1
|
Bos MK, Lam SW, Motta G, Helmijr JCA, Beaufort CM, de Jonge E, Martens JWM, Boven E, Jansen MPHM, Jager A, Sleijfer S. Plasma ESR1 mutations and outcome to first-line paclitaxel and bevacizumab in patients with advanced ER-positive/HER2-negative breast cancer. Breast Cancer Res Treat 2023:10.1007/s10549-023-06965-5. [PMID: 37226020 DOI: 10.1007/s10549-023-06965-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/02/2023] [Indexed: 05/26/2023]
Abstract
BACKGROUND ESR1 mutations have been identified as mechanism for endocrine resistance and are also associated with a decreased overall survival. We assessed ESR1 mutations in circulating tumor DNA (ctDNA) for impact on outcome to taxane-based chemotherapy in advanced breast cancer patients. METHODS ESR1 mutations were determined in archived plasma samples from patients treated with paclitaxel and bevacizumab (AT arm, N = 91) in the randomized phase II ATX study. Samples collected at baseline (n = 51) and at cycle 2 (n = 13, C2) were analyzed using a breast cancer next-generation sequencing panel. This study was powered to detect a benefit in progression-free survival (PFS) at six months for patients treated with paclitaxel/bevacizumab compared to historical trials with fulvestrant. PFS, overall survival (OS), and ctDNA dynamics were exploratory analyses. RESULTS PFS at six months was 86% (18/21) in patients with an ESR1 mutation detected and 85% (23/27) in wildtype ESR1 patients. In our exploratory analysis, median progression-free survival (PFS) was 8.2 months [95% CI, 7.6-8.8] for ESR1 mutant patients versus 8.7 months [95% confidence interval (CI), 8.3-9.2] for ESR1 wildtype patients [p = 0.47]. The median overall survival (OS) was 20.7 months [95% CI, 6.6-33.7] for ESR1 mutant patients versus 28.1 months [95% confidence interval (CI), 19.3-36.9] for ESR1 wildtype patients [p = 0.27]. Patients with ≥ two ESR1 mutations had a significantly worse OS, but not PFS, compared to those who did not [p = 0.003]. Change in ctDNA level at C2 was not different between ESR1 and other mutations. CONCLUSIONS Presence of ESR1 mutations in baseline ctDNA might not be associated with inferior PFS and OS in advanced breast cancer patients treated with paclitaxel/bevacizumab.
Collapse
Affiliation(s)
- M K Bos
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015, GD, Rotterdam, The Netherlands.
| | - S W Lam
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam/Cancer Center Amsterdam, De Boelelaan 1117, 1081 , HV, Amsterdam, The Netherlands
- Department of Radiology, The Netherlands, Cancer Institute/Antoni Van Leeuwenhoek Hospital, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands
| | - G Motta
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015, GD, Rotterdam, The Netherlands
- IOM (Mediterranean Institute of Oncology) Research, Viagrande, Catania, Italy
- Department of Clinical and Experimental Medicine, A.O.U. Policlinico-Vittorio Emanuele, Center of Experimental Oncology and Hematology, University of Catania, Catania, Italy
| | - J C A Helmijr
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015, GD, Rotterdam, The Netherlands
| | - C M Beaufort
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015, GD, Rotterdam, The Netherlands
| | - E de Jonge
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015, GD, Rotterdam, The Netherlands
| | - E Boven
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam/Cancer Center Amsterdam, De Boelelaan 1117, 1081 , HV, Amsterdam, The Netherlands
| | - M P H M Jansen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015, GD, Rotterdam, The Netherlands
| | - A Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015, GD, Rotterdam, The Netherlands
| | - S Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015, GD, Rotterdam, The Netherlands
| |
Collapse
|
2
|
Abeyakoon O, Woitek R, Wallis M, Moyle P, Morscher S, Dahlhaus N, Ford S, Burton N, Manavaki R, Mendichovszky I, Joseph J, Quiros-Gonzalez I, Bohndiek S, Gilbert F. An optoacoustic imaging feature set to characterise blood vessels surrounding benign and malignant breast lesions. PHOTOACOUSTICS 2022; 27:100383. [PMID: 36068806 PMCID: PMC9441264 DOI: 10.1016/j.pacs.2022.100383] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/21/2022] [Accepted: 07/01/2022] [Indexed: 06/15/2023]
Abstract
Combining optoacoustic (OA) imaging with ultrasound (US) enables visualisation of functional blood vasculature in breast lesions by OA to be overlaid with the morphological information of US. Here, we develop a simple OA feature set to differentiate benign and malignant breast lesions. 94 female patients with benign, indeterminate or suspicious lesions were recruited and underwent OA-US. An OA-US imaging feature set was developed using images from the first 38 patients, which contained 14 malignant and 8 benign solid lesions. Two independent radiologists blindly scored the OA-US images of a further 56 patients, which included 31 malignant and 13 benign solid lesions, with a sensitivity of 96.8% and specificity of 84.6%. Our findings indicate that OA-US can reveal vascular patterns of breast lesions that indicate malignancy using a simple feature set based on single wavelength OA data, which is therefore amenable to application in low resource settings for breast cancer management.
Collapse
Affiliation(s)
- O. Abeyakoon
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - R. Woitek
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - M.G. Wallis
- Cambridge Breast Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - P.L. Moyle
- Cambridge Breast Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - S. Morscher
- iThera Medical GmbH, Zielstattstrasse 13, Munich 81379, Germany
| | - N. Dahlhaus
- iThera Medical GmbH, Zielstattstrasse 13, Munich 81379, Germany
| | - S.J. Ford
- iThera Medical GmbH, Zielstattstrasse 13, Munich 81379, Germany
| | - N.C. Burton
- iThera Medical GmbH, Zielstattstrasse 13, Munich 81379, Germany
| | - R. Manavaki
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - I.A. Mendichovszky
- Department of Nuclear Medicine, Cambridge University Hospitals Foundation Trust, Cambridge CB2 0QQ, UK
| | - J. Joseph
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| | - I. Quiros-Gonzalez
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| | - S.E. Bohndiek
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge CB2 0RE, UK
| | - F.J. Gilbert
- Department of Radiology, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| |
Collapse
|
3
|
Circulating proteins as predictive and prognostic biomarkers in breast cancer. Clin Proteomics 2022; 19:25. [PMID: 35818030 PMCID: PMC9275040 DOI: 10.1186/s12014-022-09362-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/28/2022] [Indexed: 11/22/2022] Open
Abstract
Breast cancer (BC) is the most common cancer and among the leading causes of cancer death in women. It is a heterogeneous group of tumours with numerous morphological and molecular subtypes, making predictions of disease evolution and patient outcomes difficult. Therefore, biomarkers are needed to help clinicians choose the best treatment for each patient. For the last years, studies have increasingly focused on biomarkers obtainable by liquid biopsy. Circulating proteins (from serum or plasma) can be used for inexpensive and minimally invasive determination of disease risk, early diagnosis, treatment adjusting, prognostication and disease progression monitoring. We provide here a review of the main published studies on serum proteins in breast cancer and elaborate on the potential of circulating proteins to be predictive and/or prognostic biomarkers in breast cancer.
Collapse
|
4
|
Zhang M, Liu J, Liu G, Xing Z, Jia Z, Li J, Wang W, Wang J, Qin L, Wang X, Wang X. Anti-vascular endothelial growth factor therapy in breast cancer: Molecular pathway, potential targets, and current treatment strategies. Cancer Lett 2021; 520:422-433. [PMID: 34389434 DOI: 10.1016/j.canlet.2021.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 11/15/2022]
Abstract
As the highest incidence of female malignancy, breast cancer is likewise the leading cause of cancer-related deaths. The development of cancer relies on neo-vascularization, which provides sufficient nutrition and oxygen, and supplies a pathway for distant metastasis. Angiogenesis represents the formation of new blood vessels, and is a principal pathogenetic action in breast cancer. Vascular endothelial growth factor (VEGF) is a major angiogenesis regulator that modulates the maintenance and function of mature vascular networks. Therefore, the VEGF pathway is a promising oncotherapeutic target. This review elaborates an update on the prognostic value of VEGF in breast cancer, summarizes clinical experience and lessons of anti-VEGF therapeutics, meanwhile, provides an overview of biomarkers that predict the effectiveness of anti-angiogenic treatment.
Collapse
Affiliation(s)
- Menglu Zhang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiaqi Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Gang Liu
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zeyu Xing
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ziqi Jia
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiaxin Li
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenyan Wang
- Department of Breast Surgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Jie Wang
- Department of Ultrasound, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ling Qin
- Department of Breast Surgical Oncology, Cancer Hospital of HuanXing, Beijing, 100021, China
| | - Xin Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Xiang Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
5
|
Hoon SN, Lau PK, White AM, Bulsara MK, Banks PD, Redfern AD. Capecitabine for hormone receptor-positive versus hormone receptor-negative breast cancer. Cochrane Database Syst Rev 2021; 5:CD011220. [PMID: 34037241 PMCID: PMC8150746 DOI: 10.1002/14651858.cd011220.pub2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Retrospective analyses suggest that capecitabine may carry superior activity in hormone receptor-positive relative to hormone receptor-negative metastatic breast cancer. This review examined the veracity of that finding and explored whether this differential activity extends to early breast cancer. OBJECTIVES To assess effects of chemotherapy regimens containing capecitabine compared with regimens not containing capecitabine for women with hormone receptor-positive versus hormone receptor-negative breast cancer across the three major treatment scenarios: neoadjuvant, adjuvant, metastatic. SEARCH METHODS On 4 June 2019, we searched the Cochrane Breast Cancer Specialised Register; the Cochrane Central Register of Controlled Trials (CENTRAL; 2019, Issue 5) in the Cochrane Library; MEDLINE; Embase; the World Health Organization International Clinical Trials Registry Platform; and ClinicalTrials.gov. SELECTION CRITERIA Randomised controlled trials looking at chemotherapy regimens containing capecitabine alone or in combination with other agents versus a control or similar regimen without capecitabine for treatment of breast cancer at any stage. The primary outcome measure for metastatic and adjuvant trials was overall survival (OS), and for neoadjuvant studies pathological complete response (pCR). DATA COLLECTION AND ANALYSIS Two review authors independently extracted data and assessed risk of bias and certainty of evidence using the GRADE approach. Hazard ratios (HRs) were derived for time-to-event outcomes, and odds ratios (ORs) for dichotomous outcomes, and meta-analysis was performed using a fixed-effect model. MAIN RESULTS We included 26 studies with outcome data by hormone receptor: 12 metastatic studies (n = 4325), 6 neoadjuvant trials (n = 3152), and 8 adjuvant studies (n = 13,457). Capecitabine treatment was added in several different ways across studies. These could be classified as capecitabine alone compared to another treatment, capecitabine substituted for part of the control chemotherapy, and capecitabine added to control chemotherapy. In the metastatic setting, the effect of capecitabine was heterogenous between hormone receptor-positive and -negative tumours. For OS, no difference between capecitabine-containing and non-capecitabine-containing regimens was observed for all participants taken together (HR 1.01, 95% confidence interval (CI) 0.98 to 1.05; 12 studies, 4325 participants; high-certainty evidence), for those with hormone receptor-positive disease (HR 0.93, 95% CI 0.84 to 1.04; 7 studies, 1834 participants; high-certainty evidence), and for those with hormone receptor-negative disease (HR 1.00, 95% CI 0.88 to 1.13; 8 studies, 1577 participants; high-certainty evidence). For progression-free survival (PFS), a small improvement was seen for all people (HR 0.89, 95% CI 0.82 to 0.96; 12 studies, 4325 participants; moderate-certainty evidence). This was largely accounted for by a moderate improvement in PFS for inclusion of capecitabine in hormone receptor-positive cancers (HR 0.82, 95% CI 0.73 to 0.91; 7 studies, 1594 participants; moderate-certainty evidence) compared to no difference in PFS for hormone receptor-negative cancers (HR 0.96, 95% CI 0.83 to 1.10; 7 studies, 1122 participants; moderate-certainty evidence). Quality of life was assessed in five studies; in general there did not seem to be differences in global health scores between the two treatment groups at around two years' follow-up. Neoadjuvant studies were highly variable in design, having been undertaken to test various experimental regimens using pathological complete response (pCR) as a surrogate for disease-free survival (DFS) and OS. Across all patients, capecitabine-containing regimens resulted in little difference in pCR in comparison to non-capecitabine-containing regimens (odds ratio (OR) 1.12, 95% CI 0.94 to 1.33; 6 studies, 3152 participants; high-certainty evidence). By subtype, no difference in pCR was observed for either hormone receptor-positive (OR 1.22, 95% CI 0.76 to 1.95; 4 studies, 964 participants; moderate-certainty evidence) or hormone receptor-negative tumours (OR 1.28, 95% CI 0.61 to 2.66; 4 studies, 646 participants; moderate-certainty evidence). Four studies with 2460 people reported longer-term outcomes: these investigators detected no difference in either DFS (HR 1.02, 95% CI 0.86 to 1.21; high-certainty evidence) or OS (HR 0.97, 95% CI 0.77 to 1.23; high-certainty evidence). In the adjuvant setting, a modest improvement in OS was observed across all participants (HR 0.89, 95% CI 0.81 to 0.98; 8 studies, 13,547 participants; moderate-certainty evidence), and no difference in OS was seen in hormone receptor-positive cancers (HR 0.86, 95% CI 0.68 to 1.09; 3 studies, 3683 participants), whereas OS improved in hormone receptor-negative cancers (HR 0.72, 95% CI 0.59 to 0.89; 5 studies, 3432 participants). No difference in DFS or relapse-free survival (RFS) was observed across all participants (HR 0.93, 95% CI 0.86 to 1.01; 8 studies, 13,457 participants; moderate-certainty evidence). As was observed for OS, no difference in DFS/RFS was seen in hormone receptor-positive cancers (HR 1.03, 95% CI 0.91 to 1.17; 5 studies, 5604 participants; moderate-certainty evidence), and improvements in DFS/RFS with inclusion of capecitabine were observed for hormone receptor-negative cancers (HR 0.74, 95% CI 0.64 to 0.86; 7 studies, 3307 participants; moderate-certainty evidence). Adverse effects were reported across all three scenarios. When grade 3 or 4 febrile neutropenia was considered, no difference was seen for capecitabine compared to non-capecitabine regimens in neoadjuvant studies (OR 1.31, 95% CI 0.97 to 1.77; 4 studies, 2890 participants; moderate-certainty evidence), and a marked reduction was seen for capecitabine in adjuvant studies (OR 0.55, 95% CI 0.47 to 0.64; 5 studies, 8086 participants; moderate-certainty evidence). There was an increase in diarrhoea and hand-foot syndrome in neoadjuvant (diarrhoea: OR 1.95, 95% CI 1.32 to 2.89; 3 studies, 2686 participants; hand-foot syndrome: OR 6.77, 95% CI 4.89 to 9.38; 5 studies, 3021 participants; both moderate-certainty evidence) and adjuvant trials (diarrhoea: OR 2.46, 95% CI 2.01 to 3.01; hand-foot syndrome: OR 13.60, 95% CI 10.65 to 17.37; 8 studies, 11,207 participants; moderate-certainty evidence for both outcomes). AUTHORS' CONCLUSIONS In summary, a moderate PFS benefit by including capecitabine was seen only in hormone receptor-positive cancers in metastatic studies. No benefit of capecitabine for pCR was noted overall or in hormone receptor subgroups when included in neoadjuvant therapy. In contrast, the addition of capecitabine in the adjuvant setting led to improved outcomes for OS and DFS in hormone receptor-negative cancer. Future studies should stratify by hormone receptor and triple-negative breast cancer (TNBC) status to clarify the differential effects of capecitabine in these subgroups across all treatment scenarios, to optimally guide capecitabine inclusion.
Collapse
Affiliation(s)
- Siao-Nge Hoon
- Medical Oncology Department, St John of God Midland, Perth, Australia
- Medical Oncology Department, Sir Charles Gairdner Hospital, Perth, Australia
| | - Peter Kh Lau
- Medical Oncology Department, Sir Charles Gairdner Hospital, Perth, Australia
- Medical Oncology Department, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Alison M White
- Murdoch Community Hospice, St John of God Hospital Murdoch, Perth, Australia
- Palliative Care Department, Royal Perth Hospital, Perth, Australia
| | - Max K Bulsara
- Institute for Health Research, University of Notre Dame Australia, Fremantle, Australia
- School of Population and Global Health, The University of Western Australia, Perth, Australia
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Patricia D Banks
- Medical Oncology Department, Peter MacCallum Cancer Centre, Melbourne, Australia
- Medical Oncology Department, University Hospital Geelong, Geelong, Australia
| | - Andrew D Redfern
- School of Medicine, University of Western Australia, Perth, Australia
- Medical Oncology Department, Fiona Stanley Hospital, Perth, Australia
| |
Collapse
|
6
|
Zhang J, Gao S, Zheng Q, Kang Y, Li J, Zhang S, Shang C, Tan X, Ren W, Ma Y. A Novel Model Incorporating Tumor Stiffness, Blood Flow Characteristics, and Ki-67 Expression to Predict Responses After Neoadjuvant Chemotherapy in Breast Cancer. Front Oncol 2020; 10:603574. [PMID: 33364197 PMCID: PMC7753215 DOI: 10.3389/fonc.2020.603574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/09/2020] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE To investigate the ability of tumor stiffness, tumor blood flow, and Ki-67 expression alone or in combination in predicting the pathological response to neoadjuvant chemotherapy (NACT) in breast cancer. PATIENTS AND METHODS This prospective cohort study included 145 breast cancer patients treated with NACT. Tumor stiffness (maximum stiffness (Emax), mean stiffness (Emean)), blood score (BS), and their relative changes, were evaluated before (t0), during (t1-t5), and at the end of NACT (t6) by shear-wave elastography and optical imaging. Ki-67 expression was quantitatively evaluated by immunohistochemistry using core biopsy specimens obtained before NACT. Pathological responses were evaluated by residual cancer burden. The ability of tumor stiffness, BS, Ki-67, and predRCB-which combined ΔEmean (t2) (the relative changes in Emean after the second NACT cycle), BS2 (BS after the second NACT cycle), and Ki-67-in predicting tumor responses was compared using receiver operating characteristic curves and the Z-test. RESULTS Tumor stiffness and BS decreased during NACT. ΔEmean (t2), BS2, and Ki-67 had better predictive performance than other indexes in identifying a favorable response (AUC = 0.82, 0.81, and 0.80) and resistance responses (AUC = 0.85, 0.79, and 0.84), with no significant differences between the three (p > 0.05). PredRCB had better predictive performance than any parameter alone for a favorable response (AUC = 0.90) and resistance (AUC = 0.93). CONCLUSION Tumor stiffness, BS, and Ki-67 expression showed good and similar abilities for predicting the pathological response to NACT, and predRCB was a significantly better predictor than each index alone. These results may help design therapeutic strategies for breast cancer patients undergoing NACT.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China
| | - Song Gao
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qiaojin Zheng
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ye Kang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianyi Li
- Department of Breast Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuo Zhang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cong Shang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xueying Tan
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weidong Ren
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yan Ma
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
7
|
Aggarwal V, Miranda O, Johnston PA, Sant S. Three dimensional engineered models to study hypoxia biology in breast cancer. Cancer Lett 2020; 490:124-142. [PMID: 32569616 PMCID: PMC7442747 DOI: 10.1016/j.canlet.2020.05.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/13/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022]
Abstract
Breast cancer is the second leading cause of mortality among women worldwide. Despite the available therapeutic regimes, variable treatment response is reported among different breast cancer subtypes. Recently, the effects of the tumor microenvironment on tumor progression as well as treatment responses have been widely recognized. Hypoxia and hypoxia inducible factors in the tumor microenvironment have long been known as major players in tumor progression and survival. However, the majority of our understanding of hypoxia biology has been derived from two dimensional (2D) models. Although many hypoxia-targeted therapies have elicited promising results in vitro and in vivo, these results have not been successfully translated into clinical trials. These limitations of 2D models underscore the need to develop and integrate three dimensional (3D) models that recapitulate the complex tumor-stroma interactions in vivo. This review summarizes role of hypoxia in various hallmarks of cancer progression. We then compare traditional 2D experimental systems with novel 3D tissue-engineered models giving accounts of different bioengineering platforms available to develop 3D models and how these 3D models are being exploited to understand the role of hypoxia in breast cancer progression.
Collapse
Affiliation(s)
- Vaishali Aggarwal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Oshin Miranda
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Paul A Johnston
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; UPMC-Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
8
|
Chuma M, Uojima H, Numata K, Hidaka H, Toyoda H, Hiraoka A, Tada T, Hirose S, Atsukawa M, Itokawa N, Arai T, Kako M, Nakazawa T, Wada N, Iwasaki S, Miura Y, Hishiki S, Nishigori S, Morimoto M, Hattori N, Ogushi K, Nozaki A, Fukuda H, Kagawa T, Michitaka K, Kumada T, Maeda S. Early Changes in Circulating FGF19 and Ang-2 Levels as Possible Predictive Biomarkers of Clinical Response to Lenvatinib Therapy in Hepatocellular Carcinoma. Cancers (Basel) 2020; 12:293. [PMID: 31991869 PMCID: PMC7073176 DOI: 10.3390/cancers12020293] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 01/17/2020] [Accepted: 01/23/2020] [Indexed: 12/14/2022] Open
Abstract
Predictive biomarkers of the response of hepatocellular carcinoma (HCC) to Lenvatinib therapy have not yet been clarified. The aim of this study was to identify clinically significant biomarkers of response to Lenvatinib therapy, to target strategies against HCC. Levels of circulating angiogenic factors (CAFs) were analyzed in blood samples collected at baseline and after introducing lenvatinib, from 74 Child-Pugh class A HCC patients who received lenvatinib. As CAF biomarkers, serum vascular endothelial growth factor (VEGF), fibroblast growth factor 19 (FGF19), FGF23, and angiopoietin-2 (Ang-2) were measured using enzyme-linked immunosorbent assays. Results: Significantly increased FGF19 (FGF19-i) levels and decreased Ang-2 (Ang-2-d) levels were seen in Lenvatinib responders as compared to non-responders (ratio of FGF19 level at 4 weeks/baseline in responders vs. non-responders: 2.09 vs. 1.32, respectively, p = 0.0004; ratio of Ang-2 level at four weeks/baseline: 0.584 vs. 0.810, respectively, p = 0.0002). Changes in FGF23 and VEGF levels at four weeks versus baseline, however, were not significantly different in responders versus non-responders. In multivariate analysis, the combination of serum FGF19-i and Ang-2-d was the most independent predictive factor for Lenvatinib response (Odds ratio, 9.143; p = 0.0012). Furthermore, this combination biomarker showed the greatest independent association with progression-free survival (Hazard ratio, 0.171; p = 0.0240). Early changes in circulating FGF19 and Ang-2 levels might be useful for predicting clinical response and progression-free survival in HCC patients on Lenvatinib therapy.
Collapse
Affiliation(s)
- Makoto Chuma
- Gastroenterological Center, Yokohama City University Medical Center, Yokohama 232-0024, Japan; (K.N.); (K.O.); (A.N.); (H.F.)
| | - Haruki Uojima
- Department of Gastroenterology, Internal Medicine, Kitasato University School of Medicine, Sagamihara 252-0375, Japan; (H.U.); (H.H.); (T.N.); (N.W.); (S.I.)
| | - Kazushi Numata
- Gastroenterological Center, Yokohama City University Medical Center, Yokohama 232-0024, Japan; (K.N.); (K.O.); (A.N.); (H.F.)
| | - Hisashi Hidaka
- Department of Gastroenterology, Internal Medicine, Kitasato University School of Medicine, Sagamihara 252-0375, Japan; (H.U.); (H.H.); (T.N.); (N.W.); (S.I.)
| | - Hidenori Toyoda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki 503-8502, Japan; (H.T.); (T.T.); (T.K.)
| | - Atsushi Hiraoka
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama 790-0024, Japan; (A.H.); (K.M.)
| | - Toshifumi Tada
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki 503-8502, Japan; (H.T.); (T.T.); (T.K.)
| | - Shunji Hirose
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, Isehara 259-1193, Japan; (S.H.); (T.K.)
| | - Masanori Atsukawa
- Division of Gastroenterology and Hepatology, Nippon Medical School, Tokyo 113-8603, Japan; (M.A.); (T.A.)
| | - Norio Itokawa
- Division of Gastroenterology, Nippon Medical School Chiba Hokusoh Hospital, Inzai 270-1694, Japan;
| | - Taeang Arai
- Division of Gastroenterology and Hepatology, Nippon Medical School, Tokyo 113-8603, Japan; (M.A.); (T.A.)
- Division of Gastroenterology, Nippon Medical School Musashi Kosugi Hospital, Kawasaki 211-8533, Japan
| | - Makoto Kako
- Department of Gastroenterology, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan;
| | - Takahide Nakazawa
- Department of Gastroenterology, Internal Medicine, Kitasato University School of Medicine, Sagamihara 252-0375, Japan; (H.U.); (H.H.); (T.N.); (N.W.); (S.I.)
| | - Naohisa Wada
- Department of Gastroenterology, Internal Medicine, Kitasato University School of Medicine, Sagamihara 252-0375, Japan; (H.U.); (H.H.); (T.N.); (N.W.); (S.I.)
| | - Shuitirou Iwasaki
- Department of Gastroenterology, Internal Medicine, Kitasato University School of Medicine, Sagamihara 252-0375, Japan; (H.U.); (H.H.); (T.N.); (N.W.); (S.I.)
| | - Yuki Miura
- Gastroenterology Division, Hadano Red Cross Hospital, Hadano 257-0017, Japan;
| | - Satoshi Hishiki
- Division of Gastroenterology, Saiseikai Yokohamashi-Nanbu Hospital, Yokohama 234-0054, Japan;
| | - Shuhei Nishigori
- Department of Gastroenterology, Yokohama Minami Kyosai Hospital, Yokohama 236-0037, Japan;
| | - Manabu Morimoto
- Hepatobiliary and Pancreatic Medical Oncology, Kanagawa Cancer Center Hospital, Yokohama 241-8585, Japan;
| | - Nobuhiro Hattori
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan;
| | - Katsuaki Ogushi
- Gastroenterological Center, Yokohama City University Medical Center, Yokohama 232-0024, Japan; (K.N.); (K.O.); (A.N.); (H.F.)
| | - Akito Nozaki
- Gastroenterological Center, Yokohama City University Medical Center, Yokohama 232-0024, Japan; (K.N.); (K.O.); (A.N.); (H.F.)
| | - Hiroyuki Fukuda
- Gastroenterological Center, Yokohama City University Medical Center, Yokohama 232-0024, Japan; (K.N.); (K.O.); (A.N.); (H.F.)
| | - Tatehiro Kagawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, Isehara 259-1193, Japan; (S.H.); (T.K.)
| | - Kojiro Michitaka
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama 790-0024, Japan; (A.H.); (K.M.)
| | - Takashi Kumada
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki 503-8502, Japan; (H.T.); (T.T.); (T.K.)
| | - Shin Maeda
- Department of Gastroenterology, Yokohama City University Hospital, Yokohama 236-0004, Japan;
| |
Collapse
|
9
|
Hypoxic environment may enhance migration/penetration of endocrine resistant MCF7- derived breast cancer cells through monolayers of other non-invasive cancer cells in vitro. Sci Rep 2020; 10:1127. [PMID: 31980706 PMCID: PMC6981140 DOI: 10.1038/s41598-020-58055-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/06/2020] [Indexed: 12/12/2022] Open
Abstract
The response of cancer cells to hypoxic conditions found within the interior of a tumor mass is mediated through the hypoxia inducible factor (HIF) cascade and is thought to promote metastasis. However, given their distant proximity from blood vessels as compared to normoxic cells at the vascularised tumor periphery, it is uncertain if these cells can migrate through the tumor mass to gain access. Hypoxia was simulated by exposure to cobalt chloride or deferoxamine in normal (MCF10A) and cancerous [estrogen receptor (ER)−ve (pII), and ER +ve (YS1.2/ EII)] cells. In this report, HIF1α expression and localization was measured using western blotting, ELISA, and immunofluorescence, cell proliferation by MTT assay, motility and invasion by wound healing, live cell imaging, matrigel and co-culture in chambered slides. We found that the expression and nuclear translocation of HIF1α was significantly elevated by hypoxia, which inhibited cell proliferation, but significantly increased motility of pII cells and their penetration into and through a dense layer of adjacent EII cells, as well as their selective emergence out of a co-culture. These data suggest that endocrine resistant pII cancer cells, having undergone epithelial to mesenchymal transition are able to penetrate through other cell layers, with possible enhancement in response to hypoxia.
Collapse
|
10
|
Miyagawa Y, Yanai A, Yanagawa T, Inatome J, Egawa C, Nishimukai A, Takamoto K, Morimoto T, Kikawa Y, Suwa H, Taji T, Yamaguchi A, Okada Y, Sata A, Fukui R, Bun A, Ozawa H, Higuchi T, Fujimoto Y, Imamura M, Miyoshi Y. Baseline neutrophil-to-lymphocyte ratio and c-reactive protein predict efficacy of treatment with bevacizumab plus paclitaxel for locally advanced or metastatic breast cancer. Oncotarget 2020; 11:86-98. [PMID: 32002126 PMCID: PMC6967770 DOI: 10.18632/oncotarget.27423] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/21/2019] [Indexed: 01/05/2023] Open
Abstract
The effect of bevacizumab plus paclitaxel therapy on progression-free survival (PFS) is prominent; however, no overall survival (OS) benefit has been demonstrated. Our aim was to study the predictive efficacy of peripheral immune-related parameters, neutrophil-to-lymphocyte ratio (NLR), absolute lymphocyte count (ALC), and c-reactive protein (CRP) in locally advanced and metastatic breast cancers. A total of 179 patients treated with bevacizumab plus paclitaxel were recruited from three institutes in the test cohort. The cut-off values of NLR, ALC, and CRP were set at 3, 1500/μL, and 1.0 mg/dL, respectively, and baseline values of these factors were measured. The PFS of patients with NLR-low was significantly longer than that of patients with -high (median, 12.6 vs. 7.2 months; hazard ratio (HR), 0.48, 95% confidence interval (95% CI), 0.31–0.73; p = 0.0004). OS of patients with NLR-low was significantly better than those with-high (22.2 vs. 13.5 months; HR, 0.57, 95% CI, 0.39–0.83; p = 0.0032). Similarly, improved PFS and OS were recognized in patients with CRP-low as compared with patients with -high (HR, 0.44, 95% CI, 0.28–0.68; p = 0.0001 and HR, 0.39, 95% CI, 0.26–0.61, p < 0.0001, respectively). In the validation cohort from two institutes (n = 57), similar significant improvements in PFS and OS were confirmed for patients with NLR-low (p = 0.0344 and p = 0.0233, respectively) and CRP-low groups (p < 0.0001 and p = 0.0001, respectively). Low levels of NLR and CRP at baseline were significantly associated with improved prognosis in patients treated with bevacizumab plus paclitaxel.
Collapse
Affiliation(s)
- Yoshimasa Miyagawa
- Department of Surgery, Division of Breast and Endocrine Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Ayako Yanai
- Department of Surgery, Kansai Rosai Hospital, Amagasaki, Hyogo 660-8511, Japan
| | - Takehiro Yanagawa
- Department of Surgery, Kansai Rosai Hospital, Amagasaki, Hyogo 660-8511, Japan
| | - Junichi Inatome
- Department of Surgery, Kansai Rosai Hospital, Amagasaki, Hyogo 660-8511, Japan
| | - Chiyomi Egawa
- Department of Surgery, Kansai Rosai Hospital, Amagasaki, Hyogo 660-8511, Japan
| | - Arisa Nishimukai
- Department of Breast Surgery, Yao Municipal Hospital, Yao, Osaka 581-0069, Japan
| | - Kaori Takamoto
- Department of Breast Surgery, Yao Municipal Hospital, Yao, Osaka 581-0069, Japan
| | - Takashi Morimoto
- Department of Breast Surgery, Yao Municipal Hospital, Yao, Osaka 581-0069, Japan
| | - Yuichiro Kikawa
- Department of Breast Surgery, Kobe City Medical Center General Hospital, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hirofumi Suwa
- Department of Breast Surgery, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Hyogo 660-8550, Japan
| | - Tomoe Taji
- Department of Breast Surgery, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Hyogo 660-8550, Japan
| | - Ai Yamaguchi
- Department of Breast Surgery, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Hyogo 660-8550, Japan
| | - Yuki Okada
- Department of Breast Surgery, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Hyogo 660-8550, Japan
| | - Atsushi Sata
- Department of Surgery, Division of Breast and Endocrine Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Reiko Fukui
- Department of Surgery, Division of Breast and Endocrine Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Ayako Bun
- Department of Surgery, Division of Breast and Endocrine Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Hiromi Ozawa
- Department of Surgery, Division of Breast and Endocrine Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Tomoko Higuchi
- Department of Surgery, Division of Breast and Endocrine Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Yukie Fujimoto
- Department of Surgery, Division of Breast and Endocrine Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Michiko Imamura
- Department of Surgery, Division of Breast and Endocrine Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Yasuo Miyoshi
- Department of Surgery, Division of Breast and Endocrine Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
11
|
Hamaguchi T, Denda T, Kudo T, Sugimoto N, Ura T, Yamazaki K, Fujii H, Kajiwara T, Nakajima TE, Takahashi S, Otsu S, Komatsu Y, Nagashima F, Moriwaki T, Esaki T, Sato T, Itabashi M, Oki E, Sasaki T, Chiron M, Yoshino T. Exploration of potential prognostic biomarkers in aflibercept plus FOLFIRI in Japanese patients with metastatic colorectal cancer. Cancer Sci 2019; 110:3565-3572. [PMID: 31520559 PMCID: PMC6825011 DOI: 10.1111/cas.14198] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 11/30/2022] Open
Abstract
Aflibercept plus 5-fluorouracil/levofolinate/irinotecan (FOLFIRI) is a second-line treatment for metastatic colorectal cancer. This ancillary exploratory analysis of data in Japanese people was aimed at exploring the relationship between a set of potential prognostic biomarkers and efficacy endpoints following aflibercept plus FOLFIRI therapy. Sixty-two patients with metastatic colorectal cancer received aflibercept (4 mg/kg) plus FOLFIRI every 2 weeks. Seventy-eight potential protein biomarkers were chosen for analysis based on their roles in angiogenesis, tumor progression, and tumor-stroma interaction. Plasma levels of biomarkers at baseline and at pre-dose 3 (day 1 of treatment cycle 3) were measured in all patients by ELISA. Relationships between these levels and efficacy endpoints were assessed. Ten potential biomarkers had a ±30% change from baseline to pre-dose 3 (adjusted P < .001), with the greatest changes occurring in placental growth factor (median: +4716%) and vascular endothelial growth factor receptor 1 (+2171%). Baseline levels of eight potential biomarkers correlated with overall survival in a univariate Cox regression analysis: extracellular newly identified receptor for advanced glycation end-products binding protein, insulin-like growth factor-binding protein 1, interleukin-8, kallikrein 5, pulmonary surfactant-associated protein D, tissue inhibitor of metalloproteinases 1, tenascin-C, and tumor necrosis factor receptor 2. None correlated with progression-free survival or maximum tumor shrinkage. Pre-dose 3 levels did not correlate with any efficacy endpoints. Preliminary data show that these eight biomarkers could be associated with overall survival. ClinicalTrials.gov identifier: NCT01882868.
Collapse
Affiliation(s)
- Tetsuya Hamaguchi
- Department of Gastrointestinal Medical OncologyNational Cancer Center HospitalTokyoJapan
| | | | - Toshihiro Kudo
- Osaka University Graduate School of MedicineSuitaJapan
- Present address:
Department of Medical OncologyOsaka International Cancer InstituteOsakaJapan
| | - Naotoshi Sugimoto
- Department of Medical OncologyOsaka Medical Center for Cancer and Cardiovascular DiseaseOsakaJapan
- Present address:
Department of Medical OncologyOsaka International Cancer InstituteOsakaJapan
| | - Takashi Ura
- Department of Clinical OncologyAichi Cancer Center HospitalNagoyaJapan
- Present address:
Department of Clinical OncologyNational Hospital Organization Kyoto Medical CenterKyotoJapan
| | - Kentaro Yamazaki
- Department of Gastrointestinal OncologyShizuoka Cancer CenterShizuokaJapan
| | - Hirofumi Fujii
- Department of Clinical OncologyJichi Medical University HospitalShimotsukeJapan
| | - Takeshi Kajiwara
- Department of Gastrointestinal Medical OncologyShikoku Cancer CenterMatsuyamaJapan
| | - Takako Eguchi Nakajima
- Department of Medical OncologySt. Marianna University School of Medicine HospitalKawasakiJapan
| | - Shin Takahashi
- Department of Medical OncologyTohoku University HospitalSendaiJapan
| | - Satoshi Otsu
- Department of Medical OncologyOita University HospitalYufuJapan
| | - Yoshito Komatsu
- Department of Cancer CenterHokkaido University HospitalSapporoJapan
| | - Fumio Nagashima
- Department of Medical OncologyKyorin University HospitalMitakaJapan
| | - Toshikazu Moriwaki
- Department of GastroenterologyUniversity of Tsukuba HospitalTsukubaJapan
| | - Taito Esaki
- Department of Gastrointestinal and Medical OncologyNational Kyushu Cancer CenterFukuokaJapan
| | - Takeo Sato
- Department of Gastrointestinal SurgeryKitasato University East HospitalSagamiharaJapan
| | - Michio Itabashi
- Department of Surgery 2Tokyo Women's Medical University HospitalTokyoJapan
- Present address:
Department of SurgeryInstitute of GastroenterologyTokyo Women's Medical UniversityTokyoJapan
| | - Eiji Oki
- Department of Gastrointestinal SurgeryKyushu University HospitalFukuokaJapan
| | - Toru Sasaki
- Research and developmentSanofi K. K.TokyoJapan
| | | | - Takayuki Yoshino
- Department of Gastrointestinal OncologyNational Cancer Center Hospital EastKashiwaJapan
| |
Collapse
|
12
|
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]
|
13
|
Tiainen L, Korhonen EA, Leppänen VM, Luukkaala T, Hämäläinen M, Tanner M, Lahdenperä O, Vihinen P, Jukkola A, Karihtala P, Aho S, Moilanen E, Alitalo K, Kellokumpu-Lehtinen PL. High baseline Tie1 level predicts poor survival in metastatic breast cancer. BMC Cancer 2019; 19:732. [PMID: 31340773 PMCID: PMC6657075 DOI: 10.1186/s12885-019-5959-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 07/19/2019] [Indexed: 01/08/2023] Open
Abstract
Background Angiopoietin growth factors (Angs) regulate angiogenesis and lymphangiogenesis by binding to the endothelial Tie2 receptor. Ang2 expression is elevated in tissue hypoxia and inflammation, which also induce cleavage of the extracellular domain of the orphan Tie1 receptor. Here we have examined if the concentrations of Ang2 and the soluble extracellular domain of Tie1 in patient plasma are associated with the prognosis of patients with metastatic breast cancer. Methods Plasma Tie1 and Ang2 levels were measured in metastatic breast cancer patients treated in a phase II trial with a taxane-bevacizumab combination chemotherapy in the first-line treatment setting. They were analyzed before treatment, after 6 weeks and 6 months of treatment, and at the final study visit. Using the median concentrations as cutoffs, Tie1 and Ang2 data were dichotomized into low and high concentration groups. Additionally, we analyzed Tie1 concentrations in plasma from 10 healthy women participating in a breast cancer primary prevention study. Results Plasma samples were available from 58 (89%) of the 65 patients treated in the trial. The baseline Tie1 levels of the healthy controls were significantly lower than those of the metastatic patients (p < 0.001). The overall survival of the patients with a high baseline Tie1 level was significantly shorter (multivariate HR 3.07, 95% CI 1.39–6.79, p = 0.005). Additionally, the progression-free survival was shorter for patients with a high baseline Tie1 level (multivariate HR 3.78, 95% CI 1.57–9.09, p = 0.003). In contrast, the baseline Ang2 levels had no prognostic impact in a multivariate Cox proportional hazard regression analysis. The combined analysis of baseline Tie1 and Ang2 levels revealed that patients with both high Tie1 and high Ang2 baseline levels had a significantly shorter overall survival than the patients with low baseline levels of both markers (multivariate HR for overall survival 4.32, 95% CI 1.44–12.94, p = 0.009). Conclusions This is the first study to demonstrate the prognostic value of baseline Tie1 plasma concentration in patients with metastatic breast cancer. Combined with the results of the Ang2 analyses, the patients with both high Tie1 and Ang2 levels before treatment had the poorest survival. Trial registration Clinicaltrials.gov: NCT00979641, registration date 19-DEC-2008. The regional Ethics Committee: R08142M, registration date 18-NOV-2008.
Collapse
Affiliation(s)
- Leena Tiainen
- Department of Oncology, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, P.O. Box 100, FI-33014, Tampere, Finland. .,Department of Oncology, Tampere University Hospital, P.O. Box 2000, FI-33521, Tampere, Finland.
| | - Emilia A Korhonen
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Biomedicum Helsinki, P.O. Box 63, FI-00014, Helsinki, Finland
| | - Veli-Matti Leppänen
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Biomedicum Helsinki, P.O. Box 63, FI-00014, Helsinki, Finland
| | - Tiina Luukkaala
- Research, Development and Innovation Centre, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, P.O. Box 2000, FI-33521, Tampere, Finland
| | - Mari Hämäläinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, P.O. Box 100, FI-33014, Tampere, Finland
| | - Minna Tanner
- Department of Oncology, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, P.O. Box 100, FI-33014, Tampere, Finland.,Department of Oncology, Tampere University Hospital, P.O. Box 2000, FI-33521, Tampere, Finland
| | - Outi Lahdenperä
- Department of Oncology and Radiotherapy, Turku University Central Hospital, P.O. Box 52, 20521, Turku, Finland
| | - Pia Vihinen
- Department of Oncology and Radiotherapy, Turku University Central Hospital, P.O. Box 52, 20521, Turku, Finland
| | - Arja Jukkola
- Department of Oncology and Radiotherapy, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, P.O. Box 10, 90029 OYS, Oulu, Finland
| | - Peeter Karihtala
- Department of Oncology and Radiotherapy, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, P.O. Box 10, 90029 OYS, Oulu, Finland
| | - Sonja Aho
- Department of Oncology, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, P.O. Box 100, FI-33014, Tampere, Finland.,Department of Oncology, Tampere University Hospital, P.O. Box 2000, FI-33521, Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, P.O. Box 100, FI-33014, Tampere, Finland
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Biomedicum Helsinki, P.O. Box 63, FI-00014, Helsinki, Finland
| | - Pirkko-Liisa Kellokumpu-Lehtinen
- Department of Oncology, Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, P.O. Box 100, FI-33014, Tampere, Finland.,Department of Oncology, Tampere University Hospital, P.O. Box 2000, FI-33521, Tampere, Finland
| |
Collapse
|
14
|
Janning M, Müller V, Vettorazzi E, Cubas-Cordova M, Gensch V, Ben-Batalla I, Zu Eulenburg C, Schem C, Fasching PA, Schnappauf B, Karn T, Fehm T, Just M, Kühn T, Holms F, Overkamp F, Krabisch P, Rack B, Denkert C, Untch M, Tesch H, Rezai M, Kittel K, Pantel K, Bokemeyer C, Loibl S, von Minckwitz G, Loges S. Evaluation of soluble carbonic anhydrase IX as predictive marker for efficacy of bevacizumab: A biomarker analysis from the geparquinto phase III neoadjuvant breast cancer trial. Int J Cancer 2019; 145:857-868. [PMID: 30694523 DOI: 10.1002/ijc.32163] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/14/2018] [Accepted: 12/11/2018] [Indexed: 12/16/2022]
Abstract
We analyzed the predictive potential of pretreatment soluble carbonic anhydrase IX levels (sCAIX) for the efficacy of bevacizumab in the phase III neoadjuvant GeparQuinto trial. sCAIX was determined by enzyme-linked immunosorbent assay (ELISA). Correlations between sCAIX and pathological complete response (pCR), disease-free and overall survival (DFS, OS) were assessed with logistic and Cox proportional hazard regression models using bootstrapping for robust estimates and internal validation. 1,160 HER2-negative patient sera were analyzed, of whom 577 received bevacizumab. Patients with low pretreatment sCAIX had decreased pCR rates (12.1 vs. 20.1%, p = 0.012) and poorer DFS (adjusted 5-year DFS 71.4 vs. 80.5 months, p = 0.010) compared to patients with high sCAIX when treated with neoadjuvant chemotherapy (NCT). For patients with low sCAIX, pCR rates significantly improved upon addition of bevacizumab to NCT (12.1 vs. 20.4%; p = 0.017), which was not the case in patients with high sCAIX (20.1% for NCT vs. 17.0% for NCT-B, p = 0.913). When analyzing DFS we found that bevacizumab improved 5-year DFS for patients with low sCAIX numerically but not significantly (71.4 vs. 78.5 months; log rank 0.234). In contrast, addition of bevacizumab worsened 5-year DFS for patients with high sCAIX (81 vs. 73.6 months, log-rank 0.025). By assessing sCAIX levels we identified a patient cohort in breast cancer that is potentially undertreated with NCT alone. Bevacizumab improved pCR rates in this group, suggesting sCAIX is a predictive biomarker for bevacizumab with regards to treatment response. Our data also show that bevacizumab is not beneficial in patients with high sCAIX.
Collapse
Affiliation(s)
- Melanie Janning
- Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volkmar Müller
- Clinic and Policlinic for Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eik Vettorazzi
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Miguel Cubas-Cordova
- Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Victoria Gensch
- Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Isabel Ben-Batalla
- Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christine Zu Eulenburg
- Department of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Schem
- Department of Gynecology and Obstetrics, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Benjamin Schnappauf
- Department of Obstetrics and Gynecology, School of Medicine, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Thomas Karn
- Department of Obstetrics and Gynecology, School of Medicine, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Tanja Fehm
- Department of Gynecology and Obstetrics, University Düsseldorf, Germany
| | - Marianne Just
- Onkologische Schwerpunktpraxis Bielefeld, Bielefeld, Germany
| | - Thorsten Kühn
- Department of Gynecology and Obstetrics, Klinikum Esslingen, Esslingen, Germany
| | - Frank Holms
- Department of Gynecology and Obstetrics, St. Barbara Kliniken Heessen, Hamm, Germany
| | | | - Petra Krabisch
- Department of Gynecology and Obstetrics, Klinikum Chemnitz, Germany
| | - Brigitte Rack
- Department of Obstetrics and Gynecology, Campus Innenstadt, University Hospital Munich, Germany
| | - Carsten Denkert
- Department of Pathology, Charité Berlin Campus Mitte and German Cancer Consortium (DKTK), Berlin, Germany
| | - Michael Untch
- Department of Gynecology and Obstetrics, Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Hans Tesch
- Centre for Hematology and Oncology, Frankfurt, Germany
| | - Mahdi Rezai
- Centre for Breast Cancer, Luisenkrankenhaus Düsseldorf, Germany
| | | | - Klaus Pantel
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Sonja Loges
- Department of Oncology, Hematology and Bone Marrow Transplantation with section Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
15
|
Marisi G, Scarpi E, Passardi A, Nanni O, Pagan F, Valgiusti M, Casadei Gardini A, Neri LM, Frassineti GL, Amadori D, Ulivi P. IL-8 and thrombospondin-1 as prognostic markers in patients with metastatic colorectal cancer receiving bevacizumab. Cancer Manag Res 2018; 10:5659-5666. [PMID: 30532588 PMCID: PMC6241685 DOI: 10.2147/cmar.s181570] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Purpose Bevacizumab (B) plus chemotherapy (CT) is a common choice for first-line treatment of metastatic colorectal cancer. Molecular predictors of B efficacy have still not been identified. We analyzed the role of 22 angiogenesis-associated proteins in patient outcome. Patients and methods Serum samples collected at baseline and at the first clinical evaluation were available for 58 patients enrolled in the randomized multicenter ITACa trial and who received CT+ B. Serum protein levels were determined using multiplex ELISA. Results Patients with baseline ≥145 pg/mL IL-8 showed shorter median progression-free survival and overall survival (OS) than those with lower levels (6.5 vs 6. 12.6 months; HR 7.39, P<0.0001 and 8.7 vs 28.8 months, HR 7.68, P<0.001, respectively). Moreover, patients with baseline thrombospondin-1 levels ≥12,000 ng/mL had a better median OS than those with lower levels (34.5 vs 13.1 months, HR 0.43, P=0.007). Patients with a ≥20% reduction in IL-8 levels from baseline to first clinical evaluation showed a better progression-free survival and OS than the others (HR 0.41, P=0.005 and HR 0.43, P=0.007, respectively). Conclusion Baseline IL-8 and thrombospondin-1 levels and reduced IL-8 during B treatment could represent potential prognostic markers in metastatic colorectal cancer.
Collapse
Affiliation(s)
- Giorgia Marisi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy,
| | - Emanuela Scarpi
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Alessandro Passardi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Oriana Nanni
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Flavia Pagan
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Martina Valgiusti
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Andrea Casadei Gardini
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Luca Maria Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara 44100, Italy
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Dino Amadori
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy
| | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy,
| |
Collapse
|
16
|
Paths for Improving Bevacizumab Available in 2018: The ADZT Regimen for Better Glioblastoma Treatment. Med Sci (Basel) 2018; 6:medsci6040084. [PMID: 30274295 PMCID: PMC6313620 DOI: 10.3390/medsci6040084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 09/22/2018] [Accepted: 09/25/2018] [Indexed: 01/14/2023] Open
Abstract
During glioblastoma treatment, the pharmaceutical monoclonal antibody to vascular endothelial growth factor A, bevacizumab, has improved the quality of life and delayed progression for several months, but has not (or only marginally) prolonged overall survival. In 2017, several dramatic research papers appeared that are crucial to our understanding of glioblastoma vis-a-vis the mode of action of bevacizumab. As a consequence of these papers, a new, potentially more effective treatment protocol can be built around bevacizumab. This is the ADZT regimen, where four old drugs are added to bevacizumab. These four drugs are apremilast, marketed to treat psoriasis, dapsone, marketed to treat Hansen’s disease, zonisamide, marketed to treat seizures, and telmisartan, marketed to treat hypertension. The ancillary attributes of each of these drugs have been shown to augment bevacizumab. This paper details the research data supporting this contention. Phase three testing of AZDT addition to bevacizumab is required to establish safety and effectiveness before general use.
Collapse
|
17
|
The clinical relevance of serum vascular endothelial growth factor (VEGF) in correlation to circulating tumor cells and other serum biomarkers in patients with metastatic breast cancer. Breast Cancer Res Treat 2018; 172:93-104. [DOI: 10.1007/s10549-018-4882-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 07/09/2018] [Indexed: 01/22/2023]
|
18
|
Quiros-Gonzalez I, Tomaszewski MR, Aitken SJ, Ansel-Bollepalli L, McDuffus LA, Gill M, Hacker L, Brunker J, Bohndiek SE. Optoacoustics delineates murine breast cancer models displaying angiogenesis and vascular mimicry. Br J Cancer 2018; 118:1098-1106. [PMID: 29576623 PMCID: PMC5931091 DOI: 10.1038/s41416-018-0033-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Optoacoustic tomography (OT) of breast tumour oxygenation is a promising new technique, currently in clinical trials, which may help to determine disease stage and therapeutic response. However, the ability of OT to distinguish breast tumours displaying different vascular characteristics has yet to be established. The aim of the study is to prove OT as a sensitive technique for differentiating breast tumour models with manifestly different vasculatures. METHODS Multispectral OT (MSOT) was performed in oestrogen-dependent (MCF-7) and oestrogen-independent (MDA-MB-231) orthotopic breast cancer xenografts. Total haemoglobin (THb) and oxygen saturation (SO2MSOT) were calculated. Pathological and biochemical evaluation of the tumour vascular phenotype was performed for validation. RESULTS MCF-7 tumours show SO2MSOT similar to healthy tissue in both rim and core, despite significantly lower THb in the core. MDA-MB-231 tumours show markedly lower SO2MSOT with a significant rim-core disparity. Ex vivo analysis revealed that MCF-7 tumours contain fewer blood vessels (CD31+) that are more mature (CD31+/aSMA+) than MDA-MB-231. MCF-7 presented higher levels of stromal VEGF and iNOS, with increased NO serum levels. The vasculogenic process observed in MCF-7 was consistent with angiogenesis, while MDA-MB-231 appeared to rely more on vascular mimicry. CONCLUSIONS OT is sensitive to differences in the vascular phenotypes of our breast cancer models.
Collapse
Affiliation(s)
- Isabel Quiros-Gonzalez
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Michal R Tomaszewski
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Sarah J Aitken
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
- Department of Histopathology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
| | - Laura Ansel-Bollepalli
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Leigh-Ann McDuffus
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Michael Gill
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Lina Hacker
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Joanna Brunker
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK
| | - Sarah E Bohndiek
- Department of Physics, University of Cambridge, JJ Thomson Avenue, Cambridge, CB3 0HE, UK.
- Cancer Research UK Cambridge Institute, University of Cambridge, Robinson Way, Cambridge, CB2 0RE, UK.
| |
Collapse
|
19
|
Ueda S, Saeki T, Osaki A, Yamane T, Kuji I. Bevacizumab Induces Acute Hypoxia and Cancer Progression in Patients with Refractory Breast Cancer: Multimodal Functional Imaging and Multiplex Cytokine Analysis. Clin Cancer Res 2017; 23:5769-5778. [PMID: 28679773 DOI: 10.1158/1078-0432.ccr-17-0874] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/29/2017] [Accepted: 06/30/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Bevacizumab, an antibody against endothelial growth factor, is a key but controversial drug in the treatment of metastatic breast cancer. We, therefore, aimed to determine the intrinsic resistance to bevacizumab at the physiologic and molecular levels in advanced breast cancer using PET, dynamic contrast-enhanced MRI, diffuse optical spectroscopic imaging (DOSI), and multiplex cytokine assays.Experimental Design: In total, 28 patients diagnosed with advanced stage III/IV breast cancer receiving single-agent bevacizumab for 1 week followed by paclitaxel combined with bevacizumab underwent 18F-fluorodeoxyglucose (FDG)-PET, 18F-fluoromisonidazole (FMISO)-PET, and MRI at both baseline and two courses after treatment initiation. Hemodynamic measurement using DOSI and blood sample collection were performed at baseline and multiple times during the first week after the initiation of single-agent bevacizumab. We distinguished nonresponders from responders by serial FDG-PET based on their glycolytic changes to chemotherapy.Results: Nonresponders showed significantly higher hypoxic activity on FMISO-PET and less tumor shrinkage than responders. Hemodynamic parameters showed higher tumor blood volume and a remarkable decrease in the tissue oxygen level in nonresponders compared with responders after the infusion of single-agent bevacizumab. Multiplex cytokine assays revealed increased plasma levels of both proangiogenic and hypoxia-related inflammatory cytokines in nonresponders and decreased levels in responders.Conclusions: Nonresponders exhibited a higher degree of angiogenesis with more severe hypoxia than responders during bevacizumab treatment. These findings demonstrated that the addition of bevacizumab to paclitaxel treatment under hypoxic conditions could be ineffective and may result in acute hypoxia and increased cytokine secretion associated with cancer progression. Clin Cancer Res; 23(19); 5769-78. ©2017 AACR.
Collapse
Affiliation(s)
- Shigeto Ueda
- Department of Breast Oncology, Saitama Medical University International Medical Center, Yamane, Hidaka, Saitama, Japan.
| | - Toshiaki Saeki
- Department of Breast Oncology, Saitama Medical University International Medical Center, Yamane, Hidaka, Saitama, Japan
| | - Akihiko Osaki
- Department of Breast Oncology, Saitama Medical University International Medical Center, Yamane, Hidaka, Saitama, Japan
| | - Tomohiko Yamane
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, Yamane, Hidaka, Saitama, Japan
| | - Ichiei Kuji
- Department of Nuclear Medicine, Saitama Medical University International Medical Center, Yamane, Hidaka, Saitama, Japan
| |
Collapse
|
20
|
Abstract
Seventy percent of cancer patients have detectable metastases when they receive a diagnosis and 90% of cancer deaths result from metastases. These two facts emphasise the urgency for research to study the mechanisms and processes that enable metastasis. We need to develop a greater understanding of the cellular and molecular mechanisms that cause metastasis and also we need to do more. We must also consider the micro- and macro-environmental factors that influence this disease. Studying this environmental context has led us to update the ‘seed and soil’ hypothesis which dates back to the 19th century. This theory describes cancerous cells as seeds and the substrate as the soil in target organs though this may seem antiquated. Nonetheless, the tissue specificity that researchers have recently observed in metastatic colonisation supports the validity of the seed and soil theory. We now know that the metastatic potential of a tumour cell depends on multiple, reciprocal interactions between the primary tumour and distant sites. These interactions determine tumour progression. Studies of metastasis have allowed us to develop treatments that focus on therapeutic effectiveness. These new treatments account for the frequent metastasis of some tumours to target organs such as bones, lungs, brain, and liver. The purpose of this review is first to describe interactions between the cellular and molecular entities and the target organ tumour environment that enables metastasis. A second aim is to describe the complex mechanisms that mediate these interactions.
Collapse
Affiliation(s)
- Francisco Arvelo
- Life Sciences Centre, Institute for Advanced Studies Foundation [Fundación Instituto de Estudios Avanzado]-IDEA, Apartado 17606, Caracas 1015-A, Venezuela; Tumour Biology Culture and Tissue Laboratory, Experimental Biology Institute, Central University of Venezuela, Apartado Apartado 47114, Caracas 1041-A, Venezuela
| | - Felipe Sojo
- Life Sciences Centre, Institute for Advanced Studies Foundation [Fundación Instituto de Estudios Avanzado]-IDEA, Apartado 17606, Caracas 1015-A, Venezuela
| | - Carlos Cotte
- Tumour Biology Culture and Tissue Laboratory, Experimental Biology Institute, Central University of Venezuela, Apartado Apartado 47114, Caracas 1041-A, Venezuela
| |
Collapse
|
21
|
Zhu D, Shen Z, Liu J, Chen J, Liu Y, Hu C, Li Z, Li Y. The ROS-mediated activation of STAT-3/VEGF signaling is involved in the 27-hydroxycholesterol-induced angiogenesis in human breast cancer cells. Toxicol Lett 2016; 264:79-86. [DOI: 10.1016/j.toxlet.2016.11.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 11/10/2016] [Accepted: 11/13/2016] [Indexed: 11/22/2022]
|