1
|
Okawa M, Yamakuchi M, Bibek A, Takenouchi K, Maywar DN, Yamada S, Inoue K, Higurashi K, Nakazawa J, Kawahira M, Kodama T, Tanoue K, Oyama Y, Higashi S, Fujisaki C, Hashinokuchi H, Tabaru A, Kanda H, Tachioka S, Imoto Y, Hashiguchi T, Soga Y. Plasma and serum concentrations of VEGF-A121, but not of VEGF-A165, increase post-bevacizumab administration. PLoS One 2024; 19:e0316035. [PMID: 39700124 DOI: 10.1371/journal.pone.0316035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND VEGF-A concentrations were measured in the blood of bevacizumab-treated cancer patients in previous studies, but a consensus has not formed that would develop VEGF-A into a clinical biomarker. Recently, methods to strictly distinguish between the VEGF-A isoforms have been developed but have not yet been applied to cancer patients undergoing bevacizumab treatment. METHODS An ELISA that strictly distinguishes between VEGF-A121 and VEGF-A165-the major isoforms of VEGF-A-and a commercially available ELISA for VEGF-A are used to determine the concentration of VEGF-A121, VEGF-A165, and VEGF-A in the blood of 12 patients with advanced colorectal cancer receiving bevacizumab therapy. RESULTS The serum and plasma concentrations of VEGF-A121 increased substantially post-bevacizumab administration; the median increase in serum was 860.8 pg/mL, 95% confidence interval (CI) [468.5, 1128.9], p = 0.0024, and in plasma was 808.6 pg/mL, 95% CI [748.7, 874.0], p = 0.00049. In stark contrast, VEGF-A165 after bevacizumab administration decreased in serum by a medium change of -73.8 pg/mL, 95% CI [-149.4, -10.2], p = 0.0034, with 83.3% of the post-bevacizumab concentrations falling below the high-accuracy threshold of 38 pg/mL; in plasma, all pre and post VEGF-A165 concentrations fell below this threshold. Concentrations of VEGF-A121 and VEGF-A165 in platelets did not change to a statistically significant degree. Adding recombinant VEGF-A121 (and -A165) or bevacizumab to plasma in patients post-bevacizumab administration increased or decreased, respectively, VEGF-A121 and VEGF-A165 levels. The increase in VEGF-A121 in plasma and serum after bevacizumab administration may be due to the dissociation of the complex of tumor-derived VEGF-A121 and bevacizumab when it moves from the stroma into the blood. CONCLUSIONS The VEGF-A121 isoform has been uniquely demonstrated as a clear marker of bevacizumab therapy in both plasma and serum, motivating further research on pursuing these isoforms as biomarkers in cancer care.
Collapse
Affiliation(s)
- Masashi Okawa
- Department of Cardiovascular and Gastroenterological Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Munekazu Yamakuchi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | - Aryal Bibek
- Department of Cardiovascular and Gastroenterological Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Kazunori Takenouchi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | - Drew N Maywar
- Electrical and Computer Engineering Technology, Rochester Institute of Technology, Rochester, New York, United States of America
| | | | | | | | - Junichi Nakazawa
- Department of Medical Oncology, Kagoshima City Hospital, Kagoshima, Japan
| | - Masahiro Kawahira
- Department of Medical Oncology, Kagoshima City Hospital, Kagoshima, Japan
| | - Tomoko Kodama
- Department of Medical Oncology, Kagoshima City Hospital, Kagoshima, Japan
| | - Kiyonori Tanoue
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | - Yoko Oyama
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | - Sadayuki Higashi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | - Chieko Fujisaki
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | | | - Akito Tabaru
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Hideaki Kanda
- Department of Cardiovascular and Gastroenterological Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Shuji Tachioka
- Department of Cardiovascular and Gastroenterological Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Yutaka Imoto
- Department of Cardiovascular and Gastroenterological Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Teruto Hashiguchi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | - Yoshiharu Soga
- Department of Cardiovascular and Gastroenterological Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
2
|
Gao F, Xu T, Zang F, Luo Y, Pan D. Cardiotoxicity of Anticancer Drugs: Molecular Mechanisms, Clinical Management and Innovative Treatment. Drug Des Devel Ther 2024; 18:4089-4116. [PMID: 39286288 PMCID: PMC11404500 DOI: 10.2147/dddt.s469331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/05/2024] [Indexed: 09/19/2024] Open
Abstract
With the continuous refinement of therapeutic measures, the survival rate of tumor patients has been improving year by year, while cardiovascular complications related to cancer therapy have become increasingly prominent. Exploring the mechanism and prevention strategy of cancer therapy-related cardiovascular toxicity (CTR-CVT) remains one of the research hotspots in the field of Cardio-Oncology in recent years. Cardiotoxicity of anticancer drugs involves heart failure, myocarditis, hypertension, arrhythmias and vascular toxicity, mechanistically related to vascular endothelial dysfunction, ferroptosis, mitochondrial dysfunction and oxidative stress. To address the cardiotoxicity induced by different anticancer drugs, various therapeutic measures have been put in place, such as reducing the accumulation of anticancer drugs, shifting to drugs with less cardiotoxicity, using cardioprotective drugs, and early detection. Due to the very limited treatments available to ameliorate anticancer drugs-induced cardiotoxicity, a few innovations are being shifted from animal studies to human studies. Examples include mitochondrial transplantation. Mitochondrial transplantation has been proven to be effective in in vivo and in vitro experiments. Several recent studies have demonstrated that intercellular mitochondrial transfer can ameliorate doxorubicin(DOX)-induced cardiotoxicity, laying the foundation for innovative therapies in anticancer drugs-induced cardiotoxicity. In this review, we will discuss the current status of anticancer drugs-induced cardiotoxicity in terms of the pathogenesis and treatment, with a focus on mitochondrial transplantation, and we hope that this review will bring some inspiration to you.
Collapse
Affiliation(s)
- Feiyu Gao
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Tao Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Fangnan Zang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Yuanyuan Luo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Defeng Pan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| |
Collapse
|
3
|
Kuo HY, Khan KA, Kerbel RS. Antiangiogenic-immune-checkpoint inhibitor combinations: lessons from phase III clinical trials. Nat Rev Clin Oncol 2024; 21:468-482. [PMID: 38600370 DOI: 10.1038/s41571-024-00886-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/12/2024]
Abstract
Antiangiogenic agents, generally antibodies or tyrosine-kinase inhibitors that target the VEGF-VEGFR pathway, are currently among the few combination partners clinically proven to improve the efficacy of immune-checkpoint inhibitors (ICIs). This benefit has been demonstrated in pivotal phase III trials across different cancer types, some with practice-changing results; however, numerous phase III trials have also had negative results. The rationale for using antiangiogenic drugs as partners for ICIs relies primarily on blocking the multiple immunosuppressive effects of VEGF and inducing several different vascular-modulating effects that can stimulate immunity, such as vascular normalization leading to increased intratumoural blood perfusion and flow, and inhibition of pro-apoptotic effects of endothelial cells on T cells, among others. Conversely, VEGF blockade can also cause changes that suppress antitumour immunity, such as increased tumour hypoxia, and reduced intratumoural ingress of co-administered ICIs. As a result, the net clinical benefits from antiangiogenic-ICI combinations will be determined by the balance between the opposing effects of VEGF signalling and its inhibition on the antitumour immune response. In this Perspective, we summarize the results from the currently completed phase III trials evaluating antiangiogenic agent-ICI combinations. We also discuss strategies to improve the efficacy of these combinations, focusing on aspects that include the deleterious functions of VEGF-VEGFR inhibition on antitumour immunity, vessel co-option as a driver of non-angiogenic tumour growth, clinical trial design, or the rationale for drug selection, dosing and scheduling.
Collapse
Affiliation(s)
- Hung-Yang Kuo
- Department of Oncology, National Taiwan University Hospital, and Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Kabir A Khan
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| | - Robert S Kerbel
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
4
|
Xu T, Wang J, Wu Y, Wu J, Lu W, Liu M, Zhang S, Xie D, Xin W, Xie J. Ac4C Enhances the Translation Efficiency of Vegfa mRNA and Mediates Central Sensitization in Spinal Dorsal Horn in Neuropathic Pain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303113. [PMID: 37877615 PMCID: PMC10724395 DOI: 10.1002/advs.202303113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/25/2023] [Indexed: 10/26/2023]
Abstract
N4-Acetylcytidine (ac4C), a highly conserved post-transcriptional machinery with extensive existence for RNA modification, plays versatile roles in various cellular processes and functions. However, the molecular mechanism by which ac4C modification mediates neuropathic pain remains elusive. Here, it is found that the enhanced ac4C modification promotes the recruitment of polysome in Vegfa mRNA and strengthens the translation efficiency following SNI. Nerve injury increases the expression of NAT10 and the interaction between NAT10 and Vegfa mRNA in the dorsal horn neurons, and the gain and loss of NAT10 function further confirm that NAT10 is involved in the ac4C modification in Vegfa mRNA and pain behavior. Moreover, the ac4C-mediated VEGFA upregulation contributes to the central sensitivity and neuropathic pain induced by SNI or AAV-hSyn-NAT10. Finally, SNI promotes the binding of HNRNPK in Vegfa mRNA and subsequently recruits the NAT10. The enhanced interaction between HNRNPK and NAT10 contributes to the ac4C modification of Vegfa mRNA and neuropathic pain. These findings suggest that the enhanced interaction between HNRNPK and Vegfa mRNA upregulates the ac4C level by recruiting NAT10 and contributes to the central sensitivity and neuropathic pain following SNI. Blocking this cascade may be a novel therapeutic approach in patients with neuropathic pain.
Collapse
Affiliation(s)
- Ting Xu
- Neuroscience ProgramZhongshan School of MedicineThe Fifth Affiliated HospitalGuangdong Province Key Laboratory of Brain Function and DiseaseDepartment of Physiology and Pain Research CenterSun Yat‐Sen UniversityGuangzhou510080China
| | - Jing Wang
- Neuroscience ProgramZhongshan School of MedicineThe Fifth Affiliated HospitalGuangdong Province Key Laboratory of Brain Function and DiseaseDepartment of Physiology and Pain Research CenterSun Yat‐Sen UniversityGuangzhou510080China
- Department of Pain ManagementHenan Provincial People's HospitalZhengzhou UniversityZhengzhou450000China
| | - Yan Wu
- Department of AnesthesiologyThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdong510062China
| | - Jia‐Yan Wu
- Neuroscience ProgramZhongshan School of MedicineThe Fifth Affiliated HospitalGuangdong Province Key Laboratory of Brain Function and DiseaseDepartment of Physiology and Pain Research CenterSun Yat‐Sen UniversityGuangzhou510080China
| | - Wei‐Cheng Lu
- State Key Laboratory of Oncology in Southern ChinaCollaborative Innovation for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhou510060China
| | - Meng Liu
- Department of Anesthesia and Pain MedicineGuangzhou First People's HospitalGuangzhou510180China
| | - Su‐Bo Zhang
- State Key Laboratory of Oncology in Southern ChinaCollaborative Innovation for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhou510060China
| | - Dan Xie
- State Key Laboratory of Oncology in Southern ChinaCollaborative Innovation for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhou510060China
| | - Wen‐Jun Xin
- Neuroscience ProgramZhongshan School of MedicineThe Fifth Affiliated HospitalGuangdong Province Key Laboratory of Brain Function and DiseaseDepartment of Physiology and Pain Research CenterSun Yat‐Sen UniversityGuangzhou510080China
| | - Jing‐Dun Xie
- State Key Laboratory of Oncology in Southern ChinaCollaborative Innovation for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhou510060China
| |
Collapse
|
5
|
Saliby RM, El Zarif T, Bakouny Z, Shah V, Xie W, Flippot R, Denize T, Kane MH, Madsen KN, Ficial M, Hirsch L, Wei XX, Steinharter JA, Harshman LC, Vaishampayan UN, Severgnini M, McDermott DF, Mary Lee GS, Xu W, Van Allen EM, McGregor BA, Signoretti S, Choueiri TK, McKay RR, Braun DA. Circulating and Intratumoral Immune Determinants of Response to Atezolizumab plus Bevacizumab in Patients with Variant Histology or Sarcomatoid Renal Cell Carcinoma. Cancer Immunol Res 2023; 11:1114-1124. [PMID: 37279009 PMCID: PMC10526700 DOI: 10.1158/2326-6066.cir-22-0996] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/04/2023] [Accepted: 06/02/2023] [Indexed: 06/07/2023]
Abstract
Renal cell carcinoma (RCC) of variant histology comprises approximately 20% of kidney cancer diagnoses, yet the optimal therapy for these patients and the factors that impact immunotherapy response remain largely unknown. To better understand the determinants of immunotherapy response in this population, we characterized blood- and tissue-based immune markers for patients with variant histology RCC, or any RCC histology with sarcomatoid differentiation, enrolled in a phase II clinical trial of atezolizumab and bevacizumab. Baseline circulating (plasma) inflammatory cytokines were highly correlated with one another, forming an "inflammatory module" that was increased in International Metastatic RCC Database Consortium poor-risk patients and was associated with worse progression-free survival (PFS; P = 0.028). At baseline, an elevated circulating vascular endothelial growth factor A (VEGF-A) level was associated with a lack of response (P = 0.03) and worse PFS (P = 0.021). However, a larger increase in on-treatment levels of circulating VEGF-A was associated with clinical benefit (P = 0.01) and improved overall survival (P = 0.0058). Among peripheral immune cell populations, an on-treatment decrease in circulating PD-L1+ T cells was associated with improved outcomes, with a reduction in CD4+PD-L1+ [HR, 0.62; 95% confidence interval (CI), 0.49-0.91; P = 0.016] and CD8+PD-L1+ T cells (HR, 0.59; 95% CI, 0.39-0.87; P = 0.009) correlated with improved PFS. Within the tumor itself, a higher percentage of terminally exhausted (PD-1+ and either TIM-3+ or LAG-3+) CD8+ T cells was associated with worse PFS (P = 0.028). Overall, these findings support the value of tumor and blood-based immune assessments in determining therapeutic benefit for patients with RCC receiving atezolizumab plus bevacizumab and provide a foundation for future biomarker studies for patients with variant histology RCC receiving immunotherapy-based combinations.
Collapse
Affiliation(s)
- Renee Maria Saliby
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Talal El Zarif
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Ziad Bakouny
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
- Department of Internal Medicine, Brigham and Women’s Hospital, Boston, MA, 02215, USA
| | - Valisha Shah
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Wanling Xie
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Ronan Flippot
- Department of Cancer Medicine, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Thomas Denize
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - M. Harry Kane
- Yale Center of Cellular and Molecular Oncology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Katrine N. Madsen
- Yale Center of Cellular and Molecular Oncology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Miriam Ficial
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Laure Hirsch
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Xiao X. Wei
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - John A. Steinharter
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
- Larner College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Lauren C. Harshman
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
- Surface Oncology, Cambridge MA 02139, USA
| | - Ulka N. Vaishampayan
- University of Michigan/Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201 USA
| | - Mariano Severgnini
- Center for Immuno-Oncology Immune Assessment Laboratory at the Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - David F. McDermott
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Gwo-Shu Mary Lee
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Wenxin Xu
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Eliezer M. Van Allen
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Bradley A. McGregor
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Toni K. Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Rana R. McKay
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92037, USA
| | - David A. Braun
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
- Yale Center of Cellular and Molecular Oncology, Yale School of Medicine, New Haven, CT 06511, USA
| |
Collapse
|
6
|
Matsumoto M, Noritake H, Yamashita M, Hanaoka T, Umemura M, Kitsugi K, Takatori S, Ohta K, Ito J, Chida T, Kawata K. A case of hepatitis B virus-infected patient with bevacizumab-related severe intratumor hemorrhage of large hepatocellular carcinoma (HCC). KANZO 2023; 64:382-392. [DOI: 10.2957/kanzo.64.382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Moe Matsumoto
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine
| | - Hidenao Noritake
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine
| | - Maho Yamashita
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine
| | - Tomohiko Hanaoka
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine
| | - Masahiro Umemura
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine
| | - Kensuke Kitsugi
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine
| | - Shingo Takatori
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine
| | - Kazuyoshi Ohta
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine
| | - Jun Ito
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine
| | - Takeshi Chida
- Department of Regional Medical Care Support, Hamamatsu University School of Medicine
| | - Kazuhito Kawata
- Division of Hepatology, Department of Internal Medicine, Hamamatsu University School of Medicine
| |
Collapse
|
7
|
Antitumor effect of melatonin on breast cancer in experimental models: A systematic review. Biochim Biophys Acta Rev Cancer 2023; 1878:188838. [PMID: 36403922 DOI: 10.1016/j.bbcan.2022.188838] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 11/19/2022]
Abstract
Breast cancer is the most frequent malignant neoplasm in females. While conventional treatments such as chemotherapy and radiotherapy are available, they are highly invasive and toxic to oncological patients. Melatonin is a promising molecule for the treatment of breast cancer with antitumor effects on tumorigenesis and tumor progression. The aim of this systematic review was to synthesize knowledge about the antitumor effect of melatonin on breast cancer in experimental models and propose the main mechanisms of action already described in relation to the processes regulated by melatonin. PubMed, Web of Science, and Embase databases were used. The inclusion criteria were in vitro and in vivo experimental studies that used different formulations of melatonin as a treatment for breast cancer, without year or language restrictions. Risk of bias for studies was assessed using the Systematic Review Center for Laboratory Animal Experimentation (SYRCLE) tool. Data from selected articles were presented as narrative descriptions and tables. Seventy-five articles on different breast cancer cell lines and experimental models treated with melatonin alone, or in combination with other compounds were included. Melatonin showed antitumor effects on proliferative pathways related to the cell cycle and tumorigenesis, tumor death, angiogenesis, and tumor metastasis, as well as on oxidative stress and immune regulatory pathways. These effects were either dependent or independent of melatonin receptors. Herein, we clarify the antitumor action of melatonin on different tumorigenic processes in breast cancer in experimental models. Systematic review registration: PROSPERO database (CRD42022309822/https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022309822).
Collapse
|
8
|
Two cases showing the effects of bevacizumab on recurrent cervical cancer with pleural effusion. Int Cancer Conf J 2022; 11:165-171. [PMID: 35669902 PMCID: PMC9163267 DOI: 10.1007/s13691-022-00538-x] [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: 12/17/2021] [Accepted: 02/09/2022] [Indexed: 10/18/2022] Open
Abstract
The survival rate and quality of life of patients with recurrent cervical cancer and pleural effusion had been extremely poor until bevacizumab was approved. We report two cases of recurrent cervical cancer with remarkably decreased pleural effusion and a long survival rate after combination chemotherapy with bevacizumab. Case 1: A patient was diagnosed with stage IIB cervical adenocarcinoma and treated with concurrent chemoradiotherapy (CCRT), total hysterectomy, and paclitaxel/carboplatin (TC) therapy as the primary treatment. After the first recurrence had been treated with irinotecan-cisplatin therapy and radiotherapy, symptomatic pleural effusion emerged. Paclitaxel-cisplatin-bevacizumab (Pac-Cis-Bev) was administered during 13 cycles of chemotherapy to promptly relieve pleural effusion, respiratory distress, and back pain. She survived for more than a year and a half after starting Pac-Cis-Bev therapy. Case 2: A patient was diagnosed with stage IIIB cervical squamous cell carcinoma and pulmonary recurrence after CCRT. After 21 cycles of TC or Pac-Cis-Bev therapy, pleural effusion emerged. Topotecan-paclitaxel-bevacizumab (Topo-Pac-Bev) was administered for 12 cycles. Respiratory distress was relieved in 2 weeks and pleural effusion almost completely resolved after 2 months. We changed the treatment to ifosfamide and nedaplatin as pleural effusion exacerbated. However, this treatment was not effective; hence the patient was rechallenged with Topo-Pac-Bev therapy. Six cycles of Topo-Pac-Bev rechallenge therapy effectively suppressed pleural effusion. She survived for 2 years after pleural effusion appeared. Chemotherapy with bevacizumab is useful for both symptom relief and improvement in prognosis in patients with recurrent cervical cancer, despite being in the late phase.
Collapse
|
9
|
Ozaki Y, Tsurutani J, Mukohara T, Iwasa T, Takahashi M, Tanabe Y, Kawabata H, Masuda N, Futamura M, Minami H, Matsumoto K, Yoshimura K, Kitano S, Takano T. Safety and efficacy of nivolumab plus bevacizumab, paclitaxel for HER2-negative metastatic breast cancer: Primary results and biomarker data from a phase 2 trial (WJOG9917B). Eur J Cancer 2022; 171:193-202. [PMID: 35728379 DOI: 10.1016/j.ejca.2022.05.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/02/2022] [Accepted: 05/12/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Preclinical models revealed potential synergistic effects of programmed cell death-1 inhibitors and anti-vascular endothelial growth factor (VEGF) antibodies. Therefore, we investigated the use of nivolumab, bevacizumab, and paclitaxel triple therapy for metastatic breast cancer. METHODS This phase 2, multicentre, single-arm study (NEWBEAT) investigated the safety and efficacy of first-line nivolumab, paclitaxel, and bevacizumab in patients with human epidermal growth factor receptor 2-negative metastatic breast cancer, regardless of programmed cell death-ligand 1 expression. The primary end-point was objective response rate. Key secondary end-points included progression-free survival, overall survival, and toxicities. A biomarker study evaluated tumour programmed cell death-ligand 1 expression and serum VEGF-A levels. RESULTS Between February 2018 and October 2018, 57 patients were enrolled. An objective response rate was seen in 39/56 patients (70%, 95% confidence interval [CI]: 55.9-81.2%), meeting the primary end-point. The objective response rate was 74% in patients with hormone receptor-positive breast cancer versus 59% in patients with triple-negative breast cancer. The median progression-free survival and overall survival were 14.0 (95% CI 11.0-16.3) and 32.5 (95% CI 26.0-not evaluable) months, respectively (median follow-up: 29.5 months). Grade 3/4 adverse drug reactions occurred in 33 of 57 patients (58%). There were no grade 5 adverse events. Immune-related adverse events occurred in 43 of 57 patients (75%), with grade 3/4 events in eight patients (14%). Biomarker analysis showed that tumour programmed cell death-ligand 1 expression was not correlated with the efficacy of triple therapy. Efficacy outcomes were similar between the serum VEGF-high and VEGF-low groups. CONCLUSIONS First-line nivolumab, bevacizumab, and paclitaxel therapy showed promising efficacy and manageable toxicity in patients with human epidermal growth factor receptor 2-negative metastatic breast cancer.
Collapse
Affiliation(s)
- Yukinori Ozaki
- Department of Medical Oncology, Toranomon Hospital, Tokyo, Japan; Department of Breast Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Junji Tsurutani
- Advanced Cancer Translational Research Institute, Showa University, Tokyo, Japan
| | - Toru Mukohara
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tsutomu Iwasa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Masato Takahashi
- Department of Breast Surgery, National Hospital Organization Hokkaido Cancer Center, Hokkaido, Japan
| | - Yuko Tanabe
- Department of Medical Oncology, Toranomon Hospital, Tokyo, Japan
| | - Hidetaka Kawabata
- Department of Breast and Endocrine Surgery, Toranomon Hospital, Tokyo, Japan
| | - Norikazu Masuda
- Department of Surgery, Breast Oncology, National Hospital Organization Osaka National Hospital, Osaka, Japan; Department of Breast and Endocrine Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Hironobu Minami
- Medical Oncology/Hematology, Internal Medicine, Kobe University School of Medicine, Hyogo, Japan
| | - Koji Matsumoto
- Department of Medical Oncology, Hyogo Cancer Center, Hyogo, Japan
| | - Kenichi Yoshimura
- Center for Integrated Medical Research, Hiroshima University Hospital, Hiroshima University, Hiroshima, Japan
| | - Shigehisa Kitano
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan; Department of Advanced Medical Development, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toshimi Takano
- Department of Medical Oncology, Toranomon Hospital, Tokyo, Japan; Department of Breast Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
10
|
Chen R, Huang P, Wei S, Zhang C, Lai X, Wang H, Tang J. Methamphetamine exposure increases cardiac microvascular permeability by activating the VEGF-PI3K-Akt-eNOS signaling pathway, reversed by Bevacizumab. Hum Exp Toxicol 2022; 41:9603271221121795. [PMID: 35975811 DOI: 10.1177/09603271221121795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Methamphetamine (METH) is an illicit amphetamine-like psychostimulant that is commonly abused. However, the modulation of METH-induced cardiac microvascular permeability is still not completely known. Previously, we discovered that the vascular endothelial growth factor (VEGF) regulated the cardiotoxicity produced by METH. In this work, we looked into the effect of METH exposure on cardiac microvascular permeability via the VEGF-PI3K-Akt-eNOS signaling pathway, as well as the efficacy of Bevacizumab treatment in reducing this effect. The findings revealed that METH exposure enhanced cardiac microvascular permeability while also activating the VEGF-PI3K-Akt-eNOS signaling pathway. Furthermore, treatment with Bevacizumab has been shown to be effective in reversing the METH-induced phenomena. Briefly stated, our research may provide fresh insight into the molecular underpinnings of METH-induced cardiac microvascular permeability, and it may also provide evidence for a relationship between METH misuse and Bevacizumab medication.
Collapse
Affiliation(s)
- Rui Chen
- Department of Forensic Medicine, 12453Guangdong Medical University, Dongguan, China
| | - Peng Huang
- Women and Children Medical Research Center, Affiliated Foshan Maternity & Child Healthcare Hospital, 70570Southern Medical University, Foshan, China
| | - Songren Wei
- Department of Neuropharmacology and Novel Drug Discovery, School of Pharmaceutical Sciences, 70570Southern Medical University, Guangzhou, China
| | - Cui Zhang
- School of Forensic Medicine, 70570Southern Medical University, Guangzhou, China
| | - Xiaoping Lai
- Department of Forensic Medicine, 12453Guangdong Medical University, Dongguan, China
| | - Huijun Wang
- School of Forensic Medicine, 70570Southern Medical University, Guangzhou, China
| | - Jianpin Tang
- Department of Forensic Medicine, 12453Guangdong Medical University, Dongguan, China
| |
Collapse
|
11
|
Lee HS, Lee IH, Kang K, Park SI, Jung M, Yang SG, Kwon TW, Lee DY. Network Pharmacology-Based Dissection of the Comprehensive Molecular Mechanisms of the Herbal Prescription FDY003 Against Estrogen Receptor-Positive Breast Cancer. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211044377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Estrogen receptor-positive breast cancer (ERPBC) is the commonest subtype of breast cancer, with a high prevalence, incidence, and mortality. Herbal drugs are increasingly being used to treat ERPBC, although their mechanisms of action are not fully understood. Therefore, in this study, we aimed to analyze the therapeutic properties of FDY003, a herbal anti-ERPBC prescription, using a network pharmacology approach. FDY003 decreased the viability of human ERPBC cells and sensitized them to tamoxifen, an endocrine drug that is widely used in the treatment of ERPBC. The network pharmacology analysis revealed 18 pharmacologically active components in FDY003 that may interact with and regulate 66 therapeutic targets. The enriched gene ontology terms for the FDY003 targets were associated with the modulation of cell survival and death, cell proliferation and growth arrest, and estrogen-associated cellular processes. Analysis of the pathway enrichment of the targets showed that FDY003 may target a variety of ERPBC-associated pathways, including the PIK3-Akt, focal adhesion, MAPK, and estrogen pathways. Overall, these data provide a comprehensive mechanistic insight into the anti-ERPBC activity of FDY003.
Collapse
Affiliation(s)
- Ho-Sung Lee
- The Fore, Seoul, Republic of Korea
- Forest Hospital, Seoul, Republic of Korea
| | | | | | | | - Minho Jung
- Forest Hospital, Seoul, Republic of Korea
| | | | | | - Dae-Yeon Lee
- The Fore, Seoul, Republic of Korea
- Forest Hospital, Seoul, Republic of Korea
| |
Collapse
|
12
|
Pre-Therapeutic VEGF Level in Plasma Is a Prognostic Bio-Marker in Head and Neck Squamous Cell Carcinoma (HNSCC). Cancers (Basel) 2021; 13:cancers13153781. [PMID: 34359680 PMCID: PMC8345208 DOI: 10.3390/cancers13153781] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 11/25/2022] Open
Abstract
Simple Summary In the context of a growing variety in treatment strategies for patients with cancer, especially approaches based on antiangiogenetic pathways, we aimed to identify a useful biomarker for patients with head and neck squamous cell carcinoma (HNSCC). Our experimental results detected vascular endothelial growth factor (VEGF) in patients’ pre-therapeutic plasma, and not serum, which serves as a suitable biomarker for outcome prognostication. Results were validated in an independent cohort, confirming VEGF as an independent predictor (Pi) of outcomes in HNSCC patients. Therefore, pre-therapeutic VEGF in plasma may be an attractive biomarker in future HNSCC studies. Abstract Vascular endothelial growth factor (VEGF) is centrally involved in cancer angiogenesis. We hypothesized that pre-therapeutic VEGF levels in serum and plasma differ in their potential as biomarkers for outcomes in head and neck squamous cell carcinoma (HNSCC) patients. As prospectively defined in the study protocols of TRANSCAN-DietINT and NICEI-CIH, we measured VEGF in pretreatment serum and plasma of 75 HNSCC test cohort (TC) patients. We analyzed the prognostic value of VEGF concentrations in serum (VEGFSerum) and plasma (VEGFPlasma) for event-free survival (EFS) utilizing receiver-operating characteristics (ROC). Mean VEGF concentrations in plasma (34.6, 95% CI 26.0–43.3 ng/L) were significantly lower (p = 3.35 × 10−18) than in serum (214.8, 95% CI 179.6–250.0 ng/L) but, based on ROC (area under the curve, AUCPlasma = 0.707, 95% CI 0.573–0.840; p = 0.006 versus AUCSerum = 0.665, 95% CI 0.528–0.801; p = 0.030), superiorly correlated with event-free survival (EFS) of TC patients. Youden indices revealed optimum binary classification with VEGFPlasma 26 ng/L and VEGFSerum 264 ng/L. Kaplan–Meier plots demonstrated superiority of VEGFPlasma in discriminating patients regarding outcome. Patients with VEGFPlasma < 26 ng/L had superior nodal (NC), local (LC) and loco-regional control (LRC) leading to significant prolonged progression-free survival (PFS) and EFS. We successfully validated VEGFPlasma according the cut-off <26 ng/L as predictive for superior outcome in an independent validation cohort (iVC) of 104 HNSCC patients from the studies DeLOS-II and LIFE and found better outcomes including prolonged tumor-specific (TSS) and overall survival (OS). Outcomes in TC and iVC combined again was related to VEGFPlasma, and multivariate Cox regression revealed that VEGFPlasma was an independent outcome predictor. In HNSCC, pre-therapeutic VEGFPlasma is prognostic for outcomes.
Collapse
|
13
|
Colorectal Cancer: From Genetic Landscape to Targeted Therapy. JOURNAL OF ONCOLOGY 2021; 2021:9918116. [PMID: 34326875 PMCID: PMC8277501 DOI: 10.1155/2021/9918116] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/25/2021] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) is the third most common cancer type and the second cause of death worldwide. The advancement in understanding molecular pathways involved in CRC has led to new classifications based on the molecular characteristics of each tumor and also improved CRC management through the integration of targeted therapy into clinical practice. In this review, we will present the main molecular pathways involved in CRC carcinogenesis, the molecular classifications. The anti-VEGF and anti-EGFR therapies currently used in CRC treatment and those under clinical investigation will also be outlined, as well as the mechanisms of primary and acquired resistance to anti-EGFR monoclonal antibodies (cetuximab and panitumumab). Targeted therapy has led to great improvement in the treatment of metastatic CRC. However, there has been variability in CRC treatment outcomes due to molecular heterogeneity in colorectal tumors, which underscores the need for identifying prognostic and predictive biomarkers for CRC-targeted drugs.
Collapse
|
14
|
Han L, Korangath P, Nguyen NK, Diehl A, Cho S, Teo WW, Cope L, Gessler M, Romer L, Sukumar S. HEYL Regulates Neoangiogenesis Through Overexpression in Both Breast Tumor Epithelium and Endothelium. Front Oncol 2021; 10:581459. [PMID: 33520697 PMCID: PMC7845423 DOI: 10.3389/fonc.2020.581459] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/30/2020] [Indexed: 12/20/2022] Open
Abstract
Blocking tumor angiogenesis is an appealing therapeutic strategy, but to date, success has been elusive. We previously identified HEYL, a downstream target of Notch signaling, as an overexpressed gene in both breast cancer cells and as a tumor endothelial marker, suggesting that HEYL overexpression in both compartments may contribute to neoangiogenesis. Carcinomas arising in double transgenic Her2-neu/HeyL mice showed higher tumor vessel density and significantly faster growth than tumors in parental Her2/neu mice. Providing mechanistic insight, microarray-based mRNA profiling of HS578T-tet-off-HEYL human breast cancer cells revealed upregulation of several angiogenic factors including CXCL1/2/3 upon HEYL expression, which was validated by RT-qPCR and protein array analysis. Upregulation of the cytokines CXCL1/2/3 occurred through direct binding of HEYL to their promoter sequences. We found that vessel growth and migration of human vascular endothelial cells (HUVECs) was promoted by conditioned medium from HS578T-tet-off-HEYL carcinoma cells, but was blocked by neutralizing antibodies against CXCL1/2/3. Supporting these findings, suppressing HEYL expression using shRNA in MDA-MB-231 cells significantly reduced tumor growth. In addition, suppressing the action of proangiogenic cytokines induced by HEYL using a small molecule inhibitor of the CXCl1/2/3 receptor, CXCR2, in combination with the anti-VEGF monoclonal antibody, bevacizumab, significantly reduced tumor growth of MDA-MB-231 xenografts. Thus, HEYL expression in tumor epithelium has a profound effect on the vascular microenvironment in promoting neoangiogenesis. Furthermore, we show that lack of HEYL expression in endothelial cells leads to defects in neoangiogenesis, both under normal physiological conditions and in cancer. Thus, HeyL-/- mice showed impaired vessel outgrowth in the neonatal retina, while the growth of mammary tumor cells E0771 was retarded in syngeneic HeyL-/- mice compared to wild type C57/Bl6 mice. Blocking HEYL's angiogenesis-promoting function in both tumor cells and tumor-associated endothelium may enhance efficacy of therapy targeting the tumor vasculature in breast cancer.
Collapse
Affiliation(s)
- Liangfeng Han
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Preethi Korangath
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nguyen K Nguyen
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Adam Diehl
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Soonweng Cho
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Wei Wen Teo
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Leslie Cope
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Manfred Gessler
- Developmental Biochemistry, Comprehensive Cancer Center Mainfraken and Theodor-Boveri-Institute/Biocenter, University of Wurzburg, Wurzburg, Germany
| | - Lewis Romer
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,The Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Saraswati Sukumar
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
15
|
Li M, An W, Wang L, Zhang F, Li J, Zhang Y, Li Y, Li H, Ren W, Zhao R, Xia C, Sun L. Production of monoclonal antibodies for measuring Avastin and its biosimilar by Sandwich ELISA. J Immunol Methods 2019; 469:42-46. [DOI: 10.1016/j.jim.2019.03.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 03/26/2019] [Accepted: 03/29/2019] [Indexed: 11/28/2022]
|
16
|
Peng T, Li Z, Li D, Wang S. MACC1 promotes angiogenesis in cholangiocarcinoma by upregulating VEGFA. Onco Targets Ther 2019; 12:1893-1903. [PMID: 30881041 PMCID: PMC6415730 DOI: 10.2147/ott.s197319] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Purpose Angiogenesis actively contributes to tumor growth and metastasis. MACC1 was reported to be associated with tumor progression. In the present study, we aimed to investigate the expression and role of MACC1 in cholangiocarcinoma (CCA) and its correlation with angiogenesis. Patients and methods We investigated the expression and correlation of MACC1 and VEGFA in The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets and in 7 paired frozen CCA and matched paracarcinoma tissues. Immunohistochemistry (IHC) was used to examine MACC1 and VEGFA expression as well as microvessel density (MVD) in 122 paraffin-embedded CCA samples. Western blotting, real-time qPCR and ELISA were performed to investigate the effect of MACC1 knockdown on VEGFA expression and secretion in CCA cells. Subsequently, we collected conditioned medium from cells with MACC1 knockdown and used it in angiogenesis assays. Results The expression levels of both MACC1 and VEGFA were significantly upregulated in the TCGA and GEO datasets and in the 7 paired frozen CCA tissues compared to the matched paracarcinoma tissues, and MACC1 was significantly correlated with VEGFA. IHC showed that high expression of MACC1 and VEGFA was significantly correlated with lymph node metastasis (P<0.05 and P<0.01) and worse survival (P<0.01, P<0.05) in patients with CCA. We further verified that MACC1 was significantly correlated with VEGFA (P<0.01) and MVD (P<0.01) in clinical samples. Western blotting, real-time qPCR and ELISA results showed that MACC1 knockdown in CCA cells significantly decreased the protein and mRNA expression of VEGFA and reduced the VEGFA concentration in conditioned medium. Moreover, angiogenesis assays showed that conditioned medium from CCA cells with MACC1 knockdown decreased the number of tubes formed. Conclusion Our results indicate that MACC1 and VEGFA expression are upregulated in CCA. Moreover, MACC1 is an independent predictor of overall survival and facilitates angiogenesis in CCA by upregulating of VEGFA.
Collapse
Affiliation(s)
- Tao Peng
- Department of Hepatobiliary Surgery, Southwest Hospital, Army Medical University, Chongqing, China, ; .,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Zhonghu Li
- Department of General Surgery, General Hospital of Wuhan, People's Liberation Army, Wuhan, China
| | - Dajiang Li
- Department of Hepatobiliary Surgery, Southwest Hospital, Army Medical University, Chongqing, China, ;
| | - Shuguang Wang
- Department of Hepatobiliary Surgery, Southwest Hospital, Army Medical University, Chongqing, China, ;
| |
Collapse
|
17
|
The prognostic relevance of urokinase-type plasminogen activator (uPA) in the blood of patients with metastatic breast cancer. Sci Rep 2019; 9:2318. [PMID: 30783124 PMCID: PMC6381129 DOI: 10.1038/s41598-018-37259-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 12/04/2018] [Indexed: 12/01/2022] Open
Abstract
In breast cancer (BC), elevated levels of urokinase-type plasminogen activator (uPA) in tumor tissue have been confirmed as a strong prognostic factor in level-of-evidence-1 studies. The aim of the present study was to evaluate the clinical relevance of uPA levels in serum of metastatic BC patients and to compare uPA with other blood-based biomarkers. 252 patients were enrolled in this prospective, multicentre study. Blood samples were collected before begin of first-line or later-line systemic treatment. Serum uPA was quantified by a commercially available ELISA. Circulating tumor cells (CTCs) were detected using CellSearch; other biomarkers (EGFR, VEGF, HER2, RAS p21, TIMP1, CAIX) by ELISA. Using the ROC analysis, the optimal cut-off value (determined by the Youden index) of serum uPA was 2.52 ng/ml. Using this value, 26% of patients had elevated uPA levels. Patients with visceral metastasis and more than one metastatic site were significantly more likely to present with elevated uPA levels. CTC status, serum HER2, RAS p21, CAIX, TIMP1 and VEGF correlated significantly with uPA levels. Elevated uPA levels predicted shorter overall and progression-free survival in univariate analysis (median OS: 7.5 months [95%-CI 4.5–10.5 months] vs. not reached, p < 0.001; PFS: 4.8 [95%-CI: 3.1–6.5] vs. 9.1 [7.4–10.8] months, p < 0.001). In multivariate analysis, elevated uPA, presence of ≥5 CTCs, elevated RAS p21, higher grading and higher line of therapy were independent predictors of shorter OS, while elevated CTC counts, higher line of therapy and negative estrogen receptor status were independent predictors of shorter PFS. In conclusion, elevated uPA levels independently predict reduced overall survival and improved prognostication in patients with known CTC status. Whether high serum uPA might identify patients most likely to benefit from therapies targeting uPA, remains to be evaluated in future trials.
Collapse
|
18
|
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
|
19
|
Wang S, Xiao Z, Hong Z, Jiao H, Zhu S, Zhao Y, Bi J, Qiu J, Zhang D, Yan J, Zhang L, Huang C, Li T, Liang L, Liao W, Ye Y, Ding Y. FOXF1 promotes angiogenesis and accelerates bevacizumab resistance in colorectal cancer by transcriptionally activating VEGFA. Cancer Lett 2018; 439:78-90. [PMID: 30253191 DOI: 10.1016/j.canlet.2018.09.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/17/2018] [Accepted: 09/19/2018] [Indexed: 10/28/2022]
Abstract
Forkhead box F1 (FOXF1) has been recently implicated in the progression and metastasis of lung cancer and breast cancer. However, the biological functions and underlying mechanisms by which FOXF1 regulates the progression of colorectal cancer (CRC) are largely unknown. As shown in our previous study, FOXF1 is upregulated in 182 CRC tissues, and elevated FOXF1 expression is significantly associated with microvessel density and advanced TNM (T = primary tumour; N = regional lymph nodes; M = distant metastasis) stages. In this study, 43 CRC tissues collected from patients who underwent treatment with first-line standard chemotherapeutic regimens in combination with bevacizumab were used to explore the correlation between FOXF1 expression and resistance to bevacizumab. In addition, FOXF1 regulated angiogenesis by inducing the transcription of vascular endothelial growth factor A1 (VEGFA) in vitro and in vivo. Furthermore, upregulation of FOXF1 enhanced bevacizumab resistance in CRC, and inhibition of VEGFA attenuated angiogenesis and bevacizumab resistance in FOXF1-overexpressing CRC cells. These results suggest that FOXF1 plays critical roles in CRC angiogenesis and bevacizumab resistance by inducing VEGFA transcription and that FOXF1 represents a potentially new therapeutic strategy and biomarker for anti-angiogenic therapy against CRC.
Collapse
Affiliation(s)
- Shuyang Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Zhiyuan Xiao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Zexuan Hong
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Hongli Jiao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Shaowei Zhu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Yali Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Jiaxin Bi
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Junfeng Qiu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Dan Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Junyu Yan
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Lingjie Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Chengmei Huang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Tingting Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Wenting Liao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China.
| | - Yaping Ye
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China.
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China.
| |
Collapse
|
20
|
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]
|
21
|
Luengo-Gil G, Gonzalez-Billalabeitia E, Perez-Henarejos SA, Navarro Manzano E, Chaves-Benito A, Garcia-Martinez E, Garcia-Garre E, Vicente V, Ayala de la Peña F. Angiogenic role of miR-20a in breast cancer. PLoS One 2018; 13:e0194638. [PMID: 29617404 PMCID: PMC5884522 DOI: 10.1371/journal.pone.0194638] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 03/07/2018] [Indexed: 01/02/2023] Open
Abstract
Background Angiogenesis is a key process for tumor progression and a target for treatment. However, the regulation of breast cancer angiogenesis and its relevance for clinical resistance to antiangiogenic drugs is still incompletely understood. Recent developments on the contribution of microRNA to tumor angiogenesis and on the oncogenic effects of miR-17-92, a miRNA cluster, point to their potential role on breast cancer angiogenesis. The aim of this work was to establish the contribution of miR-20a, a member of miR-17-92 cluster, to tumor angiogenesis in patients with invasive breast carcinoma. Methods Tube-formation in vitro assays with conditioned medium from MCF7 and MDA-MB-231 breast cancer cell lines were performed after transfection with miR-20a and anti-miR20a. For clinical validation of the experimental findings, we performed a retrospective analysis of a series of consecutive breast cancer patients (n = 108) treated with neoadjuvant chemotherapy and with a full characterization of their vessel pattern and expression of angiogenic markers in pre-treatment biopsies. Expression of members of the cluster miR-17-92 and of angiogenic markers was determined by RT-qPCR after RNA purification from FFPE samples. Results In vitro angiogenesis assays with endothelial cells and conditioned media from breast cancer cell lines showed that transfection with anti-miR20a in MDA-MB-231 significantly decreased mean mesh size and total mesh area, while transfection with miR-20a in MCF7 cells increased mean mesh size. MiR-20a angiogenic effects were abrogated by treatment with aflibercept, a VEGF trap. These results were supported by clinical data showing that mir-20a expression was higher in tumors with no estrogen receptor or with more extensive nodal involvement (cN2-3). A higher miR-20a expression was associated with higher mean vessel size (p = 0.015) and with an angiogenic pattern consisting in larger vessels, higher VEGFA expression and presence of glomeruloid microvascular proliferations (p<0.001). This association was independent of tumor subtype and VEGFA expression. Conclusions Transfection of breast cancer cells with miR-20a induces vascular changes in endothelial tube-formation assays. Expression of miR-20a in breast invasive carcinomas is associated with a distinctive angiogenic pattern consisting in large vessels, anomalous glomeruloid microvascular proliferations and high VEGFA expression. Our results suggest a role for miR-20a in the regulation of breast cancer angiogenesis, and raise the possibility of its use as an angiogenic biomarker.
Collapse
Affiliation(s)
- Gines Luengo-Gil
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer y Centro Regional de Hemodonación, Murcia, Spain
- Department of Internal Medicine, University of Murcia, Murcia, Spain
- IMIB-Arrixaca, Murcia, Spain
| | - Enrique Gonzalez-Billalabeitia
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer y Centro Regional de Hemodonación, Murcia, Spain
- IMIB-Arrixaca, Murcia, Spain
- Universidad Católica San Antonio de Murcia (UCAM), Murcia, Spain
| | - Sergio Alejo Perez-Henarejos
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer y Centro Regional de Hemodonación, Murcia, Spain
- IMIB-Arrixaca, Murcia, Spain
| | - Esther Navarro Manzano
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer y Centro Regional de Hemodonación, Murcia, Spain
- IMIB-Arrixaca, Murcia, Spain
| | | | - Elena Garcia-Martinez
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer y Centro Regional de Hemodonación, Murcia, Spain
- IMIB-Arrixaca, Murcia, Spain
- Universidad Católica San Antonio de Murcia (UCAM), Murcia, Spain
| | - Elisa Garcia-Garre
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer y Centro Regional de Hemodonación, Murcia, Spain
- IMIB-Arrixaca, Murcia, Spain
| | - Vicente Vicente
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer y Centro Regional de Hemodonación, Murcia, Spain
- Department of Internal Medicine, University of Murcia, Murcia, Spain
- IMIB-Arrixaca, Murcia, Spain
| | - Francisco Ayala de la Peña
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer y Centro Regional de Hemodonación, Murcia, Spain
- Department of Internal Medicine, University of Murcia, Murcia, Spain
- IMIB-Arrixaca, Murcia, Spain
- * E-mail:
| |
Collapse
|
22
|
Mendiola M, Martínez-Marin V, Herranz J, Heredia V, Yébenes L, Zamora P, Castelo B, Pinto Á, Miguel M, Díaz E, Gámez A, Fresno JÁ, Ramírez de Molina A, Hardisson D, Espinosa E, Redondo A. Predictive value of angiogenesis-related gene profiling in patients with HER2-negative metastatic breast cancer treated with bevacizumab and weekly paclitaxel. Oncotarget 2018; 7:24217-27. [PMID: 26992213 PMCID: PMC5029696 DOI: 10.18632/oncotarget.8128] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 02/25/2016] [Indexed: 01/15/2023] Open
Abstract
Bevacizumab plus weekly paclitaxel improves progression-free survival (PFS) in HER2-negative metastatic breast cancer (mBC), but its use has been questioned due to the absence of a predictive biomarker, lack of benefit in overall survival (OS) and increased toxicity. We examined the baseline tumor angiogenic-related gene expression of 60 patients with mBC with the aim of finding a signature that predicts benefit from this drug. Multivariate analysis by Lasso-penalized Cox regression generated two predictive models: one, named G-model, including 11 genes, and the other one, named GC-model, including 13 genes plus 5 clinical covariates. Both models identified patients with improved PFS (HR (Hazard Ratio) 2.57 and 4.04, respectively) and OS (HR 3.29 and 3.43, respectively). The G-model distinguished low and high risk patients in the first 6 months, whereas the GC-model maintained significance over time.
Collapse
Affiliation(s)
- Marta Mendiola
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital - IdiPAZ, Madrid, Spain
| | - Virginia Martínez-Marin
- Department of Medical Oncology, La Paz University Hospital, Madrid, Spain.,Translational Oncology Group, La Paz University Hospital - IdiPAZ, Madrid, Spain
| | - Jesús Herranz
- IMDEA, Campus de Excelencia Internacional CEI (UAM-CSIC), Madrid, Spain
| | - Victoria Heredia
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital - IdiPAZ, Madrid, Spain
| | - Laura Yébenes
- Department of Pathology, La Paz University Hospital, Madrid, Spain
| | - Pilar Zamora
- Department of Medical Oncology, La Paz University Hospital, Madrid, Spain.,Translational Oncology Group, La Paz University Hospital - IdiPAZ, Madrid, Spain
| | - Beatriz Castelo
- Department of Medical Oncology, La Paz University Hospital, Madrid, Spain.,Translational Oncology Group, La Paz University Hospital - IdiPAZ, Madrid, Spain
| | - Álvaro Pinto
- Department of Medical Oncology, La Paz University Hospital, Madrid, Spain.,Translational Oncology Group, La Paz University Hospital - IdiPAZ, Madrid, Spain
| | - María Miguel
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital - IdiPAZ, Madrid, Spain
| | - Esther Díaz
- Translational Oncology Group, La Paz University Hospital - IdiPAZ, Madrid, Spain
| | - Angelo Gámez
- Translational Oncology Group, La Paz University Hospital - IdiPAZ, Madrid, Spain
| | - Juan Ángel Fresno
- Translational Oncology Group, La Paz University Hospital - IdiPAZ, Madrid, Spain
| | | | - David Hardisson
- Molecular Pathology and Therapeutic Targets Group, La Paz University Hospital - IdiPAZ, Madrid, Spain.,Department of Pathology, La Paz University Hospital, Madrid, Spain
| | - Enrique Espinosa
- Department of Medical Oncology, La Paz University Hospital, Madrid, Spain.,Translational Oncology Group, La Paz University Hospital - IdiPAZ, Madrid, Spain
| | - Andrés Redondo
- Department of Medical Oncology, La Paz University Hospital, Madrid, Spain.,Translational Oncology Group, La Paz University Hospital - IdiPAZ, Madrid, Spain
| |
Collapse
|
23
|
García Garre E, Luengo Gil G, Montoro García S, Gonzalez Billalabeitia E, Zafra Poves M, García Martinez E, Roldán Schilling V, Navarro Manzano E, Ivars Rubio A, Lip GYH, Ayala de la Peña F. Circulating small-sized endothelial microparticles as predictors of clinical outcome after chemotherapy for breast cancer: an exploratory analysis. Breast Cancer Res Treat 2018; 169:83-92. [PMID: 29340882 DOI: 10.1007/s10549-017-4656-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 12/30/2017] [Indexed: 01/22/2023]
Abstract
PURPOSE Therapeutic exploitation of angiogenesis in breast cancer has been limited by the lack of reliable biomarkers. Circulating small-sized endothelial microparticles (sEMP) are likely to play a significant role as messengers of angiogenesis. Higher levels of EMP have been observed in cancer patients, but their prognostic value in breast cancer is unknown. Our aim was to determine the value of circulating sEMP as a marker of response to chemotherapy in breast cancer. METHODS We included patients with breast cancer treated with neoadjuvant or first-line chemotherapy. Baseline and post-treatment circulating sEMP (CD144+) were quantified using a flow cytometer approach specifically designed for analysis of small-sized particles (0.1-0.5 μm). Small-sized EMP response was defined as a post-treatment decrease of sEMP larger than the median decrease of sEMP after chemotherapy. Baseline and post-chemotherapy VEGFA levels were determined with ELISA. RESULTS Forty-four breast cancer patients were included (19 with metastatic and 25 with locally advanced disease). Median levels of sEMP decreased after chemotherapy (P = 0.005). Response to chemotherapy showed a non-significant trend to associate with sEMP response (P = 0.056). A sEMP response was observed in 51% of patients and was associated with better overall survival (HR 0.18; 95% CI 0.04-0.87; P = 0.02) and progression free survival (HR 0.30; 95% CI 0.09-0.99; P = 0.04) in the group of women with metastatic disease. Post-chemotherapy decrease of VEGFA levels was not associated with breast cancer prognosis. CONCLUSIONS Our results did not support sEMP as a marker of response to chemotherapy. However, our exploratory analysis suggests that in patients with metastatic breast cancer, the decrease of sEMP levels after chemotherapy is associated with better overall and disease free survival and might be superior to VEGFA levels as an angiogenesis-related prognostic marker.
Collapse
Affiliation(s)
- Elisa García Garre
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Avda. Marqués de los Velez, s/n, 30008, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain
| | - Ginés Luengo Gil
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Avda. Marqués de los Velez, s/n, 30008, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain.,Universidad de Murcia, Murcia, Spain
| | - Silvia Montoro García
- Universidad Católica San Antonio de Murcia, Guadalupe, Murcia, Spain.,Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Enrique Gonzalez Billalabeitia
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Avda. Marqués de los Velez, s/n, 30008, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain.,Universidad Católica San Antonio de Murcia, Guadalupe, Murcia, Spain
| | - Marta Zafra Poves
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Avda. Marqués de los Velez, s/n, 30008, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain
| | - Elena García Martinez
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Avda. Marqués de los Velez, s/n, 30008, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain.,Universidad Católica San Antonio de Murcia, Guadalupe, Murcia, Spain
| | - Vanessa Roldán Schilling
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Avda. Marqués de los Velez, s/n, 30008, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain.,Universidad de Murcia, Murcia, Spain
| | - Esther Navarro Manzano
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Avda. Marqués de los Velez, s/n, 30008, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain.,Universidad de Murcia, Murcia, Spain
| | - Alejandra Ivars Rubio
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Avda. Marqués de los Velez, s/n, 30008, Murcia, Spain.,Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain
| | - Gregory Y H Lip
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Francisco Ayala de la Peña
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Avda. Marqués de los Velez, s/n, 30008, Murcia, Spain. .,Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain. .,Universidad de Murcia, Murcia, Spain.
| |
Collapse
|
24
|
Qu Y, Liu H, Lv X, Liu Y, Wang X, Zhang M, Zhang X, Li Y, Lou Q, Li S, Li H. MicroRNA-16-5p overexpression suppresses proliferation and invasion as well as triggers apoptosis by targeting VEGFA expression in breast carcinoma. Oncotarget 2017; 8:72400-72410. [PMID: 29069797 PMCID: PMC5641140 DOI: 10.18632/oncotarget.20398] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/07/2017] [Indexed: 01/08/2023] Open
Abstract
MicroRNAs (miRNAs), a class of small noncoding RNA molecules, can manipulate the expressions of endogenous tumor-related genes, and are implicated in the development and progression of a wide type of tumors. In this study, the investigation from real-time quantitative PCR revealed that miRNA-16-5p was downregulated in breast carcinoma tissues and cells, coupled with the elevations of HIF-α and VEGFA protein expressions, compared with normal tissues. Lentiviral armed with miR-16-5p markedly increased the miR-16-5p levels in MCF-7 and MDA-MB-231 cells, compared to blank and NC groups, and miR-16-5p overexpression significantly inhibited the proliferation and colony formation in MCF-7 and MDA-MB-231 cells. Besides, miR-16-5p upregulation markedly induced apoptosis and reduced invasion ability in MCF-7 and MDA-MB-231 cells. Notably, VEGFA was direct target of miR-16-5p. Stepwise investigation from in vitro and in vivo experiments demonstrated that miR-16-5p overexpression suppressed tumor growth and reduced HIF-α and VEGFA expressions in breast carcinoma cells and nude mice tumor tissues. These findings provide novel insights into molecular mechanism involved in the roles of miR-16-5p in tumor development and progression of breast carcinoma, and thus manipulation of miR-16-5p may be a novel potential therapeutic target for future therapies of the patients with breast carcinoma.
Collapse
Affiliation(s)
- Yunhui Qu
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University and The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, P.R. China.,Clinical Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, P.R. China
| | - Hongtao Liu
- Laboratory for Cell Biology, College of Life Sciences of Zhengzhou University, Zhengzhou, 450001, Henan, P.R. China
| | - Xinquan Lv
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University and The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, P.R. China
| | - Yuqiong Liu
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University and The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, P.R. China
| | - Xiaojuan Wang
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University and The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, P.R. China
| | - Min Zhang
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University and The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, P.R. China
| | - Xiaqing Zhang
- Laboratory for Cell Biology, College of Life Sciences of Zhengzhou University, Zhengzhou, 450001, Henan, P.R. China
| | - Yuenan Li
- Laboratory for Cell Biology, College of Life Sciences of Zhengzhou University, Zhengzhou, 450001, Henan, P.R. China
| | - Qianqian Lou
- Laboratory for Cell Biology, College of Life Sciences of Zhengzhou University, Zhengzhou, 450001, Henan, P.R. China
| | - Shenglei Li
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University and The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, P.R. China
| | - Huixiang Li
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University and The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, P.R. China
| |
Collapse
|
25
|
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
|
26
|
English WR, Lunt SJ, Fisher M, Lefley DV, Dhingra M, Lee YC, Bingham K, Hurrell JE, Lyons SK, Kanthou C, Tozer GM. Differential Expression of VEGFA Isoforms Regulates Metastasis and Response to Anti-VEGFA Therapy in Sarcoma. Cancer Res 2017; 77:2633-2646. [PMID: 28377452 DOI: 10.1158/0008-5472.can-16-0255] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/13/2016] [Accepted: 03/09/2017] [Indexed: 11/16/2022]
Abstract
Elevated plasma concentrations of soluble VEGFA isoforms are associated with poor prognosis in parallel with improved response to treatment with the anti-VEGFA antibody bevacizumab. To uncover the underlying mechanism to these observations, we administered anti-VEGFA therapy to mice bearing luminescent mouse fibrosarcomas expressing single VEGFA isoforms or their wild-type counterparts expressing all isoforms (fs120, fs164, fs188, or fsWT). Expression of the more soluble isoforms conferred an advantage for lung metastasis from subcutaneous tumors (fs120/164 vs. fs188/WT); fs120 cells also produced more lung colonies than fs188 cells when injected intravenously. Metastasis from subcutaneous fs120 tumors was more sensitive than fs188 to treatment with the anti-VEGFA antibody B20-4.1.1. Despite elevated plasma levels of VEGFA in fs120 tumor-bearing mice and a dependence on VEGF receptor 1 activity for metastasis to the lung, B20-4.1.1 did not affect survival in the lung on intravenous injection. B20-4.1.1 inhibited subcutaneous tumor growth and decreased vascular density in both fs120 and fs188 tumors. However, migration of fs120, but not fs188 cells, in vitro was inhibited by B20-4.1.1. The greater survival of fs120 cells in the lung was associated with VEGFR1-dependent accumulation of CD11b-positive myeloid cells and higher expression of the VEGFR1 ligand, PlGF2, by the fs120 cells in vitro and in the plasma and lungs of fs120 tumor-bearing mice. We conclude that soluble VEGFA isoform expression increases fibrosarcoma metastasis through multiple mechanisms that vary in their sensitivity to anti-VEGF/VEGFR inhibition, with VEGFA-targeted therapy suppressing metastasis through effects on the primary tumor rather than the metastatic site. Cancer Res; 77(10); 2633-46. ©2017 AACR.
Collapse
Affiliation(s)
- William R English
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom.
| | - Sarah Jane Lunt
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Matthew Fisher
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Diane V Lefley
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Mohit Dhingra
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Yu-Chin Lee
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Karina Bingham
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Jack E Hurrell
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Scott K Lyons
- CR-UK Cambridge Institute, The Li Ka Shing Building, Cambridge, United Kingdom
| | - Chryso Kanthou
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| | - Gillian M Tozer
- Tumor Microcirculation Group, Department of Oncology and Metabolism, University of Sheffield School of Medicine, Beech Hill Road, Sheffield, United Kingdom
| |
Collapse
|
27
|
Rai S, Nejadhamzeeigilani Z, Gutowski NJ, Whatmore JL. Loss of the endothelial glycocalyx is associated with increased E-selectin mediated adhesion of lung tumour cells to the brain microvascular endothelium. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:105. [PMID: 26407999 PMCID: PMC4582832 DOI: 10.1186/s13046-015-0223-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/14/2015] [Indexed: 12/21/2022]
Abstract
Background Arrest of metastasising lung cancer cells to the brain microvasculature maybe mediated by interactions between ligands on circulating tumour cells and endothelial E-selectin adhesion molecules; a process likely to be regulated by the endothelial glycocalyx. Using human cerebral microvascular endothelial cells and non-small cell lung cancer (NSCLC) cell lines, we describe how factors secreted by NSCLC cells i.e. cystatin C, cathepsin L, insulin-like growth factor-binding protein 7 (IGFBP7), vascular endothelial growth factor (VEGF) and tumour necrosis factor-alpha (TNF-α), damage the glycocalyx and enhance initial contacts between lung tumour and cerebral endothelial cells. Methods Endothelial cells were treated with tumour secreted-proteins or lung tumour conditioned medium (CM). Surface levels of E-selectin were quantified by ELISA. Adhesion of A549 and SK-MES-1 cells was examined under flow conditions (1 dyne/cm2). Alterations in the endothelial glycocalyx were quantified by binding of fluorescein isothiocyanate-linked wheat germ agglutinin (WGA-FITC). Results A549 and SK-MES-1 CM and secreted-proteins significantly enhanced endothelial surface E-selectin levels after 30 min and 4 h and tumour cell adhesion after 30 min, 4 and 24 h. Both coincided with significant glycocalyx degradation; A549 and SK-MES-1 CM removing 55 ± 12 % and 58 ± 18.7 % of WGA-FITC binding, respectively. Inhibition of E-selectin binding by monoclonal anti-E-selectin antibody completely attenuated tumour cell adhesion. Conclusion These data suggest that metastasising lung cancer cells facilitate their own adhesion to the brain endothelium by secreting factors that damage the endothelial glycocalyx, resulting in exposure of the previously shielded adhesion molecules and engagement of the E-selectin-mediated adhesion axis. Electronic supplementary material The online version of this article (doi:10.1186/s13046-015-0223-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Srijana Rai
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, St. Luke's campus, Exeter, EX1 2LU, UK.
| | - Zaynab Nejadhamzeeigilani
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, St. Luke's campus, Exeter, EX1 2LU, UK.
| | - Nicholas J Gutowski
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, St. Luke's campus, Exeter, EX1 2LU, UK. .,Royal Devon and Exeter NHS Foundation Trust, Barrack road, Exeter, EX2 5DW, UK.
| | - Jacqueline L Whatmore
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, St. Luke's campus, Exeter, EX1 2LU, UK.
| |
Collapse
|