1
|
Turlej E, Domaradzka A, Radzka J, Drulis-Fajdasz D, Kulbacka J, Gizak A. Cross-Talk Between Cancer and Its Cellular Environment-A Role in Cancer Progression. Cells 2025; 14:403. [PMID: 40136652 PMCID: PMC11940884 DOI: 10.3390/cells14060403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
The tumor microenvironment is a dynamic and complex three-dimensional network comprising the extracellular matrix and diverse non-cancerous cells, including fibroblasts, adipocytes, endothelial cells and various immune cells (lymphocytes T and B, NK cells, dendritic cells, monocytes/macrophages, myeloid-derived suppressor cells, and innate lymphoid cells). A constantly and rapidly growing number of studies highlight the critical role of these cells in shaping cancer survival, metastatic potential and therapy resistance. This review provides a synthesis of current knowledge on the modulating role of the cellular microenvironment in cancer progression and response to treatment.
Collapse
Affiliation(s)
- Eliza Turlej
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Aleksandra Domaradzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Justyna Radzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Dominika Drulis-Fajdasz
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Julita Kulbacka
- Departament of Molecular and Cellular Biology, Faculty of Pharmacy, Wrocław Medical University, Borowska 211A, 50-556 Wrocław, Poland;
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Agnieszka Gizak
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| |
Collapse
|
2
|
Chai F, Wang G, Shen Y, Niu Y, Huang Y, Fu T, Yang T, Jiang Y, Zhang J. KGF impedes TRIM21-enhanced stabilization of keratin 10 mediating differentiation in hypopharyngeal cancer. Cell Signal 2025; 127:111614. [PMID: 39848455 DOI: 10.1016/j.cellsig.2025.111614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/03/2025] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
KGF, also known as FGF7, is a member of the fibroblast growth factor (FGF) family that binds with high affinity to the FGF receptor 2b (FGFR2b) and regulates various cellular processes, including cell proliferation and differentiation in a variety of tumors. However, its potential role in hypopharyngeal cancer (HPC) remains largely unknown. In our study, we observed increased expression of FGFR2b in HPC. KGF treatment inhibited the expression of the differentiation marker keratin 10 (K10) protein at the post-transcriptional level in FaDu cells. Furthermore, treatment with the proteasome inhibitor MG132 was found to attenuate KGF-induced K10 reduction, suggesting the involvement of the ubiquitin-proteasome system. Using mass spectrometry and immunoprecipitation analysis, we identified the E3 ubiquitin ligase TRIM21 as a K10-interacting protein. Unexpectedly, instead of causing degradation, TRIM21 enhanced K10 protein stability through K6-linked ubiquitination of K10 at lysine 163 (K163) in the context of KGF exposure. Meanwhile, KGF treatment decreased TRIM21 protein levels, which were regulated by the p38 MAPK pathway, leading to K48-linked ubiquitination-mediated degradation of TRIM21. Notably, TRIM21 knockdown significantly promoted proliferation, inhibited differentiation and migration of FaDu cells, whereas TRIM21 overexpression had opposite effects in vitro and suppressed xenograft tumor growth in vivo. Our study demonstrates that TRIM21 may act as a tumor suppressor in HPC. However, TRIM21 overexpression decreased the sensitivity of FaDu cells to 5-fluorouracil, whereas TRIM21 knockdown or KGF administration significantly increased 5-fluorouracil sensitivity. Taken together, these findings highlight the intricate balance between protein stabilization and degradation orchestrated by KGF. This ubiquitination-mediated non-degradation mechanism of TRIM21 may provide novel therapeutic strategies for HPC and other cancers.
Collapse
Affiliation(s)
- Fangyu Chai
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China
| | - Guangyi Wang
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China
| | - Yibang Shen
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China
| | - Yanfang Niu
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Yichuan Huang
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China
| | - Tao Fu
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China
| | - Tao Yang
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Higher Education Key Laboratory of Tumor Immunology & Targeted Drug Development in Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi 030001, China; Key laboratory of Digestive Disease & Organ Transplantation in Shanxi Province, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China.
| | - Yan Jiang
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China.
| | - Jisheng Zhang
- Key Laboratory, Department of Otolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, 266003 Qingdao, China.
| |
Collapse
|
3
|
Li S, Zhao X, Fu K, Zhu S, Pan C, Yang C, Wang F, To KK, Fu L. Resistance to antibody-drug conjugates: A review. Acta Pharm Sin B 2025; 15:737-756. [PMID: 40177568 PMCID: PMC11959940 DOI: 10.1016/j.apsb.2024.12.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 04/05/2025] Open
Abstract
Antibody-drug conjugates (ADCs) are antitumor drugs composed of monoclonal antibodies and cytotoxic payload covalently coupled by a linker. Currently, 15 ADCs have been clinically approved worldwide. More than 100 clinical trials at different phases are underway to investigate the newly developed ADCs. ADCs represent one of the fastest growing classes of targeted antitumor drugs in oncology drug development. It takes advantage of the specific targeting of tumor-specific antigen by antibodies to deliver cytotoxic chemotherapeutic drugs precisely to tumor cells, thereby producing promising antitumor efficacy and favorable adverse effect profiles. However, emergence of drug resistance has severely hindered the clinical efficacy of ADCs. In this review, we introduce the structure and mechanism of ADCs, describe the development of ADCs, summarized the latest research about the mechanisms of ADC resistance, discussed the strategies to overcome ADCs resistance, and predicted biomarkers for treatment response to ADC, aiming to contribute to the development of ADCs in the future.
Collapse
Affiliation(s)
- Sijia Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xinyu Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Kai Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shuangli Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Can Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Kenneth K.W. To
- School of Pharmacy, the Chinese University of Hong Kong, Hong Kong 999077, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
4
|
Hsieh CH, Ho PS, Wang WL, Shih FH, Hong CT, Wang PW, Shieh DB, Chang WL, Wang YC. Decreased plasma gelsolin fosters a fibrotic tumor microenvironment and promotes chemoradiotherapy resistance in esophageal squamous cell carcinoma. J Biomed Sci 2024; 31:90. [PMID: 39261905 PMCID: PMC11389350 DOI: 10.1186/s12929-024-01078-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Stromal fibrosis is highly associated with therapeutic resistance and poor survival in esophageal squamous cell carcinoma (ESCC) patients. Low expression of plasma gelsolin (pGSN), a serum abundant protein, has been found to correlate with inflammation and fibrosis. Here, we evaluated pGSN expression in patients with different stages of cancer and therapeutic responses, and delineated the molecular mechanisms involved to gain insight into therapeutic strategies for ESCC. METHODS Circulating pGSN level in ESCC patients was determined by enzyme-linked immunosorbent assay analysis, and the tissue microarray of tumors was analyzed by immunohistochemistry staining. Cell-based studies were performed to investigate cancer behaviors and molecular mechanisms, and mouse models were used to examine the pGSN-induced tumor suppressive effects in vivo. RESULTS Circulating pGSN expression is distinctively decreased during ESCC progression, and low pGSN expression correlates with poor therapeutic responses and poor survival. Methylation-specific PCR analysis confirmed that decreased pGSN expression is partly attributed to the hypermethylation of the GSN promoter, the gene encoding pGSN. Importantly, cell-based immunoprecipitation and protein stability assays demonstrated that pGSN competes with oncogenic tenascin-C (TNC) for the binding and degradation of integrin αvβ3, revealing that decreased pGSN expression leads to the promotion of oncogenic signaling transduction in cancer cells and fibroblasts. Furthermore, overexpression of pGSN caused the attenuation of TNC expression and inactivation of cancer-associated fibroblast (CAF), thereby leading to tumor growth inhibition in mice. CONCLUSIONS Our results demonstrated that GSN methylation causes decreased secretion of pGSN, leading to integrin dysregulation, oncogenic TNC activation, and CAF formation. These findings highlight the role of pGSN in therapeutic resistance and the fibrotic tumor microenvironment of ESCC.
Collapse
Affiliation(s)
- Chih-Hsiung Hsieh
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Shiuan Ho
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Lun Wang
- Department of Internal Medicine, E-Da Hospital, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Fu-Hsuan Shih
- Institute of Basic Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chen-Tai Hong
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Wen Wang
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Dar-Bin Shieh
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, Taiwan
- Center for Micro/Nano Science and Technology, Advanced Optoelectronic Technology Center, Innovation Center for Advanced Medical Device Technology, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Lun Chang
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Yi-Ching Wang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Institute of Basic Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
5
|
Dunbar KJ, Wong KK, Rustgi AK. Cancer-Associated Fibroblasts in Esophageal Cancer. Cell Mol Gastroenterol Hepatol 2024; 17:687-695. [PMID: 38246591 PMCID: PMC10958110 DOI: 10.1016/j.jcmgh.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024]
Abstract
Cancer-associated fibroblasts (CAFs), a heterogenous population, can promote cancer cell proliferation, migration, invasion, immunosuppression, and therapeutic resistance in solid tumors. These effects are mediated through secretion of cytokines and growth factors, remodeling of the extracellular matrix, and providing metabolic support for cancer cells. The presence of CAFs in esophageal carcinoma are associated with reduced overall survival and increased resistance to chemotherapy and radiotherapy; thus, identifying therapeutic vulnerabilities of CAFs is a necessity. In esophageal cancer, the mechanisms for CAF recruitment, CAF-mediated promotion of tumorigenesis, metastatic dissemination, and therapeutic resistance have yet to be fully evaluated. Here, we provide an overview of the current understanding of CAFs in esophageal cancer, namely in esophageal squamous cell carcinoma and esophageal adenocarcinoma, as well as in the preneoplastic conditions that predispose to these cancers. Interestingly, there is a discrepancy in our knowledge of CAF biology between esophageal cancer subtypes, with very few studies in esophageal adenocarcinoma, and its precursor lesion Barrett's esophagus, compared with esophageal squamous cell carcinoma. We propose that although great strides have been made, certain questions remain to which answers hopefully will emerge to have an impact on biomarker diagnostics and translational therapeutics.
Collapse
Affiliation(s)
- Karen J Dunbar
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York
| | - Kwok K Wong
- Division of Hematology-Oncology, Perlmutter Cancer Center, New York University Langone Health, New York, New York
| | - Anil K Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York.
| |
Collapse
|
6
|
Al-Bzour NN, Al-Bzour AN, Ababneh OE, Al-Jezawi MM, Saeed A, Saeed A. Cancer-Associated Fibroblasts in Gastrointestinal Cancers: Unveiling Their Dynamic Roles in the Tumor Microenvironment. Int J Mol Sci 2023; 24:16505. [PMID: 38003695 PMCID: PMC10671196 DOI: 10.3390/ijms242216505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Gastrointestinal cancers are highly aggressive malignancies with significant mortality rates. Recent research emphasizes the critical role of the tumor microenvironment (TME) in these cancers, which includes cancer-associated fibroblasts (CAFs), a key component of the TME that have diverse origins, including fibroblasts, mesenchymal stem cells, and endothelial cells. Several markers, such as α-SMA and FAP, have been identified to label CAFs, and some specific markers may serve as potential therapeutic targets. In this review article, we summarize the literature on the multifaceted role of CAFs in tumor progression, including their effects on angiogenesis, immune suppression, invasion, and metastasis. In addition, we highlight the use of single-cell transcriptomics to understand CAF heterogeneity and their interactions within the TME. Moreover, we discuss the dynamic interplay between CAFs and the immune system, which contributes to immunosuppression in the TME, and the potential for CAF-targeted therapies and combination approaches with immunotherapy to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Noor N. Al-Bzour
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15232, USA; (N.N.A.-B.); (A.N.A.-B.)
| | - Ayah N. Al-Bzour
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15232, USA; (N.N.A.-B.); (A.N.A.-B.)
| | - Obada E. Ababneh
- Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan; (O.E.A.); (M.M.A.-J.)
| | - Moayad M. Al-Jezawi
- Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan; (O.E.A.); (M.M.A.-J.)
| | - Azhar Saeed
- Department of Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT 05401, USA;
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15232, USA; (N.N.A.-B.); (A.N.A.-B.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
7
|
Alotaibi A, Gadekar VP, Gundla PS, Mandarthi S, Jayendra N, Tungekar A, Lavanya BV, Bhagavath AK, Cordero MAW, Pitkaniemi J, Niazi SK, Upadhya R, Bepari A, Hebbar P. Global comparative transcriptomes uncover novel and population-specific gene expression in esophageal squamous cell carcinoma. Infect Agent Cancer 2023; 18:47. [PMID: 37641095 PMCID: PMC10463703 DOI: 10.1186/s13027-023-00525-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) has a poor prognosis and is one of the deadliest gastrointestinal malignancies. Despite numerous transcriptomics studies to understand its molecular basis, the impact of population-specific differences on this disease remains unexplored. AIMS This study aimed to investigate the population-specific differences in gene expression patterns among ESCC samples obtained from six distinct global populations, identify differentially expressed genes (DEGs) and their associated pathways, and identify potential biomarkers for ESCC diagnosis and prognosis. In addition, this study deciphers population specific microbial and chemical risk factors in ESCC. METHODS We compared the gene expression patterns of ESCC samples from six different global populations by analyzing microarray datasets. To identify DEGs, we conducted stringent quality control and employed linear modeling. We cross-compared the resulting DEG lists of each populations along with ESCC ATLAS to identify known and novel DEGs. We performed a survival analysis using The Cancer Genome Atlas Program (TCGA) data to identify potential biomarkers for ESCC diagnosis and prognosis among the novel DEGs. Finally, we performed comparative functional enrichment and toxicogenomic analysis. RESULTS Here we report 19 genes with distinct expression patterns among populations, indicating population-specific variations in ESCC. Additionally, we discovered 166 novel DEGs, such as ENDOU, SLCO1B3, KCNS3, IFI35, among others. The survival analysis identified three novel genes (CHRM3, CREG2, H2AC6) critical for ESCC survival. Notably, our findings showed that ECM-related gene ontology terms and pathways were significantly enriched among the DEGs in ESCC. We also found population-specific variations in immune response and microbial infection-related pathways which included genes enriched for HPV, Ameobiosis, Leishmaniosis, and Human Cytomegaloviruses. Our toxicogenomic analysis identified tobacco smoking as the primary risk factor and cisplatin as the main drug chemical interacting with the maximum number of DEGs across populations. CONCLUSION This study provides new insights into population-specific differences in gene expression patterns and their associated pathways in ESCC. Our findings suggest that changes in extracellular matrix (ECM) organization may be crucial to the development and progression of this cancer, and that environmental and genetic factors play important roles in the disease. The novel DEGs identified may serve as potential biomarkers for diagnosis, prognosis and treatment.
Collapse
Grants
- 43- PRFA-P-8 the Deanship of Scientific Research, Princess Nourah bint Abdulrahman University, through the Program of Research Project Funding After Publication
- 43- PRFA-P-8 the Deanship of Scientific Research, Princess Nourah bint Abdulrahman University, through the Program of Research Project Funding After Publication
- 43- PRFA-P-8 the Deanship of Scientific Research, Princess Nourah bint Abdulrahman University, through the Program of Research Project Funding After Publication
- 43- PRFA-P-8 the Deanship of Scientific Research, Princess Nourah bint Abdulrahman University, through the Program of Research Project Funding After Publication
- 43- PRFA-P-8 the Deanship of Scientific Research, Princess Nourah bint Abdulrahman University, through the Program of Research Project Funding After Publication
- 43- PRFA-P-8 the Deanship of Scientific Research, Princess Nourah bint Abdulrahman University, through the Program of Research Project Funding After Publication
- 43- PRFA-P-8 the Deanship of Scientific Research, Princess Nourah bint Abdulrahman University, through the Program of Research Project Funding After Publication
Collapse
Affiliation(s)
- Amal Alotaibi
- Basic Science Department, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | | | | | - Sumana Mandarthi
- Mbiomics LLC, 16192 Coastal Highway, Lewes, DE, 19958, USA
- Meta Biosciences Pvt Ltd, Manipal-GOK Bioincubator, Manipal, India
| | - Nidhi Jayendra
- Mbiomics LLC, 16192 Coastal Highway, Lewes, DE, 19958, USA
| | - Asna Tungekar
- Mbiomics LLC, 16192 Coastal Highway, Lewes, DE, 19958, USA
| | - B V Lavanya
- Mbiomics LLC, 16192 Coastal Highway, Lewes, DE, 19958, USA
| | - Ashok Kumar Bhagavath
- Department of Cellular and Molecular Biology, University of Texas Health Science Center, Tyler, TX, USA
| | - Mary Anne Wong Cordero
- Basic Science Department, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Janne Pitkaniemi
- Finnish Cancer Registry, Unioninkatu 22, 00130, Helsinki, Finland
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Shaik Kalimulla Niazi
- Department of Preparatory Health Sciences, Riyadh Elm University, Riyadh, Saudi Arabia
| | - Raghavendra Upadhya
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, India
| | - Asmatanzeem Bepari
- Basic Science Department, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.
| | - Prashantha Hebbar
- Mbiomics LLC, 16192 Coastal Highway, Lewes, DE, 19958, USA.
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, India.
- Meta Biosciences Pvt Ltd, Manipal-GOK Bioincubator, Manipal, India.
| |
Collapse
|
8
|
Soós AÁ, Kelemen A, Orosz A, Szvicsek Z, Tölgyes T, Dede K, Bursics A, Wiener Z. High CD142 Level Marks Tumor-Promoting Fibroblasts with Targeting Potential in Colorectal Cancer. Int J Mol Sci 2023; 24:11585. [PMID: 37511344 PMCID: PMC10381019 DOI: 10.3390/ijms241411585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Colorectal cancer (CRC) has a high incidence and is one of the leading causes of cancer-related death. The accumulation of cancer-associated fibroblasts (CAF) induces an aggressive, stem-like phenotype in tumor cells, and it indicates a poor prognosis. However, cellular heterogeneity among CAFs and the targeting of both stromal and CRC cells are not yet well resolved. Here, we identified CD142high fibroblasts with a higher stimulating effect on CRC cell proliferation via secreting more hepatocyte growth factor (HGF) compared to CD142low CAFs. We also found that combinations of inhibitors that had either a promising effect in other cancer types or are more active in CRC compared to normal colonic epithelium acted synergistically in CRC cells. Importantly, heat shock protein 90 (HSP90) inhibitor selected against CD142high fibroblasts, and both CRC cells and CAFs were sensitive to a BCL-xL inhibitor. However, targeting mitogen-activated protein kinase kinase (MEK) was ineffective in fibroblasts, and an epigenetic inhibitor selected for a tumor cell population with markers of aggressive behavior. Thus, we suggest BCL-xL and HSP90 inhibitors to eliminate cancer cells and decrease the tumor-promoting CD142high CAF population. This may be the basis of a strategy to target both CRC cells and stromal fibroblasts, resulting in the inhibition of tumor relapse.
Collapse
Affiliation(s)
- András Áron Soós
- Department of Genetics, Cell and Immunobiology, Semmelweis University, H-1089 Budapest, Hungary; (A.Á.S.); (A.K.); (A.O.); (Z.S.)
| | - Andrea Kelemen
- Department of Genetics, Cell and Immunobiology, Semmelweis University, H-1089 Budapest, Hungary; (A.Á.S.); (A.K.); (A.O.); (Z.S.)
| | - Adrián Orosz
- Department of Genetics, Cell and Immunobiology, Semmelweis University, H-1089 Budapest, Hungary; (A.Á.S.); (A.K.); (A.O.); (Z.S.)
| | - Zsuzsanna Szvicsek
- Department of Genetics, Cell and Immunobiology, Semmelweis University, H-1089 Budapest, Hungary; (A.Á.S.); (A.K.); (A.O.); (Z.S.)
| | - Tamás Tölgyes
- Uzsoki Teaching Hospital, H-1145 Budapest, Hungary; (T.T.); (K.D.); (A.B.)
| | - Kristóf Dede
- Uzsoki Teaching Hospital, H-1145 Budapest, Hungary; (T.T.); (K.D.); (A.B.)
| | - Attila Bursics
- Uzsoki Teaching Hospital, H-1145 Budapest, Hungary; (T.T.); (K.D.); (A.B.)
| | - Zoltán Wiener
- Department of Genetics, Cell and Immunobiology, Semmelweis University, H-1089 Budapest, Hungary; (A.Á.S.); (A.K.); (A.O.); (Z.S.)
| |
Collapse
|
9
|
Alotaibi A, Gadekar VP, Gundla PS, Mandarthi S, Ravi S, Mallya D, Tungekar A, Lavanya BV, Bhagavath AK, Cordero MW, Pitkaniemi J, Seetharam RN, Bepari A, Hebbar P. A comprehensive analysis of mRNA expression profiles of Esophageal Squamous Cell Carcinoma reveals downregulation of Desmoglein 1 and crucial genomic targets. Cancer Biomark 2023; 38:465-487. [PMID: 38073377 DOI: 10.3233/cbm-230145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
AIM Esophageal Squamous Cell Carcinoma (ESCC) is a histological subtype of esophageal cancer that begins in the squamous cells in the esophagus. In only 19% of the ESCC-diagnosed patients, a five-year survival rate has been seen. This necessitates the identification of high-confidence biomarkers for early diagnosis, prognosis, and potential therapeutic targets for the mitigation of ESCC. METHOD We performed a meta-analysis of 10 mRNA datasets and identified consistently perturbed genes across the studies. Then, integrated with ESCC ATLAS to segregate 'core' genes to identify consequences of primary gene perturbation events leading to gene-gene interactions and dysregulated molecular signaling pathways. Further, by integrating with toxicogenomics data, inferences were drawn for gene interaction with environmental exposures, trace elements, chemical carcinogens, and drug chemicals. We also deduce the clinical outcomes of candidate genes based on survival analysis using the ESCC related dataset in The Cancer Genome Atlas. RESULT We identified 237 known and 18 novel perturbed candidate genes. Desmoglein 1 (DSG1) is one such gene that we found significantly downregulated (Fold Change =-1.89, p-value = 8.2e-06) in ESCC across six different datasets. Further, we identified 31 'core' genes (that either harbor genetic variants or are regulated by epigenetic modifications) and found regulating key biological pathways via adjoining genes in gene-gene interaction networks. Functional enrichment analysis showed dysregulated biological processes and pathways including "Extracellular matrix", "Collagen trimmer" and "HPV infection" are significantly overrepresented in our candidate genes. Based on the toxicogenomic inferences from Comparative Toxicogenomics Database we report the key genes that interacted with risk factors such as tobacco smoking, zinc, nitroso benzylmethylamine, and drug chemicals such as cisplatin, Fluorouracil, and Mitomycin in relation to ESCC. We also point to the STC2 gene that shows a high risk for mortality in ESCC patients. CONCLUSION We identified novel perturbed genes in relation to ESCC and explored their interaction network. DSG1 is one such gene, its association with microbiota and a clinical presentation seen commonly with ESCC hints that it is a good candidate for early diagnostic marker. Besides, in this study we highlight candidate genes and their molecular connections to risk factors, biological pathways, drug chemicals, and the survival probability of ESCC patients.
Collapse
Affiliation(s)
- Amal Alotaibi
- Basic Science Department, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
- Basic Science Department, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Veerendra P Gadekar
- Mbiomics LLC, Lewes DE, USA
- Basic Science Department, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | | | | | | | | | | | | | - Ashok Kumar Bhagavath
- Department of Cellular and Molecular Biology, University of Texas Health Science Center, Tyler, Texas, TX, USA
| | - MaryAnne Wong Cordero
- Basic Science Department, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Janne Pitkaniemi
- Finnish Cancer Registry, Helsinki, Finland
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Raviraja N Seetharam
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, India
| | - Asmatanzeem Bepari
- Basic Science Department, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Prashantha Hebbar
- Mbiomics LLC, Lewes DE, USA
- Manipal Center for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, India
- Meta Biosciences Pvt Ltd., Manipal - GOK Bioincubator, Advanced Research Center, Manipal, India
| |
Collapse
|
10
|
Rizzolio S, Giordano S, Corso S. The importance of being CAFs (in cancer resistance to targeted therapies). J Exp Clin Cancer Res 2022; 41:319. [PMID: 36324182 PMCID: PMC9632140 DOI: 10.1186/s13046-022-02524-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/23/2022] [Indexed: 05/09/2023] Open
Abstract
In the last two decades, clinical oncology has been revolutionized by the advent of targeted drugs. However, the efficacy of these therapies is significantly limited by primary and acquired resistance, that relies not only on cell-autonomous mechanisms but also on tumor microenvironment cues. Cancer-associated fibroblasts (CAFs) are extremely plastic cells of the tumor microenvironment. They not only produce extracellular matrix components that build up the structure of tumor stroma, but they also release growth factors, chemokines, exosomes, and metabolites that affect all tumor properties, including response to drug treatment. The contribution of CAFs to tumor progression has been deeply investigated and reviewed in several works. However, their role in resistance to anticancer therapies, and in particular to molecular therapies, has been largely overlooked. This review specifically dissects the role of CAFs in driving resistance to targeted therapies and discusses novel CAF targeted therapeutic strategies to improve patient survival.
Collapse
Affiliation(s)
| | - Silvia Giordano
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
- Department of Oncology, University of Torino, Torino, Italy
| | - Simona Corso
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
- Department of Oncology, University of Torino, Torino, Italy.
| |
Collapse
|
11
|
Bennett AN, Huang RX, He Q, Lee NP, Sung WK, Chan KHK. Drug repositioning for esophageal squamous cell carcinoma. Front Genet 2022; 13:991842. [PMID: 36246638 PMCID: PMC9554346 DOI: 10.3389/fgene.2022.991842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Esophageal cancer (EC) remains a significant challenge globally, having the 8th highest incidence and 6th highest mortality worldwide. Esophageal squamous cell carcinoma (ESCC) is the most common form of EC in Asia. Crucially, more than 90% of EC cases in China are ESCC. The high mortality rate of EC is likely due to the limited number of effective therapeutic options. To increase patient survival, novel therapeutic strategies for EC patients must be devised. Unfortunately, the development of novel drugs also presents its own significant challenges as most novel drugs do not make it to market due to lack of efficacy or safety concerns. A more time and cost-effective strategy is to identify existing drugs, that have already been approved for treatment of other diseases, which can be repurposed to treat EC patients, with drug repositioning. This can be achieved by comparing the gene expression profiles of disease-states with the effect on gene-expression by a given drug. In our analysis, we used previously published microarray data and identified 167 differentially expressed genes (DEGs). Using weighted key driver analysis, 39 key driver genes were then identified. These driver genes were then used in Overlap Analysis and Network Analysis in Pharmomics. By extracting drugs common to both analyses, 24 drugs are predicted to demonstrate therapeutic effect in EC patients. Several of which have already been shown to demonstrate a therapeutic effect in EC, most notably Doxorubicin, which is commonly used to treat EC patients, and Ixazomib, which was recently shown to induce apoptosis and supress growth of EC cell lines. Additionally, our analysis predicts multiple psychiatric drugs, including Venlafaxine, as repositioned drugs. This is in line with recent research which suggests that psychiatric drugs should be investigated for use in gastrointestinal cancers such as EC. Our study shows that a drug repositioning approach is a feasible strategy for identifying novel ESCC therapies and can also improve the understanding of the mechanisms underlying the drug targets.
Collapse
Affiliation(s)
- Adam N. Bennett
- Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Rui Xuan Huang
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qian He
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Nikki P. Lee
- Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Wing-Kin Sung
- Department of Computer Sciences, National University of Singapore, Singapore, Singapore
| | - Kei Hang Katie Chan
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Epidemiology, Centre for Global Cardiometabolic Health, Brown University, Providence, RI, United States
| |
Collapse
|
12
|
Hassan MS, Cwidak N, Awasthi N, von Holzen U. Cytokine Interaction With Cancer-Associated Fibroblasts in Esophageal Cancer. Cancer Control 2022; 29:10732748221078470. [PMID: 35442094 PMCID: PMC9024076 DOI: 10.1177/10732748221078470] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Esophageal cancer (EC) is a highly aggressive cancer with poor outcomes under current treatment regimens. More recent findings suggest stroma elements, specifically cancer-associated fibroblasts (CAFs), play a role in disease occurrence and progression. Cancer-associated fibroblasts are largely the product of converted fibroblasts, but a variety of other local cell types including epithelial cells, endothelial cells, and mesenchymal cells have also been shown to transform to CAFs under the correct conditions. Cancer-associated fibroblasts primarily function in the communication between the tumor microenvironment and cancer cells via cytokine and chemokine secretions that accentuate immunosuppression and cancer growth. Cancer-associated fibroblasts also pose issues for EC treatment by contributing to resistance of current chemotherapeutics like cisplatin. Targeting this cell type directly proves difficult given the heterogeneity between CAFs subpopulations, but emerging research provides hope that treatment is on the horizon. This review aims to unravel some of the complexities surrounding CAFs’ impact on EC growth and therapy.
Collapse
Affiliation(s)
- Md Sazzad Hassan
- Department of Surgery, 158720Indiana University School of Medicine, South Bend, IN 46617, USA.,Harper Cancer Research Institute, South Bend, IN 46617, USA
| | - Nicholas Cwidak
- Department of Surgery, 158720Indiana University School of Medicine, South Bend, IN 46617, USA
| | - Niranjan Awasthi
- Department of Surgery, 158720Indiana University School of Medicine, South Bend, IN 46617, USA.,Harper Cancer Research Institute, South Bend, IN 46617, USA
| | - Urs von Holzen
- Department of Surgery, 158720Indiana University School of Medicine, South Bend, IN 46617, USA.,Harper Cancer Research Institute, South Bend, IN 46617, USA.,Goshen Center for Cancer Care, Goshen, Goshen, IN 46526, USA.,University of Basel, Basel, Switzerland
| |
Collapse
|
13
|
FGF/FGFR-Dependent Molecular Mechanisms Underlying Anti-Cancer Drug Resistance. Cancers (Basel) 2021; 13:cancers13225796. [PMID: 34830951 PMCID: PMC8616288 DOI: 10.3390/cancers13225796] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/13/2021] [Accepted: 11/16/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Deregulation of the FGF/FGFR axis is associated with many types of cancer and contributes to the development of chemoresistance, limiting the effectiveness of current treatment strategies. There are several mechanisms involved in this phenomenon, including cross-talks with other signaling pathways, avoidance of apoptosis, stimulation of angiogenesis, and initiation of EMT. Here, we provide an overview of current research and approaches focusing on targeting components of the FGFR/FGF signaling module to overcome drug resistance during anti-cancer therapy. Abstract Increased expression of both FGF proteins and their receptors observed in many cancers is often associated with the development of chemoresistance, limiting the effectiveness of currently used anti-cancer therapies. Malfunctioning of the FGF/FGFR axis in cancer cells generates a number of molecular mechanisms that may affect the sensitivity of tumors to the applied drugs. Of key importance is the deregulation of cell signaling, which can lead to increased cell proliferation, survival, and motility, and ultimately to malignancy. Signaling pathways activated by FGFRs inhibit apoptosis, reducing the cytotoxic effect of some anti-cancer drugs. FGFRs-dependent signaling may also initiate angiogenesis and EMT, which facilitates metastasis and also correlates with drug resistance. Therefore, treatment strategies based on FGF/FGFR inhibition (using receptor inhibitors, ligand traps, monoclonal antibodies, or microRNAs) appear to be extremely promising. However, this approach may lead to further development of resistance through acquisition of specific mutations, metabolism switching, and molecular cross-talks. This review brings together information on the mechanisms underlying the involvement of the FGF/FGFR axis in the generation of drug resistance in cancer and highlights the need for further research to overcome this serious problem with novel therapeutic strategies.
Collapse
|
14
|
DW14383 is an irreversible pan-FGFR inhibitor that suppresses FGFR-dependent tumor growth in vitro and in vivo. Acta Pharmacol Sin 2021; 42:1498-1506. [PMID: 33288861 PMCID: PMC8379184 DOI: 10.1038/s41401-020-00567-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/08/2020] [Indexed: 12/23/2022]
Abstract
Fibroblast growth factor receptor (FGFR) is a promising anticancer target. Currently, most FGFR inhibitors lack sufficient selectivity and have nonnegligible activity against kinase insert domain receptor (KDR), limiting their feasibility due to the serious side effects. Notably, compensatory activation occurs among FGFR1-4, suggesting the urgent need to develop selective pan-FGFR1-4 inhibitors. Here, we explored the antitumor activity of DW14383, a novel irreversible FGFR1-4 inhibitor. DW14383 exhibited equivalently high potent inhibition against FGFR1, 2, 3 and 4, with IC50 values of less than 0.3, 1.1, less than 0.3, and 0.5 nmol/L, respectively. It is a selective FGFR inhibitor, exhibiting more than 1100-fold selectivity for FGFR1 over recombinant KDR, making it one of the most selective FGFR inhibitors over KDR described to date. Furthermore, DW14383 significantly inhibited cellular FGFR1-4 signaling, inducing G1/S cell cycle arrest, which in turn antagonized FGFR-dependent tumor cell proliferation. In contrast, DW14383 had no obvious antiproliferative effect against cancer cell lines without FGFR aberration, further confirming its selectivity against FGFR. In representative FGFR-dependent xenograft models, DW14383 oral administration substantially suppressed tumor growth by simultaneously inhibiting tumor proliferation and angiogenesis via inhibiting FGFR signaling. In summary, DW14383 is a promising selective irreversible pan-FGFR inhibitor with pan-tumor spectrum potential in FGFR1-4 aberrant cancers, which has the potential to overcome compensatory activation among FGFR1-4.
Collapse
|
15
|
Li S. Anlotinib: A Novel Targeted Drug for Bone and Soft Tissue Sarcoma. Front Oncol 2021; 11:664853. [PMID: 34094958 PMCID: PMC8173120 DOI: 10.3389/fonc.2021.664853] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Bone and soft tissue sarcomas account for approximately 15% of pediatric solid malignant tumors and 1% of adult solid malignant tumors. There are over 50 subtypes of sarcomas, each of which is notably heterogeneous and manifested by remarkable phenotypic and morphological variability. Anlotinib is a novel oral tyrosine kinase inhibitor (TKI) targeting c-kit, platelet-derived growth factor receptors, fibroblast growth factor receptor, and vascular endothelial growth factor receptor. In comparison with the placebo, anlotinib was associated with better overall survival and progression-free survival (PFS) in a phase III trial of patients with advanced non-small cell lung cancer (NSCLC), albeit with cancer progression after two previous lines of treatment. Recently, the National Medical Products Administration approved anlotinib monotherapy as a third-line treatment for patients with advanced NSCLC. Additionally, a phase IIB randomized trial substantiated that anlotinib is associated with a significant longer median PFS in patients with advanced soft tissue sarcoma. Moreover, anlotinib is also effective in patients with advanced medullary thyroid carcinoma and metastatic renal cell carcinoma. Anlotinib has similar tolerability to other TKIs targeting vascular endothelial growth factor receptors and other tyrosine kinase-mediated pathways. However, anlotinib has a notably lower rate of side effects ≥grade 3 relative to sunitinib. This review discussed the remarkable characteristics and major dilemmas of anlotinib as a targeted therapy for sarcomas.
Collapse
Affiliation(s)
- Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China.,Department of Tissue Engineering, Center of 3D Printing & Organ Manufacturing, School of Fundamental Sciences, China Medical University (CMU), Shenyang, China
| |
Collapse
|
16
|
Luan S, Zeng X, Zhang C, Qiu J, Yang Y, Mao C, Xiao X, Zhou J, Zhang Y, Yuan Y. Advances in Drug Resistance of Esophageal Cancer: From the Perspective of Tumor Microenvironment. Front Cell Dev Biol 2021; 9:664816. [PMID: 33816512 PMCID: PMC8017339 DOI: 10.3389/fcell.2021.664816] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 02/28/2021] [Indexed: 02/05/2023] Open
Abstract
Drug resistance represents the major obstacle to get the maximum therapeutic benefit for patients with esophageal cancer since numerous patients are inherently or adaptively resistant to therapeutic agents. Notably, increasing evidence has demonstrated that drug resistance is closely related to the crosstalk between tumor cells and the tumor microenvironment (TME). TME is a dynamic and ever-changing complex biological network whose diverse cellular and non-cellular components influence hallmarks and fates of tumor cells from the outside, and this is responsible for the development of resistance to conventional therapeutic agents to some extent. Indeed, the formation of drug resistance in esophageal cancer should be considered as a multifactorial process involving not only cancer cells themselves but cancer stem cells, tumor-associated stromal cells, hypoxia, soluble factors, extracellular vesicles, etc. Accordingly, combination therapy targeting tumor cells and tumor-favorable microenvironment represents a promising strategy to address drug resistance and get better therapeutic responses for patients with esophageal cancer. In this review, we mainly focus our discussion on molecular mechanisms that underlie the role of TME in drug resistance in esophageal cancer. We also discuss the opportunities and challenges for therapeutically targeting tumor-favorable microenvironment, such as membrane proteins, pivotal signaling pathways, and cytokines, to attenuate drug resistance in esophageal cancer.
Collapse
Affiliation(s)
- Siyuan Luan
- Department of Thoracic Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxi Zeng
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chao Zhang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiajun Qiu
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yushang Yang
- Department of Thoracic Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Chengyi Mao
- Department of Thoracic Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Xiao
- Department of Thoracic Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jianfeng Zhou
- Department of Thoracic Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yonggang Zhang
- Department of Periodical Press, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Nursing Key Laboratory of Sichuan Province, Chengdu, China
| | - Yong Yuan
- Department of Thoracic Surgery, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Recent advances of dual FGFR inhibitors as a novel therapy for cancer. Eur J Med Chem 2021; 214:113205. [PMID: 33556787 DOI: 10.1016/j.ejmech.2021.113205] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/06/2021] [Accepted: 01/13/2021] [Indexed: 12/14/2022]
Abstract
Fibroblast growth factor receptor (FGFR) includes four highly conserved transmembrane receptor tyrosine kinases (FGFR1-4). FGF and FGFR regulate many biological processes, such as angiogenesis, wound healing and tissue regeneration. The abnormal expression of FGFR is related to the tumorigenesis, tumor progression and drug resistance of anti-tumor treatments in many types of tumors. Nowadays there are many anti-cancer drugs targeting FGFR. However, traditional single-target anti-tumor drugs are easy to acquire drug resistance. The therapeutic effect can be enhanced by simultaneously inhibiting FGFR and another target (such as VEGFR, EGFR, PI3K, CSF-1R, etc.). We know drug combination can bring problems such as drug interactions. Therefore, the development of FGFR dual target inhibitors is an important direction. In this paper, we reviewed the research on dual FGFR inhibitors in recent years and made brief comments on them.
Collapse
|
18
|
Alternative splicing modulates cancer aggressiveness: role in EMT/metastasis and chemoresistance. Mol Biol Rep 2021; 48:897-914. [PMID: 33400075 DOI: 10.1007/s11033-020-06094-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022]
Abstract
Enhanced metastasis and disease recurrence accounts for the high mortality rates associated with cancer. The process of Epithelial-Mesenchymal Transition (EMT) contributes towards the augmentation of cancer invasiveness along with the gain of stem-like and the subsequent drug-resistant behavior. Apart from the well-established transcriptional regulation, EMT is also controlled post-transcriptionally by virtue of alternative splicing (AS). Numerous genes including Fibroblast Growth Factor receptor (FGFR) as well as CD44 are differentially spliced during this trans-differentiation process which, in turn, governs cancer progression. These splicing alterations are controlled by various splicing factors including ESRP, RBFOX2 as well as hnRNPs. Here, we have depicted the mechanisms governing the splice isoform switching of FGFR and CD44. Moreover, the role of the splice variants generated by AS of these gene transcripts in modulating the metastatic potential and stem-like/chemoresistant behavior of cancer cells has also been highlighted. Additionally, the involvement of splicing factors in regulating EMT/invasiveness along with drug-resistance as well as the metabolic properties of the cells has been emphasized. Tumorigenesis is accompanied by a remodeling of the cellular splicing profile generating diverse protein isoforms which, in turn, control the cancer-associated hallmarks. Therefore, we have also briefly discussed about a wide variety of genes which are differentially spliced in the tumor cells and promote cancer progression. We have also outlined different strategies for targeting the tumor-associated splicing events which have shown promising results and therefore this approach might be useful in developing therapies to reduce cancer aggressiveness in a more specific manner.
Collapse
|
19
|
Zhang N, Shi J, Shi X, Chen W, Liu J. Mutational Characterization and Potential Prognostic Biomarkers of Chinese Patients with Esophageal Squamous Cell Carcinoma. Onco Targets Ther 2020; 13:12797-12809. [PMID: 33363385 PMCID: PMC7751839 DOI: 10.2147/ott.s275688] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose Esophageal squamous cell carcinoma (ESCC) is the most common type of esophageal cancer in China and the 5-year mortality rate is up to 70%. Studies on the ESCC genetic landscape are needed to further explore clinical therapeutic strategies. In this study, we evaluated the genetic landscape of ESCC to aid the search for clinical therapeutic strategies. Patients and Methods A total of 225 ESCC patients were enrolled in this study. Deep sequencing of 450 cancer genes was performed on formalin-fixed paraffin-embedded tumor biopsies and matched blood samples from patients. Tumor mutational burden (TMB) was calculated using an algorithm developed in-house. Results Our results showed that the most commonly mutated genes in ESCC were TP53 (96%), CCND1 (46%), FGF4 (44%), FGF19 (44%), FGF3 (44%), CDKN2A (31%), PIK3CA (26%), NOTCH1 (24%), KMT2D (18%), FAT1 (16%), and LRP1B (16%). We found that TMB correlated with patient drinking status. We identified mutations associated with sex, early ESCC, high TMB, and metastasis lymph nodes. KMT2D mutations associated with sex (P = 0.035), tumor stage (P = 0.016), high TMB (P = 0.0072), and overall survival of patients (P = 0.0026). SPEN mutations associated with high TMB (P = 0.0016) and metastasis-positive lymph nodes (P = 0.027). These results suggested that SPEN and KMT2D could be potential prognosis biomarkers for Chinese patients with ESCC. We also found that the number of positive lymph nodes was associated with disease-free survival. Clinical target gene analysis indicated that nearly half of Chinese ESCC patients might benefit from treatment with gene-specific target drugs. Conclusion Our study revealed the ESCC mutational landscape in 225 Chinese patients and uncovered the potential prognosis biomarker for Chinese patients with ESCC.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Junping Shi
- Department of Medicine, OrigiMed Co. Ltd, Shanghai, People's Republic of China
| | - Xiaoliang Shi
- Department of Medicine, OrigiMed Co. Ltd, Shanghai, People's Republic of China
| | - Wenting Chen
- Department of Medicine, OrigiMed Co. Ltd, Shanghai, People's Republic of China
| | - Junfeng Liu
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| |
Collapse
|
20
|
Gao Y, Liu P, Shi R. Anlotinib as a molecular targeted therapy for tumors. Oncol Lett 2020; 20:1001-1014. [PMID: 32724339 PMCID: PMC7377159 DOI: 10.3892/ol.2020.11685] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/30/2020] [Indexed: 12/24/2022] Open
Abstract
Angiogenesis has an essential role in tumor growth and metastasis, and blocking this pathway has been a successfully utilized strategy in the clinical treatment of cancer. Anlotinib (AL3818) is a novel oral receptor tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor 2 and 3, fibroblast growth factor 1-4, platelet-derived growth factor receptor α and β, c-Kit and Ret. Anlotinib exerts inhibitory effects on tumor growth and angiogenesis and received its first approval as a third-line treatment for refractory advanced non-small-cell lung cancer in May 2018 and its second approval as a second-line treatment for advanced soft-tissue sarcoma in June 2019 in the People's Republic of China. Anlotinib has encouraging efficacy and a manageable and tolerable safety profile in a broad range of malignancies, including medullary thyroid cancer, renal cell cancer, gastric cancer and esophageal squamous cell carcinoma. In the present review, the preclinical and clinical trials of anlotinib were summarized with a focus on safety evaluation and adverse event management.
Collapse
Affiliation(s)
- Yi Gao
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
- Department of Gastroenterology, The Jiangyin Clinical College of Xuzhou Medical University, Jiangyin, Jiangsu 214400, P.R. China
- Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Pengfei Liu
- Department of Gastroenterology, The Jiangyin Clinical College of Xuzhou Medical University, Jiangyin, Jiangsu 214400, P.R. China
- State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Ruihua Shi
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
- Department of Gastroenterology, Zhongda Hospital, Affiliated Hospital of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
21
|
Li X, Ren Z, Xiong C, Geng J, Li Y, Liu C, Ren C, Liu H. Minichromosome maintenance 6 complex component identified by bioinformatics analysis and experimental validation in esophageal squamous cell carcinoma. Oncol Rep 2020; 44:987-1002. [PMID: 32583000 PMCID: PMC7388536 DOI: 10.3892/or.2020.7658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC), the main subtype of esophageal cancer (EC), is a common lethal type of cancer with a high mortality rate. The aim of the present study was to select key relevant genes and identify potential mechanisms involved in the development of ESCC based on bioinformatics analysis. Minichromosome maintenance 6 complex component (MCM6) has been identified to be upregulated in multiple malignancies; however, its contributions to ESCC remain unclear. For the purposes of the present study, four datasets were downloaded from the Gene Expression Omnibus (GSE63941, GSE26886, GSE17351 and GSE77861), and the intersection of the differentially expressed genes was obtained using a Venn diagram. The protein‑protein interaction was then constructed, and the modules were verified by Cytoscape, in which the key genes have a high connectivity degree with other genes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway were subsequently filtered out to analyze the development of ESCC. MCM6, an upregulated gene, was selected and connected with most of the other genes, for further research validation. The expression levels of MCM6 were then assessed using the Oncomine, GEPIA and UALCAN databases and validated in both ESCC tissues samples and cell lines by immunohistochemistry and RT‑qPCR. Cell counting kit‑8 (CCK‑8), flow cytometry, wound healing and Transwell assays were used to determine the proliferation, apoptosis, cell cycle, migration and invasion of ESCC cells. A total of 24 genes were identified by a series of bioinformatics analyses and the results revealed that the genes were associated with DNA replication and cell cycle. Experimental validation revealed that MCM6 expression was significantly elevated in both ESCC tissues and cell lines. The results were consistent with those of bioinformatics analysis. Furthermore, the knockdown of MCM6 inhibited cell proliferation, migration and invasion and promoted cell apoptosis, and made cells arrested in S stage. In summary, the findings of bioinformatics analysis provided a novel hypothesis for ESCC progression. In particular, the aberrantly elevated expression of MCM6 is a potential biomarker for ESCC diagnosis and treatment.
Collapse
Affiliation(s)
- Xuebing Li
- Department of Medical Laboratory, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhenzhen Ren
- Department of Medical Laboratory, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chao Xiong
- Department of Medical Laboratory, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Jie Geng
- Department of Medical Laboratory, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yuqing Li
- Department of Medical Laboratory, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450000, P.R. China
| | - Cong Liu
- Department of Medical Laboratory, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chunfeng Ren
- Department of Medical Laboratory, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hongchun Liu
- Department of Medical Laboratory, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
22
|
Fernández-Nogueira P, Mancino M, Fuster G, Bragado P, Prats de Puig M, Gascón P, Casado FJ, Carbó N. Breast Mammographic Density: Stromal Implications on Breast Cancer Detection and Therapy. J Clin Med 2020; 9:jcm9030776. [PMID: 32178425 PMCID: PMC7141321 DOI: 10.3390/jcm9030776] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/21/2022] Open
Abstract
Current evidences state clear that both normal development of breast tissue as well as its malignant progression need many-sided local and systemic communications between epithelial cells and stromal components. During development, the stroma, through remarkably regulated contextual signals, affects the fate of the different mammary cells regarding their specification and differentiation. Likewise, the stroma can generate tumour environments that facilitate the neoplastic growth of the breast carcinoma. Mammographic density has been described as a risk factor in the development of breast cancer and is ascribed to modifications in the composition of breast tissue, including both stromal and glandular compartments. Thus, stroma composition can dramatically affect the progression of breast cancer but also its early detection since it is mainly responsible for the differences in mammographic density among individuals. This review highlights both the pathological and biological evidences for a pivotal role of the breast stroma in mammographic density, with particular emphasis on dense and malignant stromas, their clinical meaning and potential therapeutic implications for breast cancer patients.
Collapse
Affiliation(s)
- Patricia Fernández-Nogueira
- Institut d’Investigacions Biomèdiques Augustí Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Correspondence: (P.F.-N.); (M.M.)
| | - Mario Mancino
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain
- Department of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Correspondence: (P.F.-N.); (M.M.)
| | - Gemma Fuster
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain
- Department of Biochemistry & Physiology, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- Department of Biosciences, Faculty of Sciences and Technology, University of Vic, 08500 Vic, Spain
| | - Paloma Bragado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Health Research Institute of the Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Miquel Prats de Puig
- Department of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Breast Committee, Hospital El Pilar, Quirón salud Group, 08006 Barcelona, Spain
| | - Pere Gascón
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain
- Oncology and Multidisciplinary Knowledge, 08036 Barcelona, Spain
| | - Francisco Javier Casado
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain
| | - Neus Carbó
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain
| |
Collapse
|
23
|
Zhan H, Tu S, Zhang F, Shao A, Lin J. MicroRNAs and Long Non-coding RNAs in c-Met-Regulated Cancers. Front Cell Dev Biol 2020; 8:145. [PMID: 32219093 PMCID: PMC7078111 DOI: 10.3389/fcell.2020.00145] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/21/2020] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are components of many signaling pathways associated with tumor aggressiveness and cancer metastasis. Some lncRNAs are classified as competitive endogenous RNAs (ceRNAs) that bind to specific miRNAs to prevent interaction with target mRNAs. Studies have shown that the hepatocyte growth factor/mesenchymal-epithelial transition factor (HGF/c-Met) pathway is involved in physiological and pathological processes such as cell growth, angiogenesis, and embryogenesis. Overexpression of c-Met can lead to sustained activation of downstream signals, resulting in carcinogenesis, metastasis, and resistance to targeted therapies. In this review, we evaluated the effects of anti-oncogenic and oncogenic non-coding RNAs (ncRNAs) on c-Met, and the interactions among lncRNAs, miRNAs, and c-Met in cancer using clinical and tissue chromatin immunoprecipition (ChIP) analysis data. We summarized current knowledge of the mechanisms and effects of the lncRNAs/miR-34a/c-Met axis in various tumor types, and evaluated the potential therapeutic value of lncRNAs and/or miRNAs targeted to c-Met on drug-resistance. Furthermore, we discussed the functions of lncRNAs and miRNAs in c-Met-related carcinogenesis and potential therapeutic strategies.
Collapse
Affiliation(s)
- Hong Zhan
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Tu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Zhang
- School of Medicine, Zhejiang University Hangzhou, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Lin
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
24
|
Zhou Y, Wu C, Lu G, Hu Z, Chen Q, Du X. FGF/FGFR signaling pathway involved resistance in various cancer types. J Cancer 2020; 11:2000-2007. [PMID: 32127928 PMCID: PMC7052940 DOI: 10.7150/jca.40531] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 01/04/2020] [Indexed: 12/16/2022] Open
Abstract
Resistance becomes major clinical issue in cancer treatment, which strongly limits patients to benefit from oncotherapy. Growing evidences have been indicative of the critical role of fibroblast growth factor (FGF)/receptor (FGFR) signaling played in resistance to oncotherapy. In this review we discussed the underlying mechanisms of FGF/FGFR signaling mediated resistance to chemotherapy, radiotherapy and target therapy in various cancers. Meanwhile, we summarized the reported mechanism of FGF/FGFR inhibitors resistance in cancers.
Collapse
Affiliation(s)
- Yangyang Zhou
- Department of Rheumatology and Immunology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Chengyu Wu
- Department of Rheumatology and Immunology, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Guangrong Lu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical, Wenzhou, Zhejiang 325000, China)
| | - Zijing Hu
- College of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Qiuxiang Chen
- Department of Ultrasonic Imaging, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaojing Du
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| |
Collapse
|
25
|
Fernández-Nogueira P, Mancino M, Fuster G, López-Plana A, Jauregui P, Almendro V, Enreig E, Menéndez S, Rojo F, Noguera-Castells A, Bill A, Gaither LA, Serrano L, Recalde-Percaz L, Moragas N, Alonso R, Ametller E, Rovira A, Lluch A, Albanell J, Gascon P, Bragado P. Tumor-Associated Fibroblasts Promote HER2-Targeted Therapy Resistance through FGFR2 Activation. Clin Cancer Res 2019; 26:1432-1448. [DOI: 10.1158/1078-0432.ccr-19-0353] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 09/15/2019] [Accepted: 11/04/2019] [Indexed: 11/16/2022]
|
26
|
Peng X, Hou P, Chen Y, Dai Y, Ji Y, Shen Y, Su Y, Liu B, Wang Y, Sun D, Jiang Y, Zha C, Xie Z, Ding J, Geng M, Ai J. Preclinical evaluation of 3D185, a novel potent inhibitor of FGFR1/2/3 and CSF-1R, in FGFR-dependent and macrophage-dominant cancer models. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:372. [PMID: 31438996 PMCID: PMC6704710 DOI: 10.1186/s13046-019-1357-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/06/2019] [Indexed: 12/25/2022]
Abstract
Background The interaction between tumor cells and their immunosuppressive microenvironment promotes tumor progression and drug resistance. Thus, simultaneously targeting tumor cells and stromal cells is expected to have synergistic antitumor effects. Herein, we present for the first time a preclinical antitumor investigation of 3D185, a novel dual inhibitor targeting FGFRs, which are oncogenic drivers, and CSF-1R, which is the major survival factor for protumor macrophages. Methods The antitumor characteristics of 3D185 were assessed by a range of assays, including kinase profiling, cell viability, cell migration, immunoblotting, CD8+ T cell suppression, and in vivo antitumor efficacy, followed by flow cytometric and immunohistochemical analyses of tumor-infiltrating immune cells and endothelial cells in nude mice and immune-competent mice. Results 3D185 significantly inhibited the kinase activity of FGFR1/2/3 and CSF-1R, with equal potency and high selectivity over other kinases. 3D185 suppressed FGFR signaling and tumor cell growth in FGFR-driven models both in vitro and in vivo. In addition, 3D185 could inhibit the survival and M2-like polarization of macrophages, reversing the immunosuppressive effect of macrophages on CD8+ T cells as well as CSF1-differentiated macrophage induced-FGFR3-aberrant cancer cell migration. Furthermore, 3D185 inhibited tumor growth via remodeling the tumor microenvironment in TAM-dominated tumor models. Conclusions 3D185 is a promising antitumor candidate drug that simultaneously targets tumor cells and their immunosuppressive microenvironment and has therapeutic potential due to synergistic effects. Our study provides a solid foundation for the investigation of 3D185 in cancer patients, particularly in patients with aberrant FGFR and abundant macrophages, who respond poorly to classic pan-FGFRi treatment. Electronic supplementary material The online version of this article (10.1186/s13046-019-1357-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xia Peng
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Pengcong Hou
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yi Chen
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yang Dai
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yinchun Ji
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yanyan Shen
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yi Su
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Bo Liu
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yueliang Wang
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Deqiao Sun
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Yuchen Jiang
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Chuantao Zha
- Shanghai HaiHe Pharmaceutical Co., Ltd, No. 421 Newton Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, People's Republic of China
| | - Zuoquan Xie
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Jian Ding
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China.,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China
| | - Meiyu Geng
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China. .,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China.
| | - Jing Ai
- Division of Anti-Tumor Pharmacology, State Key Laboratory of Drug Research, Chinese Academy of Sciences, Shanghai Institute of Materia Medica, No. 555 Zuchongzhi Road, Pudong New District, Shanghai, 201203, People's Republic of China. .,University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Shijingshan District, Beijing, 100049, People's Republic of China.
| |
Collapse
|
27
|
Qu Y, Dou B, Tan H, Feng Y, Wang N, Wang D. Tumor microenvironment-driven non-cell-autonomous resistance to antineoplastic treatment. Mol Cancer 2019; 18:69. [PMID: 30927928 PMCID: PMC6441162 DOI: 10.1186/s12943-019-0992-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/28/2019] [Indexed: 12/24/2022] Open
Abstract
Drug resistance is of great concern in cancer treatment because most effective drugs are limited by the development of resistance following some periods of therapeutic administration. The tumor microenvironment (TME), which includes various types of cells and extracellular components, mediates tumor progression and affects treatment efficacy. TME-mediated drug resistance is associated with tumor cells and their pericellular matrix. Noninherent-adaptive drug resistance refers to a non-cell-autonomous mechanism in which the resistance lies in the treatment process rather than genetic or epigenetic changes, and this mechanism is closely related to the TME. A new concept is therefore proposed in which tumor cell resistance to targeted therapy may be due to non-cell-autonomous mechanisms. However, knowledge of non-cell-autonomous mechanisms of resistance to different treatments is not comprehensive. In this review, we outlined TME factors and molecular events involved in the regulation of non-cell-autonomous resistance of cancer, summarized how the TME contributes to non-cell-autonomous drug resistance in different types of antineoplastic treatment, and discussed the novel strategies to investigate and overcome the non-cell-autonomous mechanism of cancer non-cell-autonomous resistance.
Collapse
Affiliation(s)
- Yidi Qu
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Bo Dou
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Horyue Tan
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun, 130012, China.
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
28
|
Farrokhzadeh A, Akher FB, Olotu FA, Soliman MES, Van Heerden FR. Revealing the distinct mechanistic binding and activity of 5-(1-(3,5-dichloropyridin-4-yl)ethoxy)-3-(5-(4-methylpiperazin-1-yl)-1H-benzo[d]imidazol-2-yl)-1H-indazole enantiomers against FGFR1. Phys Chem Chem Phys 2019; 21:15120-15132. [DOI: 10.1039/c9cp02112d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The concept of chirality has become prominent over the years, particularly with regards to the design of therapeutic molecules.
Collapse
Affiliation(s)
- Abdolkarim Farrokhzadeh
- Molecular Bio-Computation and Drug Design Laboratory
- School of Health Sciences
- University of KwaZulu-Natal
- Westville Campus
- Durban 4001
| | - Farideh Badichi Akher
- Molecular Bio-Computation and Drug Design Laboratory
- School of Health Sciences
- University of KwaZulu-Natal
- Westville Campus
- Durban 4001
| | - Fisayo A. Olotu
- Molecular Bio-Computation and Drug Design Laboratory
- School of Health Sciences
- University of KwaZulu-Natal
- Westville Campus
- Durban 4001
| | - Mahmoud E. S. Soliman
- Molecular Bio-Computation and Drug Design Laboratory
- School of Health Sciences
- University of KwaZulu-Natal
- Westville Campus
- Durban 4001
| | - Fanie R. Van Heerden
- School of Chemistry and Physics
- University of KwaZulu-Natal
- Pietermaritzburg 3209
- South Africa
| |
Collapse
|
29
|
Badichi Akher F, Farrokhzadeh A, Olotu FA, Agoni C, Soliman MES. The irony of chirality – unveiling the distinct mechanistic binding and activities of 1-(3-(4-amino-5-(7-methoxy-5-methylbenzo[b]thiophen-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)pyrrolidin-1-yl)prop-2-en-1-one enantiomers as irreversible covalent FGFR4 inhibitors. Org Biomol Chem 2019; 17:1176-1190. [DOI: 10.1039/c8ob02811g] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Theoretical investigation of the effect of chirality on inhibitors is providing essential insights for drug design.
Collapse
Affiliation(s)
- Farideh Badichi Akher
- Molecular Bio-computation and Drug Design Laboratory
- School of Health Sciences
- University of KwaZulu-Natal
- Durban 4001
- South Africa
| | - Abdolkarim Farrokhzadeh
- Molecular Bio-computation and Drug Design Laboratory
- School of Health Sciences
- University of KwaZulu-Natal
- Durban 4001
- South Africa
| | - Fisayo A. Olotu
- Molecular Bio-computation and Drug Design Laboratory
- School of Health Sciences
- University of KwaZulu-Natal
- Durban 4001
- South Africa
| | - Clement Agoni
- Molecular Bio-computation and Drug Design Laboratory
- School of Health Sciences
- University of KwaZulu-Natal
- Durban 4001
- South Africa
| | - Mahmoud E. S. Soliman
- Molecular Bio-computation and Drug Design Laboratory
- School of Health Sciences
- University of KwaZulu-Natal
- Durban 4001
- South Africa
| |
Collapse
|
30
|
Wang Y, Li L, Fan J, Dai Y, Jiang A, Geng M, Ai J, Duan W. Discovery of Potent Irreversible Pan-Fibroblast Growth Factor Receptor (FGFR) Inhibitors. J Med Chem 2018. [PMID: 29522671 DOI: 10.1021/acs.jmedchem.7b01843] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Fibroblast growth factor receptors (FGFR1-4) are promising therapeutic targets in many cancers. With the resurgence of interest in irreversible inhibitors, efforts have been directed to the discovery of irreversible FGFR inhibitors. Currently, several selective irreversible inhibitors are being evaluated in clinical trials that could covalently target a conserved cysteine in the P-loop of FGFR. In this article, we used a structure-guided approach that is rationalized by a computer-aided simulation to discover the novel and irreversible pan-FGFR inhibitor, 9g, which provided superior FGFR in vitro activities and decent selectivity over VEGFR2 (vascular endothelia growth factor receptor 2). In in vivo studies, 9g displayed clear antitumor activities in NCI-H1581 and SNU-16 xenograft mice models. Additionally, the diluting method confirmed the irreversible binding of 9g to FGFR.
Collapse
Affiliation(s)
- Yuming Wang
- Department of Medicinal Chemistry , Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zu Chong Zhi Road , Shanghai 201203 , P. R. China.,University of Chinese Academy of Sciences , No.19A Yuquan Road , Beijing 100049 , P. R. China
| | - Lijun Li
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zu Chong Zhi Road , Shanghai 201203 , P. R. China.,University of Chinese Academy of Sciences , No.19A Yuquan Road , Beijing 100049 , P. R. China
| | - Jun Fan
- Department of Medicinal Chemistry , Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zu Chong Zhi Road , Shanghai 201203 , P. R. China.,University of Chinese Academy of Sciences , No.19A Yuquan Road , Beijing 100049 , P. R. China
| | - Yang Dai
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zu Chong Zhi Road , Shanghai 201203 , P. R. China.,University of Chinese Academy of Sciences , No.19A Yuquan Road , Beijing 100049 , P. R. China
| | - Alan Jiang
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zu Chong Zhi Road , Shanghai 201203 , P. R. China.,University of Chinese Academy of Sciences , No.19A Yuquan Road , Beijing 100049 , P. R. China
| | - Meiyu Geng
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zu Chong Zhi Road , Shanghai 201203 , P. R. China.,University of Chinese Academy of Sciences , No.19A Yuquan Road , Beijing 100049 , P. R. China
| | - Jing Ai
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research , Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zu Chong Zhi Road , Shanghai 201203 , P. R. China.,University of Chinese Academy of Sciences , No.19A Yuquan Road , Beijing 100049 , P. R. China
| | - Wenhu Duan
- Department of Medicinal Chemistry , Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences , 555 Zu Chong Zhi Road , Shanghai 201203 , P. R. China.,University of Chinese Academy of Sciences , No.19A Yuquan Road , Beijing 100049 , P. R. China
| |
Collapse
|
31
|
Tan HY, Wang N, Lam W, Guo W, Feng Y, Cheng YC. Targeting tumour microenvironment by tyrosine kinase inhibitor. Mol Cancer 2018; 17:43. [PMID: 29455663 PMCID: PMC5817793 DOI: 10.1186/s12943-018-0800-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 02/01/2018] [Indexed: 12/12/2022] Open
Abstract
Tumour microenvironment (TME) is a key determinant of tumour growth and metastasis. TME could be very different for each type and location of tumour and TME may change constantly during tumour growth. Multiple counterparts in surrounding microenvironment including mesenchymal-, hematopoietic-originated cells as well as non-cellular components affect TME. Thus, therapeutics that can disrupt the tumour-favouring microenvironment should be further explored for cancer therapy. Previous efforts in unravelling the dysregulated mechanisms of TME components has identified numerous protein tyrosine kinases, while its corresponding inhibitors have demonstrated potent modulatory effect on TME. Recent works have demonstrated that beyond the direct action on cancer cells, tyrosine kinase inhibitors (TKIs) have been implicated in inactivation or normalization of dysregulated TME components leading to cancer regression. Either through re-sensitizing the tumour cells or reversing the immunological tolerance microenvironment, the emergence of these TME modulatory mechanism of TKIs supports the combinatory use of TKIs with current chemotherapy or immunotherapy for cancer therapy. Therefore, an appropriate understanding on TME modulation by TKIs may offer another mode of action of TKIs for cancer treatment. This review highlights mode of kinase activation or paracrine ligand production from TME components and summarises the findings on the potential use of various TKIs on regulating TME components. At last, the combination use of current TKIs with immunotherapy in the perspectives of efficacy and safety are discussed.
Collapse
Affiliation(s)
- Hor-Yue Tan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Wing Lam
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Wei Guo
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, People's Republic of China.
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.
| | - Yung-Chi Cheng
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
32
|
Chen B, Liu S, Gan L, Wang J, Hu B, Xu H, Tong R, Yang H, Cristina I, Xue J, Hu X, Lu Y. FGFR1 signaling potentiates tumor growth and predicts poor prognosis in esophageal squamous cell carcinoma patients. Cancer Biol Ther 2017; 19:76-86. [PMID: 29257923 DOI: 10.1080/15384047.2017.1394541] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Fibroblast growth factor receptor-1 (FGFR1) over-expression was broadly found in squamous cancer, where it induced cellular proliferation, differentiation, and metastasis by activating various signaling pathway. However, the role of FGFR1 gene expression in predicting prognosis of Esophageal Squamous Cell Carcinoma (ESCC) and its regulatory function in the progression of ESCC are not well understood. Therefore, we performed an analysis of FGFR1 mRNA expression by quantitative RT-PCR in tumor tissue of 145 patients with ESCC. The relationships between FGFR1 gene expression and clinicopathological parameters, also the prognosis were further examined. Results suggested that higher FGFR1 gene expression predicted worse overall survival (HR = 1.502, 95%[CI] = 1.005-2.246, P = 0.045). Disease-free survival tends to be shorter in patients with higher FGFR1 expression but without statistical significance (HR = 1.398, 95%[CI] = 0.942-2.074, P = 0.096). FGFR1 was up regulated in multiple ESCC cell lines. Subsequent in vitro experiments demonstrated that anti-FGFR1 treatment by PD173074 inhibited TE-1 and EC9706 cell viability along with the attenuation of MEK-ERK signaling pathway. In vivo, PD173074 administration also had shown potent ESCC growth arresting effect. Overall, our study suggested that FGFR1 gene expression could be an independent prognosis predictive factor in patients with ESCC. Anti-FGFR1 inhibited ESCC growth and could be a potential strategy in ESCC targeted therapy.
Collapse
Affiliation(s)
- Baoqing Chen
- a Department of Thoracic Oncology , Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu , Sichuan , China.,b Huaxi Student Society of Oncology Research, West China School of Medicine, Sichuan University , Chengdu , Sichuan , China
| | - Shurui Liu
- a Department of Thoracic Oncology , Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Lu Gan
- c Laboratory of Anesthesiology & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Jingwen Wang
- a Department of Thoracic Oncology , Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Binbin Hu
- a Department of Thoracic Oncology , Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - He Xu
- a Department of Thoracic Oncology , Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Ruizhan Tong
- a Department of Thoracic Oncology , Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Hui Yang
- a Department of Thoracic Oncology , Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu , Sichuan , China.,b Huaxi Student Society of Oncology Research, West China School of Medicine, Sichuan University , Chengdu , Sichuan , China
| | - Ivan Cristina
- d Center for RNA Interference and Non-coding RNAs, The University of Texas MD Anderson Cancer Center , Houston , Texas , USA
| | - Jianxin Xue
- a Department of Thoracic Oncology , Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Xun Hu
- e Huaxi Biobank, West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - You Lu
- a Department of Thoracic Oncology , Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Chengdu , Sichuan , China.,b Huaxi Student Society of Oncology Research, West China School of Medicine, Sichuan University , Chengdu , Sichuan , China
| |
Collapse
|
33
|
|
34
|
Farrell PJ, Matuszkiewicz J, Balakrishna D, Pandya S, Hixon MS, Kamran R, Chu S, Lawson JD, Okada K, Hori A, Mizutani A, Iwata H, de Jong R, Hibner B, Vincent P. MET Tyrosine Kinase Inhibition Enhances the Antitumor Efficacy of an HGF Antibody. Mol Cancer Ther 2017; 16:1269-1278. [PMID: 28341789 DOI: 10.1158/1535-7163.mct-16-0771] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/13/2016] [Accepted: 03/15/2017] [Indexed: 11/16/2022]
Abstract
Receptor tyrosine kinase therapies have proven to be efficacious in specific cancer patient populations; however, a significant limitation of tyrosine kinase inhibitor (TKI) treatment is the emergence of resistance mechanisms leading to a transient, partial, or complete lack of response. Combination therapies using agents with synergistic activity have potential to improve response and reduce acquired resistance. Chemoreagent or TKI treatment can lead to increased expression of hepatocyte growth factor (HGF) and/or MET, and this effect correlates with increased metastasis and poor prognosis. Despite MET's role in resistance and cancer biology, MET TKI monotherapy has yielded disappointing clinical responses. In this study, we describe the biological activity of a selective, oral MET TKI with slow off-rate and its synergistic antitumor effects when combined with an anti-HGF antibody. We evaluated the combined action of simultaneously neutralizing HGF ligand and inhibiting MET kinase activity in two cancer xenograft models that exhibit autocrine HGF/MET activation. The combination therapy results in additive antitumor activity in KP4 pancreatic tumors and synergistic activity in U-87MG glioblastoma tumors. Pharmacodynamic characterization of biomarkers that correlate with combination synergy reveal that monotherapies induce an increase in the total MET protein, whereas combination therapy significantly reduces total MET protein levels and phosphorylation of 4E-BP1. These results hold promise that dual targeting of HGF and MET by combining extracellular ligand inhibitors with intracellular MET TKIs could be an effective intervention strategy for cancer patients who have acquired resistance that is dependent on total MET protein. Mol Cancer Ther; 16(7); 1269-78. ©2017 AACR.
Collapse
Affiliation(s)
- Pamela J Farrell
- Department of Biological Sciences, Takeda California, San Diego, California.
| | | | | | - Shweta Pandya
- Department of Biological Sciences, Takeda California, San Diego, California
| | - Mark S Hixon
- Department of Biological Sciences, Takeda California, San Diego, California
| | - Ruhi Kamran
- Department of Biological Sciences, Takeda California, San Diego, California
| | - Shaosong Chu
- Department of Chemistry, Takeda California, San Diego, California
| | - J David Lawson
- Department of Computational Sciences and Crystallography, Takeda California, San Diego, California
| | - Kengo Okada
- Pharmaceutical Research Division, Takeda Pharmaceutical Companies Ltd, Shonan, Japan
| | - Akira Hori
- Pharmaceutical Research Division, Takeda Pharmaceutical Companies Ltd, Shonan, Japan
| | - Akio Mizutani
- Pharmaceutical Research Division, Takeda Pharmaceutical Companies Ltd, Shonan, Japan
| | - Hidehisa Iwata
- Pharmaceutical Research Division, Takeda Pharmaceutical Companies Ltd, Shonan, Japan
| | - Ron de Jong
- Department of Biological Sciences, Takeda California, San Diego, California
| | - Barbara Hibner
- Oncology Biology, Takeda Boston, Cambridge, Massachusetts
| | - Patrick Vincent
- Department of Biological Sciences, Takeda California, San Diego, California
| |
Collapse
|
35
|
Qin S, Sun D, Li H, Li X, Pan W, Yan C, Tang R, Liu X. The Effect of SHH-Gli Signaling Pathway on the Synovial Fibroblast Proliferation in Rheumatoid Arthritis. Inflammation 2017; 39:503-12. [PMID: 26552406 DOI: 10.1007/s10753-015-0273-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by chronic synovitis. This study aims to investigate the role of sonic hedgehog (SHH)-Gli signaling pathway in synovial fibroblast proliferation in rheumatoid arthritis. The expression of serum SHH in RA patients group was significantly increased compared with the systemic lupus erythematosus (SLE), ankylosing spondylitis (AS), and healthy subject (healthy control, HC) groups, respectively; serum SHH expression of RA patients was positively correlated with rheumatoid factor (RF) and anti-cyclic citrullinated peptide antibodies (anti-CCP Ab), while there was no significant correlation between SHH expression and erythrocyte sedimentation rate (ESR). SHH, Ptch, Smo, and Gli molecules were highly expressed in rat RA-synovial fibroblast (RA-SF); after blocking the SHH-Gli signaling pathway with a Gli specific inhibitor, Gli-antagonist 61 (GANT61), RA-SF proliferation was inhibited in a dose-dependent manner and the apoptosis rate of RA-SF was increased as well; the expression levels of fibroblast growth factor receptor 1 (FGFR1) and FGFR3 declined in SF cells after GANT61 treatment. Our results suggest that SHH-Gli pathway is involved in the pathogenesis of RA, and blocking SHH-Gli pathway inhibits RA-SF cell proliferation and increases cell apoptosis, which may shed light on developing new ideas for RA treatment.
Collapse
Affiliation(s)
- Suping Qin
- Department of pathogenic biology and Immunology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Dexu Sun
- Department of Human Anatomy, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, China
| | - Hui Li
- Department of pathogenic biology and Immunology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Xiangyang Li
- Department of pathogenic biology and Immunology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Wei Pan
- Department of pathogenic biology and Immunology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Chao Yan
- Department of pathogenic biology and Immunology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Renxian Tang
- Department of pathogenic biology and Immunology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| | - Xiaomei Liu
- Department of pathogenic biology and Immunology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
36
|
Zhang H, Xie C, Yue J, Jiang Z, Zhou R, Xie R, Wang Y, Wu S. Cancer-associated fibroblasts mediated chemoresistance by a FOXO1/TGFβ1 signaling loop in esophageal squamous cell carcinoma. Mol Carcinog 2016; 56:1150-1163. [PMID: 27769097 DOI: 10.1002/mc.22581] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/14/2016] [Accepted: 10/19/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Hongfang Zhang
- Hangzhou Cancer Institution; Hangzhou Cancer Hospital; Hangzhou China
| | - Conghua Xie
- Department of Radiation and Medical Oncology; Zhongnan Hospital; Wuhan University; Wuhan Hubei China
- Hubei Key Laboratory of Tumor Biological Behaviors; Wuhan University; Wuhan Hubei China
- Hubei Cancer Clinical Study Center; Wuhan University; Wuhan Hubei China
| | - Jing Yue
- Hangzhou Cancer Institution; Hangzhou Cancer Hospital; Hangzhou China
| | - Zhenzhen Jiang
- Hangzhou Cancer Institution; Hangzhou Cancer Hospital; Hangzhou China
| | - Rongjing Zhou
- Department of Pathology; Hangzhou Cancer Hospital; Hangzhou China
| | - Ruifei Xie
- Department of Bio-Informatics; Hangzhou Cancer Hospital; Hangzhou China
| | - Yan Wang
- Hangzhou Cancer Institution; Hangzhou Cancer Hospital; Hangzhou China
| | - Shixiu Wu
- Hangzhou Cancer Institution; Hangzhou Cancer Hospital; Hangzhou China
| |
Collapse
|
37
|
Yan W, Wang X, Dai Y, Zhao B, Yang X, Fan J, Gao Y, Meng F, Wang Y, Luo C, Ai J, Geng M, Duan W. Discovery of 3-(5'-Substituted)-Benzimidazole-5-(1-(3,5-dichloropyridin-4-yl)ethoxy)-1H-indazoles as Potent Fibroblast Growth Factor Receptor Inhibitors: Design, Synthesis, and Biological Evaluation. J Med Chem 2016; 59:6690-708. [PMID: 27348537 DOI: 10.1021/acs.jmedchem.6b00056] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Fibroblast growth factor receptor (FGFR) represents an attractive oncology target for cancer therapy in view of its critical role in promoting cancer formation and progression, as well as causing resistance to approved therapies. In this article, we describe the identification of the potent pan-FGFR inhibitor (R)-21c (FGFR1-4 IC50 values of 0.9, 2.0, 2.0, and 6.1 nM, respectively). Compound (R)-21c exhibited excellent in vitro inhibitory activity against a panel of FGFR-amplified cell lines. Western blot analysis demonstrated that (R)-21c suppressed FGF/FGFR and downstream signaling pathways at nanomolar concentrations. Moreover, (R)-21c provided nearly complete inhibition of tumor growth (96.9% TGI) in NCI-H1581 (FGFR1-amplified) xenograft mice model at the dose of 10 mg/kg/qd via oral administration.
Collapse
Affiliation(s)
- Wei Yan
- School of Pharmacy, East China University of Science & Technology , 130 Mei Long Road, Shanghai 200237, P. R. China
| | - Xinyi Wang
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, P. R. China.,University of Chinese Academy of Sciences , Beijing 100049, P. R. China
| | - Yang Dai
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, P. R. China
| | - Bin Zhao
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, P. R. China
| | - Xinying Yang
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, P. R. China
| | - Jun Fan
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, P. R. China
| | - Yinglei Gao
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, P. R. China
| | - Fanwang Meng
- Department of Chemistry, College of Sciences, Shanghai University , 99 Shang Da Road, Shanghai 200444, P. R. China.,Drug Discovery & Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, P. R. China
| | - Yuming Wang
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, P. R. China
| | - Cheng Luo
- Drug Discovery & Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, P. R. China
| | - Jing Ai
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, P. R. China
| | - Meiyu Geng
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, P. R. China
| | - Wenhu Duan
- School of Pharmacy, East China University of Science & Technology , 130 Mei Long Road, Shanghai 200237, P. R. China.,Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu Chong Zhi Road, Shanghai 201203, P. R. China
| |
Collapse
|
38
|
Design, synthesis and biological evaluation of pyrazolylaminoquinazoline derivatives as highly potent pan-fibroblast growth factor receptor inhibitors. Bioorg Med Chem Lett 2016; 26:2594-9. [PMID: 27117427 DOI: 10.1016/j.bmcl.2016.04.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/25/2016] [Accepted: 04/12/2016] [Indexed: 11/20/2022]
Abstract
Fibroblast growth factor receptors (FGFRs) are important oncology targets due to the dysregulation of this signaling pathway in a wide variety of human cancers. We identified a series of pyrazolylaminoquinazoline derivatives as potent FGFR inhibitors with low nanomolar potency. The representative compound 29 strongly inhibited FGFR1-3 kinase activity and suppressed FGFR signaling transduction in FGFR-addicted cancer cells; FGFRs-driven cell proliferation was also strongly inhibited regardless of mechanistic complexity implicated in FGFR activation, which further confirmed that 29 was a potent pan-FGFR inhibitor. The flexibility of our structure offered the potential to preserve good affinity for mutant FGFR, which is important for developing TKIs with long-term efficacy.
Collapse
|
39
|
Giacomini A, Chiodelli P, Matarazzo S, Rusnati M, Presta M, Ronca R. Blocking the FGF/FGFR system as a two-compartment antiangiogenic/antitumor approach in cancer therapy. Pharmacol Res 2016; 107:172-185. [PMID: 27013279 DOI: 10.1016/j.phrs.2016.03.024] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 12/22/2022]
|
40
|
Tanner Y, Grose RP. Dysregulated FGF signalling in neoplastic disorders. Semin Cell Dev Biol 2016; 53:126-35. [DOI: 10.1016/j.semcdb.2015.10.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/08/2015] [Indexed: 12/31/2022]
|
41
|
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]
|
42
|
Akizawa H, Nagatomo H, Odagiri H, Kohri N, Yamauchi N, Yanagawa Y, Nagano M, Takahashi M, Kawahara M. Conserved roles of fibroblast growth factor receptor 2 signaling in the regulation of inner cell mass development in bovine blastocysts. Mol Reprod Dev 2016; 83:516-25. [DOI: 10.1002/mrd.22646] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 03/30/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Hiroki Akizawa
- Laboratory of Animal Breeding and Reproduction; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Hiroaki Nagatomo
- Laboratory of Animal Breeding and Reproduction; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Haruka Odagiri
- Laboratory of Animal Breeding and Reproduction; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Nanami Kohri
- Laboratory of Animal Breeding and Reproduction; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Nobuhiko Yamauchi
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture; Kyushu University; Fukuoka Japan
| | - Yojiro Yanagawa
- Laboratory of Theriogenology, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine; Hokkaido University; Sapporo Japan
| | - Masashi Nagano
- Laboratory of Theriogenology, Department of Veterinary Clinical Sciences, Graduate School of Veterinary Medicine; Hokkaido University; Sapporo Japan
| | - Masashi Takahashi
- Laboratory of Animal Breeding and Reproduction; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| | - Manabu Kawahara
- Laboratory of Animal Breeding and Reproduction; Research Faculty of Agriculture; Hokkaido University; Sapporo Japan
| |
Collapse
|
43
|
Ishii G, Ochiai A, Neri S. Phenotypic and functional heterogeneity of cancer-associated fibroblast within the tumor microenvironment. Adv Drug Deliv Rev 2016; 99:186-196. [PMID: 26278673 DOI: 10.1016/j.addr.2015.07.007] [Citation(s) in RCA: 328] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 06/26/2015] [Accepted: 07/20/2015] [Indexed: 12/30/2022]
Abstract
Cancer microenvironment is created not only by malignant epithelial cells, but also by several kinds of stromal cells. Since Paget proposed the "seed and soil" hypothesis, the biological importance of the cancer microenvironment has come to be widely accepted. The main compartment of host stromal cells are fibroblasts (Cancer-Associated Fibroblasts; CAFs), which are the main source of the collagen-producing cells. CAFs directly communicate with the cancer cells and other types of stromal cells to acquire a specific biological phenotype. CAFs play important roles in several aspects of the tumor progression process and the chemotherapeutic process. However, CAFs have heterogeneous origins, phenotypes, and functions under these conditions. A crucial challenge is to understand how much of this heterogeneity serves different biological responses to cancer cells. In this review, we highlight the issue of how diverse and heterogeneous functions given by CAFs can exert potent influences on tumor progression and therapeutic response. Furthermore, we also discuss the current advances in the development of novel therapeutic strategies against CAFs.
Collapse
Affiliation(s)
- Genichiro Ishii
- Division of Pathology Exploratory Oncology Research & Clinical Trial Center National Cancer Center 6-5-1, Kashiwanoha, Kashiwa-City, Chiba 277-8577, Japan.
| | - Atsushi Ochiai
- Division of Pathology Exploratory Oncology Research & Clinical Trial Center National Cancer Center 6-5-1, Kashiwanoha, Kashiwa-City, Chiba 277-8577, Japan
| | - Shinya Neri
- Division of Pathology Exploratory Oncology Research & Clinical Trial Center National Cancer Center 6-5-1, Kashiwanoha, Kashiwa-City, Chiba 277-8577, Japan
| |
Collapse
|
44
|
Shi H, Xu J, Zhao R, Wu H, Gu L, Chen Y. FGF2 regulates proliferation, migration, and invasion of ECA109 cells through PI3K/Akt signalling pathway in vitro. Cell Biol Int 2016; 40:524-33. [PMID: 26833879 DOI: 10.1002/cbin.10588] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 01/28/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Hui Shi
- Department of Thoracic Surgery; The First Affiliated Hospital of Nanjing Medical University; Nanjing 210000 Jiangsu China
- Department of Biochemistry and Molecular Biology; Nanjing Medical University; Nanjing 210000 Jiangsu China
- Department of Thoracic Surgery; Affiliated Hospital of Nantong University; Nantong 226001 Jiangsu China
| | - Jingjing Xu
- Nursing School of Nantong University; Nantong 226001 Jiangsu China
| | - Rui Zhao
- Nursing School of Nantong University; Nantong 226001 Jiangsu China
| | - Huiqun Wu
- Medical School of Nantong University; Nantong 226001 Jiangsu China
| | - Luo Gu
- Department of Biochemistry and Molecular Biology; Nanjing Medical University; Nanjing 210000 Jiangsu China
| | - Yijiang Chen
- Department of Thoracic Surgery; The First Affiliated Hospital of Nanjing Medical University; Nanjing 210000 Jiangsu China
| |
Collapse
|
45
|
Wang J, Zhang G, Wang J, Wang L, Huang X, Cheng Y. The role of cancer-associated fibroblasts in esophageal cancer. J Transl Med 2016; 14:30. [PMID: 26822225 PMCID: PMC4732002 DOI: 10.1186/s12967-016-0788-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 01/17/2016] [Indexed: 01/04/2023] Open
Abstract
Fibroblasts are known as critical stromal cells in wound healing by synthesizing extracellular matrix and collagen. A subpopulation of them is called cancer-associated fibroblasts (CAFs), because their production of proteins participated in various biological activities including tumor cell proliferation, invasion and metastasis. Currently some studies shed light on their role in esophageal cancer which was an aggressive cancer with a dismal survival and high rate of metastasis. Thus, to find cures for it relies on elucidating the epithelial-fibroblasts crosstalk. Herein, we reviewed the present knowledge of the CAFs’ role in esophageal premalignant condition, cancer initiation, progression, metastasis and prognosis prediction and further provided some insights into its clinical application.
Collapse
Affiliation(s)
- Jiangfeng Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, No 107 West Wenhua Road, Jinan, 250012, People's Republic of China.
| | - Guangyu Zhang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, No 107 West Wenhua Road, Jinan, 250012, People's Republic of China.
| | - Jianbo Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, No 107 West Wenhua Road, Jinan, 250012, People's Republic of China.
| | - Lu Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, No 107 West Wenhua Road, Jinan, 250012, People's Republic of China.
| | - Xiaochen Huang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, No 107 West Wenhua Road, Jinan, 250012, People's Republic of China.
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, No 107 West Wenhua Road, Jinan, 250012, People's Republic of China.
| |
Collapse
|
46
|
Garajová I, Giovannetti E, Biasco G, Peters GJ. c-Met as a Target for Personalized Therapy. TRANSLATIONAL ONCOGENOMICS 2015. [PMID: 26628860 DOI: 10.4137/togog.s30534] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
MET and its ligand HGF are involved in many biological processes, both physiological and pathological, making this signaling pathway an attractive therapeutic target in oncology. Downstream signaling effects are transmitted via mitogen-activated protein kinase (MAPK), PI3K (phosphoinositide 3-kinase protein kinase B)/AKT, signal transducer and activator of transcription proteins (STAT), and nuclear factor-κB. The final output of the terminal effector components of these pathways is activation of cytoplasmic and nuclear processes leading to increases in cell proliferation, survival, mobilization and invasive capacity. In addition to its role as an oncogenic driver, increasing evidence implicates MET as a common mechanism of resistance to targeted therapies including EGFR and VEGFR inhibitors. In the present review, we summarize the current knowledge on the role of the HGF-MET signaling pathway in cancer and its therapeutic targeting (HGF activation inhibitors, HGF inhibitors, MET antagonists and selective/nonselective MET kinase inhibitors). Recent advances in understanding the role of this pathway in the resistance to current anticancer strategies used in lung, kidney and pancreatic cancer are discussed.
Collapse
Affiliation(s)
- Ingrid Garajová
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands. ; Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands. ; Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Guido Biasco
- Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
47
|
Garajová I, Giovannetti E, Biasco G, Peters GJ. c-Met as a Target for Personalized Therapy. TRANSLATIONAL ONCOGENOMICS 2015; 7:13-31. [PMID: 26628860 PMCID: PMC4659440 DOI: 10.4137/tog.s30534] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/20/2015] [Accepted: 09/23/2015] [Indexed: 12/30/2022]
Abstract
MET and its ligand HGF are involved in many biological processes, both physiological and pathological, making this signaling pathway an attractive therapeutic target in oncology. Downstream signaling effects are transmitted via mitogen-activated protein kinase (MAPK), PI3K (phosphoinositide 3-kinase protein kinase B)/AKT, signal transducer and activator of transcription proteins (STAT), and nuclear factor-κB. The final output of the terminal effector components of these pathways is activation of cytoplasmic and nuclear processes leading to increases in cell proliferation, survival, mobilization and invasive capacity. In addition to its role as an oncogenic driver, increasing evidence implicates MET as a common mechanism of resistance to targeted therapies including EGFR and VEGFR inhibitors. In the present review, we summarize the current knowledge on the role of the HGF-MET signaling pathway in cancer and its therapeutic targeting (HGF activation inhibitors, HGF inhibitors, MET antagonists and selective/nonselective MET kinase inhibitors). Recent advances in understanding the role of this pathway in the resistance to current anticancer strategies used in lung, kidney and pancreatic cancer are discussed.
Collapse
Affiliation(s)
- Ingrid Garajová
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, Sant’Orsola-Malpighi Hospital, Bologna, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Guido Biasco
- Department of Experimental, Diagnostic and Speciality Medicine, University of Bologna, Sant’Orsola-Malpighi Hospital, Bologna, Italy
| | - Godefridus J. Peters
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
48
|
von Loga K, Kohlhaussen J, Burkhardt L, Simon R, Steurer S, Burdak-Rothkamm S, Jacobsen F, Sauter G, Krech T. FGFR1 Amplification Is Often Homogeneous and Strongly Linked to the Squamous Cell Carcinoma Subtype in Esophageal Carcinoma. PLoS One 2015; 10:e0141867. [PMID: 26555375 PMCID: PMC4640518 DOI: 10.1371/journal.pone.0141867] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 10/14/2015] [Indexed: 12/12/2022] Open
Abstract
Background and Aims Amplification of the fibroblast growth factor receptor 1 (FGFR1) is believed to predict response to multi-kinase inhibitors targeting FGFR1. Esophageal cancer is an aggressive disease, for which novel targeted therapies are highly warranted. Methods This study was designed to investigate the prevalence and clinical significance of FGFR1 amplification in a tissue microarray containing 346 adenocarcinomas and 254 squamous cell carcinomas of the esophagus, using dual-labeling fluorescence in situ hybridization (FISH) analysis. Results FGFR1 amplification, defined as a ratio of FGFR1:centromere 8 copy numbers ≥ 2.0, was more frequently seen in squamous cell carcinoma (8.9% of 202 interpretable cases) than in adenocarcinoma (1.6% of 308; p<0.0001). There was no association between FGFR1 amplification and tumor phenotype or clinical outcome. To study potential heterogeneity of FGFR1 amplification, all available tumor blocks from 23 FGFR1 amplified tumors were analyzed on conventional large sections. This analysis revealed complete homogeneity of FGFR1 amplification in 20 (86.9%) primary tumors and in all available lymph node metastases. Remarkably, FGFR1 amplification was also seen in dysplasia adjacent to tumor in 6 of 9 patients with FGFR1 amplified primary cancers. Conclusions In conclusion, FGFR1 amplification occurs in a relevant subgroup of carcinomas of the esophagus and may play a particular role for development of squamous cell cancers. The high homogeneity of FGFR1 amplification suggests that patients with FGFR1 amplified esophageal cancers may particularly benefit from anti-FGFR1 therapies and prompt for clinical studies in this tumor type.
Collapse
Affiliation(s)
- Katharina von Loga
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| | - Jule Kohlhaussen
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lia Burkhardt
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ronald Simon
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Steurer
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Frank Jacobsen
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Till Krech
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
49
|
Ueno N, Shimizu A, Kanai M, Iwaya Y, Ueda S, Nakayama J, Seo MK. Enhanced Expression of Fibroblast Growth Factor Receptor 3 IIIc Promotes Human Esophageal Carcinoma Cell Proliferation. J Histochem Cytochem 2015; 64:7-17. [PMID: 26487184 DOI: 10.1369/0022155415616161] [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] [Received: 06/06/2015] [Accepted: 10/08/2015] [Indexed: 11/22/2022] Open
Abstract
Deregulated expression of fibroblast growth factor receptors (FGFRs) and their ligands plays critical roles in tumorigenesis. The gene expression of an alternatively spliced isoforms of FGFR3, FGFR3IIIc, was analyzed by RT-PCR in samples from patients with esophageal carcinoma (EC), including esophageal squamous cell carcinoma (ESCC) and adenocarcinoma (EAC). The incidence of FGFR3IIIc was higher in EC [12/16 (75%); p=0.073] than in non-cancerous mucosa (NCM) [6/16 (38%)]. Indeed, an immunohistochemical analysis of early-stage ESCC showed that carcinoma cells expressing FGFR3IIIc stained positively with SCC-112, a tumor marker, and Ki67, a cell proliferation marker, suggesting that the expression of FGFR3IIIc promotes cell proliferation. We used EC-GI-10 cells endogenously expressing FGFR3IIIc as a model of ESCC to provide mechanistic insight into the role of FGFR3IIIc in ESCC. The knockdown of endogenous FGFR3 using siRNA treatment significantly abrogated cell proliferation and the overexpression of FGFR3IIIc in cells with enhanced cell proliferation. EC-GI-10 cells and ESCC from patients with EC showed endogenous expression of FGF2, a specific ligand for FGFR3IIIc, suggesting that the upregulated expression of FGFR3IIIc may create autocrine FGF signaling in ESCC. Taken together, FGFR3IIIc may have the potential to be an early-stage tumor marker and a molecular target for ESCC therapy.
Collapse
Affiliation(s)
- Nobuhiro Ueno
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan (NU, AS, MKS)
| | - Akio Shimizu
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan (NU, AS, MKS)
| | - Michiyuki Kanai
- Digestive Disease Center, Hanwasumiyoshi General Hospital, Osaka, Japan (MK)
| | - Yugo Iwaya
- Department of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan (YI)
| | - Shugo Ueda
- Department of Gastroenterological Surgery and Oncology, Kitano Hospital, Osaka, Japan (SU)
| | - Jun Nakayama
- Department of Molecular Pathology, Shinshu University Graduate School of Medicine, Matsumoto, Japan (JN)
| | - Misuzu Kurokawa Seo
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan (NU, AS, MKS)
| |
Collapse
|