1
|
Dix-Peek T, Dickens C, Valcárcel J, Duarte RAB. Lower FGFR2 mRNA Expression and Higher Levels of FGFR2 IIIc in HER2-Positive Breast Cancer. BIOLOGY 2024; 13:920. [PMID: 39596875 PMCID: PMC11591975 DOI: 10.3390/biology13110920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
Fibroblast growth factor receptor 2 (FGFR2) has been associated with breast cancer. We performed in silico analyses to investigate the FGFR2 mRNA expression and splice variants associated with breast cancer subtypes. Online databases, including cBioPortal and TCGA SpliceSeq, were used to examine the association between the FGFR2 expression and splice variants with breast cancer subtypes. A higher FGFR2 mRNA was significantly associated with luminal, oestrogen receptor (ER)-positive breast cancers, and invasive lobular carcinomas, whereas a lower FGFR2 was associated with human epidermal growth factor receptor 2 (HER2)-positive breast cancer and invasive ductal carcinomas. The epithelial alternatively spliced FGFR2 IIIb isoform was significantly enriched in ER+ breast cancer, while the mesenchymal FGFR2 IIIc isoform was significantly prevalent in HER2+ cancer. Increased levels of FGFR2 and IIIb splice isoforms are associated with less aggressive breast cancer phenotypes, while decreased levels of FGFR2 and increased IIIc splice isoform are associated with more aggressive phenotypes.
Collapse
Affiliation(s)
- Thérèse Dix-Peek
- Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 07 York Road, Parktown, Johannesburg 2193, South Africa;
| | - Caroline Dickens
- Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 07 York Road, Parktown, Johannesburg 2193, South Africa;
| | - Juan Valcárcel
- ICREA and Center for Genomic Regulation (CRG), Dr. Aiguader 88, 08003 Barcelona, Spain;
| | - Raquel A. B. Duarte
- Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 07 York Road, Parktown, Johannesburg 2193, South Africa;
| |
Collapse
|
2
|
Chi F, Griffiths JI, Nath A, Bild AH. Paradoxical cancer cell proliferation after FGFR inhibition through decreased p21 signaling in FGFR1-amplified breast cancer cells. Breast Cancer Res 2024; 26:54. [PMID: 38553760 PMCID: PMC10979625 DOI: 10.1186/s13058-024-01808-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
Fibroblast growth factors (FGFs) control various cellular functions through fibroblast growth factor receptor (FGFR) activation, including proliferation, differentiation, migration, and survival. FGFR amplification in ER + breast cancer patients correlate with poor prognosis, and FGFR inhibitors are currently being tested in clinical trials. By comparing three-dimensional spheroid growth of ER + breast cancer cells with and without FGFR1 amplification, our research discovered that FGF2 treatment can paradoxically decrease proliferation in cells with FGFR1 amplification or overexpression. In contrast, FGF2 treatment in cells without FGFR1 amplification promotes classical FGFR proliferative signaling through the MAPK cascade. The growth inhibitory effect of FGF2 in FGFR1 amplified cells aligned with an increase in p21, a cell cycle inhibitor that hinders the G1 to S phase transition in the cell cycle. Additionally, FGF2 addition in FGFR1 amplified cells activated JAK-STAT signaling and promoted a stem cell-like state. FGF2-induced paradoxical effects were reversed by inhibiting p21 or the JAK-STAT pathway and with pan-FGFR inhibitors. Analysis of patient ER + breast tumor transcriptomes from the TCGA and METABRIC datasets demonstrated a strong positive association between expression of FGF2 and stemness signatures, which was further enhanced in tumors with high FGFR1 expression. Overall, our findings reveal a divergence in FGFR signaling, transitioning from a proliferative to stemness state driven by activation of JAK-STAT signaling and modulation of p21 levels. Activation of these divergent signaling pathways in FGFR amplified cancer cells and paradoxical growth effects highlight a challenge in the use of FGFR inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Feng Chi
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Institute, 1218 S Fifth Ave, Monrovia, CA, 91016, USA
| | - Jason I Griffiths
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Institute, 1218 S Fifth Ave, Monrovia, CA, 91016, USA
| | - Aritro Nath
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Institute, 1218 S Fifth Ave, Monrovia, CA, 91016, USA
| | - Andrea H Bild
- Department of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Institute, 1218 S Fifth Ave, Monrovia, CA, 91016, USA.
| |
Collapse
|
3
|
Lee J, Choi SR, Cho KH. Network Dynamics Caused by Genomic Alteration Determine the Therapeutic Response to FGFR Inhibitors for Lung Cancer. Biomolecules 2022; 12:biom12091197. [PMID: 36139037 PMCID: PMC9496101 DOI: 10.3390/biom12091197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/13/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Recently, FGFR inhibitors have been highlighted as promising targeted drugs due to the high prevalence of FGFR1 amplification in cancer patients. Although various potential biomarkers for FGFR inhibitors have been suggested, their functional effects have been shown to be limited due to the complexity of the cancer signaling network and the heterogenous genomic conditions of patients. To overcome such limitations, we have reconstructed a lung cancer network model by integrating a cell line genomic database and analyzing the model in order to understand the underlying mechanism of heterogeneous drug responses. Here, we identify novel genomic context-specific candidates that can increase the efficacy of FGFR inhibitors. Furthermore, we suggest optimal targets that can induce more effective therapeutic responses than that of FGFR inhibitors in each of the FGFR-resistant lung cancer cells through computational simulations at a system level. Our findings provide new insights into the regulatory mechanism of differential responses to FGFR inhibitors for optimal therapeutic strategies in lung cancer.
Collapse
Affiliation(s)
| | | | - Kwang-Hyun Cho
- Correspondence: ; Tel.: +82-42-350-4325; Fax: +82-42-350-4310
| |
Collapse
|
4
|
Krzyscik MA, Opaliński Ł, Szymczyk J, Otlewski J. Cyclic and dimeric fibroblast growth factor 2 variants with high biomedical potential. Int J Biol Macromol 2022; 218:243-258. [PMID: 35878661 DOI: 10.1016/j.ijbiomac.2022.07.105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/29/2022] [Accepted: 07/14/2022] [Indexed: 11/05/2022]
Abstract
Fibroblast growth factor 2 (FGF2) is a pleiotropic protein engaged in the regulation of key cellular processes in a wide spectrum of cells. FGF2 is an important object of basic research as well as a molecule used in regenerative medicine, in vitro cell culture maintenance, and as an anticancer drug carrier. However, the unsatisfactory stability and pleiotropic activities of the wild-type FGF2 largely limit its use as a medical product. To overcome these limitations, we have designed a set of FGF2-based macromolecules via sortase A-mediated cyclization and oligomerization. We obtained heparin-switchable FGF2 variants with enhanced stability and improved ability to stimulate cell proliferation and migration. We have shown that stimulation of glucose uptake by adipocytes is modulated by the architecture of FGF2 oligomers. Moreover, we used hyper-stable FGF2 variants for the construction of highly effective drug carriers for selective killing of FGFR1-overproducing cancer cells. The strategy for FGF2 engineering presented in this work provides novel insights into the design of growth factor variants for regenerative and anti-cancer precise medicine.
Collapse
Affiliation(s)
- Mateusz A Krzyscik
- University of Wroclaw, Faculty of Biotechnology, Department of Protein Engineering, 50-383 Wroclaw, Poland
| | - Łukasz Opaliński
- University of Wroclaw, Faculty of Biotechnology, Department of Protein Engineering, 50-383 Wroclaw, Poland
| | - Jakub Szymczyk
- University of Wroclaw, Faculty of Biotechnology, Department of Protein Engineering, 50-383 Wroclaw, Poland
| | - Jacek Otlewski
- University of Wroclaw, Faculty of Biotechnology, Department of Protein Engineering, 50-383 Wroclaw, Poland.
| |
Collapse
|
5
|
Anti-tumor and anti-metastatic activity of the FGF2 118-126 fragment dependent on the loop structure. Biochem J 2022; 479:1285-1302. [PMID: 35638868 DOI: 10.1042/bcj20210830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/20/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022]
Abstract
Fibroblast Growth Factor /FGF Receptor 1 (FGF2/FGFR1) system regulates the growth and metastasis of different cancers. Inhibition of this signaling pathway is an attractive target for cancer therapy. Here, we aimed to reproduce the 118-126 fragment of FGF2 to interfere with the FGF2-FGFR1 interaction. To determine whether the loop structure affects the function of this fragment, we compared cyclic (disulfide-bonded) and linear peptide variants. The cyclic peptide (referred to as BGF1) effectively inhibited the FGF2-induced proliferation of HUVECs, 4T1 mammary carcinoma, U87 glioblastoma, and SKOV3 ovarian carcinoma cells. It led to apoptosis induction in HUVECs, whereas the linear peptide (referred to as BGF2) was ineffective. In a murine 4T1 tumor model, BGF1 inhibited tumor growth more effectively than Avastin and increased animals' survival without causing weight loss, but the linear peptide BGF2 had no significant anti-tumor effects. According to immunohistochemical studies, the anti-tumor properties of BGF1 were associated with suppression of tumor cell proliferation (Ki-67 expression), angiogenesis (CD31 expression), and apoptosis induction (as was shown by increased p53 expression and TUNEL staining and decreased Bcl-2 expression). The potential of BGF1 to suppress tumor invasion was indicated by quantitative analysis of the metastasis-related proteins, including FGFR1, pFGFR1, NF-κB, p-NF-κB, MMP-9, E-cadherin, N-cadherin, and Vimentin, and supported by small animal positron emission tomography (PET) used 18Fluorodeoxyglucose (18F-FDG). These results demonstrate that the functional properties of the 118-126 region of FGF2 depend on the loop structure and the peptide derived from this fragment encourages further preclinical investigations.
Collapse
|
6
|
Porębska N, Knapik A, Poźniak M, Krzyścik MA, Zakrzewska M, Otlewski J, Opaliński Ł. Intrinsically Fluorescent Oligomeric Cytotoxic Conjugates Toxic for FGFR1-Overproducing Cancers. Biomacromolecules 2021; 22:5349-5362. [PMID: 34855396 PMCID: PMC8672352 DOI: 10.1021/acs.biomac.1c01280] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
Fibroblast growth
factor receptor 1 (FGFR1) is an integral membrane
protein that transmits prolife signals through the plasma membrane.
Overexpression of FGFR1 has been reported in various tumor types,
and therefore, this receptor constitutes an attractive molecular target
for selective anticancer therapies. Here, we present a novel system
for generation of intrinsically fluorescent, self-assembling, oligomeric
cytotoxic conjugates with high affinity and efficient internalization
targeting FGFR1. In our approach, we employed FGF1 as an FGFR1 recognizing
molecule and genetically fused it to green fluorescent protein polygons
(GFPp), a fluorescent oligomerization scaffold, resulting in a set
of GFPp_FGF1 oligomers with largely improved receptor binding. To
validate the applicability of using GFPp_FGF1 oligomers as cancer
probes and drug carriers in targeted therapy of cancers with aberrant
FGFR1, we selected a trimeric variant from generated GFPp_FGF1 oligomers
and further engineered it by introducing FGF1-stabilizing mutations
and by incorporating the cytotoxic drug monomethyl auristatin E (MMAE)
in a site-specific manner. The resulting intrinsically fluorescent,
trimeric cytotoxic conjugate 3xGFPp_FGF1E_LPET_MMAE exhibits nanomolar
affinity for the receptor and very high stability. Notably, the intrinsic
fluorescence of 3xGFPp_FGF1E_LPET_MMAE allows for tracking the cellular
transport of the conjugate, demonstrating that 3xGFPp_FGF1E_LPET_MMAE
is efficiently and selectively internalized into cells expressing
FGFR1. Importantly, we show that 3xGFPp_FGF1E_LPET_MMAE displays very
high cytotoxicity against a panel of different cancer cells overproducing
FGFR1 while remaining neutral toward cells devoid of FGFR1 expression.
Our data implicate that the engineered fluorescent conjugates can
be used for imaging and targeted therapy of FGFR1-overproducing cancers.
Collapse
Affiliation(s)
- Natalia Porębska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Agata Knapik
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Marta Poźniak
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Mateusz Adam Krzyścik
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Małgorzata Zakrzewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Jacek Otlewski
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| | - Łukasz Opaliński
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, Wroclaw 50-383, Poland
| |
Collapse
|
7
|
Overexpression of glycosyltransferase 8 domain containing 2 confers ovarian cancer to CDDP resistance by activating FGFR/PI3K signalling axis. Oncogenesis 2021; 10:55. [PMID: 34294681 PMCID: PMC8298492 DOI: 10.1038/s41389-021-00343-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/15/2021] [Accepted: 06/24/2021] [Indexed: 11/09/2022] Open
Abstract
It has been reported that chemotherapy resistance mainly contributed to treatment failure and poor survival in patients with ovarian cancer. Therefore, clarifying the molecular mechanism and identifying effective strategies to overcome drug resistance may play an important clinical impact on this malignant tumor. In our study, we found that the expression of Glycosyltransferase 8 domain containing 2 (GLT8D2) was significantly upregulated in ovarian cancer samples with CDDP (Cis-dichlorodiammine-platinum) resistance. Biological experiment demonstrate that GLT8D2 overexpression confers CDDP resistance on ovarian cancer cells; however, inhibition of GLT8D2 sensitized ovarian cancer cell lines to CDDP cytotoxicity both in vitro and in vivo. By using affinity purification/mass spectrometry (IP/MS) and reciprocal co-immunoprecipitation (co-IP) analyses, we found that GLT8D2 interacts with fibroblast growth factor receptor 1(FGFR1) in ovarian cancer cells. Furthermore, overexpression of GLT8D2 activated FGFR/PI3K signaling axis and upregulated the phosphorylation levels of FRS2a and AKT (AKT serine/threonine kinase). Importantly, pharmacological inhibition of FGFR and PI3K (phosphatidylinositol 3-kinase) signaling pathway significantly counteracted GLT8D2-induced chemoresistance and enhanced platinum's therapeutic efficacy in ovarian cancer. Therefore, our findings suggest that GLT8D2 is a potential therapeutic target for the treatment of ovarian cancer; targeting GLT8D2/FGFR/PI3K/AKT signaling axis may represent a promising strategy to enhance platinum response in patients with chemoresistant ovarian cancer.
Collapse
|
8
|
Comparative Gene Signature of (-)-Oleocanthal Formulation Treatments in Heterogeneous Triple Negative Breast Tumor Models: Oncological Therapeutic Target Insights. Nutrients 2021; 13:nu13051706. [PMID: 34069906 PMCID: PMC8157589 DOI: 10.3390/nu13051706] [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] [Received: 04/13/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 12/29/2022] Open
Abstract
Triple negative breast cancer (TNBC) heterogeneity and limited therapeutic options confer its phenotypic aggressiveness. The discovery of anti-TNBC natural products with valid molecular target(s) and defined pharmacodynamic profile would facilitate their therapeutic nutraceutical use by TNBC patients. The extra-virgin olive oil (EVOO) is a key Mediterranean diet ingredient. S-(−)-Oleocanthal (OC) leads the bioactive anti-tumor EVOO phenolic ingredients. A previous study reported the solid dispersion formulated OC with (+)-xylitol (OC-X) suppressed the in vivo progression and recurrence of the TNBC MDA-MB-231 cells. This study investigates the ability of OC-X formulation to suppress the in vivo heterogeneous BC initiation and progression utilizing advanced preclinical transgenic MMTV-PyVT and TNBC PDX mouse models. Furthermore, the clustering of the gene expression profiles in MMTV-PyVT and PDX mouse tumors treated with OC-X acquired by a Clariom S microarray analysis identified the distinctly affected genes. Several affected novel signature genes identified in response to OC-X treatments and proved overlapped in both mouse and human tumor models, shedding some lights toward understanding the OC anticancer molecular mechanism and assisting in predicting prospective clinical outcomes. This study provides molecular and preclinical evidences of OC-X potential as a nutraceutical suppressing heterogeneous TNBC model and offers preliminary gene-level therapeutic mechanistic insights.
Collapse
|
9
|
Poźniak M, Porębska N, Krzyścik MA, Sokołowska-Wędzina A, Jastrzębski K, Sochacka M, Szymczyk J, Zakrzewska M, Otlewski J, Opaliński Ł. The cytotoxic conjugate of highly internalizing tetravalent antibody for targeting FGFR1-overproducing cancer cells. Mol Med 2021; 27:46. [PMID: 33962559 PMCID: PMC8103757 DOI: 10.1186/s10020-021-00306-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Antibody drug conjugates (ADCs) represent one of the most promising approaches in the current immuno-oncology research. The precise delivery of cytotoxic drugs to the cancer cells using ADCs specific for tumor-associated antigens enables sparing the healthy cells and thereby reduces unwanted side effects. Overexpression of fibroblast growth factor receptor 1 (FGFR1) has been demonstrated in numerous tumors and thereby constitutes a convenient molecular target for selective cancer treatment. We have recently engineered tetravalent anti-FGFR1 antibody, T-Fc, and have demonstrated that it displays extremely efficient internalization into FGFR1 producing cells, a feature highly desirable in the ADC approach. We have revealed that T-Fc mediates clustering of FGFR1, largely enhancing the uptake of FGFR1-T-Fc complexes by induction of clathrin-independent endocytic routes. The aim of this study was to obtain highly internalizing cytotoxic conjugate of the T-Fc for specific delivery of drugs into FGFR1-positive cancer cells. METHODS Conjugation of the T-Fc to a cytotoxic payload, vcMMAE, was carried out via maleimide chemistry, yielding the T-Fc-vcMMAE. The specific binding of the T-Fc-vcMMAE conjugate to FGFR1 was confirmed in vitro with BLI technique. Confocal microscopy and flow cytometry were applied to determine FGFR1-dependence of the T-Fc-vcMMAE internalization. Western blot analyses of FGFR1-dependent signaling were conducted to assess the impact of the T-Fc-vcMMAE on FGFR1 activation and initiation of downstream signaling cascades. Finally, using FGFR1-negative and FGFR1-possitive cell lines, the cytotoxic potential of the T-Fc-vcMMAE was evaluated. RESULTS We have performed the efficient conjugation of the tetravalent engineered antibody with a cytotoxic drug and generated FGFR1-specific ADC molecule, T-Fc-vcMMAE. We have demonstrated that T-Fc-vcMMAE conjugate exhibits high selectivity and affinity for FGFR1, similarly to T-Fc. Furthermore, we have shown that T-Fc constitutes an effective drug delivery vehicle as T-Fc-vcMMAE was efficiently and selectively internalized by FGFR1-producing cells leading to their death. Interestingly, we show that the efficiency of the uptake of T-Fc-vcMMAE corresponds well with the cytotoxicity of the conjugate, but doesn't correlate with the FGFR1expression level. CONCLUSION Our results show that T-Fc-vcMMAE fulfills the key criteria for the successful cytotoxic drug carrier in a targeted approach against FGFR1-positive cancer cells. Furthermore, our data implicate that not solely expression level of the receptor, but rather its cellular trafficking should be taken into account for selection of suitable molecular targets and cancer models for successful ADC approach.
Collapse
MESH Headings
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Antineoplastic Agents, Immunological/chemistry
- Antineoplastic Agents, Immunological/pharmacology
- Cell Line, Tumor
- Cell Survival/drug effects
- Fluorescent Antibody Technique
- Gene Expression
- Genetic Engineering
- Humans
- Immunoconjugates/chemistry
- Immunoconjugates/pharmacology
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
Collapse
Affiliation(s)
- Marta Poźniak
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Natalia Porębska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Mateusz Adam Krzyścik
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Aleksandra Sokołowska-Wędzina
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Kamil Jastrzębski
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Martyna Sochacka
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Jakub Szymczyk
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Małgorzata Zakrzewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Jacek Otlewski
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Łukasz Opaliński
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland.
| |
Collapse
|
10
|
Zhang S, Huang J, Zhang L, Gu J, Song Q, Cai Y, Zhong J, Zhong H, Deng Y, Zhu W, Zhao J, Deng N. Fermentation, Purification, and Tumor Inhibition of a Disulfide-Stabilized Diabody Against Fibroblast Growth Factor-2. Front Oncol 2021; 11:585457. [PMID: 33718141 PMCID: PMC7947002 DOI: 10.3389/fonc.2021.585457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/04/2021] [Indexed: 11/16/2022] Open
Abstract
Angiogenesis is considered one of the hallmarks of cancer and plays a critical role in the development of tumor. Fibroblast growth factor 2 (FGF-2) is a member of the FGF family and participates in excessive cancer cell proliferation and tumor angiogenesis. Thus, targeting FGF-2 was considered to be a promising anti-tumor strategy. A disulfide-stabilized diabody (ds-Diabody) against FGF-2 was produced in Pichia pastoris (GS115) by fermentation and the anti-tumor activity was analyzed. The novel 10-L fed batch fermentation with newly designed media was established, and the maximum production of the ds-Diabody against FGF-2 reached 210.4 mg/L. The ds-Diabody against FGF-2 was purified by Ni2+ affinity chromatography and DEAE anion exchange chromatography. The recombinant ds-Diabody against FGF-2 could effectively inhibit proliferation, migration, and invasion of melanoma and glioma tumor cells stimulated by FGF-2. Furthermore, xenograft tumor model assays showed that the ds-Diabody against FGF-2 had potent antitumor activity in nude mice by inhibiting tumor growth and angiogenesis. The tumor growth inhibition rate of melanoma and glioma was about 70 and 45%, respectively. The tumor angiogenesis inhibition rate of melanoma and glioma was about 64 and 51%, respectively. The results revealed that the recombinant ds-Diabody against FGF-2 may be a promising anti-tumor drug for cancer therapy.
Collapse
Affiliation(s)
- Simin Zhang
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou, China
| | - Jiahui Huang
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou, China
| | - Ligang Zhang
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou, China
| | - Jiangtao Gu
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou, China
| | - Qifang Song
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou, China
| | - Yaxiong Cai
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou, China
| | - Jiangchuan Zhong
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou, China
| | - Hui Zhong
- Biomedicine Translational Institute, Jinan University, Guangzhou, China
| | - Yanrui Deng
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou, China
| | - Wenhui Zhu
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou, China
| | - Jianfu Zhao
- Cancer Diagnosis and Therapy Research Center, Department of Oncology of the First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ning Deng
- Guangdong Province Engineering Research Center for Antibody Drug and Immunoassay, Department of Biology, Jinan University, Guangzhou, China
| |
Collapse
|
11
|
Xiao H, Wang K, Li D, Wang K, Yu M. Evaluation of FGFR1 as a diagnostic biomarker for ovarian cancer using TCGA and GEO datasets. PeerJ 2021; 9:e10817. [PMID: 33604191 PMCID: PMC7866899 DOI: 10.7717/peerj.10817] [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] [Received: 05/14/2020] [Accepted: 12/30/2020] [Indexed: 12/30/2022] Open
Abstract
Background Malignant ovarian cancer is associated with the highest mortality of all gynecological tumors. Designing therapeutic targets that are specific to OC tissue is important for optimizing OC therapies. This study aims to identify different expression patterns of genes related to FGFR1 and the usefulness of FGFR1 as diagnostic biomarker for OC. Methods We collected data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases. In the TCGA cohort we analyzed clinical information according to patient characteristics, including age, stage, grade, longest dimension of the tumor and the presence of a residual tumor. GEO data served as a validation set. We obtained data on differentially expressed genes (DEGs) from the two microarray datasets. We then used gene set enrichment analysis (GSEA) to analyze the DEG data in order to identify enriched pathways related to FGFR1. Results Differential expression analysis revealed that FGFR1 was significantly downregulated in OC specimens. 303 patients were included in the TCGA cohort. The GEO dataset confirmed these findings using information on 75 Asian patients. The GSE105437 and GSE12470 database highlighted the significant diagnostic value of FGFR1 in identifying OC (AUC = 1, p = 0.0009 and AUC = 0.8256, p = 0.0015 respectively). Conclusions Our study examined existing TCGA and GEO datasets for novel factors associated with OC and identified FGFR1 as a potential diagnostic factor. Further investigation is warranted to characterize the role played by FGFR1 in OC.
Collapse
Affiliation(s)
- Huiting Xiao
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Kun Wang
- Department of Urologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Dan Li
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Ke Wang
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Min Yu
- Department of Gynecologic Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
12
|
Sobhani N, Fassl A, Mondani G, Generali D, Otto T. Targeting Aberrant FGFR Signaling to Overcome CDK4/6 Inhibitor Resistance in Breast Cancer. Cells 2021; 10:293. [PMID: 33535617 PMCID: PMC7912842 DOI: 10.3390/cells10020293] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 01/01/2023] Open
Abstract
Breast cancer (BC) is the most common cause of cancer-related death in women worldwide. Therapies targeting molecular pathways altered in BC had significantly enhanced treatment options for BC over the last decades, which ultimately improved the lives of millions of women worldwide. Among various molecular pathways accruing substantial interest for the development of targeted therapies are cyclin-dependent kinases (CDKs)-in particular, the two closely related members CDK4 and CDK6. CDK4/6 inhibitors indirectly trigger the dephosphorylation of retinoblastoma tumor suppressor protein by blocking CDK4/6, thereby blocking the cell cycle transition from the G1 to S phase. Although the CDK4/6 inhibitors abemaciclib, palbociclib, and ribociclib gained FDA approval for the treatment of hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative BC as they significantly improved progression-free survival (PFS) in randomized clinical trials, regrettably, some patients showed resistance to these therapies. Though multiple molecular pathways could be mechanistically responsible for CDK4/6 inhibitor therapy resistance, one of the most predominant ones seems to be the fibroblast growth factor receptor (FGFR) pathway. FGFRs are involved in many aspects of cancer formation, such as cell proliferation, differentiation, and growth. Importantly, FGFRs are frequently mutated in BC, and their overexpression and/or hyperactivation correlates with CDK4/6 inhibitor resistance and shortened PFS in BC. Intriguingly, the inhibition of aberrant FGFR activity is capable of reversing the resistance to CDK4/6 inhibitors. This review summarizes the molecular background of FGFR signaling and discusses the role of aberrant FGFR signaling during cancer development in general and during the development of CDK4/6 inhibitor resistance in BC in particular, together with other possible mechanisms for resistance to CDK4/6 inhibitors. Subsequently, future directions on novel therapeutic strategies targeting FGFR signaling to overcome such resistance during BC treatment will be further debated.
Collapse
Affiliation(s)
- Navid Sobhani
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anne Fassl
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Giuseppina Mondani
- Department Breast Oncoplastic Surgery Royal Cornwall Hospital, Treliske, Truro TR13LJ, UK;
| | - Daniele Generali
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, 34149 Trieste, Italy;
| | - Tobias Otto
- Department of Internal Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| |
Collapse
|
13
|
Kähkönen TE, Toriseva M, Petruk N, Virta AR, Maher A, Eigéliené N, Kaivola J, Boström P, Koskivuo I, Nees M, Tuomela JM, Ivaska KK, Härkönen PL. Effects of FGFR inhibitors TKI258, BGJ398 and AZD4547 on breast cancer cells in 2D, 3D and tissue explant cultures. Cell Oncol (Dordr) 2020; 44:205-218. [PMID: 33119860 PMCID: PMC7907049 DOI: 10.1007/s13402-020-00562-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2020] [Indexed: 11/28/2022] Open
Abstract
Purpose Fibroblast growth factor receptors (FGFR) and pathways are important players in breast cancer (BC) development. They are commonly altered, and BCs exhibiting FGFR gene amplification are currently being studied for drug development. Here, we aimed to compare the effects of three FGFR inhibitors (FGFRis), i.e., non-selective TKI258 and selective BGJ398 and AZD4547, on different BC-derived cell lines (BCCs) and primary tissues. Methods The human BCCs MCF-7 and MDA-MB-231(SA) (wild-type FGFR) and MFM223 (amplified FGFR1 and FGFR2) were analyzed for FGFR expression using qRT-PCR, and the effects of FGFRis on FGFR signaling by Western blotting. The effects of FGFRis on proliferation, viability, migration and invasion of BCCs were assessed in 2D cultures using live-cell imaging, and in 3D cultures using phenotypic analysis of organoids. To study radio-sensitization, FGFRi treatment was combined with irradiation. Patient-derived BC samples were treated with FGFRis in explant cultures and immunostained for Ki67 and cleaved caspase 3. Results We found that all FGFRis tested decreased the growth and viability of BC cells in 2D and 3D cultures. BGJ398 and AZD4547 were found to be potent at low concentrations in FGFR-amplified MFM233 cells, whereas higher concentrations were required in non-amplified MCF7 and MDA-MB-231(SA) cells. TKI258 inhibited the migration and invasion, whereas BGJ398 and AZD4547 only inhibited the invasion of MDA-MB-231(SA) cells. FGFRi treatment of MCF7 and MFM223 cells enhanced the inhibitory effect of radiotherapy, but this effect was not observed in MDA-MB-231(SA) cells. FGFRi-treated primary BC explants with moderate FGFR levels showed a tendency towards decreased proliferation and increased apoptosis. Conclusions Our results indicate that, besides targeting FGFR-amplified BCs with selective FGFRis, also BCs without FGFR amplification/activation may benefit from FGFRi-treatment. Combination with other treatment modalities, such as radiotherapy, may allow the use of FGFRis at relatively low concentrations and, thereby, contribute to better BC treatment outcomes. Supplementary Information The online version of this article (10.1007/s13402-020-00562-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- T E Kähkönen
- University of Turku, Institute of Biomedicine, 20520, Turku, Finland
| | - M Toriseva
- University of Turku, Institute of Biomedicine, 20520, Turku, Finland.,FICAN West Cancer Centre, 20520, Turku, Finland
| | - N Petruk
- University of Turku, Institute of Biomedicine, 20520, Turku, Finland.,FICAN West Cancer Centre, 20520, Turku, Finland
| | - A-R Virta
- University of Turku, Institute of Biomedicine, 20520, Turku, Finland
| | - A Maher
- University of Turku, Institute of Biomedicine, 20520, Turku, Finland
| | - N Eigéliené
- University of Turku, Institute of Biomedicine, 20520, Turku, Finland
| | - J Kaivola
- University of Turku, Institute of Biomedicine, 20520, Turku, Finland
| | - P Boström
- Department of Pathology, Turku University Hospital, 20520, Turku, Finland
| | - I Koskivuo
- Department of Plastic and General Surgery, Turku University Hospital, 20520, Turku, Finland
| | - M Nees
- University of Turku, Institute of Biomedicine, 20520, Turku, Finland.,Department of Biomedicine and Molecular Biology II, Uniwersytet Medyczny w Lublinie, 20-095, Lublin, Poland
| | - J M Tuomela
- University of Turku, Institute of Biomedicine, 20520, Turku, Finland.,FICAN West Cancer Centre, 20520, Turku, Finland
| | - K K Ivaska
- University of Turku, Institute of Biomedicine, 20520, Turku, Finland
| | - P L Härkönen
- University of Turku, Institute of Biomedicine, 20520, Turku, Finland. .,FICAN West Cancer Centre, 20520, Turku, Finland.
| |
Collapse
|
14
|
Jendryczko K, Chudzian J, Skinder N, Opaliński Ł, Rzeszótko J, Wiedlocha A, Otlewski J, Szlachcic A. FGF2-Derived PeptibodyF2-MMAE Conjugate for Targeted Delivery of Cytotoxic Drugs into Cancer Cells Overexpressing FGFR1. Cancers (Basel) 2020; 12:E2992. [PMID: 33076489 PMCID: PMC7602595 DOI: 10.3390/cancers12102992] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 10/10/2020] [Indexed: 12/23/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) are emerging targets for directed cancer therapy. Presented here is a new FGFR1-targeting conjugate, the peptibodyF2, which employs peptibody, a fusion of peptide and the Fc fragment of human IgG as a selective targeting agent and drug carrier. Short peptide based on FGF2 sequence was used to construct a FGFR1-targeting peptibody. We have shown that this peptide ensures specific delivery of peptibodyF2 into FGFR1-expressing cells. In order to use peptibodyF2 as a delivery vehicle for cytotoxic drugs, we have conjugated it with MMAE, a drug widely used in antibody-drug conjugates for targeted therapy. Resulting conjugate shows high and specific cytotoxicity towards FGFR1-positive cells, i.e., squamous cell lung carcinoma NCI-H520, while remaining non-toxic for FGFR1-negative cells. Such peptibody-drug conjugate can serve as a basis for development of therapy for tumors with overexpressed or malfunctioning FGFRs.
Collapse
Affiliation(s)
- Karolina Jendryczko
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Julia Chudzian
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Natalia Skinder
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Łukasz Opaliński
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Jakub Rzeszótko
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Antoni Wiedlocha
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway;
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 01163 Warsaw, Poland
- Center for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
| | - Jacek Otlewski
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Anna Szlachcic
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| |
Collapse
|
15
|
Sobhani N, Fan C, O. Flores-Villanueva P, Generali D, Li Y. The Fibroblast Growth Factor Receptors in Breast Cancer: from Oncogenesis to Better Treatments. Int J Mol Sci 2020; 21:E2011. [PMID: 32188012 PMCID: PMC7139621 DOI: 10.3390/ijms21062011] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/11/2020] [Accepted: 03/14/2020] [Indexed: 01/09/2023] Open
Abstract
Breast cancer (BC) is the most frequent form of malignancy and second only to lung cancer as cause of deaths in women. Notwithstanding many progresses made in the field, metastatic BC has a very poor prognosis. As therapies are becoming more personalized to meet the needs of patients, a better knowledge of the molecular biology leading to the disease unfolds the possibility to project more precise compounds or antibodies targeting definite alteration at the molecular level and functioning on such cancer-causing molecules expressed in cancer cells of patients, or present as antigens on the surface of cancer cell membranes. Fibroblast growth factor receptor (FGFR) is one of such druggable targets, activated by its own ligands -namely the Fibroblast Growth Factors (FGFs). This pathway provides a vast range of interesting molecular targets pursued at different levels of clinical investigation. Herein we provide an update on the knowledge of genetic alterations of the receptors in breast cancer, their role in tumorigenesis and the most recent drugs against this particular receptor for the treatment of the disease.
Collapse
Affiliation(s)
- Navid Sobhani
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA; (C.F.); (P.O.F.-V.)
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada Di Fiume 447, 34149 Trieste, Italy;
| | - Chunmei Fan
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA; (C.F.); (P.O.F.-V.)
| | - Pedro O. Flores-Villanueva
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA; (C.F.); (P.O.F.-V.)
| | - Daniele Generali
- Department of Medical, Surgical and Health Sciences, University of Trieste, Cattinara Hospital, Strada Di Fiume 447, 34149 Trieste, Italy;
| | - Yong Li
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA; (C.F.); (P.O.F.-V.)
| |
Collapse
|
16
|
Karimi-Zarchi M, Moghimi M, Abbasi H, Hadadan A, Salimi E, Morovati-Sharifabad M, Akbarian-Bafghi MJ, Zare-Shehneh M, Mosavi-Jarrahi A, Neamatzadeh H. Association of MTHFR 677C>T Polymorphism with Susceptibility to Ovarian and Cervical Cancers: A Systematic Review and Meta-Analysis. Asian Pac J Cancer Prev 2019; 20:2569-2577. [PMID: 31554347 PMCID: PMC6976840 DOI: 10.31557/apjcp.2019.20.9.2569] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Indexed: 12/14/2022] Open
Abstract
Background: Previous studies have evaluated the impact of MTHFR 677C>T polymorphism on susceptibility to ovarian and cervical cancers in women, but the conclusions are still controversial. To get a more precise evaluation of the association between MTHFR 677C>T polymorphism and risk of ovarian and cervical cancers, we performed a meta-analysis of the association of all eligible studies. Methods: A comprehensive search performed in PubMed, Google Scholar, CNKI, and Web of Science databases to identify the relevant studies up to October 15, 2018. The strength of the association was estimated by odds ratios (OR) with 95% confidence interval (CI). Results: A total of 27 case-control studies including eleven studies with 4990 cases 7730 controls on ovarian cancer and 16 studies with 4990 cases and 7730 controls on cervical cancer were selected. Pooled data revealed that the MTHFR 677C>T polymorphism not significantly associated with an increased risk of ovarian and cervical cancers under all five genetic models. However, stratified analysis by ethnicity showed that the MTHFR 677C>T polymorphism was significantly associated with risk of ovarian cancer in Asians. No publication bias was found in the current meta-analysis. Conclusions: The results of this meta-analysis proposes that the MTHFR 677C>T polymorphism may not play a role in development of ovarian and cervical cancers in overall population. Further well-designed studies are necessary to clarify the precise role of the MTHFR 677C>T polymorphism on ovarian and cervical cancers risk.
Collapse
Affiliation(s)
- Mojgan Karimi-Zarchi
- Department of Gynecology and Obstetrics, Iran University of Medical Sciences, Tehran, Iran
| | - Mansour Moghimi
- Department of Pathology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Hajar Abbasi
- Department of Gynecology and Obstetrics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amaneh Hadadan
- Department of Gynecology and Obstetrics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Erfaneh Salimi
- Department of Gynecology and Obstetrics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Masoud Zare-Shehneh
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alireza Mosavi-Jarrahi
- Department of Social Medicine, Medical School, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Neamatzadeh
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
17
|
Mori M, Mori T, Yamamoto A, Takagi S, Ueda M. Proliferation of poorly differentiated endometrial cancer cells through autocrine activation of FGF receptor and HES1 expression. Hum Cell 2019; 32:367-378. [PMID: 30963412 DOI: 10.1007/s13577-019-00249-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 03/15/2019] [Indexed: 12/12/2022]
Abstract
Patients with poorly differentiated endometrial cancer show poor prognosis, and effective molecular target-based therapies are needed. Endometrial cancer cells proliferate depending on the activation of HES1 (hairy and enhancer of split-1), which is induced by several pathways, such as the Notch and fibroblast growth factor receptor (FGFR) signaling pathways. In addition, aberrant, ligand-free activation of the FGFR signaling pathway resulting from mutations in FGFR2 was also reported in endometrial cancer. However, a clinical trial showed that there was no difference in the effectiveness of FGFR inhibitors between patients with and without the FGFR2 mutation, suggesting a presence of another signaling pathway for the FGFR activation. Here, we investigated the signaling pathway regulating the expression of HES1 and proliferation of poorly and well-differentiated endometrial cancer cell lines Ishikawa and HEC-50B, respectively. Whereas Ishikawa cells proliferated and expressed HES1 in a Notch signaling-dependent manner, Notch signaling was not involved in HES1 and proliferation of HEC-50B cells. The FGFR inhibitor, NVP-BGJ398, decreased HES1 expression and proliferation of HEC-50B cells; however, HEC50B cells had no mutations in the FGFR2 gene. Instead, HEC-50B cells highly expressed ligands for FGFR2, suggesting that FGFR2 is activated by an autocrine manner, not by ligand-free activation. This autocrine pathway activated Akt downstream of FGFR for cell proliferation. Our findings suggest the usefulness of HES1 as a marker for the proliferation signaling and that FGFR inhibitor may be effective for poorly differentiated endometrial cancers that harbor wild-type FGFR.
Collapse
Affiliation(s)
- Michihiro Mori
- Department of Medical Life Science, College of Life Science, Kurashiki University of Science and the Arts, 2640 Nishinoura Tsurajima-cho Kurashiki-shi, Okayama, 712-8505, Japan. .,Kake Institute of Cytopathology, Okayama, Japan.
| | - Toshinori Mori
- Department of Clinical Laboratory, Mihara Medical Associations Hospital, Hiroshima, Japan.,Department of Chemical Technology, Graduate School of Science and Industrial Technology, Kurashiki University of Science and the Arts, Okayama, Japan
| | - Aina Yamamoto
- Department of Chemical Technology, Graduate School of Science and Industrial Technology, Kurashiki University of Science and the Arts, Okayama, Japan
| | - Shoji Takagi
- Department of Medical Life Science, College of Life Science, Kurashiki University of Science and the Arts, 2640 Nishinoura Tsurajima-cho Kurashiki-shi, Okayama, 712-8505, Japan.,Kake Institute of Cytopathology, Okayama, Japan
| | - Masatsugu Ueda
- Faculty of Health Sciences, Kio University, Nara, Japan.,Graduate School of Health Sciences, Kio University, Nara, Japan
| |
Collapse
|
18
|
Lipok M, Szlachcic A, Kindela K, Czyrek A, Otlewski J. Identification of a peptide antagonist of the FGF1-FGFR1 signaling axis by phage display selection. FEBS Open Bio 2019; 9:914-924. [PMID: 30968602 PMCID: PMC6487701 DOI: 10.1002/2211-5463.12618] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/14/2019] [Accepted: 02/17/2019] [Indexed: 12/11/2022] Open
Abstract
Overexpression of fibroblast growth factor receptor 1 (FGFR1) is a common aberration in lung and breast cancers and has necessitated the design of drugs targeting FGFR1‐dependent downstream signaling and FGFR1 ligand binding. To date, the major group of drugs being developed for treatment of FGFR1‐dependent cancers are small‐molecule tyrosine kinase inhibitors; however, the limited specificity of these drugs has led to increasing attempts to design molecules targeting the extracellular domain of FGFR1. Here, we used the phage display technique to select cyclic peptides F8 (ACSLNHTVNC) and G10 (ACSAKTTSAC) as binders of the fibroblast growth factor 1 (FGF1)–FGFR1 interface. ELISA and in vitro cell assays were performed to reveal that cyclic peptide F8 is more effective in preventing the FGF1–FGFR1 interaction, and also decreases FGF1‐induced proliferation of BA/F3 FGFR1c cells by over 40%. Such an effect was not observed for BA/F3 cells lacking FGFR1. Therefore, cyclic peptide F8 can act as a FGF1–FGFR1 interaction antagonist, and may be suitable for further development for potential use in therapies against FGFR1‐expressing cancer cells.
Collapse
Affiliation(s)
- Magdalena Lipok
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Poland.,PORT - Polish Center for Technology Development, Wroclaw, Poland
| | - Anna Szlachcic
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Poland.,PORT - Polish Center for Technology Development, Wroclaw, Poland
| | - Kinga Kindela
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Poland
| | - Aleksandra Czyrek
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Poland
| | - Jacek Otlewski
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, Poland.,PORT - Polish Center for Technology Development, Wroclaw, Poland
| |
Collapse
|
19
|
Kähkönen TE, Tuomela JM, Grönroos TJ, Halleen JM, Ivaska KK, Härkönen PL. Dovitinib dilactic acid reduces tumor growth and tumor-induced bone changes in an experimental breast cancer bone growth model. J Bone Oncol 2019; 16:100232. [PMID: 30956945 PMCID: PMC6434100 DOI: 10.1016/j.jbo.2019.100232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/17/2019] [Accepted: 03/18/2019] [Indexed: 02/06/2023] Open
Abstract
Advanced breast cancer has a high incidence of bone metastases. In bone, breast cancer cells induce osteolytic or mixed bone lesions by inducing an imbalance in bone formation and resorption. Activated fibroblast growth factor receptors (FGFRs) are important in regulation of tumor growth and bone remodeling. In this study we used FGFR1 and FGFR2 gene amplifications containing human MFM223 breast cancer cells in an experimental xenograft model of breast cancer bone growth using intratibial inoculation technique. This model mimics bone metastases in breast cancer patients. The effects of an FGFR inhibitor, dovitinib dilactic acid (TKI258) on tumor growth and tumor-induced bone changes were evaluated. Cancer-induced bone lesions were smaller in dovitinib-treated mice as evaluated by X-ray imaging. Peripheral quantitative computed tomography imaging showed higher total and cortical bone mineral content and cortical bone mineral density in dovitinib-treated mice, suggesting better preserved bone mass. CatWalk gait analysis indicated that dovitinib-treated mice experienced less cancer-induced bone pain in the tumor-bearing leg. A trend towards decreased tumor growth and metabolic activity was observed in dovitinib-treated mice quantified by positron emission tomography imaging with 2-[18F]fluoro-2-deoxy-D-glucose at the endpoint. We conclude that dovitinib treatment decreased tumor burden, cancer-induced changes in bone, and bone pain. The results suggest that targeting FGFRs could be beneficial in breast cancer patients with bone metastases.
Collapse
Affiliation(s)
- Tiina E Kähkönen
- University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland.,Pharmatest Services, Itäinen Pitkäkatu 4C, 5th floor, 20520 Turku, Finland
| | | | - Tove J Grönroos
- Turku PET Centre, University of Turku, Tykistökatu 6A, 20520 Turku, Finland.,Medicity Research Laboratory, University of Turku, Turku, Finland.,Department of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland
| | - Jussi M Halleen
- Pharmatest Services, Itäinen Pitkäkatu 4C, 5th floor, 20520 Turku, Finland
| | - Kaisa K Ivaska
- University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland
| | | |
Collapse
|
20
|
Zhao M, Zhuo ML, Zheng X, Su X, Meric-Bernstam F. FGFR1β is a driver isoform of FGFR1 alternative splicing in breast cancer cells. Oncotarget 2019; 10:30-44. [PMID: 30713601 PMCID: PMC6343755 DOI: 10.18632/oncotarget.26530] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/16/2018] [Indexed: 01/11/2023] Open
Abstract
Abnormal FGFR1 alternative splicing is correlated with tumorigenicity and poor prognosis in several tumor types. We sought to determine the roles of FGFR1α and FGFR1β variants in breast cancer. TCGA samples and cell lines were analyzed for FGFR1α/FGFR1β expression. MCF-10A cells were used to overexpress these variants. Cell growth and transformation were assessed by SRB, colony formation, 3D-Matrigel, soft agar, cell motility assays. In TCGA, compared to FGFR1 non-amplified samples, FGFR1-amplified samples had significantly higher FGFR1α but not FGFR1β levels. FGFR1β expression levels and FGFR1β/FGFR1α ratio were higher in basal subtype samples than in ER-positive/luminal samples in both TCGA and breast cancer cell lines. Both FGFR1α and FGFR1β induced transformation of MCF-10A cells. However, only FGFR1β-expressing cells, not FGFR1α, enhanced cell growth and cell motility. Cells with higher FGFR1β levels and FGFR1β/FGFR1α ratio were more sensitive to FGFR inhibitor BGJ-398. Interestingly, in ER-negative cells, FGFR inhibitors decreased FGFR1β levels, likely by increasing expression of splicing repressor PTBP1. In ER-positive cells, estrogen treatment increased FGFR1β levels by decreasing PTBP1 expression, which was blocked by 4-OHT. Lastly, combination treatment with BGJ-398 and 4-OHT synergistically inhibited cell survival. These findings suggest that FGFR1 alternative FGFR1α/FGFR1β splicing plays an important role in breast cancer.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ming-Lei Zhuo
- Key Laboratory of Carcinogenesis and Translational Research, Department of Thoracic Medical Oncology-I, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaofeng Zheng
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Institute of Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
21
|
Sadej R, Lu X, Turczyk L, Novitskaya V, Lopez-Clavijo AF, Kordek R, Potemski P, Wakelam MJO, Romanska-Knight H, Berditchevski F. CD151 regulates expression of FGFR2 in breast cancer cells via PKC-dependent pathways. J Cell Sci 2018; 131:jcs220640. [PMID: 30257985 DOI: 10.1242/jcs.220640] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/17/2018] [Indexed: 11/20/2022] Open
Abstract
Expression of the tetraspanin CD151 is frequently upregulated in epithelial malignancies and correlates with poor prognosis. Here, we report that CD151 is involved in regulation of the expression of fibroblast growth factor receptor 2 (FGFR2). Depletion of CD151 in breast cancer cells resulted in an increased level of FGFR2. Accordingly, an inverse correlation between CD151 and FGFR2 was observed in breast cancer tissues. CD151-dependent regulation of the FGFR2 expression relies on post-transcriptional mechanisms involving HuR (also known as ELAVL1), a multifunctional RNA-binding protein, and the assembly of processing bodies (P-bodies). Depletion of CD151 correlated with inhibition of PKC, a well-established downstream target of CD151. Accordingly, the levels of dialcylglycerol species were decreased in CD151-negative cells, and inhibition of PKC resulted in the increased expression of FGFR2. Whereas expression of FGFR2 itself did not correlate with any of the clinicopathological data, we found that FGFR2-/CD151+ patients were more likely to have developed lymph node metastasis. Conversely, FGFR2-/CD151- patients demonstrated better overall survival. These results illustrate functional interdependency between CD151 complexes and FGFR2, and suggest a previously unsuspected role of CD151 in breast tumorigenesis.
Collapse
Affiliation(s)
- Rafal Sadej
- Department of Molecular Enzymology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdańsk, Poland
| | - Xiaohong Lu
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Lukasz Turczyk
- Department of Molecular Enzymology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Abrahama 58, 80-307 Gdańsk, Poland
| | - Vera Novitskaya
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | | | - Radzisław Kordek
- Department of Pathology and Chemotherapy, Medical University of Łódź, 92-213 Łódź, Poland
| | - Piotr Potemski
- Department of Pathology and Chemotherapy, Medical University of Łódź, 92-213 Łódź, Poland
| | | | - Hanna Romanska-Knight
- Department of Pathology and Chemotherapy, Medical University of Łódź, 92-213 Łódź, Poland
| | - Fedor Berditchevski
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
22
|
Sobhani N, Ianza A, D'Angelo A, Roviello G, Giudici F, Bortul M, Zanconati F, Bottin C, Generali D. Current Status of Fibroblast Growth Factor Receptor-Targeted Therapies in Breast Cancer. Cells 2018; 7:76. [PMID: 30011957 PMCID: PMC6071019 DOI: 10.3390/cells7070076] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 06/30/2018] [Accepted: 07/11/2018] [Indexed: 01/08/2023] Open
Abstract
Breast cancer (BC) is the most common malignancy and second only to lung cancer in terms of mortality in women. Despite the incredible progress made in this field, metastatic breast cancer has a poor prognosis. In an era of personalized medicine, there is an urgent need for better knowledge of the biology leading to the disease, which can lead to the design of increasingly accurate drugs against patients' specific molecular aberrations. Among one of the actionable targets is the fibroblast growth factor receptor (FGFR) pathway, triggered by specific ligands. The Fibroblast Growth Factor Receptors/Fibroblast Growth Factors (FGFRs/FGFs) axis offers interesting molecular targets to be pursued in clinical development. This mini-review will focus on the current knowledge of FGFR mutations, which lead to tumor formation and summarizes the state-of-the-art therapeutic strategies for targeted treatments against the FGFRs/FGFs axis in the context of BC.
Collapse
Affiliation(s)
- Navid Sobhani
- Department of Medical, Surgical & Health Sciences, University of Trieste, Cattinara Teaching Hospital, 34149 Trieste, Italy.
- Department of Medical, Surgery & Health Sciences, University of Trieste, 34129 Trieste, Italy.
| | - Anna Ianza
- Department of Medical, Surgery & Health Sciences, University of Trieste, 34129 Trieste, Italy.
| | - Alberto D'Angelo
- Department of Medical, Surgical & Health Sciences, University of Trieste, Cattinara Teaching Hospital, 34149 Trieste, Italy.
| | - Giandomenico Roviello
- Division of Medical Oncology, Department of Onco-Hematology, IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture (PZ), 85028 Rionero, Italy.
| | - Fabiola Giudici
- Department of Medical, Surgical & Health Sciences, University of Trieste, Cattinara Teaching Hospital, 34149 Trieste, Italy.
| | - Marina Bortul
- Department of Medical, Surgical & Health Sciences, University of Trieste, Cattinara Teaching Hospital, 34149 Trieste, Italy.
| | - Fabrizio Zanconati
- Department of Medical, Surgical & Health Sciences, University of Trieste, Cattinara Teaching Hospital, 34149 Trieste, Italy.
| | - Cristina Bottin
- Department of Medical, Surgical & Health Sciences, University of Trieste, Cattinara Teaching Hospital, 34149 Trieste, Italy.
| | - Daniele Generali
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34129 Trieste, Italy; Breast Cancer Unit and Translational Research Unit, ASST Cremona, Viale Concordia 1, C.A.P. 26100 Cremona, Italy.
| |
Collapse
|
23
|
Qi L, Song W, Li L, Cao L, Yu Y, Song C, Wang Y, Zhang F, Li Y, Zhang B, Cao W. FGF4 induces epithelial-mesenchymal transition by inducing store-operated calcium entry in lung adenocarcinoma. Oncotarget 2018; 7:74015-74030. [PMID: 27677589 PMCID: PMC5342032 DOI: 10.18632/oncotarget.12187] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 08/23/2016] [Indexed: 12/21/2022] Open
Abstract
Several fibroblast growth factor (FGF) isoforms act to stimulate epithelial-mesenchymal transition (EMT) during cancer progression. FGF4 and FGF7 are two ligands of FGF receptor 2 (FGFR2). Using two lung adenocarcinoma (ADC) cell lines, A549 and H1299, we showed that FGF4, but not FGF7, altered cell morphology, promoted EMT-associated protein expression, and enhanced cell proliferation, migration/invasion and colony initiation. In addition, FGF4 increased store-operated calcium entry (SOCE) and expression of the calcium signal-associated protein Orai1. The SOCE inhibitor 2,5-di-tert-butylhydroquinone (BHQ) or Orai1 knockdown reversed all of the EMT-promoting effects of FGF4. BHQ also inhibited FGF4-induced EMT in a mouse xenograft model. Finally, 60 human lung ADC samples and 21 sets of matched specimens (primary and metastatic foci in lymph nodes from one patient) were used to confirm the clinicopathologic significance of FGF4 and its correlation with E-cadherin, Vimentin and Orai1 expression. Our study thus shows that FGF4 induces EMT by elevating SOCE in lung ADC.
Collapse
Affiliation(s)
- Lisha Qi
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin 300060, China.,National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Wangzhao Song
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,Tianjin Medical University, Tianjin 300070, China.,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin 300060, China.,National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Lingmei Li
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin 300060, China.,National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Lu Cao
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,Tianjin Medical University, Tianjin 300070, China.,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin 300060, China.,National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yue Yu
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin 300060, China.,National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Chunmin Song
- Department of Family Planning, Maternity & Child Care Center of Luoyang, Luoyang 471000, China
| | - Yalei Wang
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin 300060, China.,National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Fei Zhang
- The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin 300060, China.,National Clinical Research Center for Cancer, Tianjin 300060, China.,Research Center of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Yang Li
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,Tianjin Medical University, Tianjin 300070, China.,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin 300060, China.,National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Bin Zhang
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin 300060, China.,National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Wenfeng Cao
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.,The Key Laboratory of Tianjin Cancer Prevention and Treatment, Tianjin 300060, China.,National Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
24
|
Fearon AE, Carter EP, Clayton NS, Wilkes EH, Baker AM, Kapitonova E, Bakhouche BA, Tanner Y, Wang J, Gadaleta E, Chelala C, Moore KM, Marshall JF, Chupin J, Schmid P, Jones JL, Lockley M, Cutillas PR, Grose RP. PHLDA1 Mediates Drug Resistance in Receptor Tyrosine Kinase-Driven Cancer. Cell Rep 2018; 22:2469-2481. [PMID: 29490281 PMCID: PMC5848852 DOI: 10.1016/j.celrep.2018.02.028] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/09/2017] [Accepted: 02/06/2018] [Indexed: 11/09/2022] Open
Abstract
Development of resistance causes failure of drugs targeting receptor tyrosine kinase (RTK) networks and represents a critical challenge for precision medicine. Here, we show that PHLDA1 downregulation is critical to acquisition and maintenance of drug resistance in RTK-driven cancer. Using fibroblast growth factor receptor (FGFR) inhibition in endometrial cancer cells, we identify an Akt-driven compensatory mechanism underpinned by downregulation of PHLDA1. We demonstrate broad clinical relevance of our findings, showing that PHLDA1 downregulation also occurs in response to RTK-targeted therapy in breast and renal cancer patients, as well as following trastuzumab treatment in HER2+ breast cancer cells. Crucially, knockdown of PHLDA1 alone was sufficient to confer de novo resistance to RTK inhibitors and induction of PHLDA1 expression re-sensitized drug-resistant cancer cells to targeted therapies, identifying PHLDA1 as a biomarker for drug response and highlighting the potential of PHLDA1 reactivation as a means of circumventing drug resistance.
Collapse
Affiliation(s)
- Abbie E Fearon
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, UK
| | - Edward P Carter
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, UK
| | - Natasha S Clayton
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, UK
| | - Edmund H Wilkes
- Integrative Cell Signalling and Proteomics, Centre for Haemato-Oncology, Barts Cancer Institute, London EC1M 6BQ, UK
| | - Ann-Marie Baker
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, UK
| | - Ekaterina Kapitonova
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, UK
| | - Bakhouche A Bakhouche
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, UK
| | - Yasmine Tanner
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, UK
| | - Jun Wang
- Centre for Molecular Oncology, Barts Cancer Institute, London EC1M 6BQ, UK
| | - Emanuela Gadaleta
- Centre for Molecular Oncology, Barts Cancer Institute, London EC1M 6BQ, UK
| | - Claude Chelala
- Centre for Molecular Oncology, Barts Cancer Institute, London EC1M 6BQ, UK
| | - Kate M Moore
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, UK
| | - John F Marshall
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, UK
| | - Juliette Chupin
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, London EC1M 6BQ, UK
| | - Peter Schmid
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, London EC1M 6BQ, UK
| | - J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, UK
| | - Michelle Lockley
- Centre for Molecular Oncology, Barts Cancer Institute, London EC1M 6BQ, UK
| | - Pedro R Cutillas
- Integrative Cell Signalling and Proteomics, Centre for Haemato-Oncology, Barts Cancer Institute, London EC1M 6BQ, UK
| | - Richard P Grose
- Centre for Tumour Biology, Barts Cancer Institute-a CRUK Centre of Excellence, Queen Mary University of London, London EC1M 6BQ, UK.
| |
Collapse
|
25
|
Estienne A, Price CA. The fibroblast growth factor 8 family in the female reproductive tract. Reproduction 2018; 155:R53-R62. [DOI: 10.1530/rep-17-0542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/12/2017] [Accepted: 11/06/2017] [Indexed: 12/12/2022]
Abstract
Several growth factor families have been shown to be involved in the function of the female reproductive tract. One subfamily of the fibroblast growth factor (FGF) superfamily, namely the FGF8 subfamily (including FGF17 and FGF18), has become important as Fgf8 has been described as an oocyte-derived factor essential for glycolysis in mouse cumulus cells and aberrant expression ofFGF18has been described in ovarian and endometrial cancers. In this review, we describe the pattern of expression of these factors in normal ovaries and uteri in rodents, ruminants and humans, as well as the expression of their receptors and intracellular negative feedback regulators. Expression of these molecules in gynaecological cancers is also reviewed. The role of FGF8 and FGF18 in ovarian and uterine function is described, and potential differences between rodents and ruminants have been highlighted especially with respect to FGF18 signalling within the ovarian follicle. Finally, we identify major questions about the reproductive biology of FGFs that remain to be answered, including (1) the physiological concentrations within the ovary and uterus, (2) which cell types within the endometrial stroma and theca layer express FGFs and (3) which receptors are activated by FGF8 subfamily members in reproductive tissues.
Collapse
|
26
|
Hou N, Zhang M, Xu Y, Sun Z, Wang J, Zhang L, Zhang Q. Polysaccharides and their depolymerized fragments from Costaria costata: Molecular weight and sulfation-dependent anticoagulant and FGF/FGFR signal activating activities. Int J Biol Macromol 2017; 105:1511-1518. [PMID: 28619642 DOI: 10.1016/j.ijbiomac.2017.06.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/17/2017] [Accepted: 06/03/2017] [Indexed: 12/21/2022]
Abstract
Crude polysaccharides from Costaria costata were extracted by hot water and further fractionated by anion exchange chromatography into three polysaccharide fractions. Three low molecular weight fragments were then prepared by degradation of the polysaccharides with hydrogen peroxide and ascorbic acid. The structural features of the polysaccharides and their low molecular weight fragments were elucidated for the first time based on the HGPC, FT-IR, NMR, MS, monosaccharide composition, and other chemical analyses. Their anticoagulant and FGF-1, -2, -7, -8, -9, -10/FGFR1c signaling activation activities in BaF3 cells were also examined. Our studies showed that the polysaccharides were sulfated at different positions of galactose and fucose residues. The APTT-, PT- and TT-based anticoagulant assay results indicated that a high molecular weight and a higher degree of sulfation were essential for their anticoagulant activities. In contrast, not only the polysaccharides but also the depolymerized fragments showed significant FGF/FGFR signal activating activities in a FGF-, molecular weight-, and sulfation-dependent manner. The results presented in current study demonstrated the potential use of the polysaccharides and their fragments as anticoagulants and FGF signal regulators.
Collapse
Affiliation(s)
- Ningning Hou
- Key Laboratory of Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; College of Earth Science, University of Chinese Academy of Sciences, Beijing 100049, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China
| | - Meng Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Yingjie Xu
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Zhongmin Sun
- Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China; Department of Marine Organism Taxonomy and Phylogeny, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | - Jing Wang
- Key Laboratory of Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China
| | - Lijuan Zhang
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - Quanbin Zhang
- Key Laboratory of Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China; Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Sci. & Tech, Qingdao 266071, China.
| |
Collapse
|
27
|
Zhou C, Chen T, Xie Z, Qin Y, Ou Y, Zhang J, Li S, Chen R, Zhong N. RACK1 forms a complex with FGFR1 and PKM2, and stimulates the growth and migration of squamous lung cancer cells. Mol Carcinog 2017; 56:2391-2399. [PMID: 28418088 DOI: 10.1002/mc.22663] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 03/19/2017] [Accepted: 04/13/2017] [Indexed: 12/31/2022]
Abstract
Phosphorylation of Pyruvate Kinase M2 (PKM2) on Tyr105 by fibroblast growth factor receptor 1 (FGFR1) has been shown to promote its nuclear localization as well as cell growth in lung cancer. Better understanding the regulation of this process would benefit the clinical treatment for lung cancer. Here, it has been found that the adaptor protein receptor for activated PKC kinase (RACK1) formed a complex with FGFR1 and PKM2, and activated the FGFR1/PKM2 signaling. Knocking down the expression of RACK1 impaired the phosphorylation on Tyr105 of PKM2 and inhibited the growth and migration of lung cancer cells, while over-expression of RACK1 in lung cancer cells led to the resistance to Erdafitinib. Moreover, knocking down the expression of RACK1 impaired the tumorigenesis of lung cancer driven by LKB loss and mutated Ras (KrasG12D). Taken together, our study demonstrated the pivotal roles of RACK1 in FGFR1/PKM2 signaling, suggesting FGFR1/RACK1/PKM2 might be a therapeutic target for lung cancer treatment.
Collapse
Affiliation(s)
- Chengzhi Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Department of Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Tao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Department of Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Zhanhong Xie
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Department of Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Yinyin Qin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Department of Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Yangming Ou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Department of Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Jiexia Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Department of Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Shiyue Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Department of Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Rongchang Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Department of Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Department of Respiratory Medicine, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
28
|
Opaliński Ł, Sokołowska-Wędzina A, Szczepara M, Zakrzewska M, Otlewski J. Antibody-induced dimerization of FGFR1 promotes receptor endocytosis independently of its kinase activity. Sci Rep 2017; 7:7121. [PMID: 28769084 PMCID: PMC5540934 DOI: 10.1038/s41598-017-07479-z] [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] [Received: 11/28/2016] [Accepted: 06/27/2017] [Indexed: 12/22/2022] Open
Abstract
Fibroblast growth factors (FGFs) and their plasma membrane-localized receptors (FGFRs) play a key role in the regulation of developmental processes and metabolism. Aberrant FGFR signaling is associated with the progression of serious metabolic diseases and human cancer. Binding of FGFs to FGFRs induces receptor dimerization and transphosphorylation of FGFR kinase domains that triggers activation of intracellular signaling pathways. Following activation, FGFRs undergo internalization and subsequent lysosomal degradation, which terminates transmission of signals. Although factors that regulate FGFR endocytosis are continuously discovered, little is known about the molecular mechanism that initiates the internalization of FGFRs. Here, we analyzed the internalization of antibody fragments in various formats that target FGFR1. We show that FGFR1-specific antibody fragments in the monovalent scFv format bind to FGFR1, but are not internalized into cells that overproduce FGFR1. In contrast, the same scFv proteins in the bivalent scFv-Fc format are efficiently internalized via FGFR1-mediated, clathrin and dynamin dependent endocytosis. Interestingly, the receptor tyrosine kinase activity is dispensable for endocytosis of scFv-Fc-FGFR1 complexes, suggesting that only dimerization of receptor is required to trigger endocytosis of FGFR1 complexes.
Collapse
Affiliation(s)
- Łukasz Opaliński
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland.
| | - Aleksandra Sokołowska-Wędzina
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Martyna Szczepara
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Małgorzata Zakrzewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Jacek Otlewski
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383, Wroclaw, Poland.
| |
Collapse
|
29
|
Ravoori MK, Singh SP, Lee J, Bankson JA, Kundra V. In Vivo Assessment of Ovarian Tumor Response to Tyrosine Kinase Inhibitor Pazopanib by Using Hyperpolarized 13C-Pyruvate MR Spectroscopy and 18F-FDG PET/CT Imaging in a Mouse Model. Radiology 2017; 285:830-838. [PMID: 28707963 DOI: 10.1148/radiol.2017161772] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Purpose To assess in a mouse model whether early or late components of glucose metabolism, exemplified by fluorine 18 (18F) fluorodeoxyglucose (FDG) positron emission tomography (PET) and hyperpolarized carbon 13 (13C)-pyruvate magnetic resonance (MR) spectroscopy, can serve as indicators of response in ovarian cancer to multityrosine kinase inhibitor pazopanib. Materials and Methods In this Animal Care and Use Committee approved study, 17 days after the injection of 2 × 106 human ovarian SKOV3 tumors cells into 14 female nude mice, treatment with vehicle or pazopanib (2.5 mg per mouse peroral every other day) was initiated. Longitudinal T2-weighted MR imaging, dynamic MR spectroscopy of hyperpolarized pyruvate, and 18F-FDG PET/computed tomographic (CT) imaging were performed before treatment, 2 days after treatment, and 2 weeks after treatment. Results Pazopanib inhibited ovarian tumor growth compared with control (0.054 g ± 0.041 vs 0.223 g ± 0.112, respectively; six mice were treated with pazopanib and seven were control mice; P < .05). Significantly higher pyruvate-to-lactate conversion (lactate/pyruvate + lactate ratio) was found 2 days after treatment with pazopanib than before treatment (0.46 ± 0.07 vs 0.31 ± 0.14, respectively; P < .05; six tumors after treatment, seven tumors before treatment). This was not observed with the control group or with 18F-FDG PET/CT imaging. Conclusion The findings suggest that hyperpolarized 13C-pyruvate MR spectroscopy may serve as an early indicator of response to tyrosine kinase (angiogenesis) inhibitors such as pazopanib in ovarian cancer even when 18F-FDG PET/CT does not indicate a response. © RSNA, 2017 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Murali K Ravoori
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - Sheela P Singh
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - Jaehyuk Lee
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - James A Bankson
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| | - Vikas Kundra
- From the Departments of Cancer Systems Imaging (M.K.R., S.P.S., V.K.), Imaging Physics (J.L., J.A.B.), and Diagnostic Radiology (V.K.), University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
| |
Collapse
|
30
|
Wang S, Ding Z. Fibroblast growth factor receptors in breast cancer. Tumour Biol 2017; 39:1010428317698370. [PMID: 28459213 DOI: 10.1177/1010428317698370] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Fibroblast growth factor receptors are growth factor receptor tyrosine kinases, exerting their roles in embryogenesis, tissue homeostasis, and development of breast cancer. Recent genetic studies have identified some subtypes of fibroblast growth factor receptors as strong genetic loci associated with breast cancer. In this article, we review the recent epidemiological findings and experiment results of fibroblast growth factor receptors in breast cancer. First, we summarized the structure and physiological function of fibroblast growth factor receptors in humans. Then, we discussed the common genetic variations in fibroblast growth factor receptors that affect breast cancer risk. In addition, we also introduced the potential roles of each fibroblast growth factor receptors isoform in breast cancer. Finally, we explored the potential therapeutics targeting fibroblast growth factor receptors for breast cancer. Based on the biological mechanisms of fibroblast growth factor receptors leading to the pathogenesis in breast cancer, targeting fibroblast growth factor receptors may provide new opportunities for breast cancer therapeutic strategies.
Collapse
Affiliation(s)
- Shuwei Wang
- Department of General Surgery, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, P.R. China
| | - Zhongyang Ding
- Department of General Surgery, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, P.R. China
| |
Collapse
|
31
|
Karp JM, Sparks S, Cowburn D. Effects of FGFR2 kinase activation loop dynamics on catalytic activity. PLoS Comput Biol 2017; 13:e1005360. [PMID: 28151998 PMCID: PMC5313233 DOI: 10.1371/journal.pcbi.1005360] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 02/16/2017] [Accepted: 01/12/2017] [Indexed: 12/17/2022] Open
Abstract
The structural mechanisms by which receptor tyrosine kinases (RTKs) regulate catalytic activity are diverse and often based on subtle changes in conformational dynamics. The regulatory mechanism of one such RTK, fibroblast growth factor receptor 2 (FGFR2) kinase, is still unknown, as the numerous crystal structures of the unphosphorylated and phosphorylated forms of the kinase domains show no apparent structural change that could explain how phosphorylation could enable catalytic activity. In this study, we use several enhanced sampling molecular dynamics (MD) methods to elucidate the structural changes to the kinase's activation loop that occur upon phosphorylation. We show that phosphorylation favors inward motion of Arg664, while simultaneously favoring outward motion of Leu665 and Pro666. The latter structural change enables the substrate to bind leading to its resultant phosphorylation. Inward motion of Arg664 allows it to interact with the γ-phosphate of ATP as well as the substrate tyrosine. We show that this stabilizes the tyrosine and primes it for the catalytic phosphotransfer, and it may lower the activation barrier of the phosphotransfer reaction. Our work demonstrates the value of including dynamic information gleaned from computer simulation in deciphering RTK regulatory function.
Collapse
Affiliation(s)
- Jerome M. Karp
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Samuel Sparks
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - David Cowburn
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
32
|
Song KH, Park MS, Nandu TS, Gadad S, Kim SC, Kim MY. GALNT14 promotes lung-specific breast cancer metastasis by modulating self-renewal and interaction with the lung microenvironment. Nat Commun 2016; 7:13796. [PMID: 27982029 PMCID: PMC5171903 DOI: 10.1038/ncomms13796] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/02/2016] [Indexed: 12/23/2022] Open
Abstract
Some polypeptide N-acetyl-galactosaminyltransferases (GALNTs) are associated with cancer, but their function in organ-specific metastasis remains unclear. Here, we report that GALNT14 promotes breast cancer metastasis to the lung by enhancing the initiation of metastatic colonies as well as their subsequent growth into overt metastases. Our results suggest that GALNT14 augments the self-renewal properties of breast cancer cells (BCCs). Furthermore, GALNT14 overcomes the inhibitory effect of lung-derived bone morphogenetic proteins (BMPs) on self-renewal and therefore facilitates metastasis initiation within the lung microenvironment. In addition, GALNT14 supports continuous growth of BCCs in the lung by not only inducing macrophage infiltration but also exploiting macrophage-derived fibroblast growth factors (FGFs). Finally, we identify KRAS-PI3K-c-JUN signalling as an upstream pathway that accounts for the elevated expression of GALNT14 in lung-metastatic BCCs. Collectively, our findings uncover an unprecedented role for GALNT14 in the pulmonary metastasis of breast cancer and elucidate the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Ki-Hoon Song
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejon 305-701, Korea
| | - Mi So Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejon 305-701, Korea
| | - Tulip S. Nandu
- Cecil H. and Ida Green Center for Reproductive Biology Sciences and Division of Basic Reproductive Biology Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Shrikanth Gadad
- Cecil H. and Ida Green Center for Reproductive Biology Sciences and Division of Basic Reproductive Biology Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Sang-Cheol Kim
- Department of Biomedical Informatics, Center for Genome Science, National Institute of Health, KCDC, Choongchung-Buk-do 363-951, Korea
| | - Mi-Young Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejon 305-701, Korea
- KAIST Institute for the BioCentury, Cancer Metastasis Control Center, 291 Daehak-ro, Yuseong-gu, Daejeon 305-701, Korea
| |
Collapse
|
33
|
Tiong KH, Tan BS, Choo HL, Chung FFL, Hii LW, Tan SH, Khor NTW, Wong SF, See SJ, Tan YF, Rosli R, Cheong SK, Leong CO. Fibroblast growth factor receptor 4 (FGFR4) and fibroblast growth factor 19 (FGF19) autocrine enhance breast cancer cells survival. Oncotarget 2016; 7:57633-57650. [PMID: 27192118 PMCID: PMC5295378 DOI: 10.18632/oncotarget.9328] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 04/26/2016] [Indexed: 12/27/2022] Open
Abstract
Basal-like breast cancer is an aggressive tumor subtype with poor prognosis. The discovery of underlying mechanisms mediating tumor cell survival, and the development of novel agents to target these pathways, is a priority for patients with basal-like breast cancer. From a functional screen to identify key drivers of basal-like breast cancer cell growth, we identified fibroblast growth factor receptor 4 (FGFR4) as a potential mediator of cell survival. We found that FGFR4 mediates cancer cell survival predominantly via activation of PI3K/AKT. Importantly, a subset of basal-like breast cancer cells also secrete fibroblast growth factor 19 (FGF19), a canonical ligand specific for FGFR4. siRNA-mediated silencing of FGF19 or neutralization of extracellular FGF19 by anti-FGF19 antibody (1A6) decreases AKT phosphorylation, suppresses cancer cell growth and enhances doxorubicin sensitivity only in the FGFR4+/FGF19+ breast cancer cells. Consistently, FGFR4/FGF19 co-expression was also observed in 82 out of 287 (28.6%) primary breast tumors, and their expression is strongly associated with AKT phosphorylation, Ki-67 staining, higher tumor stage and basal-like phenotype. In summary, our results demonstrated the presence of an FGFR4/FGF19 autocrine signaling that mediates the survival of a subset of basal-like breast cancer cells and suggest that inactivation of this autocrine loop may potentially serve as a novel therapeutic intervention for future treatment of breast cancers.
Collapse
Affiliation(s)
- Kai Hung Tiong
- School of Postgraduate Studies, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
- Oral Cancer Research and Co-ordinating Center (OCRCC), Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
- Cancer Research Initiatives Foundation, Sime Darby Medical Centre, Subang Jaya, Malaysia
| | - Boon Shing Tan
- School of Postgraduate Studies, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Heng Lungh Choo
- School of Postgraduate Studies, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
- Center for Cancer and Stem Cell Research, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Felicia Fei-Lei Chung
- Center for Cancer and Stem Cell Research, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Ling-Wei Hii
- School of Postgraduate Studies, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
- Center for Cancer and Stem Cell Research, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Si Hoey Tan
- School of Postgraduate Studies, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
- Center for Cancer and Stem Cell Research, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Nelson Tze Woei Khor
- School of Medicine, Faculty of Medical and Health Sciences, The University of Auckland, New Zealand
| | - Shew Fung Wong
- School of Medicine, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Sze-Jia See
- Center for Cancer and Stem Cell Research, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Yuen-Fen Tan
- School of Postgraduate Studies, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
- Center for Cancer and Stem Cell Research, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Rozita Rosli
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Soon-Keng Cheong
- Faculty of Medicine and Health Sciences, University Tunku Abdul Rahman, Bandar Sungai Long, Selangor, Malaysia
| | - Chee-Onn Leong
- Center for Cancer and Stem Cell Research, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| |
Collapse
|
34
|
Jing Q, Wang Y, Liu H, Deng X, Jiang L, Liu R, Song H, Li J. FGFs: crucial factors that regulate tumour initiation and progression. Cell Prolif 2016; 49:438-47. [PMID: 27383016 DOI: 10.1111/cpr.12275] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 06/13/2016] [Indexed: 02/05/2023] Open
Abstract
Fibroblast growth factors (FGFs) are crucial signalling molecules involved in normal cell growth, differentiation and proliferation. Over the past few decades, a large body of research has illustrated effects of individual FGFs on tumour initiation and progression. Tumour development is commonly accompanied with generation of new blood and lymph vessels, which support enhanced cell proliferation. Moreover, acquisition of tumour cells of the epithelial-mesenchymal transition (EMT) phenotype, enhances tumour cell migration and invasion potentials, crucial steps in tumour metastasis. This review summarizes recent findings concerning roles of FGFs in angiogenesis, lymphangiogenesis and EMT.
Collapse
Affiliation(s)
- Qian Jing
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Yuanyuan Wang
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Hao Liu
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Xiaowei Deng
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Lin Jiang
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Rui Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Haixing Song
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| | - Jingyi Li
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, China
| |
Collapse
|
35
|
Liu Z, Yu S, Chen D, Shen G, Wang Y, Hou L, Lin D, Zhang J, Ye F. Design, synthesis, and biological evaluation of 3-vinyl-quinoxalin-2(1H)-one derivatives as novel antitumor inhibitors of FGFR1. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:1489-500. [PMID: 27217720 PMCID: PMC4861610 DOI: 10.2147/dddt.s88587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
FGFR1 is well known as a molecular target in anticancer drug design. TKI258 plays an important role in RTK inhibitors. Utilizing TKI258 as a lead compound that contains a quinazolinone nucleus, we synthesized four series of 3-vinyl-quinoxalin-2(1H)-one derivatives, a total of 27 compounds. We further evaluated these compounds for FGFR1 inhibition ability as well as cytotoxicity against four cancer cell lines (H460, B16-F10, Hela229, and Hct116) in vitro. Some compounds displayed good-to-excellent potency against the four tested cancer cell lines compared with TKI258. Structure–activity relationship analyses indicated that small substituents at the side chain of the 3-vinyl-quinoxalin-2(1H)-one were more effective than large substituents. Lastly, we used molecular docking to obtain further insight into the interactions between the compounds and FGFR1.
Collapse
Affiliation(s)
- Zhiguo Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Shufang Yu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Di Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Guoliang Shen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Yu Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Leping Hou
- Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Dan Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Jinsan Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Faqing Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, People's Republic of China
| |
Collapse
|
36
|
Hallinan N, Finn S, Cuffe S, Rafee S, O’Byrne K, Gately K. Targeting the fibroblast growth factor receptor family in cancer. Cancer Treat Rev 2016; 46:51-62. [DOI: 10.1016/j.ctrv.2016.03.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/23/2016] [Accepted: 03/28/2016] [Indexed: 02/08/2023]
|
37
|
Wang X, Chen D, Yu S, Zhang Z, Wang Y, Qi X, Fu W, Xie Z, Ye F. Synthesis and Evaluation of Biological and Antitumor Activities of Tetrahydrobenzothieno[2,3-d]Pyrimidine Derivatives as Novel Inhibitors of FGFR1. Chem Biol Drug Des 2016; 87:499-507. [PMID: 26575787 DOI: 10.1111/cbdd.12687] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/27/2015] [Accepted: 10/02/2015] [Indexed: 01/12/2023]
Abstract
A series of tetrahydrobenzothieno[2,3-d]pyrimidine derivatives were designed, synthesized, and evaluated as inhibitors of FGFR1. These analogs were synthesized via Gewald's reaction under mild conditions. The structures of the synthesized compounds were characterized by spectroscopic data (IR, (1) H NMR and MS). Their antitumor activities were evaluated against H460, A549 and U251 cell lines in vitro. Results revealed that the tested compounds showed moderate antitumor activities. Structure-activity relationship analyses indicated that compounds with an aromatic ring substituted in the C-2 position or with larger molecules such as 3g, 4c, and 7 were more effective than others. The compound, 3g (78.8% FGFR1 inhibition at 10 μm), was identified to have the most potent antitumor activities, with IC50 values of 7.7, 18.9, and 13.3 μm against the H460, A549, and U251 cell lines, respectively. Together, the results suggested that tetrahydrobenzothieno[2,3-d]pyrimidine derivatives may serve as a potential agent for the treatment of FGFR1-mediated cancers.
Collapse
Affiliation(s)
- Xuebao Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Di Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Shufang Yu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zaikui Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yu Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiaolu Qi
- Yichang Humanwell Pharmaceutical Co., Ltd, Yichang, 443005, China
| | - Weitao Fu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zixin Xie
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Faqing Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| |
Collapse
|
38
|
Zang C, Eucker J, Habbel P, Neumann C, Schulz CO, Bangemann N, Kissner L, Riess H, Liu H. Targeting multiple tyrosine kinase receptors with Dovitinib blocks invasion and the interaction between tumor cells and cancer-associated fibroblasts in breast cancer. Cell Cycle 2016; 14:1291-9. [PMID: 25714853 DOI: 10.4161/15384101.2014.995050] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A constitutive and dynamic interaction between tumor cells and their surrounding stroma is a prerequisite for tumor invasion and metastasis. Fibroblasts and myofibroblasts (collectively called cancer associated fibroblasts, CAFs) often represent the major cellular components of tumor stroma. Tumor cells secret different growth factors which induce CAFs proliferation and differentiation, and, consequently, CAFs secrete different chemokines, cytokines or growth factors which induce tumor cell invasion and metastasis. In this study we showed here that CAFs from breast cancer surgical specimens significantly induced the invasion of breast cancer cells in vitro. Most interestingly, the novel multiple tyrosine kinase inhibitor Dovitinib significantly blocked the CAFs-induced invasion of breast cancer cells by, at least in part, inhibition of the expression and secretion of CCL2, CCL5 and VEGF in CAFs. Inhibition of PI3K/Akt/mTOR signaling could be responsible for the effects of Dovitinib, since Dovitinib antagonized the promoted phosphorylated Akt after treatment with PDGF, FGF or breast cancer cell-conditioned media. Treatment with Dovitinib in combination with PI3K/Akt/mTOR signaling inhibitors Ly294002 or RAD001 resulted in additive inhibition of cell invasion. This is the first in vitro study to show that the multiple tyrosine kinase inhibitor has therapeutic activities against breast cancer metastasis by targeting both tumor cells and CAFs.
Collapse
Affiliation(s)
- Chuanbing Zang
- a Division of Hematology and Oncology ; Charité-University Medicine; Charitéplatz 1 ; Berlin , Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Anticoagulant and FGF/FGFR signal activating activities of the heparinoid propylene glycol alginate sodium sulfate and its oligosaccharides. Carbohydr Polym 2016; 136:641-8. [DOI: 10.1016/j.carbpol.2015.09.059] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 09/05/2015] [Accepted: 09/18/2015] [Indexed: 12/16/2022]
|
40
|
Sonoda K. Molecular biology of gynecological cancer. Oncol Lett 2015; 11:16-22. [PMID: 26834851 DOI: 10.3892/ol.2015.3862] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 09/24/2015] [Indexed: 12/20/2022] Open
Abstract
Cancer is a pathological condition in which the balance between cell growth and death is disordered. Various molecules have been reported to be involved in the oncogenic process of invasion, metastasis and resistance to treatment. An exponential growth in the collection of genomic and proteomic data in the past 20 years has provided major advances in understanding the molecular mechanisms of human cancer, which has been applied to diagnostic and treatment strategies. Targeted therapies have been developed and adopted, particularly for advanced, refractory or recurrent cancers, depending on individual molecular profiles. The aim of the present review is to provide a report of the current literature regarding the molecular biology of gynecological cancers.
Collapse
Affiliation(s)
- Kenzo Sonoda
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Fukuoka 812-8582, Japan
| |
Collapse
|
41
|
Hua G, Lv X, He C, Remmenga SW, Rodabough KJ, Dong J, Yang L, Lele SM, Yang P, Zhou J, Karst A, Drapkin RI, Davis JS, Wang C. YAP induces high-grade serous carcinoma in fallopian tube secretory epithelial cells. Oncogene 2015; 35:2247-65. [PMID: 26364602 PMCID: PMC4791205 DOI: 10.1038/onc.2015.288] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 06/11/2015] [Accepted: 07/06/2015] [Indexed: 12/12/2022]
Abstract
Accumulating evidence indicates that ovarian high-grade serous carcinoma (HGSC) originates from fallopian tube secretory epithelial cells (FTSECs). However, the molecular mechanisms underlying the initiation and progression of HGSC derived from FTSECs remains unclear. In this study, we found that the Hippo/Yes-associated protein (YAP) signaling pathway has a critical role in the initiation and progression of fallopian tube and ovarian HGSC. Importantly, YAP was overexpressed in inflammatory and cancerous fallopian tube tissues. Further, overexpression of wild-type YAP, or constitutively active YAP in immortalized FTSECs, induced cell proliferation, migration, colony formation and tumorigenesis. Moreover, the Hippo/YAP and the fibroblast growth factor (FGF) signaling pathways formed an autocrine/paracrine-positive feedback loop to drive the progression of the FTSEC-derived HGSC. Evidence in this study strongly suggests that combined therapy with inhibitors of YAP (such as verteporfin) and FGF receptors (such as BGJ398) can provide a novel therapeutic strategy to treat fallopian tube and ovarian HGSC.
Collapse
Affiliation(s)
- G Hua
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA.,The Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - X Lv
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - C He
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA.,The Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - S W Remmenga
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - K J Rodabough
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - J Dong
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - L Yang
- The Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - S M Lele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - P Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - J Zhou
- Department of Obstetrics and Gynecology, Urumuqi General Hospital of Lanzhou Military Region, Urumuqi, China
| | - A Karst
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - R I Drapkin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - J S Davis
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.,Omaha Veterans Affairs Medical Center, Omaha, NE, USA
| | - C Wang
- Olson Center for Women's Health, Department of Obstetrics/Gynecology, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
42
|
Lo AKF, Dawson CW, Young LS, Ko CW, Hau PM, Lo KW. Activation of the FGFR1 signalling pathway by the Epstein-Barr virus-encoded LMP1 promotes aerobic glycolysis and transformation of human nasopharyngeal epithelial cells. J Pathol 2015; 237:238-48. [PMID: 26096068 DOI: 10.1002/path.4575] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 06/02/2015] [Accepted: 06/12/2015] [Indexed: 12/22/2022]
Abstract
Non-keratinizing nasopharyngeal carcinoma (NPC) is closely associated with Epstein-Barr virus (EBV) infection. The EBV-encoded latent membrane protein 1 (LMP1) is believed to play an important role in NPC pathogenesis by virtue of its ability to activate multiple cell signalling pathways which collectively promote cell proliferation, transformation, angiogenesis, and invasiveness, as well as modulation of energy metabolism. In this study, we report that LMP1 increases cellular uptake of glucose and glutamine, enhances LDHA activity and lactate production, but reduces pyruvate kinase activity and pyruvate concentrations. LMP1 also increases the phosphorylation of PKM2, LDHA, and FGFR1, as well as the expression of PDHK1, FGFR1, c-Myc, and HIF-1α, regardless of oxygen availability. Collectively, these findings suggest that LMP1 promotes aerobic glycolysis. With respect to FGFR1 signalling, LMP1 not only increases FGFR1 expression, but also up-regulates FGF2, leading to constitutive activation of the FGFR1 signalling pathway. Furthermore, two inhibitors of FGFR1 (PD161570 and SU5402) attenuate LMP1-mediated aerobic glycolysis, cellular transformation (proliferation and anchorage-independent growth), cell migration, and invasion in nasopharyngeal epithelial cells, identifying FGFR1 signalling as a key pathway in LMP1-mediated growth transformation. Immunohistochemical staining revealed that high levels of phosphorylated FGFR1 are common in primary NPC specimens and that this correlated with the expression of LMP1. In addition, FGFR1 inhibitors suppress cell proliferation and anchorage-independent growth of NPC cells. Our current findings demonstrate that LMP1-mediated FGFR1 activation contributes to aerobic glycolysis and transformation of epithelial cells, thereby implicating FGF2/FGFR1 signalling activation in the EBV-driven pathogenesis of NPC.
Collapse
Affiliation(s)
- Angela Kwok-Fung Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory in Oncology in South China and Li Ka Shing Institute of Health Science, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong.,Cancer Research UK Cancer Centre, School of Cancer Sciences, University of Birmingham, Vincent Drive, Edgbaston, Birmingham, UK
| | - Christopher W Dawson
- Cancer Research UK Cancer Centre, School of Cancer Sciences, University of Birmingham, Vincent Drive, Edgbaston, Birmingham, UK
| | | | - Chuen-Wai Ko
- Department of Anatomical and Cellular Pathology, State Key Laboratory in Oncology in South China and Li Ka Shing Institute of Health Science, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Pok-Man Hau
- Department of Anatomical and Cellular Pathology, State Key Laboratory in Oncology in South China and Li Ka Shing Institute of Health Science, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Kwok-Wai Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory in Oncology in South China and Li Ka Shing Institute of Health Science, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
43
|
Ling L, Tan SK, Goh TH, Cheung E, Nurcombe V, van Wijnen AJ, Cool SM. Targeting the heparin-binding domain of fibroblast growth factor receptor 1 as a potential cancer therapy. Mol Cancer 2015. [PMID: 26201468 PMCID: PMC4511971 DOI: 10.1186/s12943-015-0391-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background Aberrant activation of fibroblast growth factor receptors (FGFRs) deregulates cell proliferation and promotes cell survival, and may predispose to tumorigenesis. Therefore, selective inactivation of FGFRs is an important strategy for cancer therapy. Here as a proof-of-concept study, we developed a FGFR1 neutralizing antisera, IMB-R1, employing a novel strategy aimed at preventing the access of essential heparan sulfate (HS) co-receptors to the heparin-binding domain on FGFR1. Methods The mRNA and protein expression level of FGFR1 and other FGFRs were examined in several lines of breast cancer and osteosarcoma cells and corresponding normal cells using Taqman real-time quantitative PCR and Western blot analysis. The specificity of IMB-R1 against FGFR1 was assessed with various ELISA-based approaches and Receptor Tyrosine Kinase array. Proliferation assay and apoptosis analysis were performed to assess the effect of IMB-R1 on cancer cell growth and apoptosis, respectively, in comparison with known FGFR1 inhibitors. The IMB-R1 induced alteration of intracellular signaling and gene expression were analysed using Western blot and microarray approaches. Immunohistochemical staining of FGFR1 using IMB-R1 were carried out in different cancer tissues from clinical patients. Throughout the study, statistical differences were determined by Student’s t test where appropriate and reported when a p value was less than 0.05. Results We demonstrate that IMB-R1 is minimally cross-reactive for other FGFRs, and that it potently and specifically inhibits binding of heparin to FGFR1. Furthermore, IMB-R1 blocks the interaction of FGF2 with FGFR1, the kinase activity of FGFR1 and activation of intracellular FGFR signaling. Cancer cells treated with IMB-R1 displayed impaired FGF2 signaling, were unable to grow and instead underwent apoptosis. IMB-R1-induced cell death correlated with a disruption of antioxidative defense networks and increased expression of several tumor suppressors and apoptotic proteins, including p53. Immunostaining with IMB-R1 was stronger in human cancer tissues in which the FGFR1 gene is amplified. Conclusion Our study suggests that blocking HS interaction with the heparin-binding domains of FGFR1 inhibited cancer cell growth, which can be an attractive strategy to inactivate cancer-related heparin-binding proteins. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0391-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ling Ling
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore, 138648, Singapore
| | - Si Kee Tan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore
| | - Ting Hwee Goh
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore, 138648, Singapore
| | - Edwin Cheung
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore, 138672, Singapore.,Faculty of Health Sciences, University of Macau, E12 Avenida da Universidade, Taipa, Macau, China
| | - Victor Nurcombe
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore, 138648, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
| | - Andre J van Wijnen
- Department of Orthopedic Surgery & Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street SW, MedSci 3-69, Rochester, MN, 55905, USA.
| | - Simon M Cool
- Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore, 138648, Singapore. .,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore.
| |
Collapse
|
44
|
Ye F, Wang Y, Nian S, Wang Y, Chen D, Yu S, Wang S. Synthesis and evaluation of biological and antitumor activities of 5,7-dimethyl- oxazolo[5,4-d]pyrimidine-4,6(5H,7H)-dione derivatives as novel inhibitors of FGFR1. J Enzyme Inhib Med Chem 2015; 30:961-6. [PMID: 25683078 DOI: 10.3109/14756366.2014.1002401] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A series of 5,7-dimethyl-oxazolo[5,4-d]pyrimidine-4,6(5H,7H)-dione derivatives, N5a-5l, was designed, synthesized and evaluated for their FGFR1-inhibition ability as well as cytotoxicity against three cancer cell lines (H460, B16F10 and A549) in vitro. Several compounds displayed good-to-excellent potency against these cancer cell lines compared to SU5402. Structure-activity relationship analyses indicated that compounds with a rigid structure and more heteroatoms at the side chain of the parent ring were more effective than those without these substitutions. The compound N5g (37.4% FGFR1 inhibition at 1.0 μM) was identified to have the most potent antitumor activities, with IC50 values of 5.472, 4.260 and 5.837 μM against H460, B16F10 and A549 cell lines, respectively. Together, our results suggest that 5,7-dimethyl-oxazolo[5,4-d]pyrimidine-4,6(5H,7H)-dione derivatives may serve as potential agents for the treatment of FGFR1-mediated cancers.
Collapse
Affiliation(s)
- Faqing Ye
- a School of Pharmacy, Health Science Center Xi'an Jiaotong University , Xi'an , China and.,b School of Pharmaceutical Sciences, Wenzhou Medical University , Wenzhou , China
| | - Yuewu Wang
- b School of Pharmaceutical Sciences, Wenzhou Medical University , Wenzhou , China
| | - Siyun Nian
- b School of Pharmaceutical Sciences, Wenzhou Medical University , Wenzhou , China
| | - Yu Wang
- b School of Pharmaceutical Sciences, Wenzhou Medical University , Wenzhou , China
| | - Di Chen
- b School of Pharmaceutical Sciences, Wenzhou Medical University , Wenzhou , China
| | - Shufang Yu
- b School of Pharmaceutical Sciences, Wenzhou Medical University , Wenzhou , China
| | - Sicen Wang
- a School of Pharmacy, Health Science Center Xi'an Jiaotong University , Xi'an , China and
| |
Collapse
|
45
|
Beesley PW, Herrera-Molina R, Smalla KH, Seidenbecher C. The Neuroplastin adhesion molecules: key regulators of neuronal plasticity and synaptic function. J Neurochem 2014; 131:268-83. [PMID: 25040546 DOI: 10.1111/jnc.12816] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 07/02/2014] [Accepted: 07/03/2014] [Indexed: 01/21/2023]
Abstract
The Neuroplastins Np65 and Np55 are neuronal and synapse-enriched immunoglobulin superfamily molecules that play important roles in a number of key neuronal and synaptic functions including, for Np65, cell adhesion. In this review we focus on the physiological roles of the Neuroplastins in promoting neurite outgrowth, regulating the structure and function of both inhibitory and excitatory synapses in brain, and in neuronal and synaptic plasticity. We discuss the underlying molecular and cellular mechanisms by which the Neuroplastins exert their physiological effects and how these are dependent upon the structural features of Np65 and Np55, which enable them to bind to a diverse range of protein partners. In turn this enables the Neuroplastins to interact with a number of key neuronal signalling cascades. These include: binding to and activation of the fibroblast growth factor receptor; Np65 trans-homophilic binding leading to activation of p38 MAPK and internalization of glutamate (GluR1) receptor subunits; acting as accessory proteins for monocarboxylate transporters, thus affecting neuronal energy supply, and binding to GABAA α1, 2 and 5 subunits, thus regulating the composition and localization of GABAA receptors. An emerging theme is the role of the Neuroplastins in regulating the trafficking and subcellular localization of specific binding partners. We also discuss the involvement of Neuroplastins in a number of pathophysiological conditions, including ischaemia, schizophrenia and breast cancer and the role of a single nucleotide polymorphism in the human Neuroplastin (NPTN) gene locus in impairment of cortical development and cognitive functions. Neuroplastins are neuronal cell adhesion molecules, which induce neurite outgrowth and play important roles in synaptic maturation and plasticity. This review summarizes the functional implications of Neuroplastins for correct synaptic membrane protein localization, neuronal energy supply, expression of LTP and LTD, animal and human behaviour, and pathophysiology and disease. It focuses particularly on Neuroplastin binding partners and signalling mechanisms, and proposes perspectives for future research on these important immunoglobulin superfamily members.
Collapse
Affiliation(s)
- Philip W Beesley
- School of Biological Sciences, Royal Holloway University of London, Egham, UK
| | | | | | | |
Collapse
|
46
|
Vera C, Tapia V, Vega M, Romero C. Role of nerve growth factor and its TRKA receptor in normal ovarian and epithelial ovarian cancer angiogenesis. J Ovarian Res 2014; 7:82. [PMID: 25296882 PMCID: PMC4245781 DOI: 10.1186/s13048-014-0082-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 08/06/2014] [Indexed: 01/18/2023] Open
Abstract
In normal ovarian function a controlled angiogenesis is essential. Several growth factors are involved in this process, such as the vascular endothelial growth factor (VEGF) and nerve growth factor (NGF). The angiogenesis process in the normal ovary is a tightly controlled process that occurs in each ovarian cycle. Also, angiogenesis is critical for ovarian cancer development and it is responsible for tumor spread, metastasis and its peritoneal dissemination. Ovarian cancer is the fifth leading cause of cancer death in women and it is distinguished as the most lethal gynecologic cancer. In recent years angiogenesis has been given considerable attention in order to identify targets for developing effective anti-tumor therapies. Several molecules have been reported to promote angiogenesis, such as platelet-derived growth factor (PDGF) and its receptors, the angiopoietin/Tie ligand/receptor system and fibroblast growth factor (FGF). Primarily, VEGF has been identified to play key roles in driving angiogenesis. The above-mentioned molecules are candidate drug targets. Used in combination with other treatments, anti-angiogenic therapies have managed to reduce disease progression. The present review is focused in NGF and its high affinity receptor tyrosine kinase A (TRKA). The expression of VEGF, proliferation and the angiogenesis process in ovarian cancer is importantly induced by NGF, among other molecules.
Collapse
Affiliation(s)
- Carolina Vera
- />Laboratorio de Endocrinología y Biología Reproductiva, Hospital Clínico Universidad de Chile, Santos Dumont # 999, Santiago, Chile
| | - Verónica Tapia
- />Laboratorio de Endocrinología y Biología Reproductiva, Hospital Clínico Universidad de Chile, Santos Dumont # 999, Santiago, Chile
- />Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Margarita Vega
- />Laboratorio de Endocrinología y Biología Reproductiva, Hospital Clínico Universidad de Chile, Santos Dumont # 999, Santiago, Chile
- />Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Carmen Romero
- />Laboratorio de Endocrinología y Biología Reproductiva, Hospital Clínico Universidad de Chile, Santos Dumont # 999, Santiago, Chile
- />Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- />Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
| |
Collapse
|
47
|
Hussain HA, Harvey AJ. Evolution of breast cancer therapeutics: Breast tumour kinase’s role in breast cancer and hope for breast tumour kinase targeted therapy. World J Clin Oncol 2014; 5:299-310. [PMID: 25114846 PMCID: PMC4127602 DOI: 10.5306/wjco.v5.i3.299] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/25/2014] [Accepted: 06/03/2014] [Indexed: 02/06/2023] Open
Abstract
There have been significant improvements in the detection and treatment of breast cancer in recent decades. However, there is still a need to develop more effective therapeutic techniques that are patient specific with reduced toxicity leading to further increases in patients’ overall survival; the ongoing progress in understanding recurrence, resistant and spread also needs to be maintained. Better understanding of breast cancer pathology, molecular biology and progression as well as identification of some of the underlying factors involved in breast cancer tumourgenesis and metastasis has led to the identification of novel therapeutic targets. Over a number of years interest has risen in breast tumour kinase (Brk) also known as protein tyrosine kinase 6; the research field has grown and Brk has been described as a desirable therapeutic target in relation to tyrosine kinase inhibition as well as disruption of its kinase independent activity. This review will outline the current “state of play” with respect to targeted therapy for breast cancer, as well as discussing Brk’s role in the processes underlying tumour development and metastasis and its potential as a therapeutic target in breast cancer.
Collapse
|
48
|
Kähkönen T, Härkönen P. The emerging role of FGF receptor as a potential target in breast cancer. BREAST CANCER MANAGEMENT 2014. [DOI: 10.2217/bmt.14.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Tiina Kähkönen
- Institute of Biomedicine, Department of Cell Biology & Anatomy, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Pirkko Härkönen
- Institute of Biomedicine, Department of Cell Biology & Anatomy, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| |
Collapse
|
49
|
|