1
|
Buchynska LG, Brieieva OV, Nespriadko SV. EXPRESSION OF HEPATOCYTE GROWTH FACTOR AND C-MET RECEPTOR IN STROMAL FIBROBLASTS AND TUMOR CELLS OF ENDOMETRIAL CARCINOMA. Exp Oncol 2023; 45:79-87. [PMID: 37417279 DOI: 10.15407/exp-oncology.2023.01.079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/26/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND HGF/c-Met is one of the main signaling pathways that ensure communication between epithelial cells and components of the tumor microenvironment determining the invasive and metastatic potential of many cancers. However, the significance of HGF and c-Met in endometrial carcinoma (ECa) progression remains unclear. AIM To evaluate copy number variations as well as expression of the c-Met receptor and its ligand HGF in endometrial carcinomas considering the clinical and morphological characteristics of ECa. MATERIALS AND METHODS The study was conducted on ECa samples of 57 patients, among which 32 had lymph nodes and/or distant metastasis. The copy number of c-MET gene was estimated by qPCR. The expression of HGF and c-Met in tissue samples was determined by the immunohistochemical method. RESULTS Amplification of c-MET gene was detected in 10.5% of the ECa cases. In most carcinomas, a combined expression pattern of HGF and c-Met was established, in which co-expression of these markers was observed in tumor cells, and the content of HGF+ fibroblasts increased in the stroma. The expression of HGF in tumor cells was associated with the tumor differentiation grade and was higher in G3 ECa (p = 0.041). The number of HGF+ fibroblasts in the stromal component increased in the ECa cases with metastasis compared to the cases without metastasis (p = 0.032). The content of stromal c-Met+ fibroblasts was higher in deeply invasive carcinomas of patients with metastases than in tumors with invasion of < 1/2 myometrium (p = 0.035). CONCLUSION Increased expression of HGF and c-Met in stromal fibroblasts of endometrial carcinomas is associated with metastasis in patients with ECa and deep invasion of the tumor into the myometrium, and can contribute to the aggressive course of the disease.
Collapse
Affiliation(s)
- L G Buchynska
- R.E.Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, Kyiv 03022, Ukraine
| | - O V Brieieva
- R.E.Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, Kyiv 03022, Ukraine
| | - S V Nespriadko
- National Cancer Institute of the Ministry of Health of Ukraine, Kyiv 03022, Ukraine
| |
Collapse
|
2
|
Sulfated glycolipid PG545 induces endoplasmic reticulum stress and augments autophagic flux by enhancing anticancer chemotherapy efficacy in endometrial cancer. Biochem Pharmacol 2020; 178:114003. [PMID: 32360360 DOI: 10.1016/j.bcp.2020.114003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/30/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022]
Abstract
The sulfated glycolipid PG545 shows promising antitumor activity in various cancers. This study was conducted to explore the effects and the mechanism of PG545 action in endometrial cancer (EC). PG545 exhibited strong synergy as assessed by the Chou-Talalay-Method in vitro when combined with cisplatin, or paclitaxel in both type I (Hec1B) and type II (ARK2) EC cell lines. While PG545 showed antitumor activity as monotherapy, a combination of PG545 with paclitaxel and cisplatin was highly effective in reducing the tumor burden and significantly prolonged survival of both Hec1B and ARK2 xenograft bearing mice. Mechanistically, PG545 elicits ER stress as an early response with resultant induction of autophagy. Our data demonstrated an increase in pERK, Bip/Grp78, IRE1α, Calnexin and CHOP/GADD153 within 6-24 hrs of PG545 treatment in EC cells. In parallel, PG545 also blocked FGF2 and HB-EGF mediated signaling in EC cells. Moreover, melatonin-mediated ER stress inhibition reduced PG545-mediated autophagy and PG545 in combination with cisplatin further heightened this stress response. Collectively these data indicate that PG545 exhibits strong synergistic effects with chemotherapeutics in vitro and showed promising antitumor activity in vivo. Our preclinical data indicates that in future studies PG545 can be a useful adjunct to chemotherapy in endometrial cancer.
Collapse
|
3
|
Dhani NC, Hirte HW, Wang L, Burnier JV, Jain A, Butler MO, Welch S, Fleming GF, Hurteau J, Matsuo K, Matei D, Jimenez W, Johnston C, Cristea M, Tonkin K, Ghatage P, Lheureux S, Mehta A, Quintos J, Tan Q, Kamel-Reid S, Ludkovski O, Tsao MS, Wright JJ, Oza AM. Phase II Trial of Cabozantinib in Recurrent/Metastatic Endometrial Cancer: A Study of the Princess Margaret, Chicago, and California Consortia (NCI9322/PHL86). Clin Cancer Res 2020; 26:2477-2486. [PMID: 31992589 DOI: 10.1158/1078-0432.ccr-19-2576] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/11/2019] [Revised: 12/10/2019] [Accepted: 01/24/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE The relevance of the MET/hepatocyte growth factor pathway in endometrial cancer tumor biology supports the clinical evaluation of cabozantinib in this disease. PATIENTS AND METHODS PHL86/NCI#9322 (NCT01935934) is a single arm study that evaluated cabozantinib (60 mg once daily) in women with endometrial cancer with progression after chemotherapy. Coprimary endpoints were response rate and 12-week progression-free-survival (PFS). Patients with uncommon histology endometrial cancer (eg, carcinosarcoma and clear cell) were enrolled in a parallel exploratory cohort. RESULTS A total of 102 patients were accrued. Among 36 endometrioid histology patients, response rate was 14%, 12-week PFS rate was 67%, and median PFS was 4.8 months. In serous cohort of 34 patients, response rate was 12%, 12-week PFS was 56%, and median PFS was 4.0 months. In a separate cohort of 32 patients with uncommon histology endometrial cancer (including carcinosarcoma), response rate was 6% and 12-week PFS was 47%. Six patients were on treatment for >12 months, including two for >30 months. Common cabozantinib-related toxicities (>30% patients) included hypertension, fatigue, diarrhea, nausea, and hand-foot syndrome. Gastrointestinal fistula/perforation occurred in four of 70 (6%) patients with serous/endometrioid cancer and five of 32 (16%) patients in exploratory cohort. We observed increased frequency of responses with somatic CTNNB1 mutation [four partial responses (PRs) in 10 patients, median PFS 7.6 months] and concurrent KRAS and PTEN/PIK3CA mutations (three PRs in 12 patients, median PFS 5.9 months). CONCLUSIONS Cabozantinib has activity in serous and endometrioid histology endometrial cancer. These results support further evaluation in genomically characterized patient cohorts.
Collapse
Affiliation(s)
- Neesha C Dhani
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada.
| | - Hal W Hirte
- Juravinski Cancer Centre, Hamilton, Ontario, Canada
| | - Lisa Wang
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | - Angela Jain
- Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | | | - Stephen Welch
- London Regional Cancer Program, London, Ontario, Canada
| | | | - Jean Hurteau
- North Shore University Health System Evanston Hospital, Evanston, Illinois
| | - Koji Matsuo
- University of Southern California/Norris Comprehensive Cancer Centre, Los Angeles, California
| | | | | | | | - Mihaela Cristea
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Katia Tonkin
- The Cross Cancer Institute, Edmonton, Alberta, Canada
| | | | | | - Anjali Mehta
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Judy Quintos
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Qian Tan
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | - Olga Ludkovski
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | - John J Wright
- NCI Cancer Therapy Evaluation Program (CTEP), Bethesda, Maryland
| | - Amit M Oza
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Hernández JE, González-Montiel A, Allos-Villalva JCC, Cantú D, Barquet S, Olivares-Mundo A, Herrera LA, Prada D. Prognostic molecular biomarkers in endometrial cancer: A review. ACTA ACUST UNITED AC 2019; 7:17-28. [PMID: 34322276 PMCID: PMC8315102 DOI: 10.14312/2052-4994.2019-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/15/2023]
Abstract
Background: Endometrial cancer (EC) is the fourth most common malignancy in women worldwide and the most common gynecological cancer in developed countries. The endometrioid subtype has an excellent prognosis with conventional treatment; however, recurrence reduces overall survival. Objective: Describe the most relevant evidence regarding selected potential molecular biomarkers that may predict overall survival (OS), relapse-free survival (RFS), and cancer-specific survival (CSS) in EC. Methods: An exhaustive search was performed in PUBMED with the search terms endometrial cancer, molecular biomarker, and survival. We selected original articles written in English about endometrial cancer, molecular biomarkers, and that included survival analysis published between January 2000 and December 2016. Results: Several molecular prognostic biomarkers have been studied in terms of survival and therapeutic response in women with endometrial cancer; hormone receptors, microRNAs, and other molecules have emerged as potentially useful biomarkers, including HER2, p21, HE4, PTEN, p27, ANCCA, and ANXA2. Conclusions: The use of biomarkers in the assessment of OS, RFS, and CSS requires large trials to expand our understanding of endometrial carcinogenesis. Several molecular markers are significantly associated with a high tumor grade and advanced clinical stage in EC and, therefore, could have additive effects when combined.
Collapse
Affiliation(s)
- J Edgardo Hernández
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080
| | - Ailyn González-Montiel
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080
| | - Jesús C Ceb Allos-Villalva
- Department of Biomedical Informatics, Faculty of Medicine, National Autonomous University of Mexico, C.U., Av. Universidad 3000, Mexico City, Mexico, 04510
| | - David Cantú
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080
| | - Salim Barquet
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080
| | - Anny Olivares-Mundo
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080
| | - Luis A Herrera
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080
| | - Diddier Prada
- Unit of Biomedical Research, National Cancer Institute- Biomedical Research Institute, National Autonomous University of Mexico. San Fernando 22, Colonia Sección XVI, Delegatión Tlalpan, Mexico City, Mexico, 14080.,Department of Biomedical Informatics, Faculty of Medicine, National Autonomous University of Mexico, C.U., Av. Universidad 3000, Mexico City, Mexico, 04510
| |
Collapse
|
5
|
Moosavi F, Giovannetti E, Saso L, Firuzi O. HGF/MET pathway aberrations as diagnostic, prognostic, and predictive biomarkers in human cancers. Crit Rev Clin Lab Sci 2019; 56:533-566. [PMID: 31512514 DOI: 10.1080/10408363.2019.1653821] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/24/2022]
Abstract
Cancer is a major cause of death worldwide. MET tyrosine kinase receptor [MET, c-MET, hepatocyte growth factor (HGF) receptor] pathway activation is associated with the appearance of several hallmarks of cancer. The HGF/MET pathway has emerged as an important actionable target across many solid tumors; therefore, biomarker discovery becomes essential in order to guide clinical intervention and patient stratification with the aim of moving towards personalized medicine. The focus of this review is on how the aberrant activation of the HGF/MET pathway in tumor tissue or the circulation can provide diagnostic and prognostic biomarkers and predictive biomarkers of drug response. Many meta-analyses have shown that aberrant activation of the MET pathway in tumor tissue, including MET gene overexpression, gene amplification, exon 14 skipping and other activating mutations, is almost invariably associated with shorter survival and poor prognosis. Most meta-analyses have been performed in non-small cell lung cancer (NSCLC), breast, head and neck cancers as well as colorectal, gastric, pancreatic and other gastrointestinal cancers. Furthermore, several studies have shown the predictive value of MET biomarkers in the identification of patients who gain the most benefit from HGF/MET targeted therapies administered as single or combination therapies. The highest predictive values have been observed for response to foretinib and savolitinib in renal cancer, as well as tivantinib in NSCLC and colorectal cancer. However, some studies, especially those based on MET expression, have failed to show much value in these stratifications. This may be rooted in lack of standardization of methodologies, in particular in scoring systems applied in immunohistochemistry determinations or absence of oncogenic addiction of cancer cells to the MET pathway, despite detection of overexpression. Measurements of amplification and mutation aberrations are less likely to suffer from these pitfalls. Increased levels of MET soluble ectodomain (sMET) in circulation have also been associated with poor prognosis; however, the evidence is not as strong as it is with tissue-based biomarkers. As a diagnostic biomarker, sMET has shown its value in distinguishing cancer patients from healthy individuals in prostate and bladder cancers and in melanoma. On the other hand, increased circulating HGF has also been presented as a valuable prognostic and diagnostic biomarker in many cancers; however, there is controversy on the predictive value of HGF as a biomarker. Other biomarkers such as circulating tumor DNA (ctDNA) and tumor HGF levels have also been briefly covered. In conclusion, HGF/MET aberrations can provide valuable diagnostic, prognostic and predictive biomarkers and represent vital assets for personalized cancer therapy.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz , Iran
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc) , Amsterdam , The Netherlands.,Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza Onlus , Pisa , Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology, "Vittorio Erspamer," Sapienza University , Rome , Italy
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz , Iran
| |
Collapse
|
6
|
Mohd Nafi SN, Idris F, Jaafar H. Cellular and Molecular Changes in MNU-Induced Breast Tumours Injected with PF4 or bFGF. Asian Pac J Cancer Prev 2017; 18:3231-3238. [PMID: 29281877 PMCID: PMC5980876 DOI: 10.22034/apjcp.2017.18.12.3231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/27/2022] Open
Abstract
Background: Angiogenic activity has been considered to reflect important molecular events during breast tumour
development. The present study concerned cellular and molecular changes of MNU-induced breast tumours subjected
to promotion and suppression of angiogenesis. Methods: Female Sprague Dawley rats at the age of 21 days received
MNU at the dose 70 mg/kg of body weight by intraperitoneal injection. Three months post-carcinogen initiation,
mammary tumours were palpated and their growth was monitored. When the tumour diameter reached 1.0 ± 0.05 cm,
rats were given bFGF or PF4 intratumourally at a dose of 10 μg/tumour. Entire palpable tumour were subsequently
excised and subjected to histology examination, IHC staining, and RT-PCR. Results: No critical morphological changes
were observed between pro-angiogenic factor, bFGF, and control groups. However, increase of tumour size with more
necrotic and diffuse areas was notable in tumours after anti-angiogenic PF4 intervention. ER and PR mRNA expression
was significantly up- and down-regulated in bFGF and PF4 groups, respectively. The trends were significantly associated
with peri- and intratumoural MVD counts. However, irrespective of whether we promoted or inhibited angiogenesis,
the expression of EGFR and ERBB2 continued to be significantly increased but this was not significantly associated
with the MVD score. No significant differences in E-cadherin and LR gene expression were noted between intervention
and control groups. Conclusion: ER and PR receptor expression shows consistent responses when tumour angiogenesis
is manipulated either positively or negatively. Our study adds to current understanding that not only do we need to
target hormonal receptors, as presently practiced, but we also need to target endothelial receptors to successfully treat
breast cancer.
Collapse
Affiliation(s)
- Siti Norasikin Mohd Nafi
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kota Bharu, Kelantan, Malaysia.
| | | | | |
Collapse
|
7
|
Nam KH, Yoon H, Lee K, Park DJ, Kim HH, Lee HS, Shin E. Predictive value for lymph node metastasis of epithelial-mesenchymal transition and cancer stem cell marker expression in early gastric cancer. Pathol Res Pract 2017; 213:1221-1226. [PMID: 28595915 DOI: 10.1016/j.prp.2017.03.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/19/2016] [Revised: 03/11/2017] [Accepted: 03/12/2017] [Indexed: 01/12/2023]
Abstract
BACKGROUND Minimally invasive therapies, including endoscopic mucosal resection or sentinel node navigation surgery, have been widely applied in early gastric cancer because of their benefits in promoting patient quality of life. However, lymph node dissection is beyond the capability of endoscopic therapy, and in sentinel node navigation surgery, the potential for skip metastasis is not negligible. Therefore, the possibility of lymph node metastasis is the most important factor to consider when deciding whether to apply the minimally invasive therapies. In the present study, the significance of epithelial mesenchymal transition and stem cell marker expression in lymph node metastasis in early gastric cancer was investigated. METHODS We evaluated the significance of the expression of 5 epithelial mesenchymal transition-related markers (E-cadherin, MMP7, S100A, Snail-1, and HGF) and 6 stem cell markers (ALDH1, SOX2, CD24, CD44, CD54, and CD133) in 119 early gastric cancer specimens using immunohistochemistry. Because protein expression is heterogeneous in gastric cancer, we analyzed the expression of these markers in two selected regions (one each at the superficial zone and the deep invasive front). RESULTS Expression of E-cadherin, MMP7, HGF, and CD133 at the deep invasive front was associated with the absence of lymph node metastasis (P=0.013, 0.018, <0.001, and 0.026, respectively). Presence of diffuse-type component, lymphatic invasion, and lack of expression of HGF and CD133 at the deep invasive front were independent predictive markers of lymph node metastasis (P=0.019, <0.001, 0.015, and 0.047, respectively). CONCLUSIONS Lymph node metastasis is strongly associated with expression status of HGF and CD133 at the deep invasive front, suggesting the usefulness of these proteins as independent predictive markers of lymph node metastasis in early gastric cancer.
Collapse
Affiliation(s)
- Kyung Han Nam
- Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, 875, Haeun-daero, Haeundae-gu, Busan 48108, Republic of Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, 173-82 Gumi-ro, Bundang-gu, Seongnam, Gyeonggi 13620, Republic of Korea
| | - Kyungbun Lee
- Department of Pathology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Do Joong Park
- Department of Surgery, Seoul National University Bundang Hospital, 173-82 Gumi-ro, Bundang-gu, Seongnam, Gyeonggi 13620, Republic of Korea
| | - Hyung-Ho Kim
- Department of Surgery, Seoul National University Bundang Hospital, 173-82 Gumi-ro, Bundang-gu, Seongnam, Gyeonggi 13620, Republic of Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, 173-82 Gumi-ro, Bundang-gu, Seongnam, Gyeonggi 13620, Republic of Korea
| | - Eun Shin
- Department of Pathology, Seoul National University Bundang Hospital, 173-82 Gumi-ro, Bundang Gu, Seongnam, Gyeonggi 13620, Republic of Korea.
| |
Collapse
|
8
|
Cao J, Zhang Y, Yang J, He S, Li M, Yan S, Chen Y, Qu C, Xu L. NEAT1 regulates pancreatic cancer cell growth, invasion and migration though mircroRNA-335-5p/c-met axis. Am J Cancer Res 2016; 6:2361-2374. [PMID: 27822425 PMCID: PMC5088299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/14/2016] [Accepted: 05/12/2016] [Indexed: 06/06/2023] Open
Abstract
NEAT1 has been reported to affect cancer progression, which was subsequently confirmed in multiple cancers. Hsa-miRNA-335-5p (miR-335-5p) has recently been identified as an anticancer agent in various organs. However, the relationship between NEAT1 and miR-335-5p remains poorly understood. In this study, we investigated the effects of NEAT1 and miR-335-5p on development of pancreatic cancer. The ectopic expression of miR-335-5p in pancreatic cancer cell lines significantly suppressed cell growth by inhibiting c-met. In addition, downregulating NEAT1 upregulates miR-335-5p. Taken together, our results demonstrate that the NEAT1/miR-335-5p/c-met axis plays a pivotal role in pancreatic cancer by regulating the proliferation, metastasis, and apoptosis of pancreatic cancer cells in vivo and in vitro.
Collapse
Affiliation(s)
- Jia Cao
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| | - Yi Zhang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| | - Jiachun Yang
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| | - Sijia He
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| | - Mingming Li
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| | - Shiyan Yan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| | - Ying Chen
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| | - Chunying Qu
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| | - Leiming Xu
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine Shanghai 200092, China
| |
Collapse
|
9
|
Woo JK, Kang JH, Kim B, Park BH, Shin KJ, Song SW, Kim JJ, Kim HM, Lee SJ, Oh SH. Humanized anti-hepatocyte growth factor (HGF) antibody suppresses innate irinotecan (CPT-11) resistance induced by fibroblast-derived HGF. Oncotarget 2016; 6:24047-60. [PMID: 26090722 PMCID: PMC4695169 DOI: 10.18632/oncotarget.4369] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/02/2015] [Accepted: 05/30/2015] [Indexed: 12/19/2022] Open
Abstract
The growth factors derived from the microenvironment create an environment conducive to tumor growth and survival. HGF deprivation using neutralizing antibody enhanced chemosensitivity in colorectal cancer cells (CRC). We determined secreted HGF in fibroblast conditioned medium (CM). Combination treatment of anti-HGF antibody and irinotecan (CPT-11) directly enhanced CPT-11 sensitivity in CRC. We generated xenograft in NOD/SCID mice inoculating HCT-116 human colorectal cancer cells subcutaneously with or without fibroblast. We found that the combination of CPT-11 and anti-HGF antibody induced marked suppression of tumor development. These results suggest that HGF produced by fibroblast induce CPT-11 resistance, and that anti-HGF antibody abrogate such resistance in vivo. fibroblast-derived HGF is important determinant of chemoresistance. Anti-HGF monoclonal antibody treatment confirmed the importance of this growth factor for chemoresistance in CRC. These results present new options toward the early diagnosis of chemoresistance and suggest novel combinations of chemotherapy and anti-HGF agents to prevent or significantly delay the onset of therapy resistance.
Collapse
Affiliation(s)
- Jong Kyu Woo
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Ju-Hee Kang
- National Cancer Center, Goyang, Republic of Korea
| | - BoRa Kim
- National Cancer Center, Goyang, Republic of Korea
| | - Byung Hee Park
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | | | | | - Jung Ju Kim
- Yooyoung Pharmaceutical Co., Seoul, Republic of Korea
| | - Hwan-Mook Kim
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Sang-Jin Lee
- National Cancer Center, Goyang, Republic of Korea
| | - Seung Hyun Oh
- College of Pharmacy, Gachon University, Incheon, Republic of Korea
| |
Collapse
|
10
|
Gayyed MF, Abd El-Maqsoud NMR, El-Hameed El-Heeny AA, Mohammed MF. c-MET expression in colorectal adenomas and primary carcinomas with its corresponding metastases. J Gastrointest Oncol 2015; 6:618-27. [PMID: 26697193 DOI: 10.3978/j.issn.2078-6891.2015.072] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND c-MET plays an important role in tumor proliferation, invasion and metastasis. In this study we examined the expression of c-MET in colorectal adenomas, primary adenocarcinomas and their corresponding lymph node, peritoneal and liver metastases. We correlated our findings with clinicopathological features. METHODS Twenty three cases of colorectal adenoma and 102 cases of primary colorectal carcinoma and their corresponding metastases (44 lymph nodes, 21 peritoneal deposits and 16 liver metastases) were studied to evaluate c-MET expression by immunohistochemistry. For comparison, 12 sections of adjacent healthy colorectal mucosa were examined. RESULTS Statistically significant differences were present among normal tissues, colorectal adenomas and primary colorectal carcinomas (P=0.011). Normal tissues showed a negative or weak reaction in 66.67% and 33.33% of cases respectively. Expression of c-MET was positive in 47.8% of adenomas. A significant positive association was identified between c-MET high expression and degree of dysplasia (P=0.024). c-MET was highly expressed in 66.7% of primary colorectal carcinoma. Significant positive correlations were detected between c-MET expression and TNM stage (P=0.036), lymph node metastasis (LNM), peritoneal deposits and liver metastasis (P=0.038, P=0.094 and P=0.045, respectively). c-MET expression in metastatic tissues was significantly higher than that of the primary tumor. CONCLUSIONS c-MET expression is gradually up-regulated in the development and progression of colorectal cancer (CRC) from normal epithelium to adenoma to colorectal carcinoma to metastases.
Collapse
Affiliation(s)
- Mariana Fathy Gayyed
- 1 Department of Pathology, 2 Department of General Surgery, Faculty of Medicine, Minia University, Minia, Egypt ; 3 Department of General Surgery, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nehad M R Abd El-Maqsoud
- 1 Department of Pathology, 2 Department of General Surgery, Faculty of Medicine, Minia University, Minia, Egypt ; 3 Department of General Surgery, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Amr Abd El-Hameed El-Heeny
- 1 Department of Pathology, 2 Department of General Surgery, Faculty of Medicine, Minia University, Minia, Egypt ; 3 Department of General Surgery, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mostafa Fuad Mohammed
- 1 Department of Pathology, 2 Department of General Surgery, Faculty of Medicine, Minia University, Minia, Egypt ; 3 Department of General Surgery, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
11
|
Zhang Y, Jain RK, Zhu M. Recent Progress and Advances in HGF/MET-Targeted Therapeutic Agents for Cancer Treatment. Biomedicines 2015; 3:149-181. [PMID: 28536405 PMCID: PMC5344234 DOI: 10.3390/biomedicines3010149] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/22/2015] [Revised: 02/25/2015] [Accepted: 03/03/2015] [Indexed: 12/31/2022] Open
Abstract
The hepatocyte growth factor (HGF): MET axis is a ligand-mediated receptor tyrosine kinase pathway that is involved in multiple cellular functions, including proliferation, survival, motility, and morphogenesis. Aberrancy in the HGF/MET pathway has been reported in multiple tumor types and is associated with tumor stage and prognosis. Thus, targeting the HGF/MET pathway has become a potential therapeutic strategy in oncology development in the last two decades. A number of novel therapeutic agents-either as therapeutic proteins or small molecules that target the HGF/MET pathway-have been tested in patients with different tumor types in clinical studies. In this review, recent progress in HGF/MET pathway-targeted therapy for cancer treatment, the therapeutic potential of HGF/MET-targeted agents, and challenges in the development of such agents will be discussed.
Collapse
Affiliation(s)
- Yilong Zhang
- Department of Clinical Pharmacology, Modeling and Simulation, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | - Rajul K Jain
- Kite Pharma, Inc., 2225 Colorado Avenue, Santa Monica, CA 90404, USA.
| | - Min Zhu
- Department of Clinical Pharmacology, Modeling and Simulation, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| |
Collapse
|
12
|
The nerve growth factor signaling and its potential as therapeutic target for glaucoma. BIOMED RESEARCH INTERNATIONAL 2014; 2014:759473. [PMID: 25250333 PMCID: PMC4164261 DOI: 10.1155/2014/759473] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 03/08/2014] [Accepted: 08/12/2014] [Indexed: 12/25/2022]
Abstract
Neuroprotective therapies which focus on factors leading to retinal ganglion cells (RGCs) degeneration have been drawing more and more attention. The beneficial effects of nerve growth factor (NGF) on the glaucoma have been recently suggested, but its effects on eye tissue are complex and controversial in various studies. Recent clinical trials of systemically and topically administrated NGF demonstrate that NGF is effective in treating several ocular diseases, including glaucoma. NGF has two receptors named high affinity NGF tyrosine kinase receptor TrkA and low affinity receptor p75NTR. Both receptors exist in cells in retina like RGC (expressing TrkA) and glia cells (expressing p75NTR). NGF functions by binding to TrkA or p75NTR alone or both together. The binding of NGF to TrkA alone in RGC promotes RGC's survival and proliferation through activation of TrkA and several prosurvival pathways. In contrast, the binding of NGF to p75NTR leads to apoptosis although it also promotes survival in some cases. Binding of NGF to both TrkA and p75NTR at the same time leads to survival in which p75NTR functions as a TrkA helping receptor. This review discusses the current understanding of the NGF signaling in retina and the therapeutic implications in the treatment of glaucoma.
Collapse
|
13
|
Kwon MJ, Kim DH, Park HR, Shin HS, Kwon JH, Lee DJ, Kim JH, Cho SJ, Nam ES. Frequent hepatocyte growth factor overexpression and low frequency of c-Met gene amplification in human papillomavirus–negative tonsillar squamous cell carcinoma and their prognostic significances. Hum Pathol 2014; 45:1327-38. [DOI: 10.1016/j.humpath.2014.03.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/03/2014] [Revised: 03/07/2014] [Accepted: 03/14/2014] [Indexed: 10/25/2022]
|
14
|
Matias-Guiu X, Davidson B. Prognostic biomarkers in endometrial and ovarian carcinoma. Virchows Arch 2014; 464:315-31. [PMID: 24504546 DOI: 10.1007/s00428-013-1509-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/20/2013] [Revised: 11/05/2013] [Accepted: 11/07/2013] [Indexed: 02/06/2023]
Abstract
This article reviews the main prognostic and predictive biomarkers of endometrial (EC) and ovarian carcinoma (OC). In EC, prognosis still relies on conventional pathological features such as histological type and grade, as well as myometrial or lymphovascular space invasion. Estrogen receptor, p53, Ki-67, and ploidy analysis are the most promising biomarkers among a long list of molecules that have been proposed. Also, a number of putative predictive biomarkers have been proposed in molecular targeted therapy. In OC, prognosis is predominantly dependent on disease stage at diagnosis and the extent of residual disease at primary operation. Diagnostic markers which aid in establishing histological type in OC are available. However, not a single universally accepted predictive or prognostic marker exists to date. Targeted therapy has been growingly focused at in recent years, in view of the frequent development of chemoresistance at recurrent disease. The present review emphasizes the crucial role of correct pathological classification and stringent selection criteria of the material studied as basis for any evaluation of biological markers. It further emphasizes the promise of targeted therapy in EC and OC, while simultaneously highlighting the difficulties remaining before this can become standard of care.
Collapse
Affiliation(s)
- Xavier Matias-Guiu
- Department of Pathology and Molecular Genetics and Research Laboratory, Hospital Universitari Arnau de Vilanova, IRBLLEIDA, University of Lleida, Av. Alcalde Rovira Roure 80, 25198, Lleida, Spain,
| | | |
Collapse
|
15
|
Li M, Yi X, Ma L, Zhou Y. Hepatocyte growth factor and basic fibroblast growth factor regulate atrial fibrosis in patients with atrial fibrillation and rheumatic heart disease via the mitogen-activated protein kinase signaling pathway. Exp Ther Med 2013; 6:1121-1126. [PMID: 24223632 PMCID: PMC3820834 DOI: 10.3892/etm.2013.1274] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/26/2013] [Accepted: 08/16/2013] [Indexed: 01/14/2023] Open
Abstract
The aim of this study was to investigate the interrelation between basic fibroblast growth factor (bFGF), hepatocyte growth factor (HGF) and atrial fibrosis in patients with atrial fibrillation (AF) and rheumatic heart disease (RHD), and to explore the possible molecular mechanisms underlying this interrelation. Twenty patients with RHD who were scheduled for valve replacement were divided into two groups, comprising 10 cases with AF and 10 cases with sinus rhythm (SR). Clinical data were collected and a small sample of aseptic left atrial appendage was collected by the surgeon. Hematoxylin and eosin (H&E) and Masson's trichrome-stained sections were used to evaluate the cross-sectional area and level of fibrosis, respectively. The expression levels of bFGF and HGF were assessed using immunohistochemistry. The phosphorylation levels of mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2 (MEK1/2), c-Jun N-terminal kinase 1/2 (JNK1/2), extracellular signal-regulated kinase 1/2 (ERK1/2) and p38 in atrial tissue were measured using western blotting. Compared with the SR group, myocardial cell diameter was significantly expanded and there was increased collagen deposition in the AF group (P<0.05). The distribution of bFGF in the AF group was significantly higher than that in the SR group (P<0.05); however, HGF levels were significantly lower in the AF group (P<0.05). The phosphorylation levels of MEK1/2, ERK1/2, JNK1/2 and p38 in the AF group were significantly higher than those in the SR group (P<0.05). The results indicated that bFGF may promote the development of atrial fibrosis, while HGF may function in an opposite manner in patients with AF and RHD. The mitogen-activated protein kinase (MAPK) signaling pathway may be the molecular basis for these roles in atrial fibrosis.
Collapse
Affiliation(s)
- Mingjiang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | | | | | | |
Collapse
|
16
|
Burbridge MF, Bossard CJ, Saunier C, Fejes I, Bruno A, Léonce S, Ferry G, Da Violante G, Bouzom F, Cattan V, Jacquet-Bescond A, Comoglio PM, Lockhart BP, Boutin JA, Cordi A, Ortuno JC, Pierré A, Hickman JA, Cruzalegui FH, Depil S. S49076 is a novel kinase inhibitor of MET, AXL, and FGFR with strong preclinical activity alone and in association with bevacizumab. Mol Cancer Ther 2013; 12:1749-62. [PMID: 23804704 DOI: 10.1158/1535-7163.mct-13-0075] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/26/2022]
Abstract
Aberrant activity of the receptor tyrosine kinases MET, AXL, and FGFR1/2/3 has been associated with tumor progression in a wide variety of human malignancies, notably in instances of primary or acquired resistance to existing or emerging anticancer therapies. This study describes the preclinical characterization of S49076, a novel, potent inhibitor of MET, AXL/MER, and FGFR1/2/3. S49076 potently blocked cellular phosphorylation of MET, AXL, and FGFRs and inhibited downstream signaling in vitro and in vivo. In cell models, S49076 inhibited the proliferation of MET- and FGFR2-dependent gastric cancer cells, blocked MET-driven migration of lung carcinoma cells, and inhibited colony formation of hepatocarcinoma cells expressing FGFR1/2 and AXL. In tumor xenograft models, a good pharmacokinetic/pharmacodynamic relationship for MET and FGFR2 inhibition following oral administration of S49076 was established and correlated well with impact on tumor growth. MET, AXL, and the FGFRs have all been implicated in resistance to VEGF/VEGFR inhibitors such as bevacizumab. Accordingly, combination of S49076 with bevacizumab in colon carcinoma xenograft models led to near total inhibition of tumor growth. Moreover, S49076 alone caused tumor growth arrest in bevacizumab-resistant tumors. On the basis of these preclinical studies showing a favorable and novel pharmacologic profile of S49076, a phase I study is currently underway in patients with advanced solid tumors. Mol Cancer Ther; 12(9); 1749-62. ©2013 AACR.
Collapse
Affiliation(s)
- Mike F Burbridge
- Corresponding Author: Mike F. Burbridge, Oncology Research and Development Unit, Institut de Recherches Servier, 125 chemin de Ronde, 78290 Croissy-sur-Seine, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Brauer HA, Makowski L, Hoadley KA, Casbas-Hernandez P, Lang LJ, Romàn-Pèrez E, D'Arcy M, Freemerman AJ, Perou CM, Troester MA. Impact of tumor microenvironment and epithelial phenotypes on metabolism in breast cancer. Clin Cancer Res 2012; 19:571-85. [PMID: 23236214 DOI: 10.1158/1078-0432.ccr-12-2123] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Cancer cells have altered metabolism, with increased glucose uptake, glycolysis, and biomass production. This study conducted genomic and metabolomic analyses to elucidate how tumor and stromal genomic characteristics influence tumor metabolism. EXPERIMENTAL DESIGN Thirty-three breast tumors and six normal breast tissues were analyzed by gene expression microarray and by mass spectrometry for metabolites. Gene expression data and clinical characteristics were evaluated in association with metabolic phenotype. To evaluate the role of stromal interactions in altered metabolism, cocultures were conducted using breast cancer cells and primary cancer-associated fibroblasts (CAF). RESULTS Across all metabolites, unsupervised clustering resulted in two main sample clusters. Normal breast tissue and a subset of tumors with less aggressive clinical characteristics had lower levels of nucleic and amino acids and glycolysis byproducts, whereas more aggressive tumors had higher levels of these Warburg-associated metabolites. While tumor-intrinsic subtype did not predict metabolic phenotype, metabolic cluster was significantly associated with expression of a wound response signature. In cocultures, CAFs from basal-like breast cancers increased glucose uptake and basal-like epithelial cells increased glucose oxidation and glycogen synthesis, suggesting interplay of stromal and epithelial phenotypes on metabolism. Cytokine arrays identified hepatocyte growth factor (HGF) as a potential mediator of stromal-epithelial interaction and antibody neutralization of HGF resulted in reduced expression of glucose transporter 1 (GLUT1) and decreased glucose uptake by epithelium. CONCLUSIONS Both tumor/epithelial and stromal characteristics play important roles in metabolism. Warburg-like metabolism is influenced by changes in stromal-epithelial interactions, including altered expression of HGF/Met pathway and GLUT1 expression.
Collapse
Affiliation(s)
- Heather Ann Brauer
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Zhou J, Roh JW, Bandyopadhyay S, Chen Z, Munkarah AR, Hussein Y, Alosh B, Jazaerly T, Hayek K, Semaan A, Sood AK, Ali-Fehmi R. Overexpression of enhancer of zeste homolog 2 (EZH2) and focal adhesion kinase (FAK) in high grade endometrial carcinoma. Gynecol Oncol 2012; 128:344-8. [PMID: 22871469 DOI: 10.1016/j.ygyno.2012.07.128] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/17/2012] [Revised: 07/24/2012] [Accepted: 07/31/2012] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The deregulation of E-cadherin is associated with Src/FAK signaling axis and histone deacetylase (HDAC)/EZH2 activity. However, the association between EZH2 and FAK and its clinical significance in endometrial carcinoma has not been reported. METHODS 202 archived cases of endometrial carcinoma (1996-2000) were reviewed and divided into two subtypes. TMAs were developed as per established procedures. EZH2, FAK, and pFAK immunohistochemical stains were performed and the expression was scored as negative (0), low (1) and high (2). Proper statistical analysis was used to assess the correlation between the expression profiles and the clinicopathological parameters and clinical outcome. RESULTS A total of 141 (69.8%) type-1 tumors and 61 (30.2%) type-2 tumors were identified. EZH2 overexpression was identified in 7.6% of type-1 tumors vs. 63% of type-2 tumors (p<0.001). FAK and pFAK overexpression was only seen in 24.8% and 1.7% of Type-1 tumors as compared to 72% and 58.8% of type-2 tumors, respectively (p<0.001). A positive correlation between the expression of EZH2, FAK, pFAK and PTEN (p<0.0001) was found. The overexpression of EZH2, FAK, and pFAK were significantly associated with high histologic grade, angiolymphatic invasion, lymph node metastasis, myometrial invasion and cervical involvement (p<0.01). Kaplan-Meier analysis demonstrates that the overexpression of EZH2 (p=0.0024), FAK and pFAK (p=0.0001) was significantly associated with decreased overall survival. CONCLUSION The overexpression of EZH2, FAK and pFAK correlates with well established pathologic risk factors and may predict a more aggressive biologic behavior in endometrial carcinoma, transforming these proteins into potential therapeutic targets for treatment of endometrial cancer.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Pathology, Karmanos Cancer Center, Wayne State University, Detroit, Michigan 48201, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|