few sparse data are available in literature concerning the origins of TNBC. Vasculogenic mimicry detected in TNBCs, seems to be determined by a population of CD133+ cells and may be stimulated by different pharmacological agents such sunitinib. Despite the fact that TNBCs do not usually metastasize through the lymphatic pathways, TNBCs may be characterized by lymphatic invasion and by an increased lymphatic microvascular density. If TNBCs treatment depends on the molecular profile of the tumor, the same statement may be postulated for TNBCs metastasis. Whether metastases have a similar phenotype as the primary tumor remains an enigma. Therefore, the question: 'Could TNBC be subject to a standardized, unanimously accepted therapeutic strategy or is it strictly subclass-dependent?' remains to be further investigated.
Collapse
Affiliation(s)
- Adriana-Andreea Jitariu
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | - Anca Maria Cîmpean
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| | - 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
| | - Marius Raica
- Department of Microscopic Morphology/Histology, Angiogenesis Research Center, Victor Babeş University of Medicine and Pharmacy, Timişoara, Romania
| |
Collapse
|
23
|
Does biomarker information impact breast cancer patients' preferences and physician recommendation for adjuvant chemotherapy? Breast Cancer Res Treat 2017. [PMID: 28646344 DOI: 10.1007/s10549-017-4338-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE This study aimed to examine how biomarker information would impact patients' preferences and physicians' recommendations for adjuvant breast cancer therapy. METHODS At the 18-month follow-up, participants in a large, double-blind randomized controlled trial of adjuvant chemotherapy with bevacizumab or placebo (E5103) were surveyed about their preferred treatment (either chemotherapy A alone or chemotherapy A+B) in two hypothetical scenarios: (1) without biomarker information; and (2) after learning that they tested positive for a "B-receptor" which modestly increased both the benefit and toxicity expected with chemotherapy A+B. We performed a cross-sectional analysis of the prospectively collected survey data and used the McNemar's test to examine changes in treatment preferences. A one-time survey of clinical investigators who enrolled patients on the trial evaluated physician recommendations in response to the same biomarker information. RESULTS 439 patients completed both scenarios on 18-month survey. Most participants preferred A+B in both scenario 1 and 2 (77 and 76% respectively). The increase in benefit and toxicity associated with the positive biomarker information in scenario 2 led 60/439 (14%) of patients to switch their treatment preference. The corresponding physician survey revealed that most physicians chose regimen A+B in scenario 1 (77%), and moreso after the biomarker information was available in scenario 2 (84%). CONCLUSIONS Information about a positive biomarker indicating increased benefit and toxicity from additional chemotherapy did not change many participants' preferred treatment. The majority preferred the most effective course in both scenarios. Similarly, most investigators discounted increased toxicity and valued increased benefit. Parent Trial Registration: NCT00433511.
Collapse
|
24
|
Yu T, Di G. Role of tumor microenvironment in triple-negative breast cancer and its prognostic significance. Chin J Cancer Res 2017; 29:237-252. [PMID: 28729775 PMCID: PMC5497211 DOI: 10.21147/j.issn.1000-9604.2017.03.10] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Breast cancer has been shown to live in the tumor microenvironment, which consists of not only breast cancer cells themselves but also a significant amount of pathophysiologically altered surrounding stroma and cells. Diverse components of the breast cancer microenvironment, such as suppressive immune cells, re-programmed fibroblast cells, altered extracellular matrix (ECM) and certain soluble factors, synergistically impede an effective anti-tumor response and promote breast cancer progression and metastasis. Among these components, stromal cells in the breast cancer microenvironment are characterized by molecular alterations and aberrant signaling pathways, whereas the ECM features biochemical and biomechanical changes. However, triple-negative breast cancer (TNBC), the most aggressive subtype of this disease that lacks effective therapies available for other subtypes, is considered to feature a unique microenvironment distinct from that of other subtypes, especially compared to Luminal A subtype. Because these changes are now considered to significantly impact breast cancer development and progression, these unique alterations may serve as promising prognostic factors of clinical outcome or potential therapeutic targets for the treatment of TNBC. In this review, we focus on the composition of the TNBC microenvironment, concomitant distinct biological alteration, specific interplay between various cell types and TNBC cells, and the prognostic implications of these findings.
Collapse
Affiliation(s)
- Tianjian Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Genhong Di
- Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
25
|
Matsumura C, Chisaki Y, Sakimoto S, Sakae H, Yano Y. Evaluation of thromboembolic events in cancer patients receiving bevacizumab according to the Japanese Adverse Drug Event Report database. J Oncol Pharm Pract 2016; 24:22-27. [PMID: 27856923 DOI: 10.1177/1078155216679025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Purpose We aimed to examine the risk factors, time of onset, incidence rates, and outcomes of thromboembolic events induced by bevacizumab in patients with cancer using the Japanese Adverse Drug Event Report (JADER) database of the Pharmaceuticals and Medical Devices Agency. Methods Adverse event data recorded in the JADER database between January 2004 and January 2015 were used. After screening the data using the generic drug name bevacizumab, patient data were classified into two groups by age and five groups by cancer type. The histories of disorders were also categorized. Arterial thromboembolic event and venous thromboembolic event were classified as "favorable" or "unfavorable" outcomes. Results In total, 6076 patients were reported to have developed adverse events during the sample period, of which 233 and 453 developed arterial thromboembolic event and venous thromboembolic event, respectively. Logistic analysis suggested that the presence of cancer was a significant risk factor for both arterial thromboembolic event and venous thromboembolic event. Age (≥70 years), histories of either hypertension or diabetes mellitus were also risk factors for arterial thromboembolic event. Median cumulative times of onset for arterial thromboembolic event and venous thromboembolic event were 60 and 80 days, respectively, and were not significantly different by the log-rank test. By the chi-square test, the rate of unfavorable outcomes was found to be higher after developing arterial thromboembolic event than after venous thromboembolic event. Conclusion Thromboembolism is a leading cause of mortality in patients with cancer. Patients should be monitored for the symptoms of thromboembolic events right from the initial stages of bevacizumab treatment.
Collapse
Affiliation(s)
- Chikako Matsumura
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yugo Chisaki
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Satoko Sakimoto
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Honoka Sakae
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yoshitaka Yano
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, Kyoto, Japan
| |
Collapse
|
26
|
Ma X, Wang X, Huang J, Chen Y, Zhang J, Zhang B, Shi C, Liu L. Bevacizumab Addition in Neoadjuvant Treatment Increases the Pathological Complete Response Rates in Patients with HER-2 Negative Breast Cancer Especially Triple Negative Breast Cancer: A Meta-Analysis. PLoS One 2016; 11:e0160148. [PMID: 27579484 PMCID: PMC5006981 DOI: 10.1371/journal.pone.0160148] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 07/14/2016] [Indexed: 02/05/2023] Open
Abstract
Background Neoadjuvant therapy is administered to breast cancer patients as an induction process before surgery or radiotherapy to reduce tumor size. Human epidermal growth factor receptor-2 (HER-2) negative breast cancer lacks effective standard target therapy. Bevacizumab has a controversial role in the treatment of breast cancer and we conduct a meta-analysis to evaluate the value of adding bevacizumab in neoadjuvant regimen. Methods Potentially eligible studies were retrieved using PubMed, EMBASE and Medline. Clinical characteristics of patients and statistical data with pathological complete response (pCR) data were collected. Then a meta-analysis model was established to investigate the correlation between administration of bevacizumab in neoadjuvant therapy and pCR rates in HER-2 negative breast cancer. Results Seven eligible studies and 5408 patients were yielded. The pCR rates for “breast” or “breast plus lymph node” were similar. In subgroup analysis, we emphasized on patients with triple-negative breast cancer (TNBC). In the criterion of “lesions in breast” the pooled ORs was 1.55 [1.29, 1.86], P<0.00001 and regarding to the evaluation criterion of “lesions in breast and lymph nodes”, the pooled ORs was 1.48 [1.23, 1.78], P<0.0001, in favor of bevacizumab administration. Conclusion According to our pooled results, we finally find that bevacizumab addition as a neoadjuvant chemotherapy component, for induction use with limited cycle to improve the pCR rates and patients may avoid long-term adverse event and long-term invalid survival improvement. Especially in subgroup analysis, pCR rates could be improved significantly and physicians could consider bevacizumab with caution. As patients could avoid the adverse event caused by long-term using of bevacizumab, long-term quality of life improvement may be achieved, especially in TNBC.
Collapse
Affiliation(s)
- Xuelei Ma
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoshan Wang
- Department of Clinical Oncology, Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Jingwen Huang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yingtai Chen
- Department of Abdominal Surgery, Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical Collage, Beijing, China
| | - Jing Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Binglan Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Changle Shi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Liu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- * E-mail:
| |
Collapse
|
27
|
Liu J, Fan L, Wang H, Sun G. Autophagy, a double-edged sword in anti-angiogenesis therapy. Med Oncol 2015; 33:10. [PMID: 26715036 DOI: 10.1007/s12032-015-0721-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 12/12/2015] [Indexed: 12/24/2022]
Abstract
Autophagy is a highly conservative cell behavior to keep the intracellular homeostasis and is frequently activated when cells encounter disgusting conditions, such as nutrition or growth factor deprive, hypoxia and cytotoxic agents. However, the precise role of autophagy under various conditions may be opposite, differ from protect cells survival to promote cells death, and the mechanism of this conditional-dependent role is still unclear. Anti-angiogenesis agents, such as bevacizumab, sorafenib and sunitinib, could reduce tumor microvascular density and increase tumor hypoxia, thus up-regulating autophagy activation of tumor cells, but the function of autophagy induced by anti-angiogenesis agents is still divergent and is considered to play a cytoprotective role in most cases. In this review, we mainly discuss the relationship between anti-angiogenesis therapy-induced hypoxia and autophagy, and pay special attention on the exact role of anti-angiogenesis agents induced autophagy in the process of anti-angiogenesis treatment.
Collapse
Affiliation(s)
- Jiatao Liu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui Province, China.,Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Lulu Fan
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Guoping Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui Province, China.
| |
Collapse
|
28
|
Lehmann BD, Pietenpol JA. Clinical implications of molecular heterogeneity in triple negative breast cancer. Breast 2015; 24 Suppl 2:S36-40. [PMID: 26253813 PMCID: PMC4641762 DOI: 10.1016/j.breast.2015.07.009] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Triple negative breast cancer (TNBC) is a molecularly heterogeneous disease lacking recurrent targetable alterations and thus therapeutic advances have been challenging. The absence of ER, PR and HER2 amplifications, leaves combination chemotherapy as the standard of care treatment option in the adjuvant, neoadjuvant and metastatic settings. Recently, multiple studies have shed some light on the heterogeneity of TNBC and identified distinct transcriptional subtypes with unique biologies. Herein we review the molecular heterogeneity and the impact on previous and future clinical trials.
Collapse
Affiliation(s)
- Brian D Lehmann
- Department of Biochemistry, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Preston Research Building, 2200 Pierce Avenue, Nashville, TN 37232, USA.
| | - Jennifer A Pietenpol
- Department of Biochemistry, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Preston Research Building, 2200 Pierce Avenue, Nashville, TN 37232, USA.
| |
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
|
Li T, Wang B, Wang Z, Ragaz J, Zhang J, Sun S, Cao J, Lv F, Wang L, Zhang S, Ni C, Wu Z, Xie J, Hu X. Bevacizumab in Combination with Modified FOLFOX6 in Heavily Pretreated Patients with HER2/Neu-Negative Metastatic Breast Cancer: A Phase II Clinical Trial. PLoS One 2015; 10:e0133133. [PMID: 26186012 PMCID: PMC4506015 DOI: 10.1371/journal.pone.0133133] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/19/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Bevacizumab combined with modified FOLFOX6 is a standard regimen for colorectal cancer. The present study was to determine the efficacy and safety of bevacizumab-modified FOLFOX6 regimen in heavily pretreated patients with human epidermal growth factor receptor 2 (HER2/neu)-negative MBC. METHODS Bevacizumab, 5 mg/kg every two weeks or 7.5 mg/kg every three weeks, was administered with modified FOLFOX6 (oxaliplatin 85 mg/m2, leucovorin 400 mg/m2, 5-FU 400 mg/m2 on day 1, followed by 5-FU 2400 mg/m2 intravenous infusion over 46 hours every 2 weeks) to patients who failed at least 1 chemotherapy regimen in the metastatic setting. The primary objective was progression free survival (PFS). Secondary objectives included objective response rate (ORR), clinical benefit rate (CBR), overall survival (OS), safety, and the change of tumor size and Eastern Cooperative Oncology Group (ECOG) performance status. RESULTS 69 patients were enrolled. The median PFS was 6.8 months (95% CI, 5.0 to 8.5 months), ORR was 50.0% and median OS was 10.5 months (95% CI, 7.9 to 13.1 months). Patients showing objective responses had a 4.2-month median PFS gain and 5.7-month median OS gain compared with those who did not (P < 0.05). Grade 3 or 4 adverse events occurring in more than one patient were neutropenia (53/69, 76.8%), leukopenia (36/69, 52.2%), thrombocytopenia (13/69, 18.8%), anemia (3/69, 4.3%) and hypertension (3/69, 4.3%). CONCLUSIONS Adding bevacizumab to modified FOLFOX6 does have significant anti-tumor activity and good safety profile in heavily pretreated HER2/neu-negative MBC patients. Further trials are required to confirm whether the high ORR can translate into a long-term PFS and even OS benefit. TRIAL REGISTRATION www.clinicaltrials.gov NCT01658033.
Collapse
Affiliation(s)
- Ting Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Biyun Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhonghua Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Joseph Ragaz
- Faculty of Medicine, School of Population & Public Health, University of British Columbia, Vancouver, B.C., V6T 1Z4, Canada
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Si Sun
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jun Cao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Fangfang Lv
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Leiping Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Sheng Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Chen Ni
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhenhua Wu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jie Xie
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xichun Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- * E-mail:
| |
Collapse
|
31
|
Hein A, Lambrechts D, von Minckwitz G, Häberle L, Eidtmann H, Tesch H, Untch M, Hilfrich J, Schem C, Rezai M, Gerber B, Dan Costa S, Blohmer JU, Schwedler K, Kittel K, Fehm T, Kunz G, Beckmann MW, Ekici AB, Hanusch C, Huober J, Liedtke C, Mau C, Moisse M, Müller V, Nekljudova V, Peuteman G, Rack B, Rübner M, Van Brussel T, Wang L, Weinshilboum RM, Loibl S, Fasching PA. Genetic variants in VEGF pathway genes in neoadjuvant breast cancer patients receiving bevacizumab: Results from the randomized phase III GeparQuinto study. Int J Cancer 2015; 137:2981-8. [PMID: 26100253 DOI: 10.1002/ijc.29656] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 03/17/2015] [Indexed: 12/28/2022]
Abstract
Studies assessing the effect of bevacizumab (BEV) on breast cancer (BC) outcome have shown different effects on progression-free and overall survival, suggesting that a subgroup of patients may benefit from this treatment. Unfortunately, no biomarkers exist to identify these patients. Here, we investigate whether single nucleotide polymorphisms (SNPs) in VEGF pathway genes correlate with pathological complete response (pCR) in the neoadjuvant GeparQuinto trial. HER2-negative patients were randomized into treatment arms receiving either BEV combined with standard chemotherapy or chemotherapy alone. In a pre-planned biomarker study, DNA was collected from 729 and 724 patients, respectively from both treatment arms, and genotyped for 125 SNPs. Logistic regression assessed interaction between individual SNPs and both treatment arms to predict pCR. Five SNPs may be associated with a better response to BEV, but none of them remained significant after correction for multiple testing. The two SNPs most strongly associated, rs833058 and rs699947, were located upstream of the VEGF-A promoter. Odds ratios for the homozygous common, heterozygous and homozygous rare rs833058 genotypes were 2.36 (95% CI, 1.49-3.75), 1.20 (95% CI, 0.88-1.64) and 0.61 (95% CI, 0.34-1.12). Notably, some SNPs in VEGF-A exhibited a more pronounced effect in the triple-negative subgroup. Several SNPs in VEGF-A may be associated with improved pCR when receiving BEV in the neoadjuvant setting. Although none of the observed effects survived correction for multiple testing, our observations are consistent with previous studies on BEV efficacy in BC. Further research is warranted to clarify the predictive value of these markers.
Collapse
Affiliation(s)
- Alexander Hein
- Department of Gynecology and Obstetrics, University Breast Center for Franconia, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen-EMN Comprehensive Cancer Center, Erlangen, Germany
| | - Diether Lambrechts
- Vesalius Research Center, VIB, Leuven, Belgium.,Laboratory of Translational Genetics, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Gunter von Minckwitz
- GBG Forschungs GmbH, Neu-Isenburg, Germany.,Department of Gynecology and Obstetrics, Frankfurt University Hospital, Goethe University of Frankfurt, Frankfurt Am Main, Germany
| | - Lothar Häberle
- Department of Gynecology and Obstetrics, University Breast Center for Franconia, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen-EMN Comprehensive Cancer Center, Erlangen, Germany.,Biostatistics Unit, Department of Gynecology and Obstetrics, Erlangen University Hospital, Erlangen, Germany
| | - Holger Eidtmann
- Department of Obstetrics and Gynecology, Christian-Albrechts-University, Kiel, Germany
| | - Hans Tesch
- CHOP Onkologie Bethanien, Frankfurt, Germany
| | | | | | - Christian Schem
- Department of Obstetrics and Gynecology, Christian-Albrechts-University, Kiel, Germany
| | - Mahdi Rezai
- Breast Center of Düsseldorf, Luisenkrankenhaus, Düsseldorf, Germany
| | - Bernd Gerber
- Department of Gynecology, University of Rostock, Rostock, Germany
| | - Serban Dan Costa
- Department of Obstetrics and Gynecology, Magdeburg University Hospital, Magdeburg, Germany
| | - Jens-Uwe Blohmer
- St. Getrauden Breast Center, St. Gertrauden Krankenhaus, Berlin, Germany
| | - Kathrin Schwedler
- Department of Gynecology and Obstetrics, Frankfurt University Hospital, Goethe University of Frankfurt, Frankfurt Am Main, Germany.,Neue Frauenklinik, Lucerne Cantonal Hospital, Lucerne, Switzerland
| | - Kornelia Kittel
- Praxisklinik, Krebsheilkunde Für Frauen, Brustzentrum, Berlin, Germany
| | - Tanja Fehm
- Department of Gynecology, University of Tübingen, Tübingen, Germany.,Department of Gynecology and Obstetrics, Düsseldorf University Hospital, Heinrich Heine University, Düsseldorf, Germany
| | - Georg Kunz
- St. Johannes Hospital, Dortmund, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, University Breast Center for Franconia, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen-EMN Comprehensive Cancer Center, Erlangen, Germany
| | - Arif B Ekici
- Institute of Human Genetics, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Claus Hanusch
- Department of Gynecology, Klinikum Zum Roten Kreuz, Munich, Germany
| | - Jens Huober
- Department of Gynecology, University of Ulm, Ulm, Germany
| | - Cornelia Liedtke
- Department of Gynecology and Obstetrics, Lübeck University Hospital, Lübeck, Germany
| | | | | | - Volkmar Müller
- Department of Gynecology and Obstetrics, Hamburg University Hospital, Hamburg, Germany
| | | | | | - Brigitte Rack
- Department of Gynecology and Obstetrics, Ludwig Maximilian University, Munich, Germany
| | - Matthias Rübner
- Department of Gynecology and Obstetrics, University Breast Center for Franconia, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen-EMN Comprehensive Cancer Center, Erlangen, Germany
| | | | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Mayo Medical School-Mayo Foundation, Rochester, MN
| | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Mayo Medical School-Mayo Foundation, Rochester, MN
| | - Sibylle Loibl
- GBG Forschungs GmbH, Neu-Isenburg, Germany.,Department of Gynecology and Obstetrics, Frankfurt University Hospital, Goethe University of Frankfurt, Frankfurt Am Main, Germany
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, University Breast Center for Franconia, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen-EMN Comprehensive Cancer Center, Erlangen, Germany.,Division of Hematology/Oncology, Department of Medicine, University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA
| |
Collapse
|
32
|
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: 1.0] [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
|
33
|
Li Y, Wei X, zhang S, Zhang J. Prognosis of invasive breast cancer after adjuvant therapy evaluated with VEGF microvessel density and microvascular imaging. Tumour Biol 2015; 36:8755-60. [DOI: 10.1007/s13277-015-3610-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/22/2015] [Indexed: 12/20/2022] Open
|
34
|
Luengo-Gil G, González-Billalabeitia E, Chaves-Benito A, García Martínez E, García Garre E, Vicente V, Ayala de la Peña F. Effects of conventional neoadjuvant chemotherapy for breast cancer on tumor angiogenesis. Breast Cancer Res Treat 2015; 151:577-87. [PMID: 25967462 DOI: 10.1007/s10549-015-3421-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 05/07/2015] [Indexed: 01/01/2023]
Abstract
The effects of breast cancer conventional chemotherapy on tumor angiogenesis need to be further characterized. Neoadjuvant chemotherapy is an ideal model to evaluate the results of chemotherapy, allowing intra-patient direct comparison of antitumor and antiangiogenic effects. We sought to analyze the effect of neoadjuvant chemotherapy on tumor angiogenesis and its clinical significance in breast cancer. Breast cancer patients (n = 108) treated with neoadjuvant sequential anthracyclines and taxanes were studied. Pre- and post-chemotherapy microvessel density (MVD) and mean vessel size (MVS) were analyzed after CD34 immunohistochemistry and correlated with tumor expression of pro- and antiangiogenic factors (VEGFA, THBS1, HIF1A, CTGF, and PDGFA) by qRT-PCR. Angiogenic measures at diagnosis varied among breast cancer subtypes. Pre-treatment higher MVS was associated with triple-negative subtype and more advanced disease. Higher MVS was correlated with higher VEGFA (p = 0.003), while higher MVD was correlated with lower antiangiogenic factors expression (THBS1, p < 0.0001; CTGF, p = 0.001). Increased angiogenesis at diagnosis (high MVS and glomeruloid microvascular proliferation) and higher VEGFA expression were associated with tumor recurrence (p = 0.048 and 0.009, respectively). Chemotherapy-induced angiogenic response (defined as decreased MVD) was present in 35.2 % of patients. This response correlated with an increase in antiangiogenic factors (THBS1) without changes in VEGFA expression, and it was associated with tumor downstaging, but not with clinical response, pathologic complete response, or prognosis. Global effects of chemotherapy mainly consisted in an increased expression of antiangiogenic factors (THBS1, CTGF), with significant changes neither of tumor VEGFA nor of MVS. Conventionally scheduled neoadjuvant chemotherapy exerts antiangiogenic effects, through an increase in antiangiogenic factors, THBS1 and CTGF, but the expression of VEGFA is maintained after treatment. Better markers of angiogenic response and a better understanding of the cooperation of chemotherapy and antiangiogenic therapy in the neoadjuvant clinical scenario are needed.
Collapse
Affiliation(s)
- Ginés Luengo-Gil
- Department of Hematology and Medical Oncology, University Hospital Morales Meseguer, Avda. Marqués de los Vélez, s/n, 30008, Murcia, Spain
| | | | | | | | | | | | | |
Collapse
|
35
|
Tonissi F, Lattanzio L, Merlano MC, Infante L, Lo Nigro C, Garrone O. The effect of paclitaxel and nab-paclitaxel in combination with anti-angiogenic therapy in breast cancer cell lines. Invest New Drugs 2015; 33:801-9. [PMID: 25947567 DOI: 10.1007/s10637-015-0249-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/30/2015] [Indexed: 02/03/2023]
Abstract
Taxanes represent a treatment of choice for metastatic breast cancer. Their combination with bevacizumab improved response rate and progression-free survival. We studied in vitro the effect on cell survival of the combination of either paclitaxel or nab-paclitaxel with bevacizumab and we investigated the biological factors involved in the response to treatments. We used two breast cancer cell lines, MCF7 (ER+/HER2-) and MDA-MB-231 (ER-/HER2-), co-cultured with or without HUVEC cells. We analysed cell survival by MTT test, VEGF secretion by ELISA and VEGFR, SPARC, MDR1 expression by western blot. Doses of both taxanes causing a 50 % growth inhibition were higher in MCF7 than MDA-MB-231, suggesting that taxanes are more effective in ER- cell lines. When both cell lines were grown as single culture, the combination bevacizumab+paclitaxel showed a similar anti-proliferative effect compared to paclitaxel alone. The association bevacizumab+nab-paclitaxel was more effective than nab-paclitaxel alone. An increased anti-proliferative effect of bevacizumab+paclitaxel was observed when MDA-MB-231 cells were cultured with HUVEC. We detected an induction of VEGF secretion when MDA-MB-231 cells were treated with either taxanes. Paclitaxel caused a reduction of VEGF in MCF7. SPARC resulted up-regulated in both cell lines treated with bevacizumab+nab-paclitaxel. Nab-paclitaxel seems to play an important role in inhibiting tumor proliferation through albumin-SPARC bound in association with bevacizumab compared to taxanes alone in both breast cancer cells. The addition of bevacizumab to paclitaxel increased its activity only in ER- cells. This difference might be due to their ER status.
Collapse
Affiliation(s)
- Federica Tonissi
- Laboratory of Cancer Genetics and Translational Oncology, Oncology Department, S. Croce & Carle Teaching Hospital, via Carle 25, 12100, Cuneo, Italy,
| | | | | | | | | | | |
Collapse
|
36
|
Manso L, Moreno F, Márquez R, Castelo B, Arcediano A, Arroyo M, Ballesteros AI, Calvo I, Echarri MJ, Enrech S, Gómez A, González Del Val R, López-Miranda E, Martín-Angulo M, Martínez-Jañez N, Olier C, Zamora P. Use of bevacizumab as a first-line treatment for metastatic breast cancer. ACTA ACUST UNITED AC 2015; 22:e51-60. [PMID: 25908921 DOI: 10.3747/co.22.2210] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVE During clinical practice, it can be challenging, given the lack of response biomarkers, to identify the patients with metastatic breast cancer (mbca) who would benefit most from the addition of bevacizumab to first-line standard chemotherapy. The aim of the present review was to summarize the relevant scientific evidence and to discuss the experience of a group of experts in using bevacizumab to treat mbca. METHODS A panel of 17 Spanish oncology experts met to discuss the literature and their experience in the use of bevacizumab as first-line treatment for mbca. During the meeting, discussions focused on three main issues: the profile of the patients who could benefit most from bevacizumab, the optimal bevacizumab treatment duration, and the safety profile of bevacizumab. RESULTS The subset of mbca patients who would benefit the most from the addition of bevacizumab to first-line standard chemotherapy are those with clinically defined aggressive disease. Treatment with bevacizumab should be maintained until disease progression or the appearance of unacceptable toxicity. In the mbca setting, the toxicity profile of bevacizumab is well known and can be managed in clinical practice after adequate training. CONCLUSIONS This expert group recommends administering bevacizumab as first-line treatment in patients with clinically aggressive disease.
Collapse
Affiliation(s)
- L Manso
- Hospital Universitario 12 de Octubre, Madrid, Spain
| | - F Moreno
- Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - R Márquez
- MD Anderson Cancer Center, Madrid, Spain
| | - B Castelo
- Hospital Universitario La Paz, Madrid, Spain
| | - A Arcediano
- Hospital General Universitario de Guadalajara, Guadalajara, Mexico
| | - M Arroyo
- Hospital Príncipe de Asturias, Alcalá de Henares, Spain
| | | | - I Calvo
- Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - M J Echarri
- Hospital Universitario Severo Ochoa, Leganés, Spain
| | - S Enrech
- Hospital Universitario de Getafe, Getafe, Spain
| | - A Gómez
- Hospital Universitario de Salamanca, Salamanca, Spain
| | | | | | | | | | - C Olier
- Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - P Zamora
- Hospital Universitario La Paz, Madrid, Spain
| |
Collapse
|
37
|
Joy A, Ghosh M, Fernandes R, Clemons M. Systemic treatment approaches in her2-negative advanced breast cancer-guidance on the guidelines. Curr Oncol 2015; 22:S29-42. [PMID: 25848337 PMCID: PMC4381789 DOI: 10.3747/co.22.2360] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Despite advancements in the treatment of early-stage breast cancer, many patients still develop disease recurrence; others present with de novo metastatic disease. For most patients with advanced breast cancer, the primary treatment intent is noncurative-that is, palliative-in nature. The goals of treatment should therefore focus on maximizing symptom control and extending survival. Treatments should be evaluated on an individualized basis in terms of evidence, but also with full respect for the wishes of the patient in terms of acceptable toxicity. Given the availability of extensive reviews on the roles of endocrine therapy and her2 (human epidermal growth factor receptor 2)-targeted therapies for advanced disease, we focus here mainly on treatment guidelines for the non-endocrine management of her2-negative advanced breast cancer in a Canadian health care context.
Collapse
Affiliation(s)
- A.A. Joy
- Department of Oncology, Division of Medical Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB
| | - M. Ghosh
- Department of Oncology, Division of Medical Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB
| | - R. Fernandes
- Division of Medical Oncology, University of Ottawa and The Ottawa Hospital Research Institute, The Ottawa Hospital Cancer Centre, Ottawa, ON
| | - M.J. Clemons
- Division of Medical Oncology, University of Ottawa and The Ottawa Hospital Research Institute, The Ottawa Hospital Cancer Centre, Ottawa, ON
| |
Collapse
|
38
|
Koutras A, Kotoula V, Fountzilas G. Prognostic and predictive role of vascular endothelial growth factor polymorphisms in breast cancer. Pharmacogenomics 2015; 16:79-94. [DOI: 10.2217/pgs.14.148] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Current evidence indicates that angiogenesis plays an important role in the pathogenesis of several malignancies, including breast cancer. The vascular endothelial growth factor (VEGF) pathway has been investigated extensively, due to its important role in angiogenesis. The major mediator of tumor angiogenesis is VEGF-A, frequently referred to as VEGF, which activates the VEGF receptor-2. The VEGF gene is located on chromosome 6 and constitutes a highly polymorphic gene. Numerous SNPs in the promoter, 5′- and 3′-untranslated regions (UTR) of VEGF gene have been recognized. This genetic variability possibly influences the production and function of VEGF. Subsequently, the VEGF SNPs may have an impact on breast cancer risk and disease outcome. Moreover, these SNPs may be of predictive value in patients receiving agents targeting the VEGF pathway. This review presents an update on the potential role of VEGF SNPs as prognostic and/or predictive markers in patients with breast cancer.
Collapse
Affiliation(s)
- Angelos Koutras
- Division of Oncology, Department of Medicine, University Hospital, University of Patras Medical School, Patras, Rion 26504, Greece
| | - Vasiliki Kotoula
- Department of Pathology, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - George Fountzilas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
- Department of Medical Oncology, ‘Papageorgiou’ Hospital, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| |
Collapse
|
39
|
Gökmen-Polar Y, Goswami CP, Toroni RA, Sanders KL, Mehta R, Sirimalle U, Tanasa B, Shen C, Li L, Ivan M, Badve S, Sledge GW. Gene Expression Analysis Reveals Distinct Pathways of Resistance to Bevacizumab in Xenograft Models of Human ER-Positive Breast Cancer. J Cancer 2014; 5:633-45. [PMID: 25157274 PMCID: PMC4142325 DOI: 10.7150/jca.8466] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Accepted: 06/20/2014] [Indexed: 12/29/2022] Open
Abstract
Bevacizumab, the recombinant antibody targeting vascular endothelial growth factor (VEGF), improves progression-free but not overall survival in metastatic breast cancer. To seek further insights in resistance mechanisms to bevacizumab at the molecular level, we developed VEGF and non-VEGF-driven ER-positive MCF7-derived xenograft models allowing comparison of tumor response at different timepoints. VEGF gene (MV165) overexpressing xenografts were initially sensitive to bevacizumab, but eventually acquired resistance. In contrast, parental MCF7 cells derived tumors were de novo insensitive to bevacizumab. Microarray analysis with qRT-PCR validation revealed that Follistatin (FST) and NOTCH were the top signaling pathways associated with resistance in VEGF-driven tumors (P<0.05). Based on the presence of VEGF, treatment with bevacizumab resulted in altered patterns of metagenes and PAM50 gene expression. In VEGF-driven model after short and long-term bevacizumab treatments, a change in the intrinsic subtype (luminal to myoepithelial/basal-like) was observed in association with increased expression of genes implicated with cancer stem cell phenotype (P<0.05). Our results show that the presence or absence of VEGF expression affects the response to bevacizumab therapy and gene pathways. In particular, long-term bevacizumab treatment shifts the cancer cells to a more aggressive myoepithelial/basal subtype in VEGF-expressing model, but not in non-VEGF model. These findings could shed light on variable results to anti-VEGF therapy in patients and emphasize the importance of patient stratification based on the VEGF expression. Our data strongly suggest consideration of patient subgroups for treatment and designing novel combinatory therapies in the clinical setting.
Collapse
Affiliation(s)
- Yesim Gökmen-Polar
- 1. Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Chirayu P Goswami
- 2. Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
| | - Rachel A Toroni
- 3. Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Kerry L Sanders
- 3. Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Rutika Mehta
- 1. Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Usha Sirimalle
- 1. Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Bogdan Tanasa
- 4. Scripps Research Institute, University of Medicine and Pharmac, La Jolla, CA
| | - Changyu Shen
- 2. Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
| | - Lang Li
- 2. Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN
| | - Mircea Ivan
- 3. Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Sunil Badve
- 1. Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN; ; 3. Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - George W Sledge
- 1. Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN; ; 3. Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
40
|
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: 3.4] [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
|
41
|
Horwitz E, Stein I, Andreozzi M, Nemeth J, Shoham A, Pappo O, Schweitzer N, Tornillo L, Kanarek N, Quagliata L, Zreik F, Porat RM, Finkelstein R, Reuter H, Koschny R, Ganten T, Mogler C, Shibolet O, Hess J, Breuhahn K, Grunewald M, Schirmacher P, Vogel A, Terracciano L, Angel P, Ben-Neriah Y, Pikarsky E. Human and mouse VEGFA-amplified hepatocellular carcinomas are highly sensitive to sorafenib treatment. Cancer Discov 2014; 4:730-43. [PMID: 24687604 DOI: 10.1158/2159-8290.cd-13-0782] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Death rates from hepatocellular carcinoma (HCC) are steadily increasing, yet therapeutic options for advanced HCC are limited. We identify a subset of mouse and human HCCs harboring VEGFA genomic amplification, displaying distinct biologic characteristics. Unlike common tumor amplifications, this one seems to work via heterotypic paracrine interactions; stromal VEGF receptors (VEGFR), responding to tumor VEGF-A, produce hepatocyte growth factor (HGF) that reciprocally affects tumor cells. VEGF-A inhibition results in HGF downregulation and reduced proliferation, specifically in amplicon-positive mouse HCCs. Sorafenib-the first-line drug in advanced HCC-targets multiple kinases, including VEGFRs, but has only an overall mild beneficial effect. We found that VEGFA amplification specifies mouse and human HCCs that are distinctly sensitive to sorafenib. FISH analysis of a retrospective patient cohort showed markedly improved survival of sorafenib-treated patients with VEGFA-amplified HCCs, suggesting that VEGFA amplification is a potential biomarker for HCC response to VEGF-A-blocking drugs. SIGNIFICANCE Using a mouse model of inflammation-driven cancer, we identified a subclass of HCC carrying VEGFA amplification, which is particularly sensitive to VEGF-A inhibition. We found that a similar amplification in human HCC identifies patients who favorably responded to sorafenib-the first-line treatment of advanced HCC-which has an overall moderate therapeutic efficacy.
Collapse
Affiliation(s)
- Elad Horwitz
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Ilan Stein
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, GermanyAuthors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Mariacarla Andreozzi
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Julia Nemeth
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Avivit Shoham
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Orit Pappo
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Nora Schweitzer
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Luigi Tornillo
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Naama Kanarek
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Luca Quagliata
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Farid Zreik
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Rinnat M Porat
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Rutie Finkelstein
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Hendrik Reuter
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Ronald Koschny
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Tom Ganten
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Carolin Mogler
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Oren Shibolet
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Jochen Hess
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, GermanyAuthors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, GermanyAuthors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University
| | - Kai Breuhahn
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Myriam Grunewald
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Peter Schirmacher
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Arndt Vogel
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Luigi Terracciano
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Peter Angel
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Yinon Ben-Neriah
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Eli Pikarsky
- Authors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, GermanyAuthors' Affiliations:The Lautenberg Center for Immunology; Department of Developmental Biology and Cancer Research, IMRIC, Hadassah Medical School, Hebrew University;Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem; Liver Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Institute of Pathology, University Hospital Basel, Basel, Switzerland; Division of Signal Transduction and Growth Control (A100), Division of Molecular Genetics (B060), and Junior Group Molecular Mechanisms of Head and Neck Tumors (A102), German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance; Institute of Pathology, University Hospital Heidelberg; Departments of Otolaryngology, Head and Neck Surgery and Internal Medicine, University Hospital Heidelberg, Heidelberg; and Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
42
|
Alcazar-González GA, Calderón-Garcidueñas AL, Garza-Rodríguez ML, Rubio-Hernández G, Escorza-Treviño S, Olano-Martin E, Cerda-Flores RM, Castruita-Avila AL, González-Guerrero JF, le Brun S, Simon-Buela L, Barrera-Saldaña HA. Comparative study of polymorphism frequencies of the CYP2D6, CYP3A5, CYP2C8 and IL-10 genes in Mexican and Spanish women with breast cancer. Pharmacogenomics 2014; 14:1583-92. [PMID: 24088129 DOI: 10.2217/pgs.13.83] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
AIM Pharmacogenetic studies in breast cancer (BC) may predict the efficacy of tamoxifen and the toxicity of paclitaxel and capecitabine. We determined the frequency of polymorphisms in the CYP2D6 gene associated with activation of tamoxifen, and those of the genes CYP2C8, CYP3A5 and DPYD associated with toxicity of paclitaxel and capecitabine. We also included a IL-10 gene polymorphism associated with advanced tumor stage at diagnosis. PATIENTS & METHODS Genomic DNAs from 241 BC patients from northeast Mexico were genotyped using DNA microarray technology. RESULTS For tamoxifen processing, CYP2D6 genotyping predicted that 90.8% of patients were normal metabolizers, 4.2% ultrarapid, 2.1% intermediate and 2.9% poor metabolizers. For paclitaxel and the CYP2C8 gene, 75.3% were normal, 23.4% intermediate and 1.3% poor metabolizers. Regarding the DPYD gene, only one patient was a poor metabolizer. For the IL-10 gene, 47.1% were poor metabolizers. CONCLUSION These results contribute valuable information towards personalizing BC chemotherapy in Mexican women.
Collapse
|
43
|
Fakhrejahani E, Toi M. Antiangiogenesis therapy for breast cancer: an update and perspectives from clinical trials. Jpn J Clin Oncol 2014; 44:197-207. [PMID: 24474817 PMCID: PMC3941646 DOI: 10.1093/jjco/hyt201] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The development of new blood vessels is a crucial step in breast cancer growth, progression and dissemination, making it a promising therapeutic target. Breast cancer has a heterogeneous nature and the diversity of responsible angiogenic pathways between different tumors has been studied for many years. Inhibiting different targets in these pathways has been under investigation in preclinical and clinical studies for more than decades, among which antibody against vascular endothelial growth factor is the most studied. However, the clinical impact from antiangiogenic treatment alone or in combination with standard chemotherapeutic regimens has been relatively small till today. In this review, we summarize the most clinically relevant data from breast cancer treatment clinical trials and discuss safety and efficacy of common antiangiogenic therapies as well as biological predictive markers.
Collapse
Affiliation(s)
- Elham Fakhrejahani
- *Department of Breast Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin-Kawaharacho, Sakyo-ku, Kyoto 606-8507, Japan.
| | | |
Collapse
|
44
|
Gerber B, Loibl S, Eidtmann H, Rezai M, Fasching PA, Tesch H, Eggemann H, Schrader I, Kittel K, Hanusch C, Kreienberg R, Solbach C, Jackisch C, Kunz G, Blohmer JU, Huober J, Hauschild M, Nekljudova V, Untch M, von Minckwitz G. Neoadjuvant bevacizumab and anthracycline-taxane-based chemotherapy in 678 triple-negative primary breast cancers; results from the geparquinto study (GBG 44). Ann Oncol 2013; 24:2978-84. [PMID: 24136883 DOI: 10.1093/annonc/mdt361] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND We evaluated the pathological complete response (pCR) rate after neoadjuvant epirubicin, (E) cyclophosphamide (C) and docetaxel containing chemotherapy with and without the addition of bevacizumab in patients with triple-negative breast cancer (TNBC). PATIENTS AND METHODS Patients with untreated cT1c-4d TNBC represented a stratified subset of the 1948 participants of the HER2-negative part of the GeparQuinto trial. Patients were randomized to receive four cycles EC (90/600 mg/m(2); q3w) followed by four cycles docetaxel (100 mg/m(2); q3w) each with or without bevacizumab (15 mg/kg; q3w) added to chemotherapy. RESULTS TNBC patients were randomized to chemotherapy without (n = 340) or with bevacizumab (n = 323). pCR (ypT0 ypN0, primary end point) rates were 27.9% without and 39.3% with bevacizumab (P = 0.003). According to other pCR definitions, the addition of bevacizumab increased the pCR rate from 30.9% to 41.8% (ypT0 ypN0/+; P = 0.004), 36.2% to 46.4% (ypT0/is ypN0/+; P = 0.009) and 32.9% to 43.3% (ypT0/is ypN0; P = 0.007). Bevacizumab treatment [OR 1.73, 95% confidence interval (CI) 1.23-2.42; P = 0.002], lower tumor stage (OR 2.38, 95% CI 1.24-4.54; P = 0.009) and grade 3 tumors (OR 1.68, 95% CI 1.14-2.48; P = 0.009) were confirmed as independent predictors of higher pCR in multivariate logistic regression analysis. CONCLUSIONS The addition of bevacizumab to chemotherapy in TNBC significantly increases pCR rates.
Collapse
Affiliation(s)
- B Gerber
- Department of Obstetrics and Gynaecology, University of Rostock, Rostock
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
VEGFA gene locus analysis across 80 human tumour types reveals gene amplification in several neoplastic entities. Angiogenesis 2013; 17:519-27. [DOI: 10.1007/s10456-013-9396-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 10/01/2013] [Indexed: 01/20/2023]
|
46
|
Koutras AK, Kotoula V, Papadimitriou C, Dionysopoulos D, Zagouri F, Kalofonos HP, Kourea HP, Skarlos DV, Samantas E, Papadopoulou K, Kosmidis P, Pectasides D, Fountzilas G. Vascular endothelial growth factor polymorphisms and clinical outcome in patients with metastatic breast cancer treated with weekly docetaxel. THE PHARMACOGENOMICS JOURNAL 2013; 14:248-55. [PMID: 24061601 DOI: 10.1038/tpj.2013.36] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 08/03/2013] [Accepted: 08/15/2013] [Indexed: 11/09/2022]
Abstract
The aim of the study was to evaluate the association of vascular endothelial growth factor (VEGF) genotypes with treatment efficacy in a phase II trial. This study evaluated weekly docetaxel, as first-line treatment for metastatic breast cancer. Existing data from in vitro and animal model experiments suggest that docetaxel at low doses has anti-angiogenic activity. DNA was extracted from blood samples of 86 patients participating in the trial. Genotyping was performed for selected single-nucleotide polymorphisms (SNPs; VEGF-2578, -1498, -1154, and +936). Moreover, due to the highly polymorphic nature of the studied areas, we were able to analyze additional registered SNPs. All candidate genotypes were evaluated for associations with overall survival (OS), progression-free survival (PFS) and response rate. The VEGF-1154 GG genotype was more frequent in patients not responding to treatment compared with responders (42.9% vs 0.0%, P=0.048). Moreover, the VEGF-2578 AA genotype was associated with longer PFS compared with CC (hazard ratio (HR)=0.40; 95% confidence interval (CI) 0.17-0.98; pairwise P=0.0457). Patients with the VEGF-1190 GG genotype demonstrated shorter PFS compared with those with the alternative genotypes (GA and AA) combined (HR=3.85; 95% CI: 1.20-12.50; P=0.0224). In addition, the VEGF-2551/-2534 homozygous del18bp and VEGF-2430/-2425 homozygous ins1bp genotypes were associated with worse PFS compared with no deletion and no insertion, respectively (HR=2.49; 95% CI: 1.02-6.07; pairwise P=0.0442 and HR=2.57; 95% CI: 1.05-6.27; pairwise P=0.0385, respectively). Furthermore, patients with the VEGF-1498 CC genotype exhibited longer median OS compared with those with the alternatives genotypes (CT and TT) combined (HR=0.27; 95% CI: 0.08-0.89; P=0.0311). In multivariate analysis, the VEGF-2578 AA genotype retained its significance (P=0.0220) for PFS. Our results support the association of specific VEGF genotypes with clinical outcome in patients with metastatic breast cancer treated with a potentially anti-angiogenic regimen, such as weekly docetaxel. However, current results should be validated prospectively in larger cohorts.
Collapse
Affiliation(s)
- A K Koutras
- Division of Oncology, Department of Medicine, University Hospital, University of Patras Medical School, Patras, Greece
| | - V Kotoula
- Department of Pathology, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - C Papadimitriou
- Department of Clinical Therapeutics, 'Alexandra' Hospital, University of Athens School of Medicine, Athens, Greece
| | - D Dionysopoulos
- Department of Medical Oncology, 'Papageorgiou' Hospital, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - F Zagouri
- Department of Clinical Therapeutics, 'Alexandra' Hospital, University of Athens School of Medicine, Athens, Greece
| | - H P Kalofonos
- Division of Oncology, Department of Medicine, University Hospital, University of Patras Medical School, Patras, Greece
| | - H P Kourea
- Department of Pathology, University Hospital of Patras, Patras, Greece
| | - D V Skarlos
- Second Department of Medical Oncology, 'Metropolitan' Hospital, Piraeus, Greece
| | - E Samantas
- Third Department of Medical Oncology, 'Agii Anargiri' Cancer Hospital, Athens, Greece
| | - K Papadopoulou
- Department of Pathology, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| | - P Kosmidis
- Second Department of Medical Oncology, Hygeia Hospital, Athens, Greece
| | - D Pectasides
- Oncology Section, Second Department of Internal Medicine, 'Hippokration' Hospital, Athens, Greece
| | - G Fountzilas
- Department of Medical Oncology, 'Papageorgiou' Hospital, Aristotle University of Thessaloniki School of Medicine, Thessaloniki, Greece
| |
Collapse
|