1
|
Li W, Zhang K, Wang W, Liu Y, Huang J, Zheng M, Li L, Zhang X, Xu M, Chen G, Wang L, Zhang S. Combined inhibition of HER2 and VEGFR synergistically improves therapeutic efficacy via PI3K-AKT pathway in advanced ovarian cancer. J Exp Clin Cancer Res 2024; 43:56. [PMID: 38403634 PMCID: PMC10895844 DOI: 10.1186/s13046-024-02981-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 02/11/2024] [Indexed: 02/27/2024] Open
Abstract
BACKGROUND Ovarian cancer (OC) is a prevalent malignancy in the female reproductive system, and developing effective targeted therapies for this disease remains challenging. The aim of this study was to use clinically-relevant OC models to evaluate the therapeutic effectiveness of RC48, an antibody-drug conjugate (ADC) targeting HER2, either alone or in combination with the VEGFR inhibitor Cediranib Maleate (CM), for the treatment of advanced OC. METHODS OC tumor specimens and cell lines were analyzed to determine HER2 and VEGFR expression by Western blot, immunocytochemistry and immunofluorescence. Moreover, the OC cell lines, cell-derived xenograft (CDX) and patient-derived xenograft (PDX) models were treated with RC48 and/or CM and then subjected to cell proliferation, viability, apoptosis, and tumor growth analyses to evaluate the feasibility of combination therapy for OC both in vitro and in vivo. Additionally, RNA-Seq was performed to investigate the critical mechanism underlying the combination therapy of RC48 and CM. RESULTS Our results demonstrated that RC48 alone effectively targeted and inhibited the growth of HER2-positive OC tumors in both cell lines and PDX models. Furthermore, the combination of RC48 and CM synergistically induced tumor regression in human OC cell lines, as well as CDX and PDX models. Mechanistically, we observed that the combination treatment inhibited the growth of OC cells involved inducing apoptosis and suppressing cell motility. RNA-seq analysis provided further mechanistic insights and revealed that co-administration of RC48 and CM downregulated multiple cancer-related pathways, including the AKT/mTOR pathway, cell cycle, and cell proliferation. Notably, our data further confirmed that the PI3K-AKT pathway played a key role in the inhibition of proliferation triggered by combinational treatment of RC48 and CM in OC cells. CONCLUSIONS These findings provide a preclinical framework supporting the potential of dual targeting HER2 and VEGFR as a promising therapeutic strategy to improve outcomes in patients with OC.
Collapse
Affiliation(s)
- Weisong Li
- Department of Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
| | - Kai Zhang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, China
| | - Wenjun Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Liu
- Department of Gynaecology and Obstetrics, Ganzhou People's Hospital (The Affiliated Ganzhou Hospital of Nanchang University), Ganzhou, 341000, China
| | - Jianming Huang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, China
| | - Meihong Zheng
- Department of Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
| | - Ling Li
- Department of Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
| | - Xinyu Zhang
- Department of Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, 341000, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China
| | - Minjuan Xu
- Department of Gynaecology and Obstetrics, Ganzhou People's Hospital (The Affiliated Ganzhou Hospital of Nanchang University), Ganzhou, 341000, China.
| | - Guofang Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Tongji University, Shanghai, 200092, China.
| | - Liefeng Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China.
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, China.
| | - Shuyong Zhang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, 1 Hexie Road, Rongjiang New District, Ganzhou, 341000, China.
- School of Basic Medicine, Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
2
|
Duan Y, Zhang P, Zhang T, Zhou L, Yin R. Characterization of global research trends and prospects on platinum-resistant ovarian cancer: a bibliometric analysis. Front Oncol 2023; 13:1151871. [PMID: 37342181 PMCID: PMC10277726 DOI: 10.3389/fonc.2023.1151871] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/22/2023] [Indexed: 06/22/2023] Open
Abstract
Background In the last decades, growing attention has been focused on identifying effective therapeutic strategies in the orphan clinical setting of women with platinum-resistant ovarian cancer (PROC), generating thousands of original articles. However, the literature involving bibliometric analysis of PROC has not been published yet. Objective This study hopes to gain a better understanding of the hot spots and trends in PROC by conducting a bibliometric analysis, as well as identify potential new research directions. Methods We searched the Web of Science Core Collection (WOSCC) for PROC-related articles published between 1990 and 2022. CiteSpace 6.1.R2 and VOS viewer 1.6.18.0 were primarily utilized to evaluate the contribution and co-occurrence relationships of various countries and regions, institutes, and journals and to identify research hotspots and promising future trends in this research field. Results A total of 3,462 Web of Science publications were retrieved that were published in 671 academic journals by 1135 authors from 844 organizations in 75 countries and regions. The United States was the leading contributor in this field, and the University of Texas MD Anderson Cancer Center was the most productive institution. Gynecologic Oncology was the most productive journal, while the Journal of Clinical Oncology was the most cited and influential. Co-citation cluster labels revealed the characteristics of seven major clusters, including synthetic lethality, salvage treatment, human ovarian-carcinoma cell line, PARP inhibitor resistance, antitumor complexes, folate receptor, and targeting platinum-resistant disease. Keywords and references burst detection indicated that biomarkers, genetic and phenotypic changes, immunotherapy, and targeted therapy were the most recent and most significant aspects of PROC research. Conclusion This study conducted a comprehensive review of PROC research using bibliometric and visual techniques. Understanding the immunological landscape of PROC and identifying the population that can benefit from immunotherapy, especially in combination with other therapeutic options (such as chemotherapy and targeted therapy), will continue to be the focal point of research.
Collapse
Affiliation(s)
- Yuanqiong Duan
- Department of Obstetrics and Gynecology, West China Second Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Peixuan Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Tianyue Zhang
- Department of Obstetrics and Gynecology, West China Second Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Lu Zhou
- Department of Obstetrics and Gynecology, West China Second Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Rutie Yin
- Department of Obstetrics and Gynecology, West China Second Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Hu X, Li D, Fu Y, Zheng J, Feng Z, Cai J, Wang P. Advances in the Application of Radionuclide-Labeled HER2 Affibody for the Diagnosis and Treatment of Ovarian Cancer. Front Oncol 2022; 12:917439. [PMID: 35785201 PMCID: PMC9240272 DOI: 10.3389/fonc.2022.917439] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/20/2022] [Indexed: 12/19/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is a highly expressed tumor marker in epithelial ovarian cancer, and its overexpression is considered to be a potential factor of poor prognosis. Therefore, monitoring the expression of HER2 receptor in tumor tissue provides favorable conditions for accurate localization, diagnosis, targeted therapy, and prognosis evaluation of cancer foci. Affibody has the advantages of high affinity, small molecular weight, and stable biochemical properties. The molecular probes of radionuclide-labeled HER2 affibody have recently shown broad application prospects in the diagnosis and treatment of ovarian cancer; the aim is to introduce radionuclides into the cancer foci, display systemic lesions, and kill tumor cells through the radioactivity of the radionuclides. This process seamlessly integrates the diagnosis and treatment of ovarian cancer. Current research and development of new molecular probes of radionuclide-labeled HER2 affibody should focus on overcoming the deficiencies of non-specific uptake in the kidney, bone marrow, liver, and gastrointestinal tract, and on reducing the background of the image to improve image quality. By modifying the amino acid sequence; changing the hydrophilicity, surface charge, and lipid solubility of the affibody molecule; and using different radionuclides, chelating agents, and labeling conditions to optimize the labeling method of molecular probes, the specific uptake of molecular probes at tumor sites will be improved, while reducing radioactive retention in non-target organs and obtaining the best target/non-target value. These measures will enable the clinical use of radionuclide-labeled HER2 affibody molecular probes as soon as possible, providing a new clinical path for tumor-specific diagnosis, targeted therapy, and efficacy evaluation. The purpose of this review is to describe the application of radionuclide-labeled HER2 affibody in the imaging and treatment of ovarian cancer, including its potential clinical value and dilemmas.
Collapse
Affiliation(s)
- Xianwen Hu
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dandan Li
- Department of Obstetrics, Zunyi Hospital of Traditional Chinese Medicine, Zunyi, China
| | - Yujie Fu
- Research and Development Department, Jiangsu Yuanben Biotechnology Co., Ltd., Zunyi, China
| | - Jiashen Zheng
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zelong Feng
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiong Cai
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Jiong Cai, ; Pan Wang,
| | - Pan Wang
- Department of Nuclear Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Jiong Cai, ; Pan Wang,
| |
Collapse
|
4
|
Chilimoniuk Z, Rocka A, Stefaniak M, Tomczyk Ż, Jasielska F, Madras D, Filip A. Molecular methods for increasing the effectiveness of ovarian cancer treatment: a systematic review. Future Oncol 2022; 18:1627-1650. [PMID: 35129396 DOI: 10.2217/fon-2021-0565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: The aim of the current study is to analyze and summarize the latest research on improving therapy in ovarian cancer. Materials & methods: Data analysis was based on a review of publications from 2011 to 2021 in the PubMed database with use of the search terms including 'EGFR ovarian cancer', 'folate receptor inhibitors ovarian cancer', 'VEGF ovarian cancer', 'PDGF ovarian cancer' and 'CTLA-4 ovarian cancer'. Results: 6643 articles were found; 238 clinical trials and randomized control trials were analyzed; 122 studies were rejected due to inconsistency with the topic of the work. Conclusion: Extensive research on the treatment of ovarian cancer increases the chance of developing the most effective therapy suited to the individual needs of the patient.
Collapse
Affiliation(s)
- Zuzanna Chilimoniuk
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Agata Rocka
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Martyna Stefaniak
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Żaklina Tomczyk
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Faustyna Jasielska
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Dominika Madras
- Students' Scientific Association at the Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| | - Agata Filip
- Department of Cancer Genetics with Cytogenetics Laboratory, Medical University of Lublin, ul. Radziwiłłowska 11, Lublin, 20-080, Poland
| |
Collapse
|
5
|
Huang Y, Xu J, Li K, Wang J, Dai Y, Kang Y. A Novel, Personalized Drug-Screening System for Platinum-Resistant Ovarian Cancer Patients: A Preliminary Clinical Report. Cancer Manag Res 2021; 13:2849-2867. [PMID: 33833569 PMCID: PMC8020460 DOI: 10.2147/cmar.s276799] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 03/11/2021] [Indexed: 12/18/2022] Open
Abstract
PURPOSE With this study, we intended to construct a personalized drug-screening system for platinum-resistant ovarian cancer patients by consulting a patient's medical history, data derived from gene mutation detection, and drug screening results derived from mini-PDX (patient-derived xenograft) models. We also aimed to evaluate the efficacy and safety of our system. PATIENTS AND METHODS We selected 12 patients with platinum-resistant ovarian cancer who were treated at our hospital from January 2018 to December 2019 to design a single-arm clinical trial. The subsequent chemotherapeutic plans were selected according to a personalized drug-screening system that circulating tumor DNA (ctDNA) testing and the establishment of mini-PDX models. We then analyzed the patients for clinical benefits side-effects in response to chemotherapy in order to evaluate the clinical effects and safety of our new personalized drug-selection system. RESULTS We successfully established an individualized and sensitive drug-screening system for the 12 patients. Mini-PDX models verified that potentially effective drugs were identified for 11 of the patients. Treatment resulted in complete remission (one patient), partial remission (five patients), and stable disease (three patients). The remaining three patients experienced disease progression. The overall clinical-benefit rate was 75.0%. Following treatment, the levels of CA125 levels decreased significantly in seven of the 12 patients. Severe side effects, arising from chemotherapy, were only observed in one case. CONCLUSION Constructing a personalized drug-screening system for platinum-resistant ovarian cancer patients can be used to guide clinical drug selection and improve the clinical-benefit rate for patients. TRIAL REGISTRATION NUMBER ChiCTR1800016766 (Chinese Clinical Trial Registry Center).
Collapse
Affiliation(s)
- Yunke Huang
- The Obstetrics & Gynecology Hospital of Fudan University, Gynecology Department, Shanghai, People’s Republic of China
| | - Jing Xu
- The Obstetrics & Gynecology Hospital of Fudan University, Gynecology Department, Shanghai, People’s Republic of China
| | - Ke Li
- The Obstetrics & Gynecology Hospital of Fudan University, Gynecology Department, Shanghai, People’s Republic of China
| | - Jing Wang
- The Obstetrics & Gynecology Hospital of Fudan University, Gynecology Department, Shanghai, People’s Republic of China
| | - Yilin Dai
- The Obstetrics & Gynecology Hospital of Fudan University, Gynecology Department, Shanghai, People’s Republic of China
| | - Yu Kang
- The Obstetrics & Gynecology Hospital of Fudan University, Gynecology Department, Shanghai, People’s Republic of China
| |
Collapse
|
6
|
Chelariu-Raicu A, Levenback CF, Slomovitz BM, Wolf J, Bodurka DC, Kavanagh JJ, Morrison C, Gershenson DM, Coleman RL. Phase Ib/II study of weekly topotecan and daily gefitinib in patients with platinum resistant ovarian, peritoneal, or fallopian tube cancer. Int J Gynecol Cancer 2020; 30:1768-1774. [PMID: 33037105 DOI: 10.1136/ijgc-2020-001863] [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: 07/16/2020] [Revised: 08/17/2020] [Accepted: 08/25/2020] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION 50-70% of epithelial ovarian cancers overexpress epidermal growth factor receptor, and its expression has been correlated with poor prognosis. We conducted a phase Ib/II trial to examine the efficacy, safety, and toxicity of gefitinib, a tyrosine kinase inhibitor, combined with topotecan in women with recurrent ovarian cancer with epidermal growth factor receptor positivity. METHODS Patients with measurable recurrent or persistent cancer after treatment with a platinum containing regimen with positive epidermal growth factor receptor expression, as determined by immunohistochemistry, were eligible for the study. Initial treatment was 250 mg/day gefitinib (oral) and 2.0 mg/m2 topotecan (intravenous) on days 1, 8, and 15, on a 28 day cycle. Dose escalations were planned for topotecan (dose levels 1-3: 2, 3, and 4 mg/m2) until the maximum tolerated dose was reached. RESULTS 19 patients received a total of 61 cycles. Median age was 59.8 years (range 42-76 years). Histologic types in treated patients included 74% serous (n=14), 11% mixed (n=2), 11% transitional (n=2), and 5% clear cell (n=1). For phase Ib, three patients were treated at dose level 1, three at dose level 2, and three at dose level 3 for topotecan. The maximum tolerated dose was 4.0 mg/m2 (days 1, 8, and 15) for topotecan and 250 mg (daily) for gefitinib. Therefore, dose level 3 was used for phase II. Among the 19 patients, 63.2% (n=12) had progressive disease, 15.8% (n=3) had stable disease, 10.5% (n=2) had a partial response, and 10.5% (n=2) were not evaluable. The most serious adverse events of any grade attributed to the therapy were anemia (89.4%), neutropenia (68.4%), abdominal pain (84%), constipation (78.9%), and diarrhea (78.9%). CONCLUSION Although the drug combination was relatively well tolerated, this prospective phase Ib/II clinical trial did not show sufficient clinical activity of topotecan combined with gefitinib in patients with epidermal growth factor receptor positive recurrent ovarian, fallopian tube, or peritoneal cancers.
Collapse
Affiliation(s)
- Anca Chelariu-Raicu
- Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Charles F Levenback
- Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Brian M Slomovitz
- Division of Gynecologic Oncology, Broward Health Medical Center, Fort Lauderdale, Florida, USA
| | - Judith Wolf
- Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Diane C Bodurka
- Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - John J Kavanagh
- Department of Obstetrics and Gynecology, Chulalongkorn University, Bangkok, Thailand
| | | | - David M Gershenson
- Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Robert L Coleman
- Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA .,The US Oncology Network, The Woodlands, Texas, USA
| |
Collapse
|
7
|
Silbermann K, Li J, Namasivayam V, Baltes F, Bendas G, Stefan SM, Wiese M. Superior Pyrimidine Derivatives as Selective ABCG2 Inhibitors and Broad-Spectrum ABCB1, ABCC1, and ABCG2 Antagonists. J Med Chem 2020; 63:10412-10432. [PMID: 32787102 DOI: 10.1021/acs.jmedchem.0c00961] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the search for highly effective modulators addressing ABCG2-mediated MDR, 23 pyrimidines were synthesized and biologically assessed. Seven derivatives with (a) nitrogen- and/or halogen-containing residue(s) had extraordinary potencies against ABCG2 (IC50 < 150 nM). The compounds competitively inhibited ABCG2-mediated Hoechst 33342 transport but were not substrates of ABCG2. The most potent MDR reverser, compound 19, concentration-dependently increased SN-38-mediated cancer cell death at 11 nM (EC50), time-dependently doubled SN-38 toxicity in a period of 7 days at 10 nM, and half-maximally accelerated cell death combined with SN-38 at 17 nM. No induction of ABCG2 was observed. Furthermore, 11 pyrimidines were revealed as triple ABCB1/ABCC1/ABCG2 inhibitors. Five possessed IC50 values below 10 μM against each transporter, classifying them as some of the 50 most potent multitarget ABC transporter inhibitors. The most promising representative, compound 37, reversed ABCB1-, ABCC1-, and ABCG2-mediated MDR, making it one of the three most potent ABC transporter inhibitors and reversers of ABC transporters-mediated MDR.
Collapse
Affiliation(s)
- Katja Silbermann
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Jiyang Li
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Vigneshwaran Namasivayam
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Fabian Baltes
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Gerd Bendas
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Sven Marcel Stefan
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Michael Wiese
- Pharmaceutical and Cellbiological Chemistry, Pharmaceutical Institute, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121 Bonn, Germany
| |
Collapse
|
8
|
Wang Q, Peng H, Qi X, Wu M, Zhao X. Targeted therapies in gynecological cancers: a comprehensive review of clinical evidence. Signal Transduct Target Ther 2020; 5:137. [PMID: 32728057 PMCID: PMC7391668 DOI: 10.1038/s41392-020-0199-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/12/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
Advanced and recurrent gynecological cancers are associated with poor prognosis and lack of effective treatment. The developments of the molecular mechanisms on cancer progression provide insight into novel targeted therapies, which are emerging as groundbreaking and promising cancer treatment strategies. In gynecologic malignancies, potential therapeutic targeted agents include antiangiogenic agents, poly (ADP-ribose) polymerase (PARP) inhibitors, tumor-intrinsic signaling pathway inhibitors, selective estrogen receptor downregulators, and immune checkpoint inhibitors. In this article, we provide a comprehensive review of the clinical evidence of targeted agents in gynecological cancers and discuss the future implication.
Collapse
Affiliation(s)
- Qiao Wang
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Hongling Peng
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Xiaorong Qi
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China.
| |
Collapse
|
9
|
M. F. Gonçalves B, S. P. Cardoso D, U. Ferreira MJ. Overcoming Multidrug Resistance: Flavonoid and Terpenoid Nitrogen-Containing Derivatives as ABC Transporter Modulators. Molecules 2020; 25:E3364. [PMID: 32722234 PMCID: PMC7435859 DOI: 10.3390/molecules25153364] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
Multidrug resistance (MDR) in cancer is one of the main limitations for chemotherapy success. Numerous mechanisms are behind the MDR phenomenon wherein the overexpression of the ATP-binding cassette (ABC) transporter proteins P-glycoprotein (P-gp), breast cancer resistance protein (BCRP) and multidrug resistance protein 1 (MRP1) is highlighted as a prime factor. Natural product-derived compounds are being addressed as promising ABC transporter modulators to tackle MDR. Flavonoids and terpenoids have been extensively explored in this field as mono or dual modulators of these efflux pumps. Nitrogen-bearing moieties on these scaffolds were proved to influence the modulation of ABC transporters efflux function. This review highlights the potential of semisynthetic nitrogen-containing flavonoid and terpenoid derivatives as candidates for the design of effective MDR reversers. A brief introduction concerning the major role of efflux pumps in multidrug resistance, the potential of natural product-derived compounds in MDR reversal, namely natural flavonoid and terpenoids, and the effect of the introduction of nitrogen-containing groups are provided. The main modifications that have been performed during last few years to generate flavonoid and terpenoid derivatives, bearing nitrogen moieties, such as aliphatic, aromatic and heterocycle amine, amide, and related functional groups, as well as their P-gp, MRP1 and BCRP inhibitory activities are reviewed and discussed.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/chemistry
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- ATP-Binding Cassette Transporters/chemistry
- ATP-Binding Cassette Transporters/metabolism
- Drug Resistance, Multiple/drug effects
- Flavonoids/chemistry
- Flavonoids/pharmacology
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Multidrug Resistance-Associated Proteins/chemistry
- Multidrug Resistance-Associated Proteins/metabolism
- Neoplasm Proteins/chemistry
- Neoplasm Proteins/metabolism
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Nitrogen/chemistry
- Terpenes/chemistry
- Terpenes/pharmacology
Collapse
Affiliation(s)
| | | | - Maria-José U. Ferreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal; (B.M.F.G.); (D.S.P.C.)
| |
Collapse
|
10
|
Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance. Drug Resist Updat 2020; 53:100715. [PMID: 32679188 DOI: 10.1016/j.drup.2020.100715] [Citation(s) in RCA: 318] [Impact Index Per Article: 63.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 05/29/2020] [Accepted: 06/07/2020] [Indexed: 12/11/2022]
Abstract
It is well established that multifactorial drug resistance hinders successful cancer treatment. Tumor cell interactions with the tumor microenvironment (TME) are crucial in epithelial-mesenchymal transition (EMT) and multidrug resistance (MDR). TME-induced factors secreted by cancer cells and cancer-associated fibroblasts (CAFs) create an inflammatory microenvironment by recruiting immune cells. CD11b+/Gr-1+ myeloid-derived suppressor cells (MDSCs) and inflammatory tumor associated macrophages (TAMs) are main immune cell types which further enhance chronic inflammation. Chronic inflammation nurtures tumor-initiating/cancer stem-like cells (CSCs), induces both EMT and MDR leading to tumor relapses. Pro-thrombotic microenvironment created by inflammatory cytokines and chemokines from TAMs, MDSCs and CAFs is also involved in EMT and MDR. MDSCs are the most common mediators of immunosuppression and are also involved in resistance to targeted therapies, e.g. BRAF inhibitors and oncolytic viruses-based therapies. Expansion of both cancer and stroma cells causes hypoxia by hypoxia-inducible transcription factors (e.g. HIF-1α) resulting in drug resistance. TME factors induce the expression of transcriptional EMT factors, MDR and metabolic adaptation of cancer cells. Promoters of several ATP-binding cassette (ABC) transporter genes contain binding sites for canonical EMT transcription factors, e.g. ZEB, TWIST and SNAIL. Changes in glycolysis, oxidative phosphorylation and autophagy during EMT also promote MDR. Conclusively, EMT signaling simultaneously increases MDR. Owing to the multifactorial nature of MDR, targeting one mechanism seems to be non-sufficient to overcome resistance. Targeting inflammatory processes by immune modulatory compounds such as mTOR inhibitors, demethylating agents, low-dosed histone deacetylase inhibitors may decrease MDR. Targeting EMT and metabolic adaptation by small molecular inhibitors might also reverse MDR. In this review, we summarize evidence for TME components as causative factors of EMT and anticancer drug resistance.
Collapse
|
11
|
Katopodis P, Chudasama D, Wander G, Sales L, Kumar J, Pandhal M, Anikin V, Chatterjee J, Hall M, Karteris E. Kinase Inhibitors and Ovarian Cancer. Cancers (Basel) 2019; 11:E1357. [PMID: 31547471 PMCID: PMC6770231 DOI: 10.3390/cancers11091357] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/08/2019] [Accepted: 09/09/2019] [Indexed: 12/22/2022] Open
Abstract
Ovarian cancer is fifth in the rankings of cancer deaths among women, and accounts for more deaths than any other gynecological malignancy. Despite some improvement in overall-(OS) and progression-free survival (PFS) following surgery and first-line chemotherapy, there is a need for development of novel and more effective therapeutic strategies. In this mini review, we provide a summary of the current landscape of the clinical use of tyrosine kinase inhibitors (TKIs) and mechanistic target of rapamycin (mTOR) inhibitors in ovarian cancer. Emerging data from phase I and II trials reveals that a combinatorial treatment that includes TKIs and chemotherapy agents seems promising in terms of PFS despite some adverse effects recorded; whereas the use of mTOR inhibitors seems less effective. There is a need for further research into the inhibition of multiple signaling pathways in ovarian cancer and progression to phase III trials for drugs that seem most promising.
Collapse
Affiliation(s)
- Periklis Katopodis
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, London UB9 6JH, UK.
| | - Dimple Chudasama
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Gurleen Wander
- Chelsea and Westminster Hospital NHS Trust, London UB9 6JH, UK.
| | - Louise Sales
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Juhi Kumar
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Manreen Pandhal
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Vladimir Anikin
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, London UB9 6JH, UK.
- Department of Oncology and Reconstructive Surgery, Sechenov First Moscow State Medical University, 119146 Moscow, Russia.
| | - Jayanta Chatterjee
- Faculty of Health and Medical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK.
| | - Marcia Hall
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
- Mount Vernon Cancer Centre, Rickmansworth Road, Northwood HA6 2RN, UK.
| | - Emmanouil Karteris
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| |
Collapse
|
12
|
Hiraga T. Hypoxic Microenvironment and Metastatic Bone Disease. Int J Mol Sci 2018; 19:ijms19113523. [PMID: 30423905 PMCID: PMC6274963 DOI: 10.3390/ijms19113523] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/07/2018] [Accepted: 11/07/2018] [Indexed: 01/07/2023] Open
Abstract
Hypoxia is a common feature of solid tumors and is associated with an increased risk of metastasis and a poor prognosis. Recent imaging techniques revealed that bone marrow contains a quite hypoxic microenvironment. Low oxygen levels activate hypoxia signaling pathways such as hypoxia-inducible factors, which play critical roles in the key stages of metastatic dissemination including angiogenesis, epithelial-mesenchymal transition, invasion, maintenance of cancer stem cells, tumor cell dormancy, release of extracellular vesicles, and generation of pre-metastatic niches. Hypoxia also affects bone cells, such as osteoblasts and osteoclasts, and immune cells, which also act to support the development and progression of bone metastases. Paradoxically, hypoxia and related signaling molecules are recognized as high-priority therapeutic targets and many candidate drugs are currently under preclinical and clinical investigation. The present review focuses on our current knowledge of the potential roles of hypoxia in cancer metastasis to bone by considering the interaction between metastatic cancer cells and the bone microenvironment. Current therapeutic approaches targeting hypoxia are also described.
Collapse
Affiliation(s)
- Toru Hiraga
- Department of Histology and Cell Biology, Matsumoto Dental University, 1780 Gobara-Hirooka, Shiojiri, Nagano 399-0781, Japan.
| |
Collapse
|
13
|
Bonello M, Sims AH, Langdon SP. Human epidermal growth factor receptor targeted inhibitors for the treatment of ovarian cancer. Cancer Biol Med 2018; 15:375-388. [PMID: 30766749 PMCID: PMC6372909 DOI: 10.20892/j.issn.2095-3941.2018.0062] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/26/2018] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is the second most lethal gynecological cancer worldwide and while most patients respond to initial therapy, they often relapse with resistant disease. Human epidermal growth factor receptors (especially HER1/EGFR and HER2/ERBB2) are involved in disease progression; hence, strategies to inhibit their action could prove advantageous in ovarian cancer patients, especially in patients resistant to first line therapy. Monoclonal antibodies and tyrosine kinase inhibitors are two classes of drugs that act on these receptors. They have demonstrated valuable antitumor activity in multiple cancers and their possible use in ovarian cancer continues to be studied. In this review, we discuss the human epidermal growth factor receptor family; review emerging clinical studies on monoclonal antibodies and tyrosine kinase inhibitors targeting these receptors in ovarian cancer patients; and propose future research possibilities in this area.
Collapse
Affiliation(s)
- Maria Bonello
- Cancer Research UK Edinburgh Center and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Andrew Harvey Sims
- Cancer Research UK Edinburgh Center and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Simon Peter Langdon
- Cancer Research UK Edinburgh Center and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| |
Collapse
|
14
|
Morrison J, Thoma C, Goodall RJ, Lyons TJ, Gaitskell K, Wiggans AJ, Bryant A. Epidermal growth factor receptor blockers for the treatment of ovarian cancer. Cochrane Database Syst Rev 2018; 10:CD007927. [PMID: 30321910 PMCID: PMC6430330 DOI: 10.1002/14651858.cd007927.pub4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND This is an update of a previously published version of the review (Issue 10, 2011).Epithelial ovarian cancer (EOC) is the seventh most common cause of cancer death among women worldwide. Treatment consists of a combination of surgical debulking and platinum-based chemotherapy. Between 55% and 75% of women who respond to first-line therapy experience relapse within two years. Second-line chemotherapy is palliative and aims to reduce symptoms and prolong survival. Improved understanding about the molecular basis of EOC has led to the development of novel agents, such as epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors and anti-EGFR antibodies. OBJECTIVES To compare the effectiveness and harmful effects of interventions that target the epidermal growth factor receptor in the treatment of epithelial ovarian cancer (EOC). SEARCH METHODS We searched the Cochrane Gynaecological Cancer Group Trials Register, the Cochrane Central Register of Controlled Trials (CENTRAL; 2010, Issue 4), MEDLINE, and Embase up to October 2010. We also searched registers of clinical trials, abstracts of scientific meetings, and reference lists of included studies, and we contacted experts in the field. This update includes further searches up to September 2017. SELECTION CRITERIA Randomised controlled trials (RCTs) comparing anti-EGFR agents with or without conventional chemotherapy versus conventional chemotherapy alone or no treatment in women with histologically proven EOC. DATA COLLECTION AND ANALYSIS Two review authors independently abstracted data, assessed risk of bias, and performed GRADE assessment. MAIN RESULTS From 6105 references obtained through the literature search and an additional 15 references derived from grey literature searches, we identified seven RCTs that met our inclusion criteria and included 1725 participants. Trial results show that after first-line chemotherapy is provided, maintenance treatment with erlotinib (EGFR tyrosine kinase inhibitor (TKI)) probably makes little or no difference in overall survival (hazard ratio (HR) 0.99, 95% confidence interval (CI) 0.81 to 1.20; one study; 835 participants; low-certainty evidence) and may make little or no difference in progression-free survival (HR 1.05, 95% CI 0.90 to 1.23; one study; 835 participants; very low-certainty evidence). Less than 50% of participants provided quality of life data, and study authors reported these results incompletely. The certainty of evidence is very low, but treatment may reduce quality of life compared to observation.Treatment with an EGFR TKI (vandetanib) for women with relapsed EOC may make little or no difference in overall survival (HR 1.25, 95% CI 0.80 to 1.95; one study; 129 participants; low-certainty evidence) and may make little or no difference in progression-free survival (HR 0.99, 95% CI 0.69 to 1.42; one study; 129 participants; very low-certainty evidence). In treating patients with relapse, giving EGFR TKI may slightly increase some toxicities, such as severe rash (risk ratio (RR) 13.63, 95% CI 0.78 to 236.87; one study; 125 participants; very low-certainty evidence). Quality of life data were not available for meta-analysis.Anti-EGFR antibody treatment in relapsed EOC may or may not make a difference to overall survival (HR 0.93, 95% CI 0.74 to 1.18; four studies; 658 participants; moderate-certainty evidence) and may or may not have any effect on progression-free survival (HR 0.90, 95% CI 0.70 to 1.16; four studies; 658 participants; low-certainty evidence). Anti-EGFR antibody treatment may or may not increase side effects, including severe nausea and/or vomiting (RR 1.27, 95% CI 0.56 to 2.89; three studies; 503 participants; low-certainty evidence), severe fatigue (RR 1.06, 95% CI 0.66 to 1.73; I² = 0%; four studies; 652 participants; low-certainty evidence), and hypokalaemia (RR 2.01, 95% CI 0.80 to 5.06; I² = 0%; three studies; 522 participants; low-certainty evidence). Severe diarrhoea rates were heterogeneous across studies (RR 2.87, 95% CI 0.59 to 13.89; four studies; 652 participants; low-certainty evidence), and subgroup analysis revealed that severe diarrhoea was more likely with pertuzumab (RR 6.37, 95% CI 1.89 to 21.45; I² = 0%; three studies; 432 participants; low-certainty evidence) than with seribantumab treatment (RR 0.38, 95% CI 0.07 to 2.23; I² = 0%; one study; 220 participants; very low-certainty evidence). Quality of life data were incompletely reported, and we were unable to combine them in a meta-analysis. AUTHORS' CONCLUSIONS Current evidence suggests that an anti-EGFR single-agent biological treatment (EGFR TKI or anti-EGFR antibody) makes little or no difference to survival, either as maintenance treatment after first-line chemotherapy or in association with chemotherapy in recurrent cancer. Anti-EGFR therapy may increase some side effects and may or may not reduce quality of life.
Collapse
Affiliation(s)
- Jo Morrison
- Musgrove Park HospitalDepartment of Gynaecological OncologyTaunton and Somerset NHS Foundation TrustTauntonSomersetUKTA1 5DA
| | | | | | - Thomas J Lyons
- University of BristolSchool of Medical Sciences38 Kings Parade AvenueBristolUKBS8 2RB
| | - Kezia Gaitskell
- University of OxfordCancer Epidemiology Unit, Nuffield Department of Population HealthRichard Doll BuildingRoosevelt DriveOxfordUKOX3 7LF
| | - Alison J Wiggans
- Musgrove Park HospitalDepartment of Obstetrics and GynaecologyTaunton and Somerset NHS Foundation TrustTauntonSomersetUKTA1 5DA
| | - Andrew Bryant
- Newcastle UniversityInstitute of Health & SocietyMedical School New BuildRichardson RoadNewcastle upon TyneUKNE2 4AX
| | | |
Collapse
|
15
|
Poonia N, Lather V, Pandita D. Mesoporous silica nanoparticles: a smart nanosystem for management of breast cancer. Drug Discov Today 2018; 23:315-332. [DOI: 10.1016/j.drudis.2017.10.022] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/26/2017] [Accepted: 10/31/2017] [Indexed: 12/22/2022]
|
16
|
Brackman DJ, Giacomini KM. Reverse Translational Research of ABCG2 (BCRP) in Human Disease and Drug Response. Clin Pharmacol Ther 2017; 103:233-242. [PMID: 29023674 DOI: 10.1002/cpt.903] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/05/2017] [Accepted: 10/07/2017] [Indexed: 12/12/2022]
Abstract
Reverse translational research takes a bedside-to-bench approach, using sophisticated basic research to explain the biological mechanisms behind observed clinical data. For transporters, which play a role in human disease and drug response, this approach offers a distinct advantage over the typical translational research, which often falters due to inadequate in vitro and preclinical animal models. Research on ABCG2, which encodes the Breast Cancer Resistance Protein, has benefited immensely from a reverse translational approach due to its broad implications for disease susceptibility and both therapeutic and adverse drug response. In this review, we describe the success of reverse translational research for ABCG2 and opportunities for further studies.
Collapse
Affiliation(s)
- Deanna J Brackman
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA.,Institute of Human Genetics, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
17
|
SYD985, a novel duocarmycin-based HER2-targeting antibody-drug conjugate, shows promising antitumor activity in epithelial ovarian carcinoma with HER2/Neu expression. Gynecol Oncol 2017; 146:179-186. [PMID: 28473206 DOI: 10.1016/j.ygyno.2017.04.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/25/2017] [Accepted: 04/27/2017] [Indexed: 01/16/2023]
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is an aggressive and heterogeneous disease. <10% of EOC demonstrate HER2/neu 3+ receptor over-expression. However, moderate to low (i.e., 2+ and 1+) HER2/neu expression is reported in up to 50% of EOC. The objective of this study was to compare the anti-tumor activity of SYD985, a novel HER2-targeting antibody-drug conjugate (ADC), to trastuzumab emtansine (T-DM1) in EOC models with differential HER2/neu expression. METHODS The cytotoxicity of SYD985 and T-DM1 was evaluated using ten primary EOC cell lines with 0/1+, 2+, and 3+ HER2/neu expression in antibody-dependent cellular cytotoxicity (ADCC), proliferation, viability and bystander killing experiments. Finally, the in vivo activity of SYD985 and T-DM1 was also studied in ovarian cancer xenografts. RESULTS SYD985 and T-DM1 induced similar ADCC in the presence of peripheral blood lymphocytes (PBL) against EOC cell lines with differential HER2/neu expression. In contrast, SYD985 was 3 to 42 fold more cytotoxic in the absence of PBL when compared to T-DM1 (p<0.0001). Unlike T-DM1, SYD985 induced efficient bystander killing of HER2/neu 0/1+ tumor cells when admixed with HER2/neu 3+ EOC cells. In vivo studies confirmed that SYD985 is significantly more active than T-DM1 against HER2/neu 3+ EOC xenografts. CONCLUSIONS SYD985 is a novel ADC with remarkable activity against EOC with strong (3+) as well as moderate to low (i.e., 2+ and 1+) HER2/neu expression. SYD985 is more potent than T-DM1 in comparative experiments and unlike T-DM1, it is active against EOC demonstrating moderate/low or heterogeneous HER2/neu expression.
Collapse
|
18
|
A Combination of Two Receptor Tyrosine Kinase Inhibitors, Canertinib and PHA665752 Compromises Ovarian Cancer Cell Growth in 3D Cell Models. Oncol Ther 2016; 4:257-274. [PMID: 28261654 PMCID: PMC5315083 DOI: 10.1007/s40487-016-0031-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Indexed: 12/25/2022] Open
Abstract
Introduction Advanced ovarian cancer is often a fatal disease as chemotherapeutic drugs have limited effectiveness. Better targeted therapy is needed to improve the survival and quality of life for these women. Receptor tyrosine kinases including EGFR, Her-2 and c-Met are associated with a poor prognosis in ovarian cancer. Therefore, the co-activation of these receptors may be crucial for growth promoting activity. In this study, we explored the effect of combining two small molecule inhibitors that target the EGFR/Her-2 and c-Met receptor tyrosine kinases in two ovarian cancer cell lines. The aim of this study was to investigate the combined inhibition activity of a dual EGFR/Her-2 inhibitor (canertinib) and a c-Met inhibitor (PHA665752) in ovarian cancer cell lines in 3D cell aggregates. Methods OVCAR-5 and SKOV-3 ovarian cancer cell lines were cultured on a non-adherent surface to produce 3D cell clusters and aggregates. Cells were exposed to canertinib and PHA665752, both individually and in combination, for 48 h. The effect on growth, metabolism and the expression/phosphorylation of selective signaling proteins associated with EGFR, Her-2 and c-Met were investigated. Results The single drug treatments significantly decreased cell growth and altered the expression of signaling proteins in OVCAR-5 and SKOV-3 cell lines. The combination treatment showed greater reduction of cell numbers for both cell lines. Total expression and phosphorylation of signaling proteins were further reduced in the combination drug treatments, compared to the single inhibitor treatments. Conclusion Our findings suggest that the concurrent targeting of more than one receptor tyrosine kinase may be useful in developing more effective targeted drug regimens for patients, who have EGFR, Her-2 and c-Met positive ovarian cancer cells.
Collapse
|
19
|
Ricci JW, Lovato DM, Severns V, Sklar LA, Larson RS. Novel ABCG2 Antagonists Reverse Topotecan-Mediated Chemotherapeutic Resistance in Ovarian Carcinoma Xenografts. Mol Cancer Ther 2016; 15:2853-2862. [PMID: 27671528 DOI: 10.1158/1535-7163.mct-15-0789] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 08/17/2016] [Accepted: 09/14/2016] [Indexed: 11/16/2022]
Abstract
Chemotherapeutic resistance remains a challenge in the treatment of ovarian carcinoma, especially in recurrent disease. Despite the fact that most patients with newly diagnosed tumors attain complete remission following cytoreductive surgery and chemotherapy, ovarian carcinoma has a recurrence rate that exceeds 75%. The ATP-binding cassette family G member 2 (ABCG2) efflux protein has been described as one mechanism that confers multiple-drug resistance to solid tumors and contributes to topotecan resistance in ovarian carcinoma. In fact, one clinical trial demonstrated ABCG2 expression in all patients with primary or recurrent ovarian carcinoma. On the basis of our previous work, we hypothesized that three compounds (CID44640177, CID1434724, and CID46245505), which represent a new piperazine-substituted pyrazolo[1,5]pyrimidine substructure class of ABCG2-specific antagonists, would restore chemosensitivity to drug-resistant ovarian cancer in vitro and in vivo To address the treatment difficulties associated with chemotherapeutic resistance in ovarian cancer, we combined each compound (CID44640177, CID1434724, and CID46245505) with topotecan and administered the mixture to chemoresistant Igrov1/T8 ovarian cancer cells in vitro and Igrov1/T8 xenografts in CB-17 SCID mice. We found that only nanomolar concentrations of each ABCG2 inhibitor in combination with topotecan were required to restore chemosensitivity to Igrov1/T8 cells in vitro In vivo, substantial tumor reduction was achieved with each compound in 4 days, with CID1434724 causing the largest reduction in excess of 60%. No signs of secondary toxic effects were observed with the ABCG2 antagonists. These novel compounds should be viewed as promising drug candidates to reverse ABCG2-mediated chemoresistance. Mol Cancer Ther; 15(12); 2853-62. ©2016 AACR.
Collapse
Affiliation(s)
- Jerec W Ricci
- Clinical and Translational Science Center, University of New Mexico, Albuquerque, New Mexico
| | - Debbie M Lovato
- Clinical and Translational Science Center, University of New Mexico, Albuquerque, New Mexico
| | - Virginia Severns
- Clinical and Translational Science Center, University of New Mexico, Albuquerque, New Mexico
| | - Larry A Sklar
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico
| | - Richard S Larson
- Clinical and Translational Science Center, University of New Mexico, Albuquerque, New Mexico.
| |
Collapse
|
20
|
Puvanenthiran S, Essapen S, Seddon AM, Modjtahedi H. Impact of the putative cancer stem cell markers and growth factor receptor expression on the sensitivity of ovarian cancer cells to treatment with various forms of small molecule tyrosine kinase inhibitors and cytotoxic drugs. Int J Oncol 2016; 49:1825-1838. [PMID: 27599579 PMCID: PMC5063458 DOI: 10.3892/ijo.2016.3678] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/22/2016] [Indexed: 12/30/2022] Open
Abstract
Increased expression and activation of human epidermal growth factor receptor (EGFR) and HER-2 have been reported in numerous cancers. The aim of this study was to determine the sensitivity of a large panel of human ovarian cancer cell lines (OCCLs) to treatment with various forms of small molecule tyrosine kinase inhibitors (TKIs) and cytotoxic drugs. The aim was to see if there was any association between the protein expression of various biomarkers including three putative ovarian cancer stem cell (CSC) markers (CD24, CD44, CD117/c-Kit), P-glycoprotein (P-gp), and HER family members and response to treatment with these agents. The sensitivity of 10 ovarian tumour cell lines to the treatment with various forms of HER TKIs (gefitinib, erlotinib, lapatinib, sapitinib, afatinib, canertinib, neratinib), as well as other TKIs (dasatinib, imatinib, NVP-AEW541, crizotinib) and cytotoxic agents (paclitaxel, cisplatin and doxorubicin), as single agents or in combination, was determined by SRB assay. The effect on these agents on the cell cycle distribution, and downstream signaling molecules and tumour migration were determined using flow cytometry, western blotting, and the IncuCyte Clear View cell migration assay respectively. Of the HER inhibitors, the irreversible pan-TKIs (canertinib, neratinib and afatinib) were the most effective TKIs for inhibiting the growth of all ovarian cancer cells, and for blocking the phosphorylation of EGFR, HER-2, AKT and MAPK in SKOV3 cells. Interestingly, while the majority of cancer cells were highly sensitive to treatment with dasatinib, they were relatively resistant to treatment with imatinib (i.e., IC50 >10 µM). Of the cytotoxic agents, paclitaxel was the most effective for inhibiting the growth of OCCLs, and of various combinations of these drugs, only treatment with a combination of NVP-AEW541 and paclitaxel produced a synergistic or additive anti-proliferative effect in all three cell lines examined (i.e., SKOV3, Caov3, ES2). Finally, of the TKIs, only treatment with afatinib, neratinib and dasatinib were able to reduce the migration of HER-2 overexpressing SKOV3 cells. We did not find any significant association between the expression of putative ovarian CSC marker, HER family members, c-MET, ALK, and IGF-IR and the response to the irreversible HER TKIs. Our results support the need for further investigations of the therapeutic potential of these irreversible HER family blockers in ovarian cancer, and the therapeutic potential of dasatinib when used in combination with the inhibitors of the HER family members in ovarian cancer.
Collapse
Affiliation(s)
- Soozana Puvanenthiran
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston-upon-Thames, Surrey KT1 2EE, UK
| | - Sharadah Essapen
- St Luke's Cancer Centre, Royal Surrey County Hospital, Guildford GU2 7XX, UK
| | - Alan M Seddon
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston-upon-Thames, Surrey KT1 2EE, UK
| | - Helmout Modjtahedi
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston-upon-Thames, Surrey KT1 2EE, UK
| |
Collapse
|
21
|
Jiang J, Dong L, Wang L, Wang L, Zhang J, Chen F, Zhang X, Huang M, Li S, Ma W, Xu Q, Huang C, Fang J, Wang C. HER2-targeted antibody drug conjugates for ovarian cancer therapy. Eur J Pharm Sci 2016; 93:274-86. [PMID: 27509865 DOI: 10.1016/j.ejps.2016.08.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/01/2016] [Accepted: 08/06/2016] [Indexed: 01/10/2023]
Abstract
HER2 targeted delivery of ovarian cancer therapy has been beneficial for some patients, although, its efficacy is yet to be confirmed in large populations. We generated a novel anti-HER2 humanized antibody (Hertuzumab) and conjugated it to a microtubule-disrupting drug monomethyl auristatin E conjugate (MMAE) with a lysosomal protease-cleavable valine-citrulline linker. The average drug to antibody ratio (DAR) of Hertuzumab-vc-MMAE was varied by conjugating Hertuzumab antibodies with increasing linker-drugs (LDs) from D0-D8. The resulting conjugates were tested for kinetic affinity for soluble HER2-ECD, cytotoxicity, and in vivo pharmacokinetics. The kinetic binding constant values (KD) were obtained by the bio-layer interference (BLI) method. The half time (t1/2) and clearance (Cl) results of the pharmacokinetic profile in rats were DAR-dependent. Hertuzumab-vc-MMAE with DAR4 was selected for further evaluation. Both Hertuzumab and Hertuzumab conjugates could bind to HER2 antigen, and exhibited significant cytotoxicity on HER2 positive tumor cells after internalization by receptor-mediated endocytosis. Hence, Hertuzumab-vc-MMAE conjugates were significantly selective both in vitro and in vivo as compared to other ovarian cancer clinical therapies that are currently used. Cell signal transduction and cell cycle were also affected, as shown by down regulation of PI3K/AKT pathway and arrested mitosis in the G2/M phase. The pharmacokinetics and pharmacodynamics (PK-PD) of the conjugates in nude mouse xenograft model demonstrated a correlation between efficacy and drug concentration. These results show that Hertuzumab-vc-MMAE is a potential therapeutic agent for HER2 positive ovarian cancer.
Collapse
Key Words
- Antibody drug conjugates (ADCs)
- Cisplatine (PubChem CID:441,203), Paclitaxel (PubChem CID: 91,885,464), Tris (2-carboxyethyl) phosphine, TCEP (PubChem CID:119,411)
- DM1 (PubChem CID:11,343,137)
- HER2/ErbB2
- MC-VC-PAB-MMAE (PubChem CID: 71,586,737)
- MMAE (PubChem CID: 53,297,465)
- Monomethyl auristatin E (MMAE)
- Ovarian cancer
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/chemistry
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Cell Line, Tumor
- Cell Survival/drug effects
- Female
- Humans
- Immunoconjugates/chemistry
- Immunoconjugates/pharmacokinetics
- Immunoconjugates/pharmacology
- Immunoconjugates/therapeutic use
- Immunoglobulin G/immunology
- Mice, Inbred BALB C
- Mice, Nude
- Oligopeptides/chemistry
- Oligopeptides/pharmacokinetics
- Oligopeptides/pharmacology
- Oligopeptides/therapeutic use
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Rats, Sprague-Dawley
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/metabolism
- Solubility
- Tumor Burden/drug effects
Collapse
Affiliation(s)
- Jing Jiang
- Department of Pharmacology, Binzhou Medical University, Yantai 256603, Shandong, China
| | - Lihou Dong
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Lei Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ling Wang
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Jing Zhang
- Department of Pharmacology, Binzhou Medical University, Yantai 256603, Shandong, China
| | - Fang Chen
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiuli Zhang
- Department of Pharmacology, Binzhou Medical University, Yantai 256603, Shandong, China
| | - Min Huang
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Shenjun Li
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Weiwei Ma
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | - Qiaoyu Xu
- RemeGen, Ltd, Yantai 264000, Shandong, China
| | | | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai 200092, China.
| | - Chunhua Wang
- Department of Pharmacology, Binzhou Medical University, Yantai 256603, Shandong, China.
| |
Collapse
|
22
|
Ntanasis-Stathopoulos I, Fotopoulos G, Tzanninis IG, Kotteas EA. The Emerging Role of Tyrosine Kinase Inhibitors in Ovarian Cancer Treatment: A Systematic Review. Cancer Invest 2016; 34:313-39. [PMID: 27486869 DOI: 10.1080/07357907.2016.1206117] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The present systematic review summarizes current evidence regarding the mechanisms of action, the efficacy, and the adverse effects of tyrosine kinase inhibitors (TKIs) in ovarian cancer patients. Phase II and III clinical trials were sought in the PubMed database and in the Clinical Trials.gov registry through September 30, 2015. Seventy-five clinical trials regarding TKIs targeting mainly vascular endothelial growth factor receptor, epidermal growth factor receptor, platelet-derived growth factor receptor, and sarcoma tyrosine kinase (Src) were yielded. The most promising results were noted with cediranib, nintedanib, and pazopanib. However, drawing universal conclusions about the potential integration of TKIs in ovarian cancer therapy remains elusive. Furthermore, emerging challenges and directions for the future research are critically discussed.
Collapse
Affiliation(s)
| | - George Fotopoulos
- a Oncology Unit, Sotiria General Hospital , Athens School of Medicine , Athens , Greece
| | | | - Elias A Kotteas
- a Oncology Unit, Sotiria General Hospital , Athens School of Medicine , Athens , Greece
| |
Collapse
|
23
|
Petrillo M, Nero C, Amadio G, Gallo D, Fagotti A, Scambia G. Targeting the hallmarks of ovarian cancer: The big picture. Gynecol Oncol 2016; 142:176-183. [PMID: 27058837 DOI: 10.1016/j.ygyno.2016.03.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 03/20/2016] [Accepted: 03/29/2016] [Indexed: 01/06/2023]
Abstract
OBJECTIVE As a result of relevant achievements in the field of translational research, several active drugs and multiple biological targets are available in ovarian cancer (OC). In this complex scenario, there is an urgent need to effectively summarize the available data in order to update conclusions, and outline perspectives. METHODS The results in terms of target identification and drug development have been summarized using the well-known hallmarks of cancer firstly described, and recently modified by Hanahan and Weinberg [1-2]. Published data from clinical trials have been retrieved from PubMed, Embase, CINAHL and Cochrane database. Ongoing clinical trials were searched using clinicaltrials.gov web platform, and identified using NCT number. RESULTS Genomic instability and angiogenesis are the most actively investigated hallmarks in high-grade serous OC, and the inhibition of tumor immune evasion appears as the emerging strategy for molecularly-driven therapy. Targeting sustained proliferative signaling through MEK and mTOR inhibitors seems the most promising approach in clear cell, and low-grade serous OC. CONCLUSIONS This substantial amount of data suggests that targeted therapies are already part of the clinical and therapeutic management of OC patients. The expectations of getting from translational research a better knowledge of tumor biology and therefore personalized drugs are high and worthy of maximum effort from referral centers.
Collapse
Affiliation(s)
- M Petrillo
- Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy.
| | - C Nero
- Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - G Amadio
- Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| | - D Gallo
- Department of Obstetrics and Gynecology, Centre for Translational Medicine for Women and Children Health, Catholic University of the Sacred Heart, Rome, Italy
| | - A Fagotti
- Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy; University of Perugia, Italy
| | - G Scambia
- Department of Obstetrics and Gynecology, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
24
|
Hasanabady MH, Kalalinia F. ABCG2 inhibition as a therapeutic approach for overcoming multidrug resistance in cancer. J Biosci 2016; 41:313-24. [DOI: 10.1007/s12038-016-9601-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Bull-Hansen B, Berstad MB, Berg K, Cao Y, Skarpen E, Fremstedal AS, Rosenblum MG, Peng Q, Weyergang A. Photochemical activation of MH3-B1/rGel: a HER2-targeted treatment approach for ovarian cancer. Oncotarget 2016; 6:12436-51. [PMID: 26002552 PMCID: PMC4494949 DOI: 10.18632/oncotarget.3814] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 03/11/2015] [Indexed: 11/28/2022] Open
Abstract
HER2-targeted therapy has been shown to have limited efficacy in ovarian cancer despite frequent overexpression of this receptor. Photochemical internalization (PCI) is a modality for cytosolic drug delivery, currently undergoing clinical evaluation. In the present project we studied the application of PCI in combination with the HER2-targeted recombinant fusion toxin, MH3-B1/rGel, for the treatment of ovarian cancer. The SKOV-3 cell line, resistant to trastuzumab- and MH3-B1/rGel- monotherapy, was shown to respond strongly to PCI of MH3-B1/rGel to a similar extent as observed for the treatment-sensitive SK-BR-3 breast cancer cells. Extensive hydrolytic degradation of MH3-B1/rGel in acidic endocytic vesicles was indicated as the mechanism of MH3-B1/rGel resistance in SKOV-3 cells. This was shown by the positive Pearson's correlation coefficient between Alexa488-labeled MH3-B1/rGel and Lysotracker in SKOV-3 cells in contrast to the negative Pearson's correlation coefficient in SK-BR-3 cells. The application of PCI to induce the release of MH3-B1/rGel was also demonstrated to be effective on SKOV-3 xenografts. Application of PCI with MH3-B1/rGel was further found highly effective in the HER2 expressing HOC-7 and NuTu-19 ovarian cancer cell lines. The presented results warrant future development of PCI in combination with MH3-B1/rGel as a novel therapeutic approach in preclinical models of ovarian cancer.
Collapse
Affiliation(s)
- Bente Bull-Hansen
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Maria B Berstad
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Kristian Berg
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Yu Cao
- Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX, USA.,Current address: The Scripps Research Institute, Department of Chemistry, La Jolla, CA, USA
| | - Ellen Skarpen
- Department of Biochemistry, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Ane Sofie Fremstedal
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Michael G Rosenblum
- Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX, USA
| | - Qian Peng
- Department of Pathology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Anette Weyergang
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
26
|
McLachlan J, Lima JPDSN, Dumas L, Banerjee S. Targeted agents and combinations in ovarian cancer: where are we now? Expert Rev Anticancer Ther 2016; 16:441-54. [DOI: 10.1586/14737140.2016.1162101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
27
|
Wang Z, Fu S. An overview of tyrosine kinase inhibitors for the treatment of epithelial ovarian cancer. Expert Opin Investig Drugs 2015; 25:15-30. [PMID: 26560712 DOI: 10.1517/13543784.2016.1117071] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy and the fifth most common cause of cancer-related deaths in women. Initial treatment with surgery and chemotherapy has improved survival significantly. However, the disease progresses or recurs in most patients. Thus, there is an urgent need to develop more effective treatment strategies. AREAS COVERED This article provides an overview of tyrosine kinase inhibitors (TKIs) for the treatment of EOC, which is based on English peer-reviewed articles on MEDLINE and related abstracts presented at major conferences. The authors highlight the data from the published clinical trials in EOC patients who were treated with TKIs or TKI-based regimens. EXPERT OPINION EOC is responsive to most chemotherapeutic drugs and/or biological agents and represents an ideal disease model for investigating novel anti-cancer agents. Numerous small-molecule TKIs targeting the VEGFR, PARP, PI3K-AKT-mTOR, MAPK, Src, PKC, Wee1 and HER1/2 signaling pathways are currently being tested in clinical trials. Research is needed for devising regimens combining TKIs with other agents in an optimal timing schedule and for identifying potential biomarkers predictive of response and survival.
Collapse
Affiliation(s)
- Zhijie Wang
- a Department of Investigational Cancer Therapeutics , The University of Texas MD Anderson Cancer Center , 1515 Holcombe Boulevard, Houston , TX 77030 , USA.,b Department of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) , Peking University Cancer Hospital & Beijing Institute for Cancer Research , Beijing , China
| | - Siqing Fu
- a Department of Investigational Cancer Therapeutics , The University of Texas MD Anderson Cancer Center , 1515 Holcombe Boulevard, Houston , TX 77030 , USA
| |
Collapse
|
28
|
Trumpi K, Emmink BL, Prins AM, van Oijen MGH, van Diest PJ, Punt CJA, Koopman M, Kranenburg O, Rinkes IHMB. ABC-Transporter Expression Does Not Correlate with Response to Irinotecan in Patients with Metastatic Colorectal Cancer. J Cancer 2015; 6:1079-86. [PMID: 26516354 PMCID: PMC4615342 DOI: 10.7150/jca.12606] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/12/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Active efflux of irinotecan by ATP-binding cassette (ABC)-transporters, in particular ABCB1 and ABCG2, is a well-established drug resistance mechanism in vitro and in pre-clinical mouse models, but its relevance in colorectal cancer (CRC) patients is unknown. Therefore, we assessed the association between ABC-transporter expression and tumour response to irinotecan in patients with metastatic CRC. METHODS Tissue microarrays of a large cohort of metastatic CRC patients treated with irinotecan in a prospective study (CAIRO study; n=566) were analysed for expression of ABCB1 and ABCG2 by immunohistochemistry. Kaplan-Meier and Cox proportional hazard regression analyses were performed to assess the association of ABC transporter expression with irinotecan response. Gene expression profiles of 17 paired tumours were used to assess the concordance of ABCB1/ABCG2 expression in primary CRC and corresponding metastases. RESULTS The response to irinotecan was not significantly different between primary tumours with positive versus negative expression of ABCB1 (5.8 vs 5.7 months, p=0.696) or ABCG2 (5.7 vs 6.1 months, p=0.811). Multivariate analysis showed neither ABCB1 nor ABCG2 were independent predictors for progression free survival. There was a mediocre to poor concordance between ABC-transporter expression in paired tumours. CONCLUSION In metastatic CRC, ABC-transporter expression in the primary tumour does not predict irinotecan response.
Collapse
Affiliation(s)
- K Trumpi
- 1. Department of Surgery, University Medical Centre Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - B L Emmink
- 1. Department of Surgery, University Medical Centre Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - A M Prins
- 1. Department of Surgery, University Medical Centre Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - M G H van Oijen
- 2. Department of Medical Oncology, University Medical Centre Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands ; 4. Department of Medical Oncology, Academic Medical Centre, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - P J van Diest
- 3. Department of Pathology, University Medical Centre Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - C J A Punt
- 4. Department of Medical Oncology, Academic Medical Centre, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - M Koopman
- 2. Department of Medical Oncology, University Medical Centre Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - O Kranenburg
- 1. Department of Surgery, University Medical Centre Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| | - I H M Borel Rinkes
- 1. Department of Surgery, University Medical Centre Utrecht, Heidelberglaan 100, 3584CX Utrecht, The Netherlands
| |
Collapse
|
29
|
Ween MP, Armstrong MA, Oehler MK, Ricciardelli C. The role of ABC transporters in ovarian cancer progression and chemoresistance. Crit Rev Oncol Hematol 2015; 96:220-56. [PMID: 26100653 DOI: 10.1016/j.critrevonc.2015.05.012] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 04/08/2015] [Accepted: 05/18/2015] [Indexed: 02/06/2023] Open
Abstract
Over 80% of ovarian cancer patients develop chemoresistance which results in a lethal course of the disease. A well-established cause of chemoresistance involves the family of ATP-binding cassette transporters, or ABC transporters that transport a wide range of substrates including metabolic products, nutrients, lipids, and drugs across extra- and intra-cellular membranes. Expressions of various ABC transporters, shown to reduce the intracellular accumulation of chemotherapy drugs, are increased following chemotherapy and impact on ovarian cancer survival. Although clinical trials to date using ABC transporter inhibitors have been disappointing, ABC transporter inhibition remains an attractive potential adjuvant to chemotherapy. A greater understanding of their physiological functions and role in ovarian cancer chemoresistance will be important for the development of more effective targeted therapies. This article will review the role of the ABC transporter family in ovarian cancer progression and chemoresistance as well as the clinical attempts used to date to reverse chemoresistance.
Collapse
Affiliation(s)
- M P Ween
- Lung Research, Hanson Institute and Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide
| | - M A Armstrong
- Data Management and Analysis Centre, University of Adelaide, Australia
| | - M K Oehler
- Gynaecological Oncology Department, Royal Adelaide Hospital, Australia; School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, Australia
| | - C Ricciardelli
- School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, Australia.
| |
Collapse
|
30
|
Analysis of wntless (WLS) expression in gastric, ovarian, and breast cancers reveals a strong association with HER2 overexpression. Mod Pathol 2015; 28:428-36. [PMID: 25258105 DOI: 10.1038/modpathol.2014.114] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/20/2014] [Accepted: 07/20/2014] [Indexed: 12/31/2022]
Abstract
The oncogenic role of WNT is well characterized. Wntless (WLS) (also known as GPR177, or Evi), a key modulator of WNT protein secretion, was recently found to be highly overexpressed in malignant astrocytomas. We hypothesized that this molecule may be aberrantly expressed in other cancers known to possess aberrant WNT signaling such as ovarian, gastric, and breast cancers. Immunohistochemical analysis using a TMA platform revealed WLS overexpression in a subset of ovarian, gastric, and breast tumors; this overexpression was associated with poorer clinical outcomes in gastric cancer (P=0.025). In addition, a strong correlation was observed between WLS expression and human epidermal growth factor receptor 2 (HER2) overexpression. Indeed, 100% of HER2-positive intestinal gastric carcinomas, 100% of HER2-positive serous ovarian carcinomas, and 64% of HER2-positive breast carcinomas coexpressed WLS protein. Although HER2 protein expression or gene amplification is an established predictive biomarker for trastuzumab response in breast and gastric cancers, a significant proportion of HER2-positive tumors display resistance to trastuzumab, which may be in part explainable by a possible mechanistic link between WLS and HER2.
Collapse
|
31
|
Natarajan K, Baer MR, Ross DD. Role of Breast Cancer Resistance Protein (BCRP, ABCG2) in Cancer Outcomes and Drug Resistance. RESISTANCE TO TARGETED ANTI-CANCER THERAPEUTICS 2015. [DOI: 10.1007/978-3-319-09801-2_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
32
|
Comprehensive profiling of EGFR/HER receptors for personalized treatment of gynecologic cancers. Mol Diagn Ther 2014; 18:137-51. [PMID: 24403167 DOI: 10.1007/s40291-013-0070-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The primary gynecologic cancers include cancers of the endometrium, ovary, and cervix. Worldwide, cervical cancer is the most common gynecologic cancer, whereas endometrial cancer is the most common in the US. Ovarian cancer is the fifth most deadly cancer in women, with 5-year survival rates for advanced disease at only 27 %. As such, there is an urgent need for reliable screening tools and novel targeted therapeutic regimens for these malignancies. The epidermal growth factor receptor (EGFR)/human EGFR (HER) family of receptors has been associated with the development and progression of many solid tumors. Despite clear roles for these receptors in other cancers, the expression of HER family members in gynecologic cancers and their relationship with disease stage, grade, and response to treatment remain controversial. In this review, we describe the existing evidence for the use of HER family members as diagnostic and prognostic indicators as well as their potential as therapeutic targets in gynecologic cancers.
Collapse
|
33
|
Missaoui N, Abdelkarim SB, Ayachi M, Hmissa S, Yaacoubi MT. HER2 expression in ovarian mucinous carcinomas in Tunisia. Asian Pac J Cancer Prev 2014; 15:8121-5. [PMID: 25338994 DOI: 10.7314/apjcp.2014.15.19.8121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ovarian mucinous carcinoma has a poor prognosis in advanced stages and a poor response to conventional chemotherapy. An efficient treatment is not yet available. We heere investigated HER2 expression and the potential for trastuzumab therapy in ovarian mucinous tumors. MATERIALS AND METHODS Immunohistochemistry was performed in formalin-fixed, paraffin-embedded tissue from 27 ovarian mucinous tumors including 14 carcinomas and 13 borderline tumors diagnosed in the Pathology Department, Farhet Hached Hospital, Sousse, between 1993 and 2013. The HercepTest (DAKO) was used for immunohistochemistry. RESULTS HER2 expression was observed in only one borderline tumor (7.7%) and in 14.3% of mucinous carcinomas of the ovary. CONCLUSIONS Our results suggest that trastuzumab therapy would be an option for patients with mucinous carcinoma when the tumor has HER2 overexpression.
Collapse
Affiliation(s)
- Nabiha Missaoui
- Research Unit03/UR/08-13, Cancer Epidemiology and Cytopathology in Tunisian Center, Faculty of Medicine, Sousse, Tunisia E-mail :
| | | | | | | | | |
Collapse
|
34
|
Zhang X, Li F, Guo S, Chen X, Wang X, Li J, Gan Y. Biofunctionalized polymer-lipid supported mesoporous silica nanoparticles for release of chemotherapeutics in multidrug resistant cancer cells. Biomaterials 2014; 35:3650-65. [DOI: 10.1016/j.biomaterials.2014.01.013] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 01/08/2014] [Indexed: 10/25/2022]
|
35
|
Yu J, Ma Y, Drisko J, Chen Q. Antitumor Activities of Rauwolfia vomitoria Extract and Potentiation of Carboplatin Effects Against Ovarian Cancer. CURRENT THERAPEUTIC RESEARCH 2014; 75:8-14. [PMID: 24465036 PMCID: PMC3898180 DOI: 10.1016/j.curtheres.2013.04.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 04/26/2013] [Indexed: 11/17/2022]
Abstract
Background Tumor resistance to platinum-based drugs has been an obstacle to the treatment of ovarian cancer. Extract of the plant Rauwolfia vomitoria has long been used by cancer patients. However, there have not been systematic studies of its anticancer activity. Objective In an effort to enhance the effectiveness of platinum-based drugs, we investigated the anticancer effect of a Rauwolfia vomitoria extract (Rau), both alone and in combination with carboplatin (Cp). Methods In vitro cytotoxicity and colony formation were evaluated in several ovarian cancer cell lines. In vivo effects were evaluated in an intraperitoneal ovarian cancer mouse model. The combination of Rau and Cp was assessed using Chou-Talalay’s constant ratio design and median effect analysis based on the isobologram principle to determine the combination index values. Results Rau decreased cell growth in all 3 tested ovarian cancer cell lines dose dependently and completely inhibited formation of colonies in soft agar. Apoptosis was induced in a time- and dose-dependent manner and was the predominant form of Rau-induced cell death. Synergy of Rau with Cp was detected, with combination index values <1 and dose reduction index values for Cp ranging from 1.7- to 7-fold. Tumor growth in mice was significantly suppressed by 36% or 66% with Rau treatment alone at a low (20 mg/kg) or a high dose (50 mg/kg), respectively, an effect comparable to that of Cp alone. The volume of ascitic fluid and the number of nonblood cells in ascites were also significantly decreased. Combining Rau with Cp remarkably enhanced the effect of Cp and reduced tumor burden by 87% to 90% and ascites volume by 89% to 97%. Conclusions Rau has potent antitumor activity and in combination significantly enhances the effect of Cp against ovarian cancer.
Collapse
Affiliation(s)
- Jun Yu
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas ; KU Integrative Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Yan Ma
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas ; KU Integrative Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Jeanne Drisko
- KU Integrative Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Qi Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas ; KU Integrative Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
36
|
Phase I dosage finding and pharmacokinetic study of intravenous topotecan and oral erlotinib in adults with refractory solid tumors. Cancer Chemother Pharmacol 2014; 73:561-8. [PMID: 24448640 DOI: 10.1007/s00280-014-2385-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/10/2014] [Indexed: 12/22/2022]
Abstract
PURPOSE Topotecan is widely used for refractory solid tumors but multi-drug resistance may occur due to tumor expression of ATP-binding cassette (ABC) transporters. Since erlotinib, an inhibitor of the epidermal growth factor receptor, also inhibits several ABC transporters, we performed a phase I study to evaluate the safety, efficacy, and pharmacokinetics of intravenous topotecan given in combination with erlotinib. METHODS Patients received 150 mg of oral erlotinib daily and a 30 min intravenous infusion of topotecan on days 1-5 of a 21-day cycle. Dosage escalation of topotecan occurred with a starting dosage of 0.75 mg/m(2). The pharmacokinetics of topotecan was evaluated on day 1 of cycle 1 without erlotinib and on day 1 of cycle 2 or 3 with erlotinib. RESULTS Twenty-nine patients were enrolled. The maximum tolerated dosage was determined to be 1.0 mg/m(2). Dose-limiting toxicities included neutropenia and thrombocytopenia. The average duration of treatment was 97 days. Two partial responses were observed. Topotecan clearance and exposure were similar with and without erlotinib. CONCLUSIONS The combination of topotecan and erlotinib is tolerable at clinically effective doses. Erlotinib does not affect the disposition of topotecan to a clinically significant extent.
Collapse
|
37
|
Yu J, Chen Q. The plant extract of Pao pereira potentiates carboplatin effects against ovarian cancer. PHARMACEUTICAL BIOLOGY 2014; 52:36-43. [PMID: 24033267 DOI: 10.3109/13880209.2013.808232] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
CONTEXT Herbal preparation of Pao pereira [Geissospermum vellosii Allem (Apocynaceae)] has long been used by oncologic patients and Integrative Medicine practitioners in South America. However, its anticancer activities have not been systematically studied. OBJECTIVE To investigate the anticancer effects of β-carboline alkaloids-enriched extract from Pao pereira (Pao), either alone or in combination with carboplatin, in preclinical ovarian cancer models. MATERIALS AND METHODS Cytotoxicity of Pao (0-800 µg/ml) against different ovarian cancer cell lines and an immortalized epithelial cell line was detected by flow cytometry, MTT assay and colony formation in soft agar. Combination of Pao and carboplatin, a primary chemotherapeutic drug for ovarian cancer, was evaluated using Chou-Talalay's methods. Mice bearing intraperitoneally spread ovarian cancer were treated with 20 or 50 mg/kg/day Pao by i.p. injection. Carboplatin at 15 mg/kg/week i.p. was compared and combined to Pao treatments. RESULTS Pao selectively inhibited ovarian cancer cell growth with IC₅₀ values of 180-235 µg/ml, compared to 537 µg/ml in normal cells. Pao induced apoptosis dose- and time-dependently and completely inhibited colony formation of tumor cells in soft agar at 400 µg/ml. Pao greatly enhanced carboplatin cytotoxicity, with dose reduction (DRIs) for carboplatin at 1.2-10 fold. In vivo, Pao alone suppressed tumor growth by 79% and decreased volume of ascites by 55%. When Pao was combined with carboplatin, tumor inhibition reached 97% and ascites was completely eradicated. DISCUSSION AND CONCLUSION Pao possess potent antitumor activity and could enhance carboplatin effect, and therefore holds therapeutic potential in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Jun Yu
- Department of Pharmacology, Toxicology and Therapeutics, KU Integrative Medicine, University of Kansas Medical Center , Kansas City, KS , USA
| | | |
Collapse
|
38
|
Becker MA, Farzan T, Harrington SC, Krempski JW, Weroha SJ, Hou X, Kalli KR, Wong TW, Haluska P. Dual HER/VEGF receptor targeting inhibits in vivo ovarian cancer tumor growth. Mol Cancer Ther 2013; 12:2909-16. [PMID: 24130056 DOI: 10.1158/1535-7163.mct-13-0547] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Ovarian cancer mortality ranks highest among all gynecologic cancers with growth factor pathways playing an integral role in tumorigenesis, metastatic dissemination, and therapeutic resistance. The HER and VEGF receptor (VEGFR) are both overexpressed and/or aberrantly activated in subsets of ovarian tumors. While agents targeting either the HER or VEGF pathways alone have been investigated, the impact of these agents have not led to overall survival benefit in ovarian cancer. We tested the hypothesis that cotargeting HER and VEGFR would maximize antitumor efficacy at tolerable doses. To this end, ovarian cancer xenografts grown intraperitoneally in athymic nude mice were tested in response to AC480 (pan-HER inhibitor, "HERi"), cediranib (pan-VEGFR inhibitor "VEGFRi"), or BMS-690514 (combined HER/VEGFR inhibitor "EVRi"). EVRi was superior to both HERi and VEGFRi in terms of tumor growth, final tumor weight, and progression-free survival. Correlative tumor studies employing phosphoproteomic antibody arrays revealed distinct agent-specific alterations, with EVRi inducing the greatest overall effect on growth factor signaling. These data suggest that simultaneous inhibition of HER and VEGFR may benefit select subsets of ovarian cancer tumors. To this end, we derived a novel HER/VEGF signature that correlated with poor overall survival in high-grade, late stage, serous ovarian cancer patient tumors.
Collapse
Affiliation(s)
- Marc A Becker
- Corresponding Author: Paul Haluska, Division of Medical Oncology, Mayo Clinic College of Medicine, 200 First St. SW, Rochester, MN 55905.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Leone Roberti Maggiore U, Valenzano Menada M, Venturini PL, Ferrero S. The potential of sunitinib as a therapy in ovarian cancer. Expert Opin Investig Drugs 2013; 22:1671-86. [PMID: 24070205 DOI: 10.1517/13543784.2013.841138] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Sunitinib malate (SU11248; Sutent®; Pfizer, Inc., New York) is a multi-kinase inhibitor currently approved for use in advanced renal cell carcinoma (RCC), imatinib-resistant/-intolerant gastrointestinal stromal tumours and progressive, well-differentiated pancreatic neuroendocrine tumours in patients with unresectable, locally advanced or metastatic disease. AREAS COVERED This article describes the mechanism of action and of the pharmacokinetics of sunitinib; further, it summarizes Phase I and II trials on the clinical efficacy, tolerability and safety of this agent in the setting of ovarian cancer (OC) treatment. EXPERT OPINION On the basis of the current literature, sunitinib has shown modest antitumour activity and acceptable toxicity. Studies investigating the impact of horizontal and vertical combinations should represent a priority of future research. Although clinical Phase II trials on the use of sunitinib in the treatment of OC demonstrated an acceptable profile of AEs, a greater comprehension of the toxicity of this compound is recommended.
Collapse
Affiliation(s)
- Umberto Leone Roberti Maggiore
- University of Genoa, San Martino Hospital and National Institute for Cancer Research, Department of Obstetrics and Gynecology , Largo R. Benzi 1, 16132 Genoa , Italy +01139 010511525 ; +01139 010511525 ;
| | | | | | | |
Collapse
|
40
|
Shaw HM, Hall M. Emerging treatment options for recurrent ovarian cancer: the potential role of olaparib. Onco Targets Ther 2013; 6:1197-206. [PMID: 24043945 PMCID: PMC3772750 DOI: 10.2147/ott.s30748] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Olaparib has shown promising anticancer activity as a single agent in the treatment and maintenance of recurrent ovarian cancer in early clinical trials, but it is far from standard therapy. This article outlines the problem of relapsed ovarian cancer and the mechanisms of poly(ADP-ribose) polymerase inhibitors and reviews the recent literature pertaining to olaparib in ovarian cancer.
Collapse
Affiliation(s)
- Heather M Shaw
- Mount Vernon Cancer Centre, Rickmansworth Road, Northwood, Middlesex, United Kingdom
| | | |
Collapse
|
41
|
Morotti M, Becker CM, Menada MV, Ferrero S. Targeting tyrosine-kinases in ovarian cancer. Expert Opin Investig Drugs 2013; 22:1265-79. [PMID: 23815710 DOI: 10.1517/13543784.2013.816282] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Epithelial ovarian cancer (EOC) is the leading cause of gynaecologic cancer death. Although in some cases initial treatment is effective, most of the women diagnosed with EOC will probably need medical treatment for their disease. There is a critical need to develop effective new strategies for the management of patients with advanced or recurrent EOC, and targeted therapy with tyrosine kinase inhibitors (TKIs) has continued to be an area of active research and development in this setting. AREAS COVERED This review summarises the available evidence on the use of TKIs in the clinical management of women with EOC. This article consists of material obtained via Medline, PubMed and EMBASE literature searches up to March 2013. EXPERT OPINION Several Phase I/II and III trials evaluated TKIs in EOC; however, it is difficult to draw conclusions on the efficacy of TKI regimens in these patients. TKIs seem to be better tolerated than conventional chemotherapy with a different toxicity profile. A better understanding of the signalling pathways, the toxicity profiles, the potential pharmacokinetic interactions as well as the identification of predictive biomarkers are needed to better identify a targeted patient population before these agents become part of routine treatment.
Collapse
Affiliation(s)
- Matteo Morotti
- University of Genoa, San Martino Hospital, Department of Obstetrics and Gynaecology , Largo R. Benzi 1, 16132 Genoa , Italy +01139010511525 ; +01139010511525 ;
| | | | | | | |
Collapse
|
42
|
Smolle E, Taucher V, Pichler M, Petru E, Lax S, Haybaeck J. Targeting signaling pathways in epithelial ovarian cancer. Int J Mol Sci 2013; 14:9536-55. [PMID: 23644885 PMCID: PMC3676798 DOI: 10.3390/ijms14059536] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 04/13/2013] [Accepted: 04/22/2013] [Indexed: 12/16/2022] Open
Abstract
Ovarian carcinoma (OC) is the most lethal gynecological malignancy. Response to platinum-based chemotherapy is poor in some patients and, thus, current research is focusing on new therapy options. The various histological types of OC are characterized by distinctive molecular genetic alterations that are relevant for ovarian tumorigenesis. The understanding of these molecular pathways is essential for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Elisabeth Smolle
- Institute of Pathology, Medical University Graz, Auenbruggerplatz 25, A-8036 Graz, Austria; E-Mails: (E.S.); (V.T.)
| | - Valentin Taucher
- Institute of Pathology, Medical University Graz, Auenbruggerplatz 25, A-8036 Graz, Austria; E-Mails: (E.S.); (V.T.)
| | - Martin Pichler
- Department of Internal Medicine, Division of Clinical Oncology, Medical University Graz, A-8036 Graz, Austria; E-Mail:
| | - Edgar Petru
- Department of Obstetrics and Gynecology, Medical University Graz, A-8036 Graz, Austria; E-Mail:
| | - Sigurd Lax
- Department of Pathology, General Hospital Graz West, Goestinger Straße 22, A-8020 Graz, Austria
- Authors to whom correspondence should be addressed; E-Mails: (S.L.); (J.H.); Tel.: +43-316-5466-4652 (S.L.); +43-316-385-80594 (J.H.); Fax: +43-316-5466-74652 (S.L.); +43-316-384-329 (J.H.)
| | - Johannes Haybaeck
- Institute of Pathology, Medical University Graz, Auenbruggerplatz 25, A-8036 Graz, Austria; E-Mails: (E.S.); (V.T.)
- Authors to whom correspondence should be addressed; E-Mails: (S.L.); (J.H.); Tel.: +43-316-5466-4652 (S.L.); +43-316-385-80594 (J.H.); Fax: +43-316-5466-74652 (S.L.); +43-316-384-329 (J.H.)
| |
Collapse
|
43
|
Expected Benefits of Topotecan Combined With Lapatinib in Recurrent Ovarian Cancer According to Biological Profile. Int J Gynecol Cancer 2012; 22:1483-8. [DOI: 10.1097/igc.0b013e31826d1438] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
44
|
Hillig T, Thode J, Breinholt MF, Franzmann MB, Pedersen C, Lund F, Mygind H, Sölétormos G, Rudnicki M. Assessing HER2 amplification by IHC, FISH, and real-time polymerase chain reaction analysis (real-time PCR) following LCM in formalin-fixed paraffin embedded tissue from 40 women with ovarian cancer. APMIS 2012; 120:1000-7. [PMID: 23030524 PMCID: PMC3533780 DOI: 10.1111/j.1600-0463.2012.02929.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 05/07/2012] [Indexed: 12/21/2022]
Abstract
We compare HER2 receptor amplification analysis by immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), and real-time polymerase chain reaction (real-time PCR) DNA copy-number assay following laser capture microdissection (LCM) in formalin-fixed paraffin embedded tissue from 40 women with verified ovarian cancer. We speculate that LCM should result in a more accurate assessment of HER2 amplification in our real-time PCR assay compared with IHC and FISH. HER2 overexpression measured by IHC, FISH, or real-time PCR was found in 5.0%, 5.0%, and 22.5%, respectively. HER2 negative results measured by IHC, FISH, or real-time PCR were found in 95%, 92.5%, and 60.0%, respectively. Analysis failed for IHC, FISH, or real-time PCR in 0%, 2.5%, or 17.5% of cases. Concordance between IHC and FISH, IHC and real-time PCR, or FISH and real-time PCR were 89.7%, 72.7%, or 78.1%, respectively. Only few ovarian cancer patients were HER2 overexpressed measured by IHC or FISH and thus could be eligible for antibody-based therapy with trastuzumab (Herceptin). Interestingly, we find an increased number of HER2 positive patients by real-time PCR analysis on microdissected cancer cells, suggesting a number of HER2 positive patients not detected by current methods. Thus, the concept of quantitative measurement of HER2 on microdissected cancer cells should be explored further.
Collapse
Affiliation(s)
- Thore Hillig
- Department of Clinical Biochemistry, Hillerød Hospital University of Copenhagen, 3400 Hillerød, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Shigetomi H, Higashiura Y, Kajihara H, Kobayashi H. Targeted molecular therapies for ovarian cancer: an update and future perspectives (Review). Oncol Rep 2012; 28:395-408. [PMID: 22641286 DOI: 10.3892/or.2012.1833] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 05/03/2012] [Indexed: 11/06/2022] Open
Abstract
Identification of the potential gene expression profiles of epithelial ovarian cancer and the arrival of newly targeted therapies have advanced the strategies used for treatment of this disease. This review focuses on the design of ongoing and planned clinical trials and offers a synopsis of the English-language literature for preclinical and clinical targeted therapies for epithelial ovarian cancer. Among many targeted agents, a promising, novel class of targeted drugs for special patient populations expected to improve the effectiveness of current therapy include inhibitors of angiogenesis, poly (ADP ribose) polymerase (PARP) and DNA repair mechanisms. Inhibition of PARP or homologous recombination (HR) repair mediated by Chk1 (checkpoint kinase 1) would selectively sensitize p53 mutation, BRCAness phenotype (serous type ovarian cancer) or HNF (hepatocyte nuclear factor)-1β-overexpressing tumor cells (clear cell type ovarian cancer) to chemotherapeutic agents. The therapeutic response is likely to be limited to a targeted patient, but not to the broad population. This review discusses some of the key current developments and existing challenges.
Collapse
Affiliation(s)
- Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| | | | | | | |
Collapse
|
46
|
Wilken JA, Badri T, Cross S, Raji R, Santin AD, Schwartz P, Branscum AJ, Baron AT, Sakhitab AI, Maihle NJ. EGFR/HER-targeted therapeutics in ovarian cancer. Future Med Chem 2012; 4:447-69. [PMID: 22416774 PMCID: PMC4620931 DOI: 10.4155/fmc.12.11] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Despite decades of research and evolving treatment modalities, survival among patients with epithelial ovarian cancer has improved only incrementally. During this same period, the development of biologically targeted therapeutics has improved survival for patients with diverse malignancies. Many of these new drugs target the human epidermal growth factor receptor (EGFR/HER/ErbB) family of tyrosine kinases, which play a major role in the etiology and progression of many carcinomas, including epithelial ovarian cancer. While several HER-targeted therapeutics are US FDA approved for the treatment of various malignancies, none have gained approval for the treatment of ovarian cancer. Here, we review the published literature on HER-targeted therapeutics for the treatment of ovarian cancer, including novel HER-targeted therapeutics in various stages of clinical development, as well as the challenges that have limited the use of these inhibitors in clinical settings.
Collapse
Affiliation(s)
- Jason A Wilken
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Tayf Badri
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Sarah Cross
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Rhoda Raji
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Alessandro D Santin
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Peter Schwartz
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Adam J Branscum
- Oregon State University, School of Biological & Population Health Sciences
| | - Andre T Baron
- University of Kentucky, Departments of Epidemiology, & Obstetrics & Gynecology
| | - Adam I Sakhitab
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Nita J Maihle
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
- Yale University, Departments of Pathology & Pharmacology
- PO Box 208063, 333 Cedar Street, New Haven, CT 06520, USA
| |
Collapse
|
47
|
Ovarian cancer: opportunity for targeted therapy. JOURNAL OF ONCOLOGY 2011; 2012:682480. [PMID: 22235203 PMCID: PMC3253450 DOI: 10.1155/2012/682480] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Accepted: 11/01/2011] [Indexed: 11/30/2022]
Abstract
Ovarian cancer is a common cause of cancer mortality in women with limited treatment effectiveness in advanced stages. The limitation to treatment is largely the result of high rates of cancer recurrence despite chemotherapy and eventual resistance to existing chemotherapeutic agents. The objective of this paper is to review current concepts of ovarian carcinogenesis. We will review existing hypotheses of tumor origin from ovarian epithelial cells, Fallopian tube, and endometrium. We will also review the molecular pathogenesis of ovarian cancer which results in two specific pathways of carcinogenesis: (1) type I low-grade tumor and (2) type II high-grade tumor. Improved understanding of the molecular basis of ovarian carcinogenesis has opened new opportunities for targeted therapy. This paper will also review these potential therapeutic targets and will explore new agents that are currently being investigated.
Collapse
|