1
|
Sato S, Shintani D, Kaneda Y, Nakamura R, Katoh T, Yano M, Hanaoka M, Yagishita S, Yasuda M, Nagata M, Hasegawa K. Human epidermal growth factor receptor 3 expression in patients with epithelial ovarian cancer: a potential target for ovarian mucinous and clear cell carcinoma. Int J Clin Oncol 2025; 30:805-813. [PMID: 39937426 DOI: 10.1007/s10147-024-02658-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/09/2024] [Indexed: 02/13/2025]
Abstract
BACKGROUND Human epidermal growth factor receptor 3 (HER3), a tyrosine kinase belonging to the HER family, is a known target for cancer therapy; recently, an anti-HER3 antibody-drug conjugate (ADC) is developing. To understand HER3 expression in epithelial ovarian cancer (EOC), this study was conducted. METHODS We investigated the expression of HER3 in 202 patients with EOC using immunohistochemistry (IHC), and the association between HER3 expression, clinicopathological features, prognosis, and treatment timing. RESULTS Of all the cases, 55.4% had a HER3 IHC score ≥ 1 + . In particular, 78.0% of the patients with clear cell carcinoma (CCC) and 87.9% of the patients with mucinous carcinoma (MC) had a HER3 IHC score ≥ 1 + . Regarding clinicopathological features, early disease stage, feasibility of primary debulking surgery, no residual tumor, and low CA125 levels were more frequently observed in patients with a HER3 IHC score ≥ 1 + . Furthermore, a HER3 no-expression showed a significant association with a relatively short progression-free survival (PFS). And, for patients with mucinous carcinoma, those with a HER3 IHC score ≥ 1 + had poorer PFS and overall survival than those with a HER3 no-expression (no statistically significant difference). In addition, we analyzed HER3 expression at primary tumor and recurrence tumor in same patients. Thus, we observed the HER3 IHC score tended to change from 0 to ≥ 1 + in recurrence cases compared with primary cases. CONCLUSIONS These observations suggested that patients with MC, CCC and recurrence of all histological type may potentially benefit from future clinical trials of HER3-directed therapies.
Collapse
MESH Headings
- Humans
- Female
- Middle Aged
- Carcinoma, Ovarian Epithelial/pathology
- Adenocarcinoma, Clear Cell/pathology
- Adenocarcinoma, Clear Cell/metabolism
- Adenocarcinoma, Clear Cell/drug therapy
- Adenocarcinoma, Clear Cell/genetics
- Aged
- Adenocarcinoma, Mucinous/pathology
- Adenocarcinoma, Mucinous/metabolism
- Adenocarcinoma, Mucinous/drug therapy
- Adult
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/drug therapy
- Receptor, ErbB-3/metabolism
- Receptor, ErbB-3/genetics
- Receptor, ErbB-3/biosynthesis
- Prognosis
- Aged, 80 and over
- Biomarkers, Tumor
- Immunohistochemistry
- Progression-Free Survival
Collapse
Affiliation(s)
- Sho Sato
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Daisuke Shintani
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Yuki Kaneda
- Translational Science Department I, Daiichi Sankyo Co., Ltd., Shinagawa, Tokyo, 140-8710, Japan
| | - Ryuichi Nakamura
- Translational Science Department I, Daiichi Sankyo Co., Ltd., Shinagawa, Tokyo, 140-8710, Japan
| | - Tomomi Katoh
- Department of Pathology, Saitama Medical University International Medical, Center, Hidaka, Saitama, 350-1298, Japan
| | - Mitsutake Yano
- Department of Pathology, Saitama Medical University International Medical, Center, Hidaka, Saitama, 350-1298, Japan
| | - Mieko Hanaoka
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Shigehiro Yagishita
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tsukiji, Tokyo, Japan
- Department of Pharmacology and Therapeutics, National Cancer Center Research Institute, Tsukiji, Tokyo, Japan
| | - Masanori Yasuda
- Department of Pathology, Saitama Medical University International Medical, Center, Hidaka, Saitama, 350-1298, Japan
| | - Motoko Nagata
- Translational Science Department I, Daiichi Sankyo Co., Ltd., Shinagawa, Tokyo, 140-8710, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka, Saitama, 350-1298, Japan.
| |
Collapse
|
2
|
Abdelsalam RM, Hamam HW, Eissa NM, El-Sahar AE, Essam RM. Empagliflozin Dampens Doxorubicin-Induced Chemobrain in Rats: The Possible Involvement of Oxidative Stress and PI3K/Akt/mTOR/NF-κB/TNF-α Signaling Pathways. Mol Neurobiol 2025; 62:3480-3492. [PMID: 39302617 DOI: 10.1007/s12035-024-04499-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Chemobrain is a cognitive impairment observed in up to 75% of cancer patients treated with doxorubicin (DOX). Cognitive deficits associated with DOX are complex, and multiple interplay pathways contribute to memory impairment and the loss of concentration. Empagliflozin (EMPA), a sodium-glucose co-transporter-2 (SGLT-2) inhibitor with neuroprotective potential, has recently been elucidated because of its regulatory effects on oxidative stress and neuroinflammation. Thus, this study aimed to explore the protective mechanisms of EMPA in DOX-induced chemobrain. Rats were allocated to four groups: normal (NC), EMPA, DOX, and EMPA + DOX. Chemobrain was induced in the third and fourth groups by DOX (2 mg/kg, IP) on the 0th, 7th, 14th, and 21st days of the study, while EMPA was administered (10 mg/kg, PO) for 28 consecutive days in both the EMPA and EMPA + DOX groups. Behavioral and biochemical assessments were then performed. Rats treated with DOX exhibited significant memory, learning, and muscle coordination dysfunctions. Moreover, DOX boosted oxidative stress in the brain, as evidenced by elevated malondialdehyde (MDA) content together with decreased levels of nuclear factor-erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) and reduced glutathione (GSH). Neuroinflammation was also observed as an upsurge of tumor necrosis factor-alpha (TNF-α) and nuclear factor kappa B (NF-κB) (p65). Additionally, DOX diminished the expression of brain-derived neurotrophic factor (BDNF) and increased phosphoinositol-3-kinase (PI3K), phosphorylated-Akt (pAkt), and mammalian target of rapamycin (mTOR) content. EMPA exhibited potent neuroprotective potential in DOX-induced cognitive impairment, attributed to its antioxidant and neuroplasticity-enhancing properties and suppression of the PI3K/Akt/mTOR/NF-κB/TNF-α signaling pathway.
Collapse
Affiliation(s)
- Rania M Abdelsalam
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo, 11562, Egypt
| | - Hatem W Hamam
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt
| | - Noha M Eissa
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt
| | - Ayman E El-Sahar
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo, 11562, Egypt
| | - Reham M Essam
- Biology Department, School of Pharmacy, Newgiza University, Giza, Egypt.
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini St., Cairo, 11562, Egypt.
| |
Collapse
|
3
|
Bates M, Mohamed BM, Lewis F, O'Toole S, O'Leary JJ. Biomarkers in high grade serous ovarian cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189224. [PMID: 39581234 DOI: 10.1016/j.bbcan.2024.189224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024]
Abstract
High-grade serous ovarian cancer (HGSC) is the most common subtype of ovarian cancer. HGSC patients typically present with advanced disease, which is often resistant to chemotherapy and recurs despite initial responses to therapy, resulting in the poor prognosis associated with this disease. There is a need to utilise biomarkers to manage the various aspects of HGSC patient care. In this review we discuss the current state of biomarkers in HGSC, focusing on the various available immunohistochemical (IHC) and blood-based biomarkers, which have been examined for their diagnostic, prognostic and theranostic potential in HGSC. These include various routine clinical IHC biomarkers such as p53, WT1, keratins, PAX8, Ki67 and p16 and clinical blood-borne markers and algorithms such as CA125, HE4, ROMA, RMI, ROCA, and others. We also discuss various components of the liquid biopsy as well as a number of novel IHC biomarkers and non-routine blood-borne biomarkers, which have been examined in various ovarian cancer studies. We also discuss the future of ovarian cancer biomarker research and highlight some of the challenges currently facing the field.
Collapse
Affiliation(s)
- Mark Bates
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland.
| | - Bashir M Mohamed
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Faye Lewis
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland
| | - Sharon O'Toole
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin, Ireland
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland; Emer Casey Molecular Pathology Research Laboratory, Coombe Women & Infants University Hospital, Dublin, Ireland; Trinity St James's Cancer Institute, Dublin, Ireland; Department of Pathology, Coombe Women & Infants University Hospital, Dublin, Ireland
| |
Collapse
|
4
|
Bermúdez-Abreut E, Bergado Báez G, Martínez Pestano M, Attanasio G, Gonzales Castillo CY, Hernández Fernández DR, Alvarez-Arzola R, Alimonti A, Sánchez-Ramírez B. Antitumor activity of PAbs generated by immunization with a novel HER3-targeting protein-based vaccine candidate in preclinical models. Front Oncol 2024; 14:1472607. [PMID: 39479017 PMCID: PMC11521786 DOI: 10.3389/fonc.2024.1472607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Despite the cumulative evidence supporting HER3 as a target for antitumor therapies, no agents targeting HER3 have been approved for cancer treatment. Most of the agents evaluated in preclinical and clinical trials have been specific monoclonal antibodies (MAbs), with few examples of active immunotherapy directed against this receptor. However, some cancer vaccine formats may generate polyclonal antibodies (PAbs) that replicate the diverse effector mechanisms of MAbs, including ligand neutralization and receptor degradation. In this study, we developed a protein subunit-based monovalent vaccine candidate targeting the extracellular domain (ECD) of HER3. Immunization of mice with a formulation targeting murine ErbB3-ECD successfully overcome tolerance to this self-antigen, inducing high titers of ErbB3-specific PAbs. The antitumor potential of this formulation and the induced PAbs was demonstrated in vivo and in vitro in an ErbB3-overexpressing 3LL-D122-derived tumor model. The immunogenicity of the HER3-ECD-based vaccine candidate was confirmed by the induction of high titers of HER3-specific PAbs. Consistent with the initial results, HER3-ECD-targeting PAbs were cytotoxic in several human epithelial tumor cell lines and exerted antitumor effects in vivo. These results support the value of HER3 as a tumor antigen and the effector mechanisms of HER3-specific therapeutic MAbs, while suggesting the potential of the proposed vaccine candidate for the treatment of HER3-expressing carcinomas.
Collapse
Affiliation(s)
| | - Gretchen Bergado Báez
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | | | - Giuseppe Attanasio
- Department of Molecular Oncology, Institute of Oncology Research (IOR), Bellinzona, Switzerland
| | | | | | - Rydell Alvarez-Arzola
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Andrea Alimonti
- Department of Molecular Oncology, Institute of Oncology Research (IOR), Bellinzona, Switzerland
- Faculty of Medicine, Università della Svizzera Italiana, Lugano, Switzerland
- Department of Medicine, University of Padua, Padua, Italy
- Medical Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | | |
Collapse
|
5
|
Zeng H, Wang W, Zhang L, Lin Z. HER3-targeted therapy: the mechanism of drug resistance and the development of anticancer drugs. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:14. [PMID: 38835349 PMCID: PMC11149107 DOI: 10.20517/cdr.2024.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/04/2024] [Accepted: 04/23/2024] [Indexed: 06/06/2024]
Abstract
Human epidermal growth factor receptor 3 (HER3), which is part of the HER family, is aberrantly expressed in various human cancers. Since HER3 only has weak tyrosine kinase activity, when HER3 ligand neuregulin 1 (NRG1) or neuregulin 2 (NRG2) appears, activated HER3 contributes to cancer development and drug resistance by forming heterodimers with other receptors, mainly including epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2). Inhibition of HER3 and its downstream signaling, including PI3K/AKT, MEK/MAPK, JAK/STAT, and Src kinase, is believed to be necessary to conquer drug resistance and improve treatment efficiency. Until now, despite multiple anti-HER3 antibodies undergoing preclinical and clinical studies, none of the HER3-targeted therapies are licensed for utilization in clinical cancer treatment because of their safety and efficacy. Therefore, the development of HER3-targeted drugs possessing safety, tolerability, and sensitivity is crucial for clinical cancer treatment. This review summarizes the progress of the mechanism of HER3 in drug resistance, the HER3-targeted therapies that are conducted in preclinical and clinical trials, and some emerging molecules that could be used as future designed drugs for HER3, aiming to provide insights for future research and development of anticancer drugs targeting HER3.
Collapse
Affiliation(s)
- Huilan Zeng
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Wei Wang
- Department of Cancer Center, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China
| | - Lin Zhang
- Department of Gastroenterology, Chongqing University Jiangjin Hospital, Chongqing 402260, China
| | - Zhenghong Lin
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| |
Collapse
|
6
|
Majumder A. HER3: Toward the Prognostic Significance, Therapeutic Potential, Current Challenges, and Future Therapeutics in Different Types of Cancer. Cells 2023; 12:2517. [PMID: 37947595 PMCID: PMC10648638 DOI: 10.3390/cells12212517] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/14/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023] Open
Abstract
Human epidermal growth factor receptor 3 (HER3) is the only family member of the EGRF/HER family of receptor tyrosine kinases that lacks an active kinase domain (KD), which makes it an obligate binding partner with other receptors for its oncogenic role. When HER3 is activated in a ligand-dependent (NRG1/HRG) or independent manner, it can bind to other receptors (the most potent binding partner is HER2) to regulate many biological functions (growth, survival, nutrient sensing, metabolic regulation, etc.) through the PI3K-AKT-mTOR pathway. HER3 has been found to promote tumorigenesis, tumor growth, and drug resistance in different cancer types, especially breast and non-small cell lung cancer. Given its ubiquitous expression across different solid tumors and role in oncogenesis and drug resistance, there has been a long effort to target HER3. As HER3 cannot be targeted through its KD with small-molecule kinase inhibitors via the conventional method, pharmaceutical companies have used various other approaches, including blocking either the ligand-binding domain or extracellular domain for dimerization with other receptors. The development of treatment options with anti-HER3 monoclonal antibodies, bispecific antibodies, and different combination therapies showed limited clinical efficiency for various reasons. Recent reports showed that the extracellular domain of HER3 is not required for its binding with other receptors, which raises doubt about the efforts and applicability of the development of the HER3-antibodies for treatment. Whereas HER3-directed antibody-drug conjugates showed potentiality for treatment, these drugs are still under clinical trial. The currently understood model for dimerization-induced signaling remains incomplete due to the absence of the crystal structure of HER3 signaling complexes, and many lines of evidence suggest that HER family signaling involves more than the interaction of two members. This review article will significantly expand our knowledge of HER3 signaling and shed light on developing a new generation of drugs that have fewer side effects than the current treatment regimen for these patients.
Collapse
Affiliation(s)
- Avisek Majumder
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
7
|
Gandullo-Sánchez L, Ocaña A, Pandiella A. HER3 in cancer: from the bench to the bedside. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:310. [PMID: 36271429 PMCID: PMC9585794 DOI: 10.1186/s13046-022-02515-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/07/2022] [Indexed: 11/15/2022]
Abstract
The HER3 protein, that belongs to the ErbB/HER receptor tyrosine kinase (RTK) family, is expressed in several types of tumors. That fact, together with the role of HER3 in promoting cell proliferation, implicate that targeting HER3 may have therapeutic relevance. Furthermore, expression and activation of HER3 has been linked to resistance to drugs that target other HER receptors such as agents that act on EGFR or HER2. In addition, HER3 has been associated to resistance to some chemotherapeutic drugs. Because of those circumstances, efforts to develop and test agents targeting HER3 have been carried out. Two types of agents targeting HER3 have been developed. The most abundant are antibodies or engineered antibody derivatives that specifically recognize the extracellular region of HER3. In addition, the use of aptamers specifically interacting with HER3, vaccines or HER3-targeting siRNAs have also been developed. Here we discuss the state of the art of the preclinical and clinical development of drugs aimed at targeting HER3 with therapeutic purposes.
Collapse
Affiliation(s)
- Lucía Gandullo-Sánchez
- grid.428472.f0000 0004 1794 2467Instituto de Biología Molecular y Celular del Cáncer, CSIC, IBSAL and CIBERONC, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | - Alberto Ocaña
- grid.411068.a0000 0001 0671 5785Hospital Clínico San Carlos and CIBERONC, 28040 Madrid, Spain
| | - Atanasio Pandiella
- grid.428472.f0000 0004 1794 2467Instituto de Biología Molecular y Celular del Cáncer, CSIC, IBSAL and CIBERONC, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| |
Collapse
|
8
|
Zhang W, Liu C, Li J, Lu Y, Li H, Zhuang J, Ren X, Wang M, Sun C. Tanshinone IIA: New Perspective on the Anti-Tumor Mechanism of A Traditional Natural Medicine. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:209-239. [PMID: 34983327 DOI: 10.1142/s0192415x22500070] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The search for natural and efficacious antineoplastic drugs, with minimal toxicity and side effects, is an important part of antitumor drug research and development. Tanshinone IIA is the most evaluated lipophilic active component of Salvia miltiorrhiza. Tanshinone IIA is a path-breaking traditional drug applied in cardiovascular treatment. It has also been found that tanshinone IIA plays an important role in the digestive, respiratory and circulatory systems, as well as in other tumor diseases. Tanshinone IIA significantly inhibits the proliferation of several types of tumors, blocks the cell cycle, induces apoptosis and autophagic death, in addition to inhibiting cell migration and invasion. Among these, the regulation of tumor-cell apoptosis signaling pathways is the key breakthrough point in several modes of antitumor therapy. The PI3K/AKT/MTOR signaling pathway and the JNK pathway are the key pathways for tanshinone IIA to induce tumor cell apoptosis. In addition to glycolysis, reactive oxygen species and signal transduction all play an active role with the participation of tanshinone IIA. Endogenous apoptosis is considered the main mechanism of tumor apoptosis induced by tanshinone IIA. Multiple pathways and targets play a role in the process of endogenous apoptosis. Tanshinone IIA can protect chemotherapy drugs, which is mainly reflected in the protection of the side effects of chemotherapy drugs, such as neurotoxicity and inhibition of the hematopoietic system. Tanshinone IIA also has a certain regulatory effect on tumor angiogenesis, which is mainly manifested in the control of hypoxia. Our findings indicated that tanshinone IIA is an effective treatment agent in the cardiovascular field and plays a significant role in antitumor therapeutics. This paper reviews the pharmacological potential and inhibitory effect of tanshinone IIA on cancer. It is greatly anticipated that tanshinone IIA will be employed as an adjuvant in the treatment of various cancers.
Collapse
Affiliation(s)
- Wenfeng Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P. R. China.,School of Traditional Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, P. R. China
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P. R. China
| | - Jie Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P. R. China
| | - Yiping Lu
- Integrated Traditional Chinese and Western Medicine Center, Department of Medicine, Qingdao University, Qingdao Shandong 266000, P. R. China
| | - Huayao Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P. R. China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Medicine Hospital, Weifang, Shandong 261041, P. R. China
| | - Xin Ren
- Clinical Medical Colleges, Weifang Medical University, Weifang, Shandong 261000, P. R. China
| | - Mengmeng Wang
- Clinical Medical Colleges, Weifang Medical University, Weifang, Shandong 261000, P. R. China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Medicine Hospital, Weifang, Shandong 261041, P. R. China.,Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, P. R. China
| |
Collapse
|
9
|
Basu A, Albert GK, Awshah S, Datta J, Kodumudi KN, Gallen C, Beyer A, Smalley KS, Rodriguez PC, Duckett DR, Forsyth PA, Soyano A, Koski GK, Lima Barros Costa R, Han H, Soliman H, Lee MC, Kalinski P, Czerniecki BJ. Identification of Immunogenic MHC Class II Human HER3 Peptides that Mediate Anti-HER3 CD4 + Th1 Responses and Potential Use as a Cancer Vaccine. Cancer Immunol Res 2022; 10:108-125. [PMID: 34785506 PMCID: PMC9414303 DOI: 10.1158/2326-6066.cir-21-0454] [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: 06/11/2021] [Revised: 09/01/2021] [Accepted: 11/16/2021] [Indexed: 01/11/2023]
Abstract
The HER3/ERBB3 receptor is an oncogenic receptor tyrosine kinase that forms heterodimers with EGFR family members and is overexpressed in numerous cancers. HER3 overexpression associates with reduced survival and acquired resistance to targeted therapies, making it a potential therapeutic target in multiple cancer types. Here, we report on immunogenic, promiscuous MHC class II-binding HER3 peptides, which can generate HER3-specific CD4+ Th1 antitumor immune responses. Using an overlapping peptide screening methodology, we identified nine MHC class II-binding HER3 epitopes that elicited specific Th1 immune response in both healthy donors and breast cancer patients. Most of these peptides were not identified by current binding algorithms. Homology assessment of amino acid sequence BLAST showed >90% sequence similarity between human and murine HER3/ERBB3 peptide sequences. HER3 peptide-pulsed dendritic cell vaccination resulted in anti-HER3 CD4+ Th1 responses that prevented tumor development, significantly delayed tumor growth in prevention models, and caused regression in multiple therapeutic models of HER3-expressing murine tumors, including mammary carcinoma and melanoma. Tumors were robustly infiltrated with CD4+ T cells, suggesting their key role in tumor rejection. Our data demonstrate that class II HER3 promiscuous peptides are effective at inducing HER3-specific CD4+ Th1 responses and suggest their applicability in immunotherapies for human HER3-overexpressing tumors.
Collapse
Affiliation(s)
- Amrita Basu
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Gabriella K. Albert
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Sabrina Awshah
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jashodeep Datta
- Department of Surgery, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, Florida
| | - Krithika N. Kodumudi
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Department of Oncological Sciences, University of South Florida, Tampa, Florida
| | - Corey Gallen
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Amber Beyer
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Keiran S.M. Smalley
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Paulo C. Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Derek R. Duckett
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Peter A. Forsyth
- Department of NeuroOncology and the NeuroOncology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aixa Soyano
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Gary K. Koski
- Department of Biological Sciences, Kent State University, Kent, Ohio
| | | | - Heather Han
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Hatem Soliman
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Marie Catherine Lee
- Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Pawel Kalinski
- Department of Immunology, Roswell Park Comprehensive Cancer Center, New York, New York
| | - Brian J. Czerniecki
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.,Corresponding Author: Brian J. Czerniecki, Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612. E-mail:
| |
Collapse
|
10
|
Zhu Y, Zhang H, Han X, Wang Z, Cui Y, Tian R, Wang Z, Han B, Tian J, Zhang F, Niu R. STAT3 mediated upregulation of C-MET signaling acts as a compensatory survival mechanism upon EGFR family inhibition in chemoresistant breast cancer cells. Cancer Lett 2021; 519:328-342. [PMID: 34348188 DOI: 10.1016/j.canlet.2021.07.048] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 07/10/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022]
Abstract
Chemotherapy remains the most common treatment for all types of breast cancer. Chemoresistance in tumors is still a major obstacle for treating late-stage breast cancer. In the process of acquiring resistance, tumor cells dynamically evolve to adapt to the challenge of anti-cancer drugs. Besides the upregulation of drug-pumps, signal pathways related to proliferation and survival undergo adaptive evolution. Thus, these drug-resistant cells are more conducive to proliferation, even in stressful conditions. Nevertheless, the detailed mechanism that drives cancer cells to sustain their proliferation ability is unclear. Herein, we reported that the upregulated C-MET signaling acts as a compensatory mechanism that sustains the proliferation of chemoresistant cells in which EGFR family signaling was attenuated. Both C-MET and EGFR family are essential for cell proliferation due to their activation of the STAT3 signaling. Different from other cell models in which C-MET interacts with and phosphorylates EGFR family members, our cell model showed no direct interaction between C-MET and EGFR family members. Therefore, C-MET and EGFR family signaling pathways function independently to sustain the proliferation of resistant cells. Moreover, chemoresistant cells have evolved a novel, STAT3-C-MET feed-forward loop that plays a vital role in sustaining cell proliferation. The activated STAT3 interacts with the MET gene promoter to upregulate its transcription. Most importantly, the combined inhibition of C-MET and EGFR family synergistically inhibits the proliferation of drug-resistant cells in vitro and in xenograft tumor models. This work provides a new strategy for treating drug-resistant breast cancer.
Collapse
Affiliation(s)
- Yuying Zhu
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - He Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Xingxing Han
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Zhiyong Wang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Yanfen Cui
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Ran Tian
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Zhaosong Wang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Baoai Han
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Jianfei Tian
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Fei Zhang
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Ruifang Niu
- Public Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| |
Collapse
|
11
|
Peng X, Yu M, Chen J. Transcriptome sequencing identifies genes associated with invasion of ovarian cancer. J Int Med Res 2021; 48:300060520950912. [PMID: 32878513 PMCID: PMC7780583 DOI: 10.1177/0300060520950912] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To identify key genes in ovarian cancer using transcriptome sequencing in two cell lines: MCV152 (benign ovarian epithelial tumour) and SKOV-3 (ovarian serous carcinoma). METHODS Differentially expressed genes (DEGs) between SKOV-3 and MCV152 were identified. Candidate genes were assessed for enrichment in gene ontology function and Kyoto Encyclopaedia of Genes and Genomes pathway. Candidate gene expression in SKOV-3 and MCV152 cells was validated using Western blots. RESULTS A total of 2020 upregulated and 1673 downregulated DEGs between SKOV3 and MCV152 cells were identified that were significantly enriched in the cell adhesion function. Upregulated DEGs, such as angiopoietin 2 (ANGPT2), CD19 molecule (CD19), collagen type IV alpha 3 chain (COL4A3), fibroblast growth factor 18 (FGF18), integrin subunit beta 4 (ITGB4), integrin subunit beta 8 (ITGB8), laminin subunit alpha 3 (LAMA3), laminin subunit gamma 2 (LAMC2), protein phosphatase 2 regulatory subunit Bgamma (PPP2R2C) and spleen associated tyrosine kinase (SYK) were significantly involved in the extracellular matrix-receptor interaction pathway. Downregulated DEGs, such as AKT serine/threonine kinase 3 (AKT3), collagen type VI alpha 1 chain (COL6A1), colony stimulating factor 3 (CSF3), fibroblast growth factor 1 (FGF1), integrin subunit alpha 2 (ITGA2), integrin subunit alpha 11 (ITGA11), MYB proto-oncogene, transcription factor (MYB), phosphoenolpyruvate carboxykinase 2, mitochondrial (PCK2), placental growth factor (PGF), phosphoinositide-3-kinase adaptor protein 1 (PIK3AP1), serum/glucocorticoid regulated kinase 1 (SGK1), toll like receptor 4 (TLR4) and tumour protein p53 (TP53) were involved in PI3K-Akt signalling. Expression of these DEGs was confirmed by Western blot analyses. CONCLUSION Candidate genes enriched in cell adhesion, extracellular matrix-receptor interaction and PI3K-Akt signalling pathways were identified that may be closely associated with ovarian cancer invasion and potential targets for ovarian cancer treatment.
Collapse
Affiliation(s)
- Xiandong Peng
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Min Yu
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Jiazhou Chen
- Shanghai Ji Ai Genetics and IVF Institute, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
12
|
Wang X, Zhang Y, Li W, Liu X. Knockdown of cir_RNA PVT1 Elevates Gastric Cancer Cisplatin Sensitivity via Sponging miR-152-3p. J Surg Res 2021; 261:185-195. [PMID: 33444948 DOI: 10.1016/j.jss.2020.12.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 11/08/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cisplatin (DDP) resistance is a key problem for effective treatment of gastric cancer (GC). Circular RNA PVT1 (circPVT1) acts as a vital regulator in the progression and development of various cancers. However, the in-depth mechanism of circPVT1 in GC resistance to DDP is still unclear. MATERIALS AND METHODS Quantitative real-time polymerase chain reaction was executed for the detection of the expression of circPVT1, miR-152-3p, and hepatoma-derived growth factor (HDGF) mRNA in GC tissues and cells. Western blot was used to detect the levels of HDGF protein, Bax, cleaved-casp-3, Bcl-2, p-PI3K, and p-AKT in tissue samples and/or cells. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and flow cytometry assays were performed to determine the viability, proliferation, and apoptosis of DDP-resistant GC cells. The relationship between miR-152-3p and circPVT1 or HDGF was confirmed by dual-luciferase reporter assay. The biological role of circPVT1 in vivo was confirmed with a xenograft tumor model. RESULTS CircPVT1 and HDGF mRNA were upregulated while miR-152-3p was downregulated in chemoresistance tissues and DDP-resistant GC cells. Both circPVT1 and HDGF inhibition elevated cell sensitivity to DDP, suppressed cell viability, proliferation, and induced cell apoptosis in DDP-resistant GC cells. The MiR-152-3p inhibitor reversed the influence of circPVT1 silencing on DDP sensitivity, viability, proliferation, and apoptosis of DDP-resistant GC cells. Moreover, circPVT1 regulated the HDGF/PI3K/AKT pathway through sponging miR-152-3p. In addition, circPVT1 knockdown reduced the malignancy of DDP-resistant GC cells in vivo. CONCLUSIONS CircPVT1 regulated the chemoresistance and malignancy of GC through modulating HDGF expression via sponging miR-152-3p, providing a theoretical basis for the development of effective therapeutic strategies for GC.
Collapse
Affiliation(s)
- Xiaojie Wang
- Department of Oncology, Dongying People's Hospital, Dongying, Shandong Province, China
| | - Ying Zhang
- Department of Blood Transfusion, Dongying People's Hospital, Dongying, Shandong Province, China
| | - Wei Li
- Department of Clinical Laboratory, Dongying People's Hospital, Dongying, Shandong Province, China
| | - Xiaolei Liu
- Department of Clinical Laboratory, Dongying People's Hospital, Dongying, Shandong Province, China.
| |
Collapse
|
13
|
Haikala HM, Jänne PA. Thirty Years of HER3: From Basic Biology to Therapeutic Interventions. Clin Cancer Res 2021; 27:3528-3539. [PMID: 33608318 DOI: 10.1158/1078-0432.ccr-20-4465] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/13/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022]
Abstract
HER3 is a pseudokinase member of the EGFR family having a role in both tumor progression and drug resistance. Although HER3 was discovered more than 30 years ago, no therapeutic interventions have reached clinical approval to date. Because the evidence of the importance of HER3 is accumulating, increased amounts of preclinical and clinical trials with HER3-targeting agents are emerging. In this review article, we discuss the most recent HER3 biology in tumorigenic events and drug resistance and provide an overview of the current and emerging strategies to target HER3.
Collapse
Affiliation(s)
- Heidi M Haikala
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
14
|
Abstract
Neuregulins, members of the largest subclass of growth factors of the epidermal growth factor family, mediate a myriad of cellular functions including survival, proliferation, and differentiation in normal tissues through binding to receptor tyrosine kinases of the ErbB family. However, aberrant neuregulin signaling in the tumor microenvironment is increasingly recognized as a key player in initiation and malignant progression of human cancers. In this chapter, we focus on the role of neuregulin signaling in the hallmarks of cancer, including cancer initiation and development, metastasis, as well as therapeutic resistance. Moreover, role of neuregulin signaling in the regulation of tumor microenvironment and targeting of neuregulin signaling in cancer from the therapeutic perspective are also briefly discussed.
Collapse
|
15
|
Ni J, Miao T, Su M, Khan NU, Ju X, Chen H, Liu F, Han L. PSMA-targeted nanoparticles for specific penetration of blood-brain tumor barrier and combined therapy of brain metastases. J Control Release 2021; 329:934-947. [DOI: 10.1016/j.jconrel.2020.10.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/12/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023]
|
16
|
Mizuno T, Kojima Y, Yonemori K, Yoshida H, Sugiura Y, Ohtake Y, Okuma HS, Nishikawa T, Tanioka M, Sudo K, Shimomura A, Noguchi E, Kato T, Shimoi T, Uno M, Ishikawa M, Fujiwara Y, Ohe Y, Tamura K. Neoadjuvant chemotherapy promotes the expression of HER3 in patients with ovarian cancer. Oncol Lett 2020; 20:336. [PMID: 33123247 PMCID: PMC7583842 DOI: 10.3892/ol.2020.12200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/03/2020] [Indexed: 01/08/2023] Open
Abstract
HER3 (erbB3) signaling serves an important role in the development and chemoresistance of ovarian cancer, and is activated by chemotherapy. To evaluate the influence of neoadjuvant chemotherapy and other clinical factors on the expression of HER3, as well as to examine its role as a prognostic marker, the present study evaluated archived tissues from patients who underwent surgery for ovarian cancer between 2011 and 2018 at our hospital. Immunohistochemical staining for HER3 was performed using formalin-fixed paraffin-embedded surgical specimens and biopsy samples. In total, data from 111 patients with sufficient surgically resected tumor samples were extracted. A total of 28 patients with histology type high-grade serous carcinoma (HGSC) had specimens available from both pre-chemotherapy biopsies and post-chemotherapy surgery. High HER3 expression (HER3-high) was observed in 64 patients (58%), whereas low HER3 expression (HER3-low) was observed in 47 patients (42%). Multivariate logistic regression analysis identified neoadjuvant chemotherapy [odds ratio (OR), 7.49; 95% confidence interval (CI), 2.48–22.64; P<0.001) and non-HGSC histology (OR, 5.42; 95% CI, 1.99–14.78; P<0.001) as significant predictive factors for HER3-high. In pre-chemotherapy biopsy specimens, 15 patients were HER3-high and 13 were HER3-low. After chemotherapy, eight of 13 patients with HER3-low exhibited a change in status to HER3-high, with a trend toward poorer progression-free survival compared to that of patients whose status remained HER3-low. In conclusion, HER3 overexpression was revealed to be common among patients with ovarian cancer, especially in those with non-HGSC histology. In addition, HER3 expression may be promoted by chemotherapy. These findings suggested that patients with ovarian cancer are good candidates for emerging HER3-targeting therapies.
Collapse
Affiliation(s)
- Takaaki Mizuno
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan.,The Jikei University Graduate School of Medicine, Tokyo 105-8461, Japan
| | - Yuki Kojima
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Kan Yonemori
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Hiroshi Yoshida
- Department of Pathology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yukiko Sugiura
- Department of Gynecology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yohei Ohtake
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Hitomi S Okuma
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Tadaaki Nishikawa
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Maki Tanioka
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Kazuki Sudo
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Akihiko Shimomura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Emi Noguchi
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Tomoyasu Kato
- Department of Gynecology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Tatsunori Shimoi
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Masaya Uno
- Department of Gynecology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Mitsuya Ishikawa
- Department of Gynecology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yasuhiro Fujiwara
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yuichiro Ohe
- The Jikei University Graduate School of Medicine, Tokyo 105-8461, Japan
| | - Kenji Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| |
Collapse
|
17
|
Kumar R, George B, Campbell MR, Verma N, Paul AM, Melo-Alvim C, Ribeiro L, Pillai MR, da Costa LM, Moasser MM. HER family in cancer progression: From discovery to 2020 and beyond. Adv Cancer Res 2020; 147:109-160. [PMID: 32593399 DOI: 10.1016/bs.acr.2020.04.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The human epidermal growth factor receptor (HER) family of receptor tyrosine kinases (RTKs) are among the first layer of molecules that receive, interpret, and transduce signals leading to distinct cancer cell phenotypes. Since the discovery of the tooth-lid factor-later characterized as the epidermal growth factor (EGF)-and its high-affinity binding EGF receptor, HER kinases have emerged as one of the commonly upregulated or hyperactivated or mutated kinases in epithelial tumors, thus allowing HER1-3 family members to regulate several hallmarks of cancer development and progression. Each member of the HER family exhibits shared and unique structural features to engage multiple receptor activation modes, leading to a range of overlapping and distinct phenotypes. EGFR, the founding HER family member, provided the roadmap for the development of the cell surface RTK-directed targeted cancer therapy by serving as a prototype/precursor for the currently used HER-directed cancer drugs. We herein provide a brief account of the discoveries, defining moments, and historical context of the HER family and guidepost advances in basic, translational, and clinical research that solidified a prominent position of the HER family in cancer research and treatment. We also discuss the significance of HER3 pseudokinase in cancer biology; its unique structural features that drive transregulation among HER1-3, leading to a superior proximal signaling response; and potential role of HER3 as a shared effector of acquired therapeutic resistance against diverse oncology drugs. Finally, we also narrate some of the current drawbacks of HER-directed therapies and provide insights into postulated advances in HER biology with extensive implications of these therapies in cancer research and treatment.
Collapse
Affiliation(s)
- Rakesh Kumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India; Department of Medicine, Division of Hematology & Oncology, Rutgers New Jersey Medical School, Newark, NJ, United States; Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| | - Bijesh George
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| | - Marcia R Campbell
- Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States
| | - Nandini Verma
- Advanced Centre for Treatment, Research and Education in Cancer, Mumbai, India
| | - Aswathy Mary Paul
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| | - Cecília Melo-Alvim
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Leonor Ribeiro
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - M Radhakrishna Pillai
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| | - Luis Marques da Costa
- Medical Oncology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal; Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Mark M Moasser
- Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, United States.
| |
Collapse
|
18
|
Liang L, Amin A, Cheung WY, Xu R, Yu R, Tang J, Yao X, Liang C. Parameritannin A-2 from Urceola huaitingii enhances doxorubicin-induced mitochondria-dependent apoptosis by inhibiting the PI3K/Akt, ERK1/2 and p38 pathways in gastric cancer cells. Chem Biol Interact 2019; 316:108924. [PMID: 31843629 DOI: 10.1016/j.cbi.2019.108924] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/28/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022]
Abstract
Parameritannin A-2 (PA-2) is a natural product extracted from the stems of the plant Urceola huaitingii. Our previous studies have shown that PA-2 exhibits significant synergistic anticancer effects with doxorubicin (DOX) in HGC27 gastric cancer cell lines. Here we report that our isobolographic analysis confirms the synergistic cytotoxic effects of PA-2 and DOX in HGC27 cells. Flow cytometry and immunoblotting indicate that PA-2 enhances DOX-mediated apoptosis. Importantly, PA-2 enhances the intracellular accumulation of DOX in HGC27 cells. The combination of DOX and PA-2 remarkably increases the release of cytochrome C and the activation of caspase-3 and caspase-9, compared with DOX treatment alone. Moreover, PA-2 attenuates the DOX-induced activation of Akt, ERK1/2 and p38 signaling pathways, providing a molecular mechanism for the synergistic effects of DOX and PA-2 in the induction of apoptosis. In conclusion, our studies demonstrate that PA-2 and DOX synergistically induce mitochondria-dependent apoptosis as PA-2 inhibits the PI3K/Akt, ERK1/2 and p38 pathways in HGC27 cells. These findings suggest that the combination treatment with PA-2 and DOX may represent a potent therapy for gastric cancer.
Collapse
Affiliation(s)
- Lu Liang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China; Division of Life Science, Center for Cancer Research and State Key Lab for Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Aftab Amin
- Division of Life Science, Center for Cancer Research and State Key Lab for Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China; Guangzhou HKUST Fok Ying Tung Research Institute, Guangzhou, China
| | - Wing-Yan Cheung
- Division of Life Science, Center for Cancer Research and State Key Lab for Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Rui Xu
- Division of Life Science, Center for Cancer Research and State Key Lab for Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Rujian Yu
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jinshan Tang
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Xinsheng Yao
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, Jinan University, Guangzhou, China
| | - Chun Liang
- Division of Life Science, Center for Cancer Research and State Key Lab for Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China; Guangzhou HKUST Fok Ying Tung Research Institute, Guangzhou, China; Intelgen Limited, Hong Kong, Guangzhou, Foshan, China; EnKang-EnZhi, Limited, Guangzhou, China.
| |
Collapse
|
19
|
Temozolomide induces activation of Wnt/β-catenin signaling in glioma cells via PI3K/Akt pathway: implications in glioma therapy. Cell Biol Toxicol 2019; 36:273-278. [DOI: 10.1007/s10565-019-09502-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 11/05/2019] [Indexed: 12/16/2022]
|
20
|
Inhibition of TFF3 Enhances Sensitivity-and Overcomes Acquired Resistance-to Doxorubicin in Estrogen Receptor-Positive Mammary Carcinoma. Cancers (Basel) 2019; 11:cancers11101528. [PMID: 31658702 PMCID: PMC6826976 DOI: 10.3390/cancers11101528] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/01/2019] [Accepted: 10/05/2019] [Indexed: 02/07/2023] Open
Abstract
Dose-dependent toxicity and acquired resistance are two major challenges limiting the efficacious treatment of mammary carcinoma (MC) with doxorubicin. Herein, we investigated the function of Trefoil Factor 3 (TFF3) in the sensitivity and acquired resistance of estrogen receptor positive (ER+) MC cells to doxorubicin. Doxorubicin treatment of ER+MC cells increased TFF3 expression. The depletion of TFF3 by siRNA or inhibition with a small molecule TFF3 inhibitor (AMPC) synergistically enhanced the efficacy of doxorubicin in ER+MC through the suppression of doxorubicin-induced AKT activation and enhancement of doxorubicin-induced apoptosis. Elevated expression of TFF3 and increased activation of AKT were also observed using a model of acquired doxorubicin resistance in ER+MC cells. AMPC partially re-sensitized the doxorubicin resistant cells to doxorubicin-induced apoptosis. Indeed, doxorubicin resistant ER + MC cells exhibited increased sensitivity to AMPC as a single agent compared to doxorubicin sensitive cells. In vivo, AMPC attenuated growth of doxorubicin sensitive ER+MC xenografts whereas it produced regression of xenografts generated by doxorubicin resistant ER+MC cells. Hence, TFF3 inhibition may improve the efficacy and reduce required doses of doxorubicin in ER+MC. Moreover, inhibition of TFF3 may also be an effective therapeutic strategy to eradicate doxorubicin resistant ER+MC.
Collapse
|
21
|
Zhai C, Lv J, Wang K, Li Q, Qu Y. HSP70 silencing aggravates apoptosis induced by hypoxia/reoxygenation in vitro. Exp Ther Med 2019; 18:1013-1020. [PMID: 31363363 PMCID: PMC6614734 DOI: 10.3892/etm.2019.7697] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 02/28/2019] [Indexed: 01/09/2023] Open
Abstract
Lung ischemia-reperfusion can cause acute lung injury, which is closely associated with apoptosis. Heat shock protein 70 (HSP70) is an anti-apoptotic protein that promotes cell survival under a variety of different stress conditions. However, the role and mechanism of HSP70 in lung ischemia-reperfusion injury is yet to be fully elucidated. In the present study, an in vitro hypoxia/reoxygenation model of A549 cells was established to simulate lung ischemia-reperfusion and HSP70 was silenced by transfecting A549 cells with an shRNA sequence targeting HSP70. Western blotting, reverse transcription-quantitative polymerase chain reaction, Cell Counting kit-8 and flow cytometry were used to detect protein levels, RNA expression, cell activity and apoptosis. The results revealed that silencing HSP70 reduced cell viability, aggravated apoptosis, increased lactate dehydrogenase levels and induced a G2/M blockade in a hypoxia-reoxygenation A549 cell model. Furthermore, silencing HSP70 decreased the phosphorylation levels of protein kinase B (AKT) and extracellular signal-regulated kinase (ERK); however, the total AKT and ERK levels did not change significantly. Pretreating A549 cells with the AKT pathway inhibitor, LY294002 and the ERK pathway inhibitor, U0216 led to a decrease in HSP70 expression. These results indicate that silencing HSP70 may aggravate apoptosis in hypoxia-reoxygenation cell models, potentially via the mitogen-activated protein kinase/ERK and phosphoinositide 3-kinase/AKT signaling pathways.
Collapse
Affiliation(s)
- Congying Zhai
- Intensive Care Unit, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong 266071, P.R. China.,Department of Respiratory Medicine, Zibo First Hospital, Zibo, Shandong 255200, P.R. China
| | - Jiling Lv
- Department of Respiratory Medicine, Zibo First Hospital, Zibo, Shandong 255200, P.R. China
| | - Keke Wang
- Department of Emergency, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Qingshu Li
- Intensive Care Unit, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Yan Qu
- Intensive Care Unit, Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
22
|
Vancsik T, Forika G, Balogh A, Kiss E, Krenacs T. Modulated electro-hyperthermia induced p53 driven apoptosis and cell cycle arrest additively support doxorubicin chemotherapy of colorectal cancer in vitro. Cancer Med 2019; 8:4292-4303. [PMID: 31183995 PMCID: PMC6675742 DOI: 10.1002/cam4.2330] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 03/20/2019] [Accepted: 03/20/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Modulated electro-hyperthermia (mEHT), a noninvasive complementary treatment of human chemo- and radiotherapy, can generate selective ~42°C heat in cancer due to elevated glycolysis (Warburg-effect) and electric conductivity in malignant tissues. Here we tested the molecular background of mEHT and its combination with doxorubicin chemotherapy using an in vitro model. METHODS C26 mouse colorectal adenocarcinoma cultures were mEHT treated at 42°C for 2 × 60 minutes (with 120 minutes interruption) either alone or in combination with 1 µmol/L doxorubicin (mEHT + Dox). Cell stress response, apoptosis, and cell cycle regulation related markers were detected using qPCR and immunocytochemistry supported with resazurin cell viability assay, cell death analysis using flow-cytometry and clonogenic assay. RESULT Cell-stress by mEHT alone was indicated by the significant upregulation and release of hsp70 and calreticulin proteins 3 hours posttreatment. Between 3 and 9 hours after treatment significantly reduced anti-apoptotic XIAP, BCL-2, and BCL-XL and elevated pro-apoptotic BAX and PUMA, as well as the cyclin dependent kinase inhibitor p21waf1 mRNA levels were detected. After 24 hours, major elevation and nuclear translocation of phospho-p53(Ser15) protein levels and reduced phospho-Akt(Ser473) levels were accompanied by a significant caspase-3-mediated programmed cell death response. While mEHT dominantly induced apoptosis, Dox administration primarily led to tumor cell necrosis, and both significantly reduced the number of tumor progenitor colonies 10 days post-treatment. Furthermore, mEHT promoted the uptake of Dox by tumor cells and the combined treatment additively reduced tumor cell viability and augmented cell death near to synergy. CONCLUSION In C26 colorectal adenocarcinoma mEHT-induced irreversible cell stress can activate both caspase-dependent apoptosis and p21waf1 mediated growth arrest pathways, likely to be driven by the upregulated nuclear p53 protein. Elevated phospho-p53(Ser15) might contribute to p53 escape from mdm2 control, which was further supported by reduced phospho-Akt(Ser473) protein levels. In combinations, mEHT could promote the uptake and significantly potentiate the cytotoxic effect of doxorubicin.
Collapse
Affiliation(s)
- Tamas Vancsik
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gertrud Forika
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Andrea Balogh
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Eva Kiss
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Tibor Krenacs
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
23
|
James NE, Beffa L, Oliver MT, Borgstadt AD, Emerson JB, Chichester CO, Yano N, Freiman RN, DiSilvestro PA, Ribeiro JR. Inhibition of DUSP6 sensitizes ovarian cancer cells to chemotherapeutic agents via regulation of ERK signaling response genes. Oncotarget 2019; 10:3315-3327. [PMID: 31164954 PMCID: PMC6534361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 04/14/2019] [Indexed: 11/05/2022] Open
Abstract
Dual specificity phosphatase 6 (DUSP6) is a protein phosphatase that deactivates extracellular-signal-regulated kinase (ERK). Since the ovarian cancer biomarker human epididymis protein 4 (HE4) interacts with the ERK pathway, we sought to determine the relationship between DUSP6 and HE4 and elucidate DUSP6's role in epithelial ovarian cancer (EOC). Viability assays revealed a significant decrease in cell viability with pharmacological inhibition of DUSP6 using (E/Z)-BCI hydrochloride in ovarian cancer cells treated with carboplatin or paclitaxel, compared to treatment with either agent alone. Quantitative PCR was used to evaluate levels of ERK pathway response genes to BCI in combination with recombinant HE4 (rHE4), carboplatin, and paclitaxel. Expression of EGR1, a promoter of apoptosis, was higher in cells co-treated with BCI and paclitaxel or carboplatin than in cells treated with chemotherapeutic agents alone, while expression of the proto-oncogene c-JUN was decreased with co-treatment. The effect of BCI on the expression of these two genes opposed that of rHE4. Pathway focused quantitative PCR also revealed suppression of ERBB3 in cells co-treated with BCI plus carboplatin or paclitaxel. Finally, expression levels of DUSP6 in EOC tissue were evaluated by immunohistochemistry, revealing significantly increased levels of DUSP6 in serous EOC tissue compared to adjacent normal tissue. A positive correlation between HE4 and DUSP6 levels was determined by Spearman Rank correlation. In conclusion, DUSP6 inhibition sensitizes ovarian cancer cells to chemotherapeutic agents and alters gene expression of ERK response genes, suggesting that DUSP6 could plausibly function as a novel therapeutic target to reduce chemoresistance in EOC.
Collapse
Affiliation(s)
- Nicole E. James
- Women and Infants Hospital, Department of Obstetrics and Gynecology, Program in Women’s Oncology, Providence, RI, USA
- Department of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Lindsey Beffa
- Women and Infants Hospital, Department of Obstetrics and Gynecology, Program in Women’s Oncology, Providence, RI, USA
| | - Matthew T. Oliver
- Women and Infants Hospital, Department of Obstetrics and Gynecology, Program in Women’s Oncology, Providence, RI, USA
| | - Ashley D. Borgstadt
- Women and Infants Hospital, Department of Obstetrics and Gynecology, Program in Women’s Oncology, Providence, RI, USA
| | - Jenna B. Emerson
- Women and Infants Hospital, Department of Obstetrics and Gynecology, Program in Women’s Oncology, Providence, RI, USA
| | | | - Naohiro Yano
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA
| | - Richard N. Freiman
- Department of Molecular and Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Paul A. DiSilvestro
- Women and Infants Hospital, Department of Obstetrics and Gynecology, Program in Women’s Oncology, Providence, RI, USA
| | - Jennifer R. Ribeiro
- Women and Infants Hospital, Department of Obstetrics and Gynecology, Program in Women’s Oncology, Providence, RI, USA
| |
Collapse
|
24
|
Inhibition of DUSP6 sensitizes ovarian cancer cells to chemotherapeutic agents via regulation of ERK signaling response genes. Oncotarget 2019. [DOI: 10.18632/oncotarget.26915] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
25
|
Sequist LV, Gray JE, Harb WA, Lopez-Chavez A, Doebele RC, Modiano MR, Jackman DM, Baggstrom MQ, Atmaca A, Felip E, Provencio M, Cobo M, Adiwijaya B, Kuesters G, Kamoun WS, Andreas K, Pipas JM, Santillana S, Cho BC, Park K, Shepherd FA. Randomized Phase II Trial of Seribantumab in Combination with Erlotinib in Patients with EGFR Wild-Type Non-Small Cell Lung Cancer. Oncologist 2019; 24:1095-1102. [PMID: 30975923 DOI: 10.1634/theoncologist.2018-0695] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 02/08/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Seribantumab (MM-121) is a fully human IgG2 monoclonal antibody that binds to human epidermal growth factor receptor 3 (HER3/ErbB3) to block heregulin (HRG/NRG)-mediated ErbB3 signaling and induce receptor downregulation. This open-label, randomized phase 1/2 study evaluated safety and efficacy of seribantumab plus erlotinib in advanced non-small cell lung cancer (NSCLC). Here, we report the activity of seribantumab plus erlotinib, versus erlotinib alone, in patients with EGFR wild-type tumors and describe the potential predictive power of HRG. MATERIALS AND METHODS Patients with EGFR wild-type NSCLC were assigned randomly to receive seribantumab + erlotinib or erlotinib alone. Patients underwent pretreatment core needle biopsy and archived tumor samples were collected to support prespecified biomarker analyses. RESULTS One hundred twenty-nine patients received seribantumab + erlotinib (n = 85) or erlotinib alone (n = 44). Median estimated progression-free survival (PFS) in the unselected intent-to-treat (ITT) population was 8.1 and 7.7 weeks in the experimental and control arm, respectively (hazard ratio [HR], 0.822; 95% confidence interval [CI], 0.37-1.828; p = 0.63), and median estimated overall survival was 27.3 and 40.3 weeks in the experimental and control arm, respectively (HR, 1.395; 95% CI, 0.846 to 2.301; p = .1898) In patients whose tumors had detectable HRG mRNA expression, treatment benefit was observed in the seribantumab + erlotinib combination (HR, 0.35; 95% CI, 0.16-0.76; p = .008). In contrast, in patients whose tumors were HRG negative, the HR was 2.15 (95% CI, 0.97-4.76; p = .059, HRG-by-treatment interaction, p value = .0016). CONCLUSION The addition of seribantumab to erlotinib did not result in improved PFS in unselected patients. However, predefined retrospective exploratory analyses suggest that detectable HRG mRNA levels identified patients who might benefit from seribantumab. An ongoing clinical trial of seribantumab, in combination with docetaxel, is underway in patients with advanced NSCLC and high HRG mRNA expression (NCT02387216). IMPLICATIONS FOR PRACTICE The poor prognosis of patients with non-small cell lung cancer (NSCLC) underscores the need for more effective treatment options, highlighting the unmet medical need in this patient population. The results of this study show that a novel biomarker, heregulin, may help to identify patients with advanced NSCLC who could benefit from treatment with seribantumab. On the basis of the observed safety profile and promising clinical efficacy, a prospective, randomized, open-label, international, multicenter phase II trial (SHERLOC, NCT02387216) is under way to investigate the efficacy and safety of seribantumab in combination with docetaxel in patients with heregulin-positive advanced adenocarcinoma.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/genetics
- Erlotinib Hydrochloride/pharmacology
- Erlotinib Hydrochloride/therapeutic use
- Female
- Follow-Up Studies
- Humans
- Lung/pathology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Male
- Middle Aged
- Neuregulin-1/analysis
- Neuregulin-1/antagonists & inhibitors
- Patient Selection
- Progression-Free Survival
- Receptor, ErbB-3/analysis
- Receptor, ErbB-3/antagonists & inhibitors
- Retrospective Studies
Collapse
Affiliation(s)
| | | | - Wael A Harb
- Horizon Oncology Center, Lafayette, Indiana, USA
| | - Ariel Lopez-Chavez
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, USA
| | | | | | | | | | - Akin Atmaca
- Department of Hematology and Oncology, Institute of Clinical Research at Krankenhaus Nordwest, UCT-University Cancer Center, Frankfurt, Germany
| | | | | | - Manuel Cobo
- Hospital Regional Universitario Málaga, Instituto de Investigación Biomédica de Málaga, Málaga, Spain
| | | | | | - Walid S Kamoun
- Merrimack Pharmaceuticals, Inc., Cambridge, Massachusetts, USA
| | - Karen Andreas
- Merrimack Pharmaceuticals, Inc., Cambridge, Massachusetts, USA
| | - J Marc Pipas
- Merrimack Pharmaceuticals, Inc., Cambridge, Massachusetts, USA
| | | | | | - Keunchil Park
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | |
Collapse
|
26
|
Loh AHP, Stewart E, Bradley CL, Chen X, Daryani V, Stewart CF, Calabrese C, Funk A, Miller G, Karlstrom A, Krafcik F, Goshorn DR, Vogel P, Bahrami A, Shelat A, Dyer MA. Combinatorial screening using orthotopic patient derived xenograft-expanded early phase cultures of osteosarcoma identify novel therapeutic drug combinations. Cancer Lett 2018; 442:262-270. [PMID: 30395907 PMCID: PMC6342199 DOI: 10.1016/j.canlet.2018.10.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 10/17/2018] [Accepted: 10/24/2018] [Indexed: 11/28/2022]
Abstract
Lead discovery in osteosarcoma has been hampered by the lack of new agents, limited representative clinical samples and paucity of accurate preclinical models. We developed orthotopic patient-derived xenografts (PDXs) that recapitulated the molecular, cellular and histologic features of primary tumors, and screened PDX-expanded short-term cultures and commercial cell lines of osteosarcoma against focused drug libraries. Osteosarcoma cells were most sensitive to HDAC, proteasome, and combination PI3K/MEK and PI3K/mTOR inhibitors, and least sensitive to PARP, RAF, ERK and MEK inhibitors. Correspondingly, PI3K signaling pathway genes were up-regulated in metastatic tumors compared to primary tumors. In combinatorial screens, as a class, HDAC inhibitors showed additive effects when combined with standard-of-care agents gemcitabine and doxorubicin. This lead discovery strategy afforded a means to perform high-throughput drug screens of tumor cells that accurately recapitulated those from original human tumors, and identified classes of novel and repurposed drugs with activity against osteosarcoma.
Collapse
Affiliation(s)
- Amos H P Loh
- Department of Paediatric Surgery, KK Women's and Children's Hospital, Singapore; Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Elizabeth Stewart
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Cori L Bradley
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Vinay Daryani
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Clinton F Stewart
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Christopher Calabrese
- Animal Resources Center, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Amy Funk
- Animal Resources Center, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Greg Miller
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Asa Karlstrom
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Fred Krafcik
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - David R Goshorn
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Peter Vogel
- Animal Resources Center, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Armita Bahrami
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Anang Shelat
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA; Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
27
|
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
|
28
|
Obounchoey P, Tabtimmai L, Suphakun P, Thongkhao K, Eurtivong C, Gleeson MP, Choowongkomon K. In silico identification and in vitro validation of nogalamycin N-oxide (NSC116555) as a potent anticancer compound against non-small-cell lung cancer cells. J Cell Biochem 2018; 120:3353-3361. [PMID: 30324706 DOI: 10.1002/jcb.27605] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 08/07/2018] [Indexed: 12/20/2022]
Abstract
The epidermal growth factor receptor (EGFR) was found to be overexpressed in several cancers, especially in lung cancers. Finding new effective drug against EGFR is the key to cancer treatment. In this study, the GOLD docking algorithm was used to virtually screen for novel human EGFR inhibitors from the NCI database. Thirty-four hit compounds were tested for EGFR-tyrosine kinase (TK) inhibition. Two potent compounds, 1-amino-4-(4-[4-amino-2-sulfophenyl]anilino)-9,10-dioxoanthracene-2-sulfonic acid (NSC125910), and nogalamycin N-oxide (NSC116555) were identified with IC50 values against EGFR-TK comparable to gefitinib; 16.14 and 37.71 nM, respectively. However, only NSC116555 demonstrated cytotoxic effects against non-small-cell lung cancer, A549, shown in the cell cytotoxicity assay with an IC50 of 0.19 + 0.01 µM, which was more potent than gefitinib. Furthermore, NSC116555 showed cytotoxicity against A549 via apoptosis in a dose-dependent manner.
Collapse
Affiliation(s)
- Phongphat Obounchoey
- Interdisciplinary Program in Genetic Engineering, Graduate School, Kasetsart University, Chatuchak, Bangkok, Thailand
| | - Lueacha Tabtimmai
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Praphasri Suphakun
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Kannika Thongkhao
- Interdisciplinary Program in Genetic Engineering, Graduate School, Kasetsart University, Chatuchak, Bangkok, Thailand
| | - Chatchakorn Eurtivong
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
| | - Matthew Paul Gleeson
- Department of Biomedical Engineering, Faculty of Engineering, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Kiattawee Choowongkomon
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand.,Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok, Thailand
| |
Collapse
|
29
|
Alessandrini F, Pezzè L, Menendez D, Resnick MA, Ciribilli Y. ETV7-Mediated DNAJC15 Repression Leads to Doxorubicin Resistance in Breast Cancer Cells. Neoplasia 2018; 20:857-870. [PMID: 30025229 PMCID: PMC6077117 DOI: 10.1016/j.neo.2018.06.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/26/2018] [Accepted: 06/26/2018] [Indexed: 12/20/2022]
Abstract
Breast cancer treatment often includes Doxorubicin as adjuvant as well as neoadjuvant chemotherapy. Despite its cytotoxicity, cells can develop drug resistance to Doxorubicin. Uncovering pathways and mechanisms involved in drug resistance is an urgent and critical aim for breast cancer research oriented to improve treatment efficacy. Here we show that Doxorubicin and other chemotherapeutic drugs induce the expression of ETV7, a transcriptional repressor member of ETS family of transcription factors. The ETV7 expression led to DNAJC15 down-regulation, a co-chaperone protein whose low expression was previously associated with drug resistance in breast and ovarian cancer. There was a corresponding reduction in Doxorubicin sensitivity of MCF7 and MDA-MB-231 breast cancer cells. We identified the binding site for ETV7 within DNAJC15 promoter and we also found that DNA methylation may be a factor in ETV7-mediated DNAJC15 transcriptional repression. These findings of an inverse correlation between ETV7 and DNAJC15 expression in MCF7 cells in terms of Doxorubicin resistance, correlated well with treatment responses of breast cancer patients with recurrent disease, based on our analyses of reported genome-wide expression arrays. Moreover, we demonstrated that ETV7-mediated Doxorubicin-resistance involves increased Doxorubicin efflux via nuclear pumps, which could be rescued in part by DNAJC15 up-regulation. With this study, we propose a novel role for ETV7 in breast cancer, and we identify DNAJC15 as a new target gene responsible for ETV7-mediated Doxorubicin-resistance. A better understanding of the opposing impacts of Doxorubicin could improve the design of combinatorial adjuvant regimens with the aim of avoiding resistance and relapse.
Collapse
Affiliation(s)
- Federica Alessandrini
- Laboratory of Molecular Cancer Genetics, Centre for Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Povo (TN), Italy.
| | - Laura Pezzè
- Laboratory of Molecular Cancer Genetics, Centre for Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Povo (TN), Italy
| | - Daniel Menendez
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences (NIHES), NIH, Research Triangle Park, NC 27709, USA
| | - Michael A Resnick
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences (NIHES), NIH, Research Triangle Park, NC 27709, USA
| | - Yari Ciribilli
- Laboratory of Molecular Cancer Genetics, Centre for Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Povo (TN), Italy.
| |
Collapse
|
30
|
Mishra R, Patel H, Alanazi S, Yuan L, Garrett JT. HER3 signaling and targeted therapy in cancer. Oncol Rev 2018; 12:355. [PMID: 30057690 PMCID: PMC6047885 DOI: 10.4081/oncol.2018.355] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/27/2018] [Indexed: 12/27/2022] Open
Abstract
ERBB family members including epidermal growth factor receptor (EGFR) also known as HER1, ERBB2/HER2/Neu, ERBB3/HER3 and ERBB4/HER4 are aberrantly activated in multiple cancers and hence serve as drug targets and biomarkers in modern precision therapy. The therapeutic potential of HER3 has long been underappreciated, due to impaired kinase activity and relatively low expression in tumors. However, HER3 has received attention in recent years as it is a crucial heterodimeric partner for other EGFR family members and has the potential to regulate EGFR/HER2-mediated resistance. Upregulation of HER3 is associated with several malignancies where it fosters tumor progression via interaction with different receptor tyrosine kinases (RTKs). Studies also implicate HER3 contributing significantly to treatment failure, mostly through the activation of PI3K/AKT, MAPK/ERK and JAK/STAT pathways. Moreover, activating mutations in HER3 have highlighted the role of HER3 as a direct therapeutic target. Therapeutic targeting of HER3 includes abrogating its dimerization partners’ kinase activity using small molecule inhibitors (lapatinib, erlotinib, gefitinib, afatinib, neratinib) or direct targeting of its extracellular domain. In this review, we focus on HER3-mediated signaling, its role in drug resistance and discuss the latest advances to overcome resistance by targeting HER3 using mono- and bispecific antibodies and small molecule inhibitors.
Collapse
Affiliation(s)
- Rosalin Mishra
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Hima Patel
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Samar Alanazi
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Long Yuan
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Joan T Garrett
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
31
|
Huang L, Zeng L, Chu J, Xu P, Lv M, Xu J, Wen J, Li W, Wang L, Wu X, Fu Z, Xie H, Wang S. Chemoresistance‑related long non‑coding RNA expression profiles in human breast cancer cells. Mol Med Rep 2018; 18:243-253. [PMID: 29749447 PMCID: PMC6059676 DOI: 10.3892/mmr.2018.8942] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 03/07/2017] [Indexed: 12/21/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer and the leading cause of cancer death in females worldwide. Chemoresistance has been a major reason for the drug therapy failure. The present study performed a microarray analysis between MCF-7 and MCF-7/adriamycin (ADR) cells, and intended to identify long non-coding (lnc)RNA expression character in drug resistant breast cancer cells. MCF-7/ADR cells were induced from MCF-7 cells via pulse-selection with doxorubicin for 4 weeks, and the resistance to doxorubicin of ADR cells was confirmed by MTT assay. Microarray analysis was performed between MCF-7 and MCF-7/ADR cells. Total RNA was extracted from the two cell lines respectively and was transcribed into cDNA. The results of the microarray were verified by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Gene Ontology (GO) and pathways analysis were conducted to enrich the dysregulated lncRNAs presented in the microarray results. Compared to the MCF-7 cells, 8,892 lncRNAs were differentially expressed in MCF/ADR cells (absolute fold-change >2.0). A total of 32 lncRNAs were selected for RT-qPCR by fold-change filtering, standard Student's t-test, and multiple hypothesis testing. Among the dysregulated lncRNAs, AX747207 was prominent because its associated gene RUNX3 was previously reported to be relative to malignant tumor chemoresistance. GO analysis results also indicated some biological processes and molecular functions linked to chemoresistance. The pathway enrichment results provided some potential pathways associated with chemoresistance. In the present study, the authors intended to identify lncRNA expression character in drug resistant cell line MCF-7/ADR, corresponding to the parental MCF-7 cell line. In addition, the study identified the lncRNA AX747207, and its potential targeted gene RUNX3, may be related to chemoresistance in breast cancer. These results may new insights into exploring the mechanisms of chemoresistance in breast cancer.
Collapse
Affiliation(s)
- Lei Huang
- Department of Breast Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lihua Zeng
- Nanjing Maternity and Child Health Medical Institute, Affiliated Nanjing Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Jiahui Chu
- Department of Breast Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Pengfei Xu
- Nanjing Maternity and Child Health Medical Institute, Affiliated Nanjing Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Mingming Lv
- Nanjing Maternity and Child Health Medical Institute, Affiliated Nanjing Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Juan Xu
- Nanjing Maternity and Child Health Medical Institute, Affiliated Nanjing Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Juan Wen
- Nanjing Maternity and Child Health Medical Institute, Affiliated Nanjing Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Wenqu Li
- Nanjing Maternity and Child Health Medical Institute, Affiliated Nanjing Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Luyu Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Xiaowei Wu
- Department of Pharmacology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Ziyi Fu
- Nanjing Maternity and Child Health Medical Institute, Affiliated Nanjing Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Hui Xie
- Department of Breast Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Shui Wang
- Department of Breast Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
32
|
Li S, Yang L, Wang J, Liang F, Chang B, Gu H, Wang H, Yang G, Chen Y. Analysis of the chemotherapeutic effects of a propadiene compound on malignant ovarian cancer cells. Oncotarget 2018; 7:57145-57159. [PMID: 27494891 PMCID: PMC5302979 DOI: 10.18632/oncotarget.11012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 07/19/2016] [Indexed: 12/31/2022] Open
Abstract
Epithelial ovarian cancer is most lethal in female reproductive carcinomas owing to the high chemoresistance and metastasis, so more efficient therapeutic agents are terribly needed. A propadiene compound: 1-phenylpropadienyl phosphine oxide (PHPO), was employed to test the chemotherapeutic efficacy against ovarian cancer cell lines. MTT assay showed that PHPO displayed a much lower IC50 than cisplatin and paclitaxel, while combination treatment of cells with PHPO + cisplatin induced more apoptosis than with PHPO + paclitaxel or with cisplatin + paclitaxel (p < 0.05). Animal assays demonstrated that subcutaneous tumor growth was highly inhibited by PHPO + cisplatin, compared with that inhibited by PHPO or by cisplatin treatment alone, indicating PHPO and cisplatin may have synergistic effects against ovarian cancer growth. We also found that PHPO induced few side effects on animals, compared with cisplatin. Mechanistic studies suggested that treatment of cells with PHPO or with PHPO + cisplatin differentially inhibited the PI3K/Akt, MAPK and ATM/Chk2 pathways, which consequently suppressed the anti-apoptotic factors Bcl-xL, Bcl-2 and XIAP, but activated the pro-apoptotic factors Bad, Bax, p53, caspase 9, caspase 8, caspase 7 and PARP. Taken together, PHPO may induce cell apoptosis through multiple signal pathways, especially when used along with cisplatin. Therefore, PHPO may be explored as a prospective agent to effectively treat ovarian cancer.
Collapse
Affiliation(s)
- Shuqing Li
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, China
| | - Lina Yang
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, China
| | - Jingshu Wang
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, China
| | - Fan Liang
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, China
| | - Bin Chang
- Department of Pathology, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Huafen Gu
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, China
| | - Honglin Wang
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, China
| | - Gong Yang
- Cancer Institute, Fudan University Shanghai Cancer Center, and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Central laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, China
| | - Yaping Chen
- Department of Obstetrics and Gynecology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, China
| |
Collapse
|
33
|
Park CM, Kawasaki Y, Refaat A, Sakurai H. Mechanisms for DNA-damaging agent-induced inactivation of ErbB2 and ErbB3 via the ERK and p38 signaling pathways. Oncol Lett 2018; 15:1758-1762. [PMID: 29434871 DOI: 10.3892/ol.2017.7532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 02/23/2017] [Indexed: 11/06/2022] Open
Abstract
Cisplatin (CDDP) and doxorubicin (DOX) are chemotherapeutic drugs that trigger apoptosis by inducing DNA-damage. A previous study using breast cancer cells demonstrated the negative feedback modulation of the epidermal growth factor receptor (EGFR) and receptor tyrosine-protein kinase erbB-2 (ErbB2) via extracellular signal-regulated kinase (ERK)-mediated phosphorylation of conserved Thr-669 and Thr-677 residues, respectively, in the juxtamembrane domain. In addition, CDDP has been identified to cause negative feedback inhibition of activated EGFR in lung cancer cells. In the present study, the role of phosphorylation in the feedback control of the ErbB2/ErbB3 heterodimer in human breast and gastric cancer cells was investigated. Phosphorylation of ErbB2 at Thr-677 was induced by CDDP and DOX, which in turn reduced tyrosine autophosphorylation of ErbB2 and ErbB3. Treatment with trametinib, a mitogen-activated protein kinase inhibitor that blocks ERK-mediated Thr-677 phosphorylation, and substitution of Thr-677 to alanine, blocked the feedback inhibition of ErbB2 and ErbB3. In addition, these agents caused the degradation of ErbB proteins through the activation of p38 mitogen-activated protein kinase (p38) and ERK. These results demonstrate that chemotherapeutic agents trigger ERK- and p38-mediated post-translational downregulation of ErbB receptors.
Collapse
Affiliation(s)
- Chul Min Park
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Yuki Kawasaki
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Alaa Refaat
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Hiroaki Sakurai
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
34
|
Mota JM, Collier KA, Barros Costa RL, Taxter T, Kalyan A, Leite CA, Chae YK, Giles FJ, Carneiro BA. A comprehensive review of heregulins, HER3, and HER4 as potential therapeutic targets in cancer. Oncotarget 2017; 8:89284-89306. [PMID: 29179520 PMCID: PMC5687690 DOI: 10.18632/oncotarget.18467] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 04/17/2017] [Indexed: 12/30/2022] Open
Abstract
Heregulins (HRGs) bind to the receptors HER3 or HER4, induce receptor dimerization, and trigger downstream signaling that leads to tumor progression and resistance to targeted therapies. Increased expression of HRGs has been associated with worse clinical prognosis; therefore, attempts to block HRG-dependent tumor growth have been pursued. This manuscript summarizes the function and signaling of HRGs and review the preclinical evidence of its involvement in carcinogenesis, prognosis, and treatment resistance in several malignancies such as colorectal cancer, non-small cell lung cancer, ovarian cancer, and breast cancer. Agents in preclinical development and clinical trials of novel therapeutics targeting HRG-dependent signaling are also discussed, including anti-HER3 and -HER4 antibodies, anti-metalloproteinase agents, and HRG fusion proteins. Although several trials have indicated an acceptable safety profile, translating preclinical findings into clinical practice remains a challenge in this field, possibly due to the complexity of downstream signaling and patterns of HRG, HER3 and HER4 expression in different cancer subtypes. Improving patient selection through biomarkers and understanding the resistance mechanisms may translate into significant clinical benefits in the near future.
Collapse
Affiliation(s)
- Jose Mauricio Mota
- Instituto do Câncer do Estado de São Paulo, Division of Oncology, Universidade de São Paulo, São Paulo, Brazil
| | - Katharine Ann Collier
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ricardo Lima Barros Costa
- Developmental Therapeutics Program, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Timothy Taxter
- Developmental Therapeutics Program, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Aparna Kalyan
- Developmental Therapeutics Program, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Caio A. Leite
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Young Kwang Chae
- Developmental Therapeutics Program, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Francis J. Giles
- Developmental Therapeutics Program, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Benedito A. Carneiro
- Developmental Therapeutics Program, Division of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
35
|
Targeting tumor multicellular aggregation through IGPR-1 inhibits colon cancer growth and improves chemotherapy. Oncogenesis 2017; 6:e378. [PMID: 28920928 PMCID: PMC5623903 DOI: 10.1038/oncsis.2017.77] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 12/11/2022] Open
Abstract
Adhesion to extracellular matrix (ECM) is crucially important for survival of normal epithelial cells as detachment from ECM triggers specific apoptosis known as anoikis. As tumor cells lose the requirement for anchorage to ECM, they rely on cell-cell adhesion 'multicellular aggregation' for survival. Multicellular aggregation of tumor cells also significantly determines the sensitivity of tumor cells to the cytotoxic effects of chemotherapeutics. In this report, we demonstrate that expression of immunoglobulin containing and proline-rich receptor-1 (IGPR-1) is upregulated in human primary colon cancer. Our study demonstrates that IGPR-1 promotes tumor multicellular aggregation, and interfering with its adhesive function inhibits multicellular aggregation and, increases cell death. IGPR-1 supports colon carcinoma tumor xenograft growth in mouse, and inhibiting its activity by shRNA or blocking antibody inhibits tumor growth. More importantly, IGPR-1 regulates sensitivity of tumor cells to the chemotherapeutic agent, doxorubicin/adriamycin by a mechanism that involves doxorubicin-induced AKT activation and phosphorylation of IGPR-1 at Ser220. Our findings offer novel insight into IGPR-1's role in colorectal tumor growth, tumor chemosensitivity, and as a possible novel anti-cancer target.
Collapse
|
36
|
Momeny M, Zarrinrad G, Moghaddaskho F, Poursheikhani A, Sankanian G, Zaghal A, Mirshahvaladi S, Esmaeili F, Eyvani H, Barghi F, Sabourinejad Z, Alishahi Z, Yousefi H, Ghasemi R, Dardaei L, Bashash D, Chahardouli B, Dehpour AR, Tavakkoly-Bazzaz J, Alimoghaddam K, Ghavamzadeh A, Ghaffari SH. Dacomitinib, a pan-inhibitor of ErbB receptors, suppresses growth and invasive capacity of chemoresistant ovarian carcinoma cells. Sci Rep 2017. [PMID: 28646172 PMCID: PMC5482808 DOI: 10.1038/s41598-017-04147-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynaecological malignancy worldwide. Development of chemoresistance and peritoneal dissemination of EOC cells are the major reasons for low survival rate. Targeting signal transduction pathways which promote therapy resistance and metastatic dissemination is the key to successful treatment. Members of the ErbB family of receptors are over-expressed in EOC and play key roles in chemoresistance and invasiveness. Despite this, single-targeted ErbB inhibitors have demonstrated limited activity in chemoresistant EOC. In this report, we show that dacomitinib, a pan-ErbB receptor inhibitor, diminished growth, clonogenic potential, anoikis resistance and induced apoptotic cell death in therapy-resistant EOC cells. Dacominitib inhibited PLK1-FOXM1 signalling pathway and its down-stream targets Aurora kinase B and survivin. Moreover, dacomitinib attenuated migration and invasion of the EOC cells and reduced expression of epithelial-to-mesenchymal transition (EMT) markers ZEB1, ZEB2 and CDH2 (which encodes N-cadherin). Conversely, the anti-tumour activity of single-targeted ErbB agents including cetuximab (a ligand-blocking anti-EGFR mAb), transtuzumab (anti-HER2 mAb), H3.105.5 (anti-HER3 mAb) and erlotinib (EGFR small-molecule tyrosine kinase inhibitor) were marginal. Our results provide a rationale for further investigation on the therapeutic potential of dacomitinib in treatment of the chemoresistant EOC.
Collapse
Affiliation(s)
- Majid Momeny
- Haematology/Oncology and Stem Cell Transplantation Research Centre, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Zarrinrad
- Haematology/Oncology and Stem Cell Transplantation Research Centre, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farima Moghaddaskho
- Haematology/Oncology and Stem Cell Transplantation Research Centre, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Poursheikhani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Sankanian
- Haematology/Oncology and Stem Cell Transplantation Research Centre, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Azam Zaghal
- Haematology/Oncology and Stem Cell Transplantation Research Centre, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahab Mirshahvaladi
- Department of Molecular Systems Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fatemeh Esmaeili
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Haniyeh Eyvani
- Haematology/Oncology and Stem Cell Transplantation Research Centre, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farinaz Barghi
- Haematology/Oncology and Stem Cell Transplantation Research Centre, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Sabourinejad
- Haematology/Oncology and Stem Cell Transplantation Research Centre, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zivar Alishahi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Yousefi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Ghasemi
- Section of Stem Cell Biology, Division of Oncology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Leila Dardaei
- Massachusetts General Hospital Cancer Centre, Charlestown, MA, USA
| | - Davood Bashash
- Department of Haematology and Blood Banking, Faculty of Allied Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahram Chahardouli
- Haematology/Oncology and Stem Cell Transplantation Research Centre, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad R Dehpour
- Experimental Medicine Research Centre, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Javad Tavakkoly-Bazzaz
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kamran Alimoghaddam
- Haematology/Oncology and Stem Cell Transplantation Research Centre, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ardeshir Ghavamzadeh
- Haematology/Oncology and Stem Cell Transplantation Research Centre, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Haematology/Oncology and Stem Cell Transplantation Research Centre, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Smolensky D, Rathore K, Bourn J, Cekanova M. Inhibition of the PI3K/AKT Pathway Sensitizes Oral Squamous Cell Carcinoma Cells to Anthracycline-Based Chemotherapy In Vitro. J Cell Biochem 2017; 118:2615-2624. [PMID: 27649518 DOI: 10.1002/jcb.25747] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/19/2016] [Indexed: 01/21/2023]
Abstract
Anthracycline-based chemotherapy, such as doxorubicin (Dox), while effective against many solid tumors, is not widely used for head and neck cancers. In this study, we evaluated the efficacy of Dox, and its derivative AD198 in human, canine, and feline oral squamous cell carcinomas cells (OSCC) in vitro. Dox and AD198 had significant an anti-proliferative effect on human, canine, and feline OSCC cells in dose-dependent manner. AD198 inhibited cell proliferation more effectively than Dox in tested OSCC cells. In the human oral squamous cell carcinoma SCC25 cells, Dox and AD198 increased the production of reactive oxygen species and subsequently increased apoptosis through activation of caspase signaling pathway. Dox and AD198 increased activation of AKT, ERK1/2, and p38 MAPK signaling pathways in tested OSCC cells by dose-dependent manner. The efficacy of Dox and AD198 treatments in inhibition of cell proliferation was increased in tested OSCC when combined with PI3K/AKT inhibitor, LY294002 treatment. Inhibition of PI3K/AKT reduced Dox- and AD198-induced activation of ERK1/2 and further increased Dox- and AD198-induced phosphorylation of p38 MAPK in OSCC. Our results suggest that the anthracycline therapies, such as Dox or AD198, can be more effective for treatment of OSCC when combined with inhibitors of the PI3K/AKT pathway. J. Cell. Biochem. 118: 2615-2624, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Dmitriy Smolensky
- UT-ORNL Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, 37996, Tennessee.,Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, 37996, Tennessee
| | - Kusum Rathore
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, 37996, Tennessee
| | - Jennifer Bourn
- UT-ORNL Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, 37996, Tennessee.,Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, 37996, Tennessee
| | - Maria Cekanova
- UT-ORNL Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, 37996, Tennessee.,Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, 37996, Tennessee
| |
Collapse
|
38
|
ErbB Family Signalling: A Paradigm for Oncogene Addiction and Personalized Oncology. Cancers (Basel) 2017; 9:cancers9040033. [PMID: 28417948 PMCID: PMC5406708 DOI: 10.3390/cancers9040033] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 02/07/2023] Open
Abstract
ErbB family members represent important biomarkers and drug targets for modern precision therapy. They have gained considerable importance as paradigms for oncoprotein addiction and personalized medicine. This review summarizes the current understanding of ErbB proteins in cell signalling and cancer and describes the molecular rationale of prominent cases of ErbB oncoprotein addiction in different cancer types. In addition, we have highlighted experimental technologies for the development of innovative cancer cell models that accurately predicted clinical ErbB drug efficacies. In the future, such cancer models might facilitate the identification and validation of physiologically relevant novel forms of oncoprotein and non-oncoprotein addiction or synthetic lethality. The identification of genotype-drug response relationships will further advance personalized oncology and improve drug efficacy in the clinic. Finally, we review the most important drugs targeting ErbB family members that are under investigation in clinical trials or that made their way already into clinical routine. Taken together, the functional characterization of ErbB oncoproteins have significantly increased our knowledge on predictive biomarkers, oncoprotein addiction and patient stratification and treatment.
Collapse
|
39
|
Liu JF, Ray-Coquard I, Selle F, Poveda AM, Cibula D, Hirte H, Hilpert F, Raspagliesi F, Gladieff L, Harter P, Siena S, Del Campo JM, Tabah-Fisch I, Pearlberg J, Moyo V, Riahi K, Nering R, Kubasek W, Adiwijaya B, Czibere A, Naumann RW, Coleman RL, Vergote I, MacBeath G, Pujade-Lauraine E. Randomized Phase II Trial of Seribantumab in Combination With Paclitaxel in Patients With Advanced Platinum-Resistant or -Refractory Ovarian Cancer. J Clin Oncol 2016; 34:4345-4353. [PMID: 27998236 DOI: 10.1200/jco.2016.67.1891] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Seribantumab is a fully human immunoglobulin G2 monoclonal antibody that binds to human epidermal growth factor receptor (HER) 3 (ErbB3), blocking heregulin (HRG) -mediated ErbB3 signaling and inducing ErbB3 receptor downregulation. This open-label randomized phase II study evaluated progression-free survival (PFS) with seribantumab in combination with once-per-week paclitaxel compared with paclitaxel alone in patients with platinum-resistant or -refractory ovarian cancer. A key secondary objective was to determine if any of five prespecified biomarkers predicted benefit from seribantumab. Patients and Methods Patients with platinum-resistant or -refractory epithelial ovarian, fallopian tube, or primary peritoneal cancer were randomly assigned at a ratio of two to one to receive seribantumab plus paclitaxel or paclitaxel alone. Patients underwent pretreatment core needle biopsy; archival tumor samples were also obtained to support biomarker analyses. Results A total of 223 patients were randomly assigned (seribantumab plus paclitaxel, n = 140; paclitaxel alone, n = 83). Median PFS in the unselected intent-to-treat population was 3.75 months with seribantumab plus paclitaxel compared with 3.68 months with paclitaxel alone (hazard ratio [HR], 1.027; 95% CI, 0.741 to 1.425; P = .864). Among patients whose tumors had detectable HRG mRNA and low HER2 (n = 57 [38%] of 151 with available biomarker data), increased treatment benefit was observed in those receiving seribantumab plus paclitaxel compared with paclitaxel alone (PFS HR, 0.37; 95% CI, 0.18 to 0.76; P = .007). The HR in patients not meeting these criteria was 1.80 (95% CI, 1.08 to 2.98; P = .023). Conclusion The addition of seribantumab to paclitaxel did not result in improved PFS in unselected patients. Exploratory analyses suggest that detectable HRG and low HER2, biomarkers that link directly to the mechanism of action of seribantumab, identified patients who might benefit from this combination. Future clinical trials are needed to validate this finding and should preselect for HRG expression and focus on cancers with low HER2 levels.
Collapse
Affiliation(s)
- Joyce F Liu
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Isabelle Ray-Coquard
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Frederic Selle
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Andrés M Poveda
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - David Cibula
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Hal Hirte
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Felix Hilpert
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Francesco Raspagliesi
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Laurence Gladieff
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Philipp Harter
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Salvatore Siena
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Josep Maria Del Campo
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Isabelle Tabah-Fisch
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Joseph Pearlberg
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Victor Moyo
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Kaveh Riahi
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Rachel Nering
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - William Kubasek
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Bambang Adiwijaya
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Akos Czibere
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - R Wendel Naumann
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Robert L Coleman
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Ignace Vergote
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Gavin MacBeath
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - Eric Pujade-Lauraine
- Joyce F. Liu, Dana-Farber Cancer Institute, Boston; Isabelle Tabah-Fisch and Joseph Pearlberg, Sanofi Oncology; Victor Moyo, Kaveh Riahi, Rachel Nering, William Kubasek, Bambang Adiwijaya, Akos Czibere, and Gavin MacBeath, Merrimack Pharmaceuticals, Cambridge, MA; R. Wendel Naumann, Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC; Robert L. Coleman, MD Anderson Cancer Center, Houston, TX; Isabelle Ray-Coquard, Centre Léon Bérard, University Claude Bernard, and Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Lyon; Frederic Selle, Hôpital Tenon and GINECO; Eric Pujade-Lauraine, Hôpital Hotel-Dieu, Université Paris Descartes, and GINECO, Paris; Laurence Gladieff, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse Oncopole, and GINECO, Toulouse, France; Andrés M. Poveda, Instituto Valenciano de Oncologia and Grupo Español de Investigación en Cáncer de Ovario (GEICO), Valencia; Josep Maria del Campo, Hospital Universitari Vall d'Hebron and GEICO, Barcelona, Spain; David Cibula, General University Hospital Prague and Charles University, Prague, Czech Republic; Hal Hirte, Hamilton Health Sciences-Juravinski Cancer Centre, Hamilton, Ontario, Canada; Felix Hilpert, Universitätsklinikum Schleswig-Holstein, Kiel; Philipp Harter, Kliniken Essen Mitte and Arbeitsgemeinschaft Gynäkologische Onkologie, Essen, Germany; Francesco Raspagliesi, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori; Salvatore Siena, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, and Università degli Studi di Milano, Milano, Italy; and Ignace Vergote, University Hospital Katholieke Universiteit Leuven and Leuven Cancer Institute, Leuven, Belgium
| |
Collapse
|
40
|
Schram AM, Iasonos A, Hyman DM. Picking the Right Patient for Human Epidermal Growth Factor Receptor 3-Targeted Therapy in Platinum-Resistant Ovarian Cancer. J Clin Oncol 2016; 34:4312-4314. [PMID: 27998226 PMCID: PMC5455312 DOI: 10.1200/jco.2016.69.7169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Alison M Schram
- Alison M. Schram, Alexia Iasonos, and David M. Hyman, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexia Iasonos
- Alison M. Schram, Alexia Iasonos, and David M. Hyman, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David M Hyman
- Alison M. Schram, Alexia Iasonos, and David M. Hyman, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
41
|
Malm M, Frejd FY, Ståhl S, Löfblom J. Targeting HER3 using mono- and bispecific antibodies or alternative scaffolds. MAbs 2016; 8:1195-1209. [PMID: 27532938 PMCID: PMC5058629 DOI: 10.1080/19420862.2016.1212147] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The human epidermal growth factor receptor 3 (HER3) has in recent years been recognized as a key node in the complex signaling network of many different cancers. It is implicated in de novo and acquired resistance against therapies targeting other growth factor receptors, e.g., EGFR, HER2, and it is a major activator of the PI3K/Akt signaling pathway. Consequently, HER3 has attracted substantial attention, and is today a key target for drugs in clinical development. Sophisticated protein engineering approaches have enabled the generation of a range of different affinity proteins targeting this receptor, including antibodies and alternative scaffolds that are either mono- or bispecific. Here, we describe HER3 and its role as a key tumor target, and give a comprehensive review of HER3-targeted proteins currently in development, including discussions on the opportunities and challenges of targeting this receptor.
Collapse
Affiliation(s)
- Magdalena Malm
- a Division of Protein Technology, School of Biotechnology, KTH-Royal Institute of Technology, SE , Stockholm
| | - Fredrik Y Frejd
- b Affibody AB, SE, Stockholm , Sweden.,c Department of Immunology , Genetics and Pathology, Uppsala University , Uppsala , Sweden
| | - Stefan Ståhl
- a Division of Protein Technology, School of Biotechnology, KTH-Royal Institute of Technology, SE , Stockholm
| | - John Löfblom
- a Division of Protein Technology, School of Biotechnology, KTH-Royal Institute of Technology, SE , Stockholm
| |
Collapse
|
42
|
Kurzeder C, Bover I, Marmé F, Rau J, Pautier P, Colombo N, Lorusso D, Ottevanger P, Bjurberg M, Marth C, Barretina-Ginesta P, Vergote I, Floquet A, del Campo JM, Mahner S, Bastière-Truchot L, Martin N, Oestergaard MZ, Kiermaier A, Schade-Brittinger C, Polleis S, du Bois A, Gonzalez-Martin A. Double-Blind, Placebo-Controlled, Randomized Phase III Trial Evaluating Pertuzumab Combined With Chemotherapy for Low Tumor Human Epidermal Growth Factor Receptor 3 mRNA–Expressing Platinum-Resistant Ovarian Cancer (PENELOPE). J Clin Oncol 2016; 34:2516-25. [DOI: 10.1200/jco.2015.66.0787] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose The AGO-OVAR 2.29/ENGOT-ov14/PENELOPE prospectively randomized phase III trial evaluated the addition of pertuzumab to chemotherapy in patients with platinum-resistant ovarian carcinoma with low tumor human epidermal growth factor receptor 3 (HER3) mRNA expression. We report the results of the primary efficacy analysis. Patients and Methods Eligible patients had ovarian carcinoma that progressed during or within 6 months of completing four or more platinum cycles, centrally tested low tumor HER3 mRNA expression (concentration ratio ≤ 2.81 by quantitative reverse transcriptase polymerase chain reaction on cobas z480 [Roche Molecular Diagnostics, Pleasanton, CA]), and no more than two prior lines of chemotherapy. After investigators’ selection of the chemotherapy backbone (single-agent topotecan, weekly paclitaxel, or gemcitabine), patients were randomly assigned to also receive either placebo or pertuzumab (840-mg loading dose followed by 420 mg every 3 weeks). Stratification factors were selected chemotherapy, prior antiangiogenic therapy, and platinum-free interval. The primary end point was independent review committee–assessed progression-free survival (PFS). Additional end points included overall survival, investigator-assessed PFS, objective response rate, safety, patient-reported outcomes, and translational research. Results Overall, 156 patients were randomly assigned. Adding pertuzumab to chemotherapy did not significantly improve independent review committee–assessed PFS for the primary analysis (stratified hazard ratio, 0.74; 95% CI, 0.50 to 1.11; P = .14; median PFS, 4.3 months for pertuzumab plus chemotherapy v 2.6 months for placebo plus chemotherapy). Sensitivity analyses and secondary efficacy end point results were consistent with the primary analysis. The effect on PFS favoring pertuzumab was more pronounced in the gemcitabine and paclitaxel cohorts. No new safety signals were seen. Conclusion Although the primary objective was not met, subgroup analyses showed trends in PFS favoring pertuzumab in the gemcitabine and paclitaxel cohorts, meriting further exploration of pertuzumab in ovarian cancer.
Collapse
Affiliation(s)
- Christian Kurzeder
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Isabel Bover
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Frederik Marmé
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Joern Rau
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Patricia Pautier
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Nicoletta Colombo
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Domenica Lorusso
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Petronella Ottevanger
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Maria Bjurberg
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Christian Marth
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Pilar Barretina-Ginesta
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Ignace Vergote
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Anne Floquet
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Josep M. del Campo
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Sven Mahner
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Lydie Bastière-Truchot
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Nicolas Martin
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Mikkel Z. Oestergaard
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Astrid Kiermaier
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Carmen Schade-Brittinger
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Sandra Polleis
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Andreas du Bois
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| | - Antonio Gonzalez-Martin
- Christian Kurzeder and Andreas du Bois, Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and Kliniken Essen Mitte, Essen; Frederik Marmé, AGO and University Hospital Heidelberg, Heidelberg; Joern Rau and Carmen Schade-Brittinger, Coordinating Center for Clinical Trials, Philipps-University of Marburg, Marburg; Sven Mahner, AGO and University Medical Center Hamburg-Eppendorf, Hamburg; Sandra Polleis, AGO Study Group, Wiesbaden, Germany; Isabel Bover, Grupo Español de Investigación en Cáncer de Ovario
| |
Collapse
|
43
|
Smolensky D, Rathore K, Cekanova M. Phosphatidylinositol- 3-kinase inhibitor induces chemosensitivity to a novel derivative of doxorubicin, AD198 chemotherapy in human bladder cancer cells in vitro. BMC Cancer 2015; 15:927. [PMID: 26597249 PMCID: PMC4657321 DOI: 10.1186/s12885-015-1930-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 11/13/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Doxorubicin (Dox) is widely used to treat progressed bladder cancer after transurethral resection. The use of Dox-chemotherapy has been limited due to induced drug resistance and cumulative cardiotoxic effects. N-benzyladriamycin-14-valerate (AD198), a novel derivative of Dox, has a potential to become a more effective treatment than Dox by overcoming drug resistance and cardio-toxicity as shown in the rodent model of lymphoma in vivo. The purpose of this study was to compare the efficacy of Dox and AD198 and explore their mechanisms in inhibition on human bladder cancer cells in vitro. METHODS We evaluated the effects of Dox and AD198 on cell viability of human transitional cell carcinoma (TCC) cell lines T24 and UMUC3 by MTS assay in vitro. The effects of Dox and AD198 on cell apoptosis were determined by caspase 3/7 assay, generation of reactive oxygen species (ROS), and Western Blotting (WB) analysis. RESULTS AD198 was more effective than Dox in inhibition of cell viability of T24 and UMUC3 cells in vitro. Both Dox and AD198 significantly increased the generation of ROS and induced apoptosis in caspase-dependent and -independent manner in T24 and UMUC3 cells. AD 198 induced significantly higher production of ROS as compared to Dox in human TCC cells. Dox and AD198 activated the pro-apoptotic p38 MAPK pathway; however, on the other hand also increased phosphorylation of AKT, an anti-apoptotic signaling pathway, in T24 and UMUC3 cells. Combined treatment of PI3K inhibitor (LY294002) with Dox or AD198 inhibited cell viability of T24 and UMUC3 cells more effectively than any of drug treatments alone. CONCLUSIONS These data suggest that AD198 as novel derivative of Dox, could be a used as effective treatment for bladder cancer. Dox and AD198 induced PI3K/AKT signaling pathway that is a one of the indicators of pro-survival and possible drug-resistance mechanisms of chemotherapies in bladder cancer. Combined therapies of Dox or AD198 with inhibitors of PI3K/AKT signaling pathway might lead to more effective treatment outcome for patients diagnosed with bladder cancer based on our in vitro experiments.
Collapse
Affiliation(s)
- Dmitriy Smolensky
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, The University of Tennessee, 2407 River Drive A122, Knoxville, TN, 37996, USA. .,UT-ORNL Graduate School of Genome Science and Technology, The University of Tennessee, Knoxville, TN, 37996, USA.
| | - Kusum Rathore
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, The University of Tennessee, 2407 River Drive A122, Knoxville, TN, 37996, USA.
| | - Maria Cekanova
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, The University of Tennessee, 2407 River Drive A122, Knoxville, TN, 37996, USA. .,UT-ORNL Graduate School of Genome Science and Technology, The University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
44
|
Effects of silibinin on growth and invasive properties of human ovarian carcinoma cells through suppression of heregulin/HER3 pathway. Tumour Biol 2015; 37:3913-23. [DOI: 10.1007/s13277-015-4220-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 10/12/2015] [Indexed: 10/22/2022] Open
|
45
|
Riethdorf S, Frey S, Santjer S, Stoupiec M, Otto B, Riethdorf L, Koop C, Wilczak W, Simon R, Sauter G, Pantel K, Assmann V. Diverse expression patterns of the EMT suppressor grainyhead-like 2 (GRHL2) in normal and tumour tissues. Int J Cancer 2015; 138:949-63. [PMID: 26355710 DOI: 10.1002/ijc.29841] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 09/03/2015] [Indexed: 12/16/2022]
Abstract
The transcription factor grainyhead-like 2 (GRHL2) plays a crucial role in various developmental processes. Although GRHL2 recently has attracted considerable interest in that it could be identified as a novel suppressor of the epithelial-to-mesenchymal transition, evidence is emerging that GRHL2 also exhibits tumour-promoting activities. Aim of the present study therefore was to help defining the relevance of GRHL2 for human cancers by performing a comprehensive immunohistochemical analysis of GRHL2 expression in normal (n = 608) and (n = 3,143) tumour tissues using tissue microarrays. Consistent with its accepted role in epithelial morphogenesis, GRHL2 expression preferentially but not exclusively was observed in epithelial cells. Regenerative and proliferating epithelial cells with stem cell features showed a strong GRHL2 expression. Highly complex GRHL2 expression patterns indicative of both reduced and elevated GRHL2 expression in tumours, possibly reflecting potential tumour-suppressing as well as oncogenic functions of GRHL2 in distinct human tumours, were observed. A dysregulation of GRHL2 expression for the first time was found in tumours of non-epithelial origin (e.g., astrocytomas, melanomas). We also report GRHL2 copy number gains which, however, did not necessarily translate into increased GRHL2 expression levels in cancer cells. Results obtained by meta-analysis of gene expression microarray data in conjunction with functional assays demonstrating a direct regulation of HER3 expression further point to a potential therapeutic relevance of GRHL2 in ovarian cancer. Hopefully, the results presented in this study may pave the way for a better understanding of the yet largely unknown function of GRHL2 in the initiation, progression and also therapy of cancers.
Collapse
Affiliation(s)
- Sabine Riethdorf
- Department of Tumour Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabrina Frey
- Department of Tumour Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sonja Santjer
- Department of Tumour Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malgorzata Stoupiec
- Department of Tumour Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Otto
- Department of Internal Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Christina Koop
- Department of Pathology, Center for Diagnostic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Waldemar Wilczak
- Department of Pathology, Center for Diagnostic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Department of Pathology, Center for Diagnostic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Department of Pathology, Center for Diagnostic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumour Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volker Assmann
- Department of Tumour Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
46
|
Durrant DE, Das A, Dyer S, Tavallai S, Dent P, Kukreja RC. Targeted Inhibition of Phosphoinositide 3-Kinase/Mammalian Target of Rapamycin Sensitizes Pancreatic Cancer Cells to Doxorubicin without Exacerbating Cardiac Toxicity. Mol Pharmacol 2015; 88:512-23. [PMID: 26101222 PMCID: PMC4551046 DOI: 10.1124/mol.115.099143] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/16/2015] [Indexed: 12/22/2022] Open
Abstract
Pancreatic cancer has the lowest 5-year survival rate of all major cancers despite decades of effort to design and implement novel, more effective treatment options. In this study, we tested whether the dual phosphoinositide 3-kinase/mechanistic target of rapamycin inhibitor BEZ235 (BEZ) potentiates the antitumor effects of doxorubicin (DOX) against pancreatic cancer. Cotreatment of BEZ235 with DOX resulted in dose-dependent inhibition of the phosphoinositide 3-kinase/mechanistic target of rapamycin survival pathway, which corresponded with an increase in poly ADP ribose polymerase cleavage. Moreover, BEZ cotreatment significantly improved the effects of DOX toward both cell viability and cell death in part through reduced Bcl-2 expression and increased expression of the shorter, more cytotoxic forms of BIM. BEZ also facilitated intracellular accumulation of DOX, which led to enhanced DNA damage and reactive oxygen species generation. Furthermore, BEZ in combination with gemcitabine reduced MiaPaca2 cell proliferation but failed to increase reactive oxygen species generation or BIM expression, resulting in reduced necrosis and apoptosis. Treatment with BEZ and DOX in mice bearing tumor xenographs significantly repressed tumor growth as compared with BEZ, DOX, or gemcitabine. Additionally, in contrast to the enhanced expression seen in MiaPaca2 cells, BEZ and DOX cotreatment reduced BIM expression in H9C2 cardiomyocytes. Also, the Bcl-2/Bax ratio was increased, which was associated with a reduction in cell death. In vivo echocardiography showed decreased cardiac function with DOX treatment, which was not improved by combination treatment with BEZ. Thus, we propose that combining BEZ with DOX would be a better option for patients than current standard of care by providing a more effective tumor response without the associated increase in toxicity.
Collapse
Affiliation(s)
- David E Durrant
- Department of Biochemistry and Molecular Biology (D.E.D., S.T., P.D.), Department of Internal Medicine, Division of Cardiology, Pauley Heart Center (A.D., S.D., R.C.K.), and Department of Physiology and Biophysics (R.C.K.), Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Anindita Das
- Department of Biochemistry and Molecular Biology (D.E.D., S.T., P.D.), Department of Internal Medicine, Division of Cardiology, Pauley Heart Center (A.D., S.D., R.C.K.), and Department of Physiology and Biophysics (R.C.K.), Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Samya Dyer
- Department of Biochemistry and Molecular Biology (D.E.D., S.T., P.D.), Department of Internal Medicine, Division of Cardiology, Pauley Heart Center (A.D., S.D., R.C.K.), and Department of Physiology and Biophysics (R.C.K.), Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Seyedmehrad Tavallai
- Department of Biochemistry and Molecular Biology (D.E.D., S.T., P.D.), Department of Internal Medicine, Division of Cardiology, Pauley Heart Center (A.D., S.D., R.C.K.), and Department of Physiology and Biophysics (R.C.K.), Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Paul Dent
- Department of Biochemistry and Molecular Biology (D.E.D., S.T., P.D.), Department of Internal Medicine, Division of Cardiology, Pauley Heart Center (A.D., S.D., R.C.K.), and Department of Physiology and Biophysics (R.C.K.), Virginia Commonwealth University Medical Center, Richmond, Virginia
| | - Rakesh C Kukreja
- Department of Biochemistry and Molecular Biology (D.E.D., S.T., P.D.), Department of Internal Medicine, Division of Cardiology, Pauley Heart Center (A.D., S.D., R.C.K.), and Department of Physiology and Biophysics (R.C.K.), Virginia Commonwealth University Medical Center, Richmond, Virginia
| |
Collapse
|
47
|
Brown KK, Montaser-Kouhsari L, Beck AH, Toker A. MERIT40 Is an Akt Substrate that Promotes Resolution of DNA Damage Induced by Chemotherapy. Cell Rep 2015; 11:1358-66. [PMID: 26027929 DOI: 10.1016/j.celrep.2015.05.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 03/12/2015] [Accepted: 05/01/2015] [Indexed: 11/30/2022] Open
Abstract
Resistance to cytotoxic chemotherapy drugs, including doxorubicin, is a significant obstacle to the effective treatment of breast cancer. Here, we have identified a mechanism by which the PI3K/Akt pathway mediates resistance to doxorubicin. In addition to inducing DNA damage, doxorubicin triggers sustained activation of Akt signaling in breast cancer cells. We show that Akt contributes to chemotherapy resistance such that PI3K or Akt inhibitors sensitize cells to doxorubicin. We identify MERIT40, a component of the BRCA1-A DNA damage repair complex, as an Akt substrate that is phosphorylated following doxorubicin treatment. MERIT40 phosphorylation facilitates assembly of the BRCA1-A complex in response to DNA damage and contributes to DNA repair and cell survival following doxorubicin treatment. Finally, MERIT40 phosphorylation in human breast cancers is associated with estrogen receptor positivity. Our findings suggest that combination therapy with PI3K or Akt inhibitors and doxorubicin may constitute a successful strategy for overcoming chemotherapy resistance.
Collapse
Affiliation(s)
- Kristin K Brown
- Departments of Pathology and Medicine and Cancer Center, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA 02215, USA
| | - Laleh Montaser-Kouhsari
- Departments of Pathology and Medicine and Cancer Center, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA 02215, USA
| | - Andrew H Beck
- Departments of Pathology and Medicine and Cancer Center, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA 02215, USA
| | - Alex Toker
- Departments of Pathology and Medicine and Cancer Center, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
48
|
Ding L, He S, Sun X. HSP70 desensitizes osteosarcoma cells to baicalein and protects cells from undergoing apoptosis. Apoptosis 2015; 19:1269-80. [PMID: 24846187 DOI: 10.1007/s10495-014-0995-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Baicalein is a new drug that has shown promising anti-cancer effects against a broad spectrum of tumors. However, the potential effect on osteosarcoma cells and the mechanisms involved are still largely unknown. Resistance to chemotherapy remains a major obstacle in cancer therapy. Therefore, the aim of the present study was to investigate the anti-tumor effect of baicalein on human osteosarcoma cancer cells and the molecular mechanism involved, as well as identify possible mechanisms of drug resistance. Our results revealed that baicalein-induced apoptosis in osteosarcoma cells was via a mitochondrial pathway involving both caspase-dependent and independent mechanisms. Notably, baicalein treatment upregulated the expression of HSP70, which partially prevented human osteosarcoma cells from undergoing apoptosis. Moreover, it was revealed that HSP70 expression decreased the sensitivity of osteosarcoma cells to baicalein via activation of PI3K/AKT and MAPK/ERK pathways. These results suggest that targeting HSP70-mediated drug resistance, in combination with chemotherapy drugs, may provide novel therapeutic opportunities.
Collapse
Affiliation(s)
- Lianghua Ding
- Department of Orthopedics, The Third Affiliated Hospital of Suzhou University, No 185 Juqian Street, Changzhou, 213003, China
| | | | | |
Collapse
|
49
|
Mei L, Liu Y, Zhang Q, Gao H, Zhang Z, He Q. Enhanced antitumor and anti-metastasis efficiency via combined treatment with CXCR4 antagonist and liposomal doxorubicin. J Control Release 2014; 196:324-31. [PMID: 25450404 DOI: 10.1016/j.jconrel.2014.10.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 10/01/2014] [Accepted: 10/16/2014] [Indexed: 12/18/2022]
Abstract
Metastasis is the main cause of cancer treatment failure and death. However, current therapies are designed to impair carcinoma metastasis mainly by impairing initial dissemination events. CXCR4 is a G-protein coupled receptor that exclusively binds its ligand CXCL12, which can stimulate cells to metastasize to distant sites. As the antagonist of chemokine receptor CXCR4, Peptide S exhibited anti-metastasis effect. In order to enhance treatment efficiency through destroying primary tumors and inhibiting their metastases, we combined PEGylated doxorubicin-loaded liposomes (DOX-Lip) with anti-metastasis Peptide S for tumor therapy for the first time. DOX-Lip exhibited similar cytotoxic activity compared to free DOX in vitro, and Peptide S showed no toxic effect on cell viability. However, the Peptide S sensitized CXCR4-positive B16F10 melanoma cells to DOX-Lip (5 μM) when cocultured with stromal cells (50.18±0.29% of viable cells in the absence of Peptide S vs 33.70±3.99% of viable cells in the presence of Peptide S). Both Peptide S and DOX-Lip inhibited the adhesion of B16F10 cells to stromal cells. We further confirmed that the inhibition of phosphorylated Akt (pAkt) by Peptide S played a key role due to the fact that activation of pAkt by DOX-Lip promoted resistance to chemotherapy. Migration and invasion assays showed that DOX-Lip enhanced anti-metastasis effect of Peptide S in vitro because of the cytotoxicity of doxorubicin. In vivo studies also showed that the combined treatment with DOX-Lip and Peptide S not only retarded primary tumor growth, but also reduced lung metastasis. Both the DOX-Lip and DOX-Lip+Peptide S exhibited even more outstanding tumor inhibition effect (with tumor growth inhibition rates of 32.1% and 37.9% respectively). In conclusion, our combined treatment with CXCR4 antagonist and liposomal doxorubicin was proved to be promising for antitumor and anti-metastasis therapy.
Collapse
Affiliation(s)
- Ling Mei
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, PR China
| | - Yayuan Liu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, PR China
| | - Qianyu Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, PR China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, PR China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, PR China
| | - Qin He
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, and State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu 610041, PR China.
| |
Collapse
|
50
|
|