201
|
Masoumi-Moghaddam S, Amini A, Wei AQ, Robertson G, Morris DL. Intratumoral interleukin-6 predicts ascites formation in patients with epithelial ovarian cancer: A potential tool for close monitoring. J Ovarian Res 2015; 8:58. [PMID: 26282935 PMCID: PMC4539669 DOI: 10.1186/s13048-015-0183-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 07/29/2015] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The implication of IL-6 in the pathogenesis of epithelial ovarian cancer (EOC) is well documented. Accordingly, the clinicopathological significance of this cytokine in patients' ascites fluid or serum has largely been investigated. Since the main source of IL-6 secreted into the biological fluids is the tumor tissue, this study was designed to investigate the status and possible clinical relevance of the IL-6 expression in an array of EOC tissue specimens. METHODS Tissue samples obtained from ninety-eight consecutive patients with EOC were studied using immunohistochemistry. Clinicopathological characteristics and treatment related factors were collected from patient files. The relationship between the expression of the protein of interest and the study endpoints of disease-free survival (DFS) and overall survival (OS) were analyzed using the Kaplan-Meier method. For evaluating the predictive value of IL-6, logistic regression and cox proportional hazards models were employed. RESULTS An upregulation of IL-6 expression was observed in EOC tissues as compared with the normal samples (p < 0.0001). As regards the clinical relevance, IL-6 failed to predict OS, DFS and response to the platinum-based chemotherapy in EOC patients. In multivariate analysis, however, IL-6 was identified as an independent predictive factor for the development of post-treatment ascites (p:0.033). CONCLUSIONS Having the capability to predict the ascites formation, IL-6 might serve as a biomarker and a useful tool in EOC for monitoring purposes. IL-6 targeting for the prevention of the ascites development is a potential avenue for further investigation.
Collapse
Affiliation(s)
- Samar Masoumi-Moghaddam
- Department of Surgery, St George Hospital, The University of New South Wales, Gray Street, Kogarah, Sydney, NSW 2217, Australia.
| | - Afshin Amini
- Department of Surgery, St George Hospital, The University of New South Wales, Gray Street, Kogarah, Sydney, NSW 2217, Australia.
| | - Ai-Qun Wei
- Department of Orthopedic Surgery, St. George Hospital, The University of New South Wales, Gray Street, Kogarah, Sydney, NSW 2217, Australia.
| | - Gregory Robertson
- Department of Gynecology Oncology, St George Hospital, The University of New South Wales, Gray Street, Kogarah, Sydney, NSW 2217, Australia.
| | - David L Morris
- Department of Surgery, St George Hospital, The University of New South Wales, Gray Street, Kogarah, Sydney, NSW 2217, Australia.
| |
Collapse
|
202
|
Osuala KO, Sameni M, Shah S, Aggarwal N, Simonait ML, Franco OE, Hong Y, Hayward SW, Behbod F, Mattingly RR, Sloane BF. Il-6 signaling between ductal carcinoma in situ cells and carcinoma-associated fibroblasts mediates tumor cell growth and migration. BMC Cancer 2015; 15:584. [PMID: 26268945 PMCID: PMC4535667 DOI: 10.1186/s12885-015-1576-3] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 07/27/2015] [Indexed: 12/14/2022] Open
Abstract
Background Ductal carcinoma in situ (DCIS) is a non-obligate precursor lesion of invasive breast cancer in which approximately half the patients will progress to invasive cancer. Gaining a better understanding of DCIS progression may reduce overtreatment of patients. Expression of the pro-inflammatory cytokine interleukin-6 increases with pathological stage and grade, and is associated with poorer prognosis in breast cancer patients. Carcinoma associated fibroblasts (CAFs), which are present in the stroma of DCIS patients are known to secrete pro-inflammatory cytokines and promote tumor progression. Methods We hypothesized that IL-6 paracrine signaling between DCIS cells and CAFs mediates DCIS proliferation and migration. To test this hypothesis, we utilized the mammary architecture and microenvironment engineering or MAME model to study the interactions between human breast CAFs and human DCIS cells in 3D over time. We specifically inhibited autocrine and paracrine IL-6 signaling to determine its contribution to early stage tumor progression. Results Here, DCIS cells formed multicellular structures that exhibited increased proliferation and migration when cultured with CAFs. Treatment with an IL-6 neutralizing antibody inhibited growth and migration of the multicellular structures. Moreover, selective knockdown of IL-6 in CAFs, but not in DCIS cells, abrogated the migratory phenotype. Conclusion Our results suggest that paracrine IL-6 signaling between preinvasive DCIS cells and stromal CAFs represent an important factor in the initiation of DCIS progression to invasive breast carcinoma. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1576-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kingsley O Osuala
- Department of Pharmacology, Wayne State University, 540 East Canfield, Detroit, MI, 48201, USA. .,School of Medicine, Wayne State University, 540 East Canfield, Detroit, MI, 48201, USA.
| | - Mansoureh Sameni
- Department of Pharmacology, Wayne State University, 540 East Canfield, Detroit, MI, 48201, USA. .,School of Medicine, Wayne State University, 540 East Canfield, Detroit, MI, 48201, USA.
| | - Seema Shah
- Cancer Biology Program, Wayne State University, 540 East Canfield, Detroit, MI, 48201, USA. .,School of Medicine, Wayne State University, 540 East Canfield, Detroit, MI, 48201, USA.
| | - Neha Aggarwal
- Department of Physiology, Wayne State University, 540 East Canfield, Detroit, MI, 48201, USA. .,School of Medicine, Wayne State University, 540 East Canfield, Detroit, MI, 48201, USA.
| | - Michelle L Simonait
- School of Medicine, Wayne State University, 540 East Canfield, Detroit, MI, 48201, USA.
| | - Omar E Franco
- Department of Surgery, NorthShore University Health System Research Institute, 1001 University Place, Evanston, IL, 60201, USA.
| | - Yan Hong
- Division of Cancer and Developmental Biology, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.
| | - Simon W Hayward
- Department of Surgery, NorthShore University Health System Research Institute, 1001 University Place, Evanston, IL, 60201, USA.
| | - Fariba Behbod
- Division of Cancer and Developmental Biology, The University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA.
| | - Raymond R Mattingly
- Department of Pharmacology, Wayne State University, 540 East Canfield, Detroit, MI, 48201, USA. .,School of Medicine, Wayne State University, 540 East Canfield, Detroit, MI, 48201, USA.
| | - Bonnie F Sloane
- Department of Pharmacology, Wayne State University, 540 East Canfield, Detroit, MI, 48201, USA. .,School of Medicine, Wayne State University, 540 East Canfield, Detroit, MI, 48201, USA.
| |
Collapse
|
203
|
SPECT imaging of interleukin-6 receptor in ovarian tumor xenografts with a novel radiotracer of 99mTc-HYNIC-Aca-LSLITRL. Amino Acids 2015; 48:91-101. [DOI: 10.1007/s00726-015-2060-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/24/2015] [Indexed: 12/19/2022]
|
204
|
Gopinathan G, Milagre C, Pearce OMT, Reynolds LE, Hodivala-Dilke K, Leinster DA, Zhong H, Hollingsworth RE, Thompson R, Whiteford JR, Balkwill F. Interleukin-6 Stimulates Defective Angiogenesis. Cancer Res 2015; 75:3098-107. [PMID: 26081809 PMCID: PMC4527186 DOI: 10.1158/0008-5472.can-15-1227] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 05/13/2015] [Indexed: 12/19/2022]
Abstract
The cytokine IL6 has a number of tumor-promoting activities in human and experimental cancers, but its potential as an angiogenic agent has not been fully investigated. Here, we show that IL6 can directly induce vessel sprouting in the ex vivo aortic ring model, as well as endothelial cell proliferation and migration, with similar potency to VEGF. However, IL6-stimulated aortic ring vessel sprouts had defective pericyte coverage compared with VEGF-stimulated vessels. The mechanism of IL6 action on pericytes involved stimulation of the Notch ligand Jagged1 as well as angiopoietin2 (Ang2). When peritoneal xenografts of ovarian cancer were treated with an anti-IL6 antibody, pericyte coverage of vessels was restored. In addition, in human ovarian cancer biopsies, there was an association between levels of IL6 mRNA, Jagged1, and Ang2. Our findings have implications for the use of cancer therapies that target VEGF or IL6 and for understanding abnormal angiogenesis in cancers, chronic inflammatory disease, and stroke.
Collapse
Affiliation(s)
- Ganga Gopinathan
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Carla Milagre
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Oliver M T Pearce
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Louise E Reynolds
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Kairbaan Hodivala-Dilke
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - David A Leinster
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Haihong Zhong
- MedImmune, One MedImmune Way, Gaithersburg, Maryland
| | | | - Richard Thompson
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - James R Whiteford
- William Harvey Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Frances Balkwill
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom.
| |
Collapse
|
205
|
Matsuo K, Hasegawa K, Yoshino K, Murakami R, Hisamatsu T, Stone RL, Previs RA, Hansen JM, Ikeda Y, Miyara A, Hiramatsu K, Enomoto T, Fujiwara K, Matsumura N, Konishi I, Roman LD, Gabra H, Fotopoulou C, Sood AK. Venous thromboembolism, interleukin-6 and survival outcomes in patients with advanced ovarian clear cell carcinoma. Eur J Cancer 2015; 51:1978-88. [PMID: 26238017 DOI: 10.1016/j.ejca.2015.07.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/13/2015] [Accepted: 07/13/2015] [Indexed: 01/07/2023]
Abstract
BACKGROUND We compared survival outcomes and risk of venous thromboembolism (VTE) among patients with advanced and early-stage ovarian clear cell carcinoma (OCCC) and serous ovarian carcinoma (SOC), as well as potential links with interleukin-6 (IL-6) levels. METHODS A multicenter case-control study was conducted in 370 patients with OCCC and 938 with SOC. In a subset of 200 cases, pretreatment plasma IL-6 levels were examined. FINDINGS Patients with advanced OCCC had the highest 2-year cumulative VTE rates (advanced OCCC 43.1%, advanced SOC 16.2%, early-stage OCCC 11.9% and early-stage SOC 6.4%, P<0.0001) and the highest median levels of IL-6 (advanced OCCC 17.8 pg/mL, advanced SOC 9.0 pg/mL, early-stage OCCC 4.2 pg/mL and early-stage SOC 5.0 pg/mL, P=0.006). Advanced OCCC (hazard ratio [HR] 3.38, P<0.0001), thrombocytosis (HR 1.42, P=0.032) and elevated IL-6 (HR 8.90, P=0.046) were independent predictors of VTE. In multivariate analysis, patients with advanced OCCC had significantly poorer 5-year progression-free and overall survival rates than those with advanced SOC (P<0.01), and thrombocytosis was an independent predictor of decreased survival outcomes (P<0.01). Elevated IL-6 levels led to poorer 2-year progression-free survival rates in patients with OCCC (50% versus 87.5%, HR 4.89, P=0.016) than in those with SOC (24.9% versus 40.8%, HR 1.40, P=0.07). INTERPRETATION Advanced OCCC is associated with an increased incidence of VTE and decreased survival outcomes, which has major implications for clinical management of OCCC.
Collapse
Affiliation(s)
- Koji Matsuo
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA.
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Kiyoshi Yoshino
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takeshi Hisamatsu
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan; Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rebecca L Stone
- Department of Obstetrics and Gynecology, The Johns Hopkins University, Baltimore, MD, USA
| | - Rebecca A Previs
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jean M Hansen
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuji Ikeda
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan; Department of Obstetrics and Gynecology, The University of Tokyo Faculty of Medicine, Tokyo, Japan
| | - Akiko Miyara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Kosuke Hiramatsu
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medicine, Niigata, Japan
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Noriomi Matsumura
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ikuo Konishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Lynda D Roman
- Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Hani Gabra
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Christina Fotopoulou
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
206
|
Masoumi-Moghaddam S, Amini A, Wei AQ, Robertson G, Morris DL. Sprouty2 protein in prediction of post-treatment ascites in epithelial ovarian cancer treated with adjuvant carbotaxol chemotherapy. Am J Cancer Res 2015; 5:2498-2507. [PMID: 26396926 PMCID: PMC4568786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/19/2015] [Indexed: 06/05/2023] Open
Abstract
Ascites development and resistance to chemotherapy with carbotaxol are common clinical problems in epithelial ovarian cancer, partly due to the activation of MAPK/ERK signaling. Sprouty proteins are negative modulators of MAPK/ERK pathway, but their role in predicting resistance to carbotaxol chemotherapy and ascites development is unknown. In this study, we evaluated the expression of Sprouty protein isoforms by immunohistochemistry. The associations between the Sprouty expression and the clinicopathological features, including chemoresistance and the presence of ascites, were then explored. We found that the decreased expression of Spry2 was correlated with the post-treatment development of ascites and represented an independent predictor of this condition in carbotaxol-treated patients. However, no association was observed between the Sprouty expression and chemoresistance. In conclusion, our results suggest that Spry2 may be useful for patient follow-up and monitoring as it predicts the development of ascites in epithelial ovarian cancer cases treated with carbotaxol.
Collapse
Affiliation(s)
- Samar Masoumi-Moghaddam
- Department of Surgery, St George Hospital, The University of New South WalesKogarah, Sydney NSW 2217, Australia
| | - Afshin Amini
- Department of Surgery, St George Hospital, The University of New South WalesKogarah, Sydney NSW 2217, Australia
| | - Ai-Qun Wei
- Department of Orthopedic Surgery, St George Hospital, The University of New South WalesKogarah, Sydney NSW 2217, Australia
| | - Gregory Robertson
- Department of Gynecology Oncology, St George Hospital, The University of New South WalesKogarah, Sydney NSW 2217, Australia
| | - David L Morris
- Department of Surgery, St George Hospital, The University of New South WalesKogarah, Sydney NSW 2217, Australia
| |
Collapse
|
207
|
Chen R, Chen B. Siltuximab (CNTO 328): a promising option for human malignancies. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3455-8. [PMID: 26170629 PMCID: PMC4494175 DOI: 10.2147/dddt.s86438] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Siltuximab (CNTO 328) is a promising antibody-drug conjugate targeting cytokine interleukin-6 (IL-6). It is highly binding to IL-6 and thus neutralizing IL-6 bioactivity and promoting death of tumor cell. In this review, we mainly focus on the mechanisms, clinical studies, and adverse effect of siltuximab in the treatment of human malignancies. We also provide our recommendations for siltuximab treatment in the future.
Collapse
Affiliation(s)
- Runzhe Chen
- Department of Hematology and Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| |
Collapse
|
208
|
Signaling Circuits and Regulation of Immune Suppression by Ovarian Tumor-Associated Macrophages. Vaccines (Basel) 2015; 3:448-66. [PMID: 26343197 PMCID: PMC4494355 DOI: 10.3390/vaccines3020448] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 05/24/2015] [Accepted: 05/26/2015] [Indexed: 12/25/2022] Open
Abstract
The barriers presented by immune suppression in the ovarian tumor microenvironment present one of the biggest challenges to development of successful tumor vaccine strategies for prevention of disease recurrence and progression following primary surgery and chemotherapy. New insights gained over the last decade have revealed multiple mechanisms of immune regulation, with ovarian tumor-associated macrophages/DC likely to fulfill a central role in creating a highly immunosuppressive milieu that supports disease progression and blocks anti-tumor immunity. This review provides an appraisal of some of the key signaling pathways that may contribute to immune suppression in ovarian cancer, with a particular focus on the potential involvement of the c-KIT/PI3K/AKT, wnt/β-catenin, IL-6/STAT3 and AhR signaling pathways in regulation of indoleamine 2,3-dioxygenase expression in tumor-associated macrophages. Knowledge of intercellular and intracellular circuits that shape immune suppression may afford insights for development of adjuvant treatments that alleviate immunosuppression in the tumor microenvironment and enhance the clinical efficacy of ovarian tumor vaccines.
Collapse
|
209
|
Chandra D, Jahangir A, Cornelis F, Rombauts K, Meheus L, Jorcyk CL, Gravekamp C. Cryoablation and Meriva have strong therapeutic effect on triple-negative breast cancer. Oncoimmunology 2015; 5:e1049802. [PMID: 26942057 DOI: 10.1080/2162402x.2015.1049802] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 05/01/2015] [Accepted: 05/04/2015] [Indexed: 02/08/2023] Open
Abstract
Interleukin-6, a cytokine produced particularly by triple-negative breast cancers, strongly inhibits T cell responses in the tumor microenvironment. Here we tested cryoablation combined with Meriva (a lecithin delivery system of curcumin with improved bioavailability) in mice with metastatic breast cancer (4T1). Cryoablation involves killing of tumor cells through freezing and thawing, resulting in recruitment of tumor-specific T cells, while curcumin stimulates T cells through the reduction of IL-6 in the TME. Cryoablation plus Meriva accumulated and activated CD8+ T cells to multiple tumor-associated antigens such as Mage-b and Survivin (both expressed by 4T1 tumors). This correlated with a nearly complete reduction of 4T1 primary tumors and lung metastases while little effect was observed from saline or Meriva alone (28 d after tumor cell injection). The survival rate in the group of cryoablation plus Meriva was significantly improved compared to all control groups. Using a less aggressive 4T1 model expressing luciferase (4T1.2luc3), we demonstrated that all mice receiving saline or Meriva developed metastases in the lungs and a primary tumor (38 d after tumor cell injection; and died soon after that), but not the mice receiving cryoablation or cryoablation plus Meriva. However, on day 58 the mice receiving cryoablation developed 4T1.2luc3 metastases in the lungs, while mice receiving cryoablation plus Meriva were free of metastases. These results strongly suggest that cryoablation delayed the development of lung metastases on the short-term, but Meriva administered after cryoablation was significantly better in delaying the development of lung metastases and survival on the long-term.
Collapse
Affiliation(s)
- Dinesh Chandra
- Department of Microbiology and Immunology; Albert Einstein College of Medicine ; Bronx, NY USA
| | - Arthee Jahangir
- Department of Microbiology and Immunology; Albert Einstein College of Medicine ; Bronx, NY USA
| | | | - Klara Rombauts
- Anticancer Fund ; Boechoutlaan 221 ; Strombeek-Bever, Belgium
| | - Lydie Meheus
- Anticancer Fund ; Boechoutlaan 221 ; Strombeek-Bever, Belgium
| | - Cheryl L Jorcyk
- Department of Biological Sciences; Boise State University ; Boise, ID USA
| | - Claudia Gravekamp
- Department of Microbiology and Immunology; Albert Einstein College of Medicine ; Bronx, NY USA
| |
Collapse
|
210
|
Browne A, Tookman LA, Ingemarsdotter CK, Bouwman RD, Pirlo K, Wang Y, McNeish IA, Lockley M. Pharmacological Inhibition of β3 Integrin Reduces the Inflammatory Toxicities Caused by Oncolytic Adenovirus without Compromising Anticancer Activity. Cancer Res 2015; 75:2811-21. [PMID: 25977332 DOI: 10.1158/0008-5472.can-14-3761] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 04/07/2015] [Indexed: 01/28/2023]
Abstract
Adenoviruses have been clinically tested as anticancer therapies but their utility has been severely limited by rapid, systemic cytokine release and consequent inflammatory toxicity. Here, we describe a new approach to tackling these dangerous side effects. Using human ovarian cancer cell lines as well as malignant epithelial cells harvested from the ascites of women with ovarian cancer, we show that tumor cells do not produce cytokines in the first 24 hours following in vitro infection with the oncolytic adenovirus dl922-947. In contrast, dl922-947 does induce inflammatory cytokines at early time points following intraperitoneal delivery in mice with human ovarian cancer intraperitoneal xenografts. In these animals, cytokines originate predominantly in murine tissues, especially in macrophage-rich organs such as the spleen. We use a nonreplicating adenovirus to confirm that early cytokine production is independent of adenoviral replication. Using β3 integrin knockout mice injected intraperitoneally with dl922-947 and β3 null murine peritoneal macrophages, we confirm a role for macrophage cell surface β3 integrin in this dl922-947-induced inflammation. We present new evidence that co-administration of a cyclic RGD-mimetic-specific inhibitor of β3 integrin significantly attenuates the cytokine release and inflammatory hepatic toxicity induced by dl922-947 in an intraperitoneal murine model of ovarian cancer. Importantly, we find no evidence that β3 inhibition compromises viral infectivity and oncolysis in vitro or anticancer efficacy in vivo. By enabling safe, systemic delivery of replicating adenoviruses, this novel approach could have a major impact on the future development of these effective anticancer agents.
Collapse
Affiliation(s)
- Ashley Browne
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Laura A Tookman
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Carin K Ingemarsdotter
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Russell D Bouwman
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Katrina Pirlo
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Yaohe Wang
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Iain A McNeish
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom. Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Michelle Lockley
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom.
| |
Collapse
|
211
|
Yanaihara N, Hirata Y, Yamaguchi N, Noguchi Y, Saito M, Nagata C, Takakura S, Yamada K, Okamoto A. Antitumor effects of interleukin-6 (IL-6)/interleukin-6 receptor (IL-6R) signaling pathway inhibition in clear cell carcinoma of the ovary. Mol Carcinog 2015; 55:832-41. [DOI: 10.1002/mc.22325] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 03/06/2015] [Accepted: 03/16/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Nozomu Yanaihara
- Department of Obstetrics and Gynecology; The Jikei University School of Medicine; Tokyo Japan
| | - Yukihiro Hirata
- Department of Obstetrics and Gynecology; The Jikei University School of Medicine; Tokyo Japan
| | - Noriko Yamaguchi
- Department of Obstetrics and Gynecology; The Jikei University School of Medicine; Tokyo Japan
| | - Yukiko Noguchi
- Department of Obstetrics and Gynecology; The Jikei University School of Medicine; Tokyo Japan
| | - Misato Saito
- Department of Obstetrics and Gynecology; The Jikei University School of Medicine; Tokyo Japan
| | - Chie Nagata
- Department of Obstetrics and Gynecology; The Jikei University School of Medicine; Tokyo Japan
- Department of Education for Clinical Research; National Center for Child Health and Development; Tokyo Japan
| | - Satoshi Takakura
- Department of Obstetrics and Gynecology; The Jikei University School of Medicine; Tokyo Japan
| | - Kyosuke Yamada
- Department of Obstetrics and Gynecology; The Jikei University School of Medicine; Tokyo Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology; The Jikei University School of Medicine; Tokyo Japan
| |
Collapse
|
212
|
IL-6-mediated environmental conditioning of defective Th1 differentiation dampens antitumour immune responses in old age. Nat Commun 2015; 6:6702. [PMID: 25850032 PMCID: PMC4396369 DOI: 10.1038/ncomms7702] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 02/20/2015] [Indexed: 12/14/2022] Open
Abstract
Decline in immune function and inflammation concomitantly develop with ageing. Here we focus on the impact of this inflammatory environment on T cells, and demonstrate that in contrast to successful tumour elimination in young mice, replenishment of tumour-specific CD4+ T cells fails to induce tumour regression in aged hosts. The impaired antitumour effect of CD4+ T cells with their defective Th1 differentiation in an aged environment is restored by interleukin (IL)-6 blockade or IL-6 deficiency. IL-6 blockade also restores the impaired ability of CD4+ T cells to promote CD8+ T-cell-dependent tumour elimination in aged mice, which requires IFN-γ. Furthermore, IL-6-stimulated production of IL-4/IL-21 through c-Maf induction is responsible for impaired Th1 differentiation. IL-6 also contributes to IL-10 production from CD4+ T cells in aged mice, causing attenuated responses of CD8+ T cells. These findings suggest that IL-6 serves as an extrinsic factor counteracting CD4+ T-cell-mediated immunity against tumour in old age. T-cell responses are dysregulated in aged humans and mice, which leads to poor antitumour responses. Here, the authors demonstrate that this phenomenon is at least partially due to an overproduction of IL-6 caused by ageing and its inhibitory effect on Th1 differentiation of tumour-specific CD4 T cells.
Collapse
|
213
|
Milagre CS, Gopinathan G, Everitt G, Thompson RG, Kulbe H, Zhong H, Hollingsworth RE, Grose R, Bowtell DDL, Hochhauser D, Balkwill FR. Adaptive Upregulation of EGFR Limits Attenuation of Tumor Growth by Neutralizing IL6 Antibodies, with Implications for Combined Therapy in Ovarian Cancer. Cancer Res 2015; 75:1255-64. [PMID: 25670170 PMCID: PMC4384986 DOI: 10.1158/0008-5472.can-14-1801] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 01/16/2015] [Indexed: 01/05/2023]
Abstract
Excess production of the proinflammatory IL6 has both local and systemic tumor-promoting activity in many cancers, including ovarian cancer. However, treatment of advanced ovarian cancer patients with a neutralizing IL6 antibody yielded little efficacy in a previous phase II clinical trial. Here, we report results that may explain this outcome, based on the finding that neutralizing antibodies to IL6 and STAT3 inhibition are sufficient to upregulate the EGFR pathway in high-grade serous and other ovarian cancer cells. Cell treatment with the EGFR inhibitor gefitinib abolished upregulation of the EGFR pathway. Combining neutralizing IL6 antibodies and gefitinib inhibited malignant cell growth in 2D and 3D culture. We found that ErbB-1 was localized predominantly in the nucleus of ovarian cancer cells examined, contrasting with plasma membrane localization in lung cancer cells. Treatment with anti-IL6, gefitinib, or their combination all led to partial restoration of ErbB-1 on the plasma membrane. In vivo experiments confirmed the effects of IL6 inhibition on the EGFR pathway and the enhanced activity of a combination of anti-IL6 antibodies and gefitinib on malignant cell growth. Taken together, our results offer a preclinical rationale to combine anti-IL6 and gefitinib to treat patients with advanced stage ovarian cancer.
Collapse
Affiliation(s)
- Carla S Milagre
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Ganga Gopinathan
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Gemma Everitt
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Richard G Thompson
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Hagen Kulbe
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Haihong Zhong
- MedImmune, One MedImmune Way, Gaithersburg, Maryland
| | | | - Richard Grose
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - David D L Bowtell
- Cancer Genomics and Genetics Program, Peter MacCallum Cancer Centre, Research Division, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | - Frances R Balkwill
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom.
| |
Collapse
|
214
|
Transcriptional regulation of chemokine expression in ovarian cancer. Biomolecules 2015; 5:223-43. [PMID: 25790431 PMCID: PMC4384120 DOI: 10.3390/biom5010223] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/04/2015] [Accepted: 03/09/2015] [Indexed: 12/14/2022] Open
Abstract
The increased expression of pro-inflammatory and pro-angiogenic chemokines contributes to ovarian cancer progression through the induction of tumor cell proliferation, survival, angiogenesis, and metastasis. The substantial potential of these chemokines to facilitate the progression and metastasis of ovarian cancer underscores the need for their stringent transcriptional regulation. In this Review, we highlight the key mechanisms that regulate the transcription of pro-inflammatory chemokines in ovarian cancer cells, and that have important roles in controlling ovarian cancer progression. We further discuss the potential mechanisms underlying the increased chemokine expression in drug resistance, along with our perspective for future studies.
Collapse
|
215
|
Pan D, Xu L, Liu H, Zhang W, Liu W, Liu Y, Fu Q, Xu J. High expression of interleukin-11 is an independent indicator of poor prognosis in clear-cell renal cell carcinoma. Cancer Sci 2015; 106:592-7. [PMID: 25702890 PMCID: PMC4452160 DOI: 10.1111/cas.12638] [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] [Scholar Register] [Received: 12/13/2014] [Revised: 02/05/2015] [Accepted: 02/12/2015] [Indexed: 01/16/2023] Open
Abstract
Interleukin-11 (IL-11), a member of the IL-6 family of cytokines, exerts pleiotropic oncogenic activities by stimulating angiogenesis and metastasis in many cancer types. The present study aims to evaluate the impact of IL-11 expression on recurrence and mortality of patients with clear-cell renal cell carcinoma (ccRCC). We retrospectively enrolled 193 ccRCC patients undergoing nephrectomy at a single center. Clinicopathologic features, recurrence-free survival (RFS) and overall survival (OS) were recorded. IL-11 intensity was assessed by immunohistochemistry in tumor specimens. The Kaplan–Meier method was applied to compare survival curves. Cox regression models were used to analyze the impact of prognostic factors on RFS and OS. The concordance index (C-index) was calculated to assess predictive accuracy. High IL-11 expression is associated with increased risk of recurrence and poor survival for ccRCC patients (P < 0.001 and P < 0.001, respectively), especially those with early-stage disease (TNM stage I + II). Multivariate analyses confirmed that IL-11 expression was an independent prognostic factor for RFS and OS (P = 0.006 and P = 0.008, respectively). The predictive accuracy of well-established prognostic models was improved when IL-11 expression was integrated. In conclusion, high IL-11 expression is an independent predictor of poor prognosis in ccRCC patients. It may help identify patients who could benefit from additional treatments and closer follow up.
Collapse
Affiliation(s)
- Deng Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Le Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haiou Liu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Weijuan Zhang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Weisi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yidong Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qiang Fu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
216
|
Block MS, Maurer MJ, Goergen K, Kalli KR, Erskine CL, Behrens MD, Oberg AL, Knutson KL. Plasma immune analytes in patients with epithelial ovarian cancer. Cytokine 2015; 73:108-13. [PMID: 25743245 DOI: 10.1016/j.cyto.2015.01.035] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 01/21/2015] [Accepted: 01/28/2015] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Inflammation is a common feature of epithelial ovarian cancer (EOC), and measurement of plasma markers of inflammation might identify candidate markers for use in screening or presurgical evaluation of patients with adnexal masses. METHODS Plasma specimens from cohorts of 100 patients with advanced EOC (AJCC Stage III and IV), 50 patients with early stage EOC (Stage I and II), and 50 patients with benign surgical conditions were assayed for concentrations of multiple cytokines, toll-like receptor agonists, and vascular growth factors via ELISA and electrochemiluminescence. Immune proteins were then analyzed for association with EOC. Differences in plasma protein levels between benign, early, and advanced EOC patient groups were assessed with and without adjustment for plasma cancer antigen 125 (CA-125) levels. RESULTS Out of 23 proteins tested, six-including interferon gamma (IFNγ), interleukin 6 (IL-6), IL-8, IL-10, tumor necrosis factor alpha (TNFα), and placental growth factor (PlGF)-were univariately associated with EOC (all p<0.005), and one-IL-6-was associated with early stage EOC (p<0.0001). Heat shock protein 90kDa beta member 1 (HSP90B1, gp96) was associated with EOC and early stage EOC with borderline statistical significance (p=0.039 and p=0.026, respectively). However, when adjusted for (CA-125), only HSP90B1 independently predicted EOC (p=0.008), as well as early stage EOC (p=0.014). CONCLUSIONS Multiple plasma cytokines, including IFNγ, IL-6, IL-8, IL-10, TNFα, PlGF, and HSP90B1 are associated with EOC. Of these, HSP90B1 is associated with EOC independent from the biomarker CA-125.
Collapse
Affiliation(s)
- Matthew S Block
- Department of Oncology, Mayo Clinic, Rochester, MN, United States; Department of Immunology, Mayo Clinic, Rochester, MN, United States.
| | - Matthew J Maurer
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, United States
| | - Krista Goergen
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, United States
| | - Kimberly R Kalli
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | | | | | - Ann L Oberg
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, United States
| | - Keith L Knutson
- Vaccine and Gene Therapy Institute, Port Saint Lucie, FL, United States; Department of Immunology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
217
|
Abstract
Clinical outcomes, such as recurrence-free survival and overall survival, in ovarian cancer are quite variable, independent of common characteristics such as stage, response to therapy, and grade. This disparity in outcomes warrants further exploration and therapeutic targeting into the interaction between the tumor and host. One compelling host characteristic that contributes both to the initiation and progression of ovarian cancer is the immune system. Hundreds of studies have confirmed a prominent role for the immune system in modifying the clinical course of the disease. Recent studies also show that anti-tumor immunity is often negated by immune regulatory cells present in the tumor microenvironment. Regulatory immune cells also directly enhance the pathogenesis through the release of various cytokines and chemokines, which together form an integrated pathological network. Thus, in the future, research into immunotherapy targeting ovarian cancer will probably become increasingly focused on combination approaches that simultaneously augment immunity while preventing local immune suppression. In this article, we summarize important immunological targets that influence ovarian cancer outcome as well as include an update on newer immunotherapeutic strategies.
Collapse
Affiliation(s)
- Keith L Knutson
- Cancer Vaccines and Immune Therapies Program, The Vaccine and Gene Therapy Institute of Florida, 9801 SW Discovery Way, Port St. Lucie, FL, 34949, USA,
| | | | | | | |
Collapse
|
218
|
Isobe A, Sawada K, Kinose Y, Ohyagi-Hara C, Nakatsuka E, Makino H, Ogura T, Mizuno T, Suzuki N, Morii E, Nakamura K, Sawada I, Toda A, Hashimoto K, Mabuchi S, Ohta T, Morishige KI, Kurachi H, Kimura T. Interleukin 6 receptor is an independent prognostic factor and a potential therapeutic target of ovarian cancer. PLoS One 2015; 10:e0118080. [PMID: 25658637 PMCID: PMC4319819 DOI: 10.1371/journal.pone.0118080] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 01/05/2015] [Indexed: 01/01/2023] Open
Abstract
Ovarian cancer remains the most lethal gynecologic cancer and new targeted molecular therapies against this miserable disease continue to be challenging. In this study, we analyzed the expressional patterns of Interleukin-6 (IL-6) and its receptor (IL-6R) expression in ovarian cancer tissues, evaluated the impact of these expressions on clinical outcomes of patients, and found that a high-level of IL-6R expression but not IL-6 expression in cancer cells is an independent prognostic factor. In in vitro analyses using ovarian cell lines, while six (RMUG-S, RMG-1, OVISE, A2780, SKOV3ip1 and OVCAR-3) of seven overexpressed IL-6R compared with a primary normal ovarian surface epithelium, only two (RMG-1, OVISE) of seven cell lines overexpressed IL-6, suggesting that IL-6/IL-6R signaling exerts in a paracrine manner in certain types of ovarian cancer cells. Ovarian cancer ascites were collected from patients, and we found that primary CD11b+CD14+ cells, which were predominantly M2-polarized macrophages, are the major source of IL-6 production in an ovarian cancer microenvironment. When CD11b+CD14+ cells were co-cultured with cancer cells, both the invasion and the proliferation of cancer cells were robustly promoted and these promotions were almost completely inhibited by pretreatment with anti-IL-6R antibody (tocilizumab). The data presented herein suggest a rationale for anti-IL-6/IL-6R therapy to suppress the peritoneal spread of ovarian cancer, and represent evidence of the therapeutic potential of anti-IL-6R therapy for ovarian cancer treatment.
Collapse
Affiliation(s)
- Aki Isobe
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kenjiro Sawada
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- * E-mail:
| | - Yasuto Kinose
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Chifumi Ohyagi-Hara
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Obstetrics and Gynecology, JCHO Osaka Hospital, Osaka, Japan
| | - Erika Nakatsuka
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiroshi Makino
- Department of Obstetrics and Gynecology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tomonori Ogura
- Department of Obstetrics and Gynecology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tomoko Mizuno
- Department of Obstetrics and Gynecology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Noriko Suzuki
- Department of Obstetrics and Gynecology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Koji Nakamura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ikuko Sawada
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Aska Toda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kae Hashimoto
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Seiji Mabuchi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tsuyoshi Ohta
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Ken-ichirou Morishige
- Department of Obstetrics and Gynecology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hirohisa Kurachi
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
219
|
Coward JIG, Middleton K, Murphy F. New perspectives on targeted therapy in ovarian cancer. Int J Womens Health 2015; 7:189-203. [PMID: 25678824 PMCID: PMC4324539 DOI: 10.2147/ijwh.s52379] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Epithelial ovarian cancer remains the most lethal gynecologic malignancy. During the last 15 years, there has been only marginal improvement in 5 year overall survival. These daunting statistics are compounded by the fact that despite all subtypes exhibiting striking heterogeneity, their systemic management remains identical. Although changes to the scheduling and administration of chemotherapy have improved outcomes to a degree, a therapeutic ceiling is being reached with this approach, resulting in a number of trials investigating the efficacy of targeted therapies alongside standard treatment algorithms. Furthermore, there is an urge to develop subtype-specific studies in an attempt to improve outcomes, which currently remain poor. This review summarizes the key studies with antiangiogenic agents, poly(adenosine diphosphate [ADP]-ribose) inhibitors, and epidermal growth factor receptor/human epidermal growth factor receptor family targeting, in addition to folate receptor antagonists and insulin growth factor receptor inhibitors. The efficacy of treatment paradigms used in non-ovarian malignancies for type I tumors is also highlighted, in addition to recent advances in appropriate patient stratification for targeted therapies in epithelial ovarian cancer.
Collapse
Affiliation(s)
- Jermaine IG Coward
- Mater Health Services, Raymond Terrace, South Brisbane, QLD, Australia
- Inflammtion and Cancer Therapeutics Group, Mater Research, University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, QLD, Australia
- School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Kathryn Middleton
- Mater Health Services, Raymond Terrace, South Brisbane, QLD, Australia
| | - Felicity Murphy
- Mater Health Services, Raymond Terrace, South Brisbane, QLD, Australia
| |
Collapse
|
220
|
Diakos CI, Charles KA, McMillan DC, Clarke SJ. Cancer-related inflammation and treatment effectiveness. Lancet Oncol 2015; 15:e493-503. [PMID: 25281468 DOI: 10.1016/s1470-2045(14)70263-3] [Citation(s) in RCA: 1604] [Impact Index Per Article: 160.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Inflammation is a recognised hallmark of cancer that substantially contributes to the development and progression of malignancies. In established cancers, there is increasing evidence for the roles that local immune response and systemic inflammation have in progression of tumours and survival of patients with cancer. This knowledge provides an opportunity to target these inflammatory responses to improve patient outcomes. In this Review, we examine the complex interplay between local immune responses and systemic inflammation, and their influence on clinical outcomes, and propose potential anti-inflammatory interventions for patients with cancer.
Collapse
Affiliation(s)
- Connie I Diakos
- Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Kellie A Charles
- School of Medical Sciences (Pharmacology), Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Donald C McMillan
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Royal Infirmary, Glasgow, UK
| | - Stephen J Clarke
- Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital, Sydney, NSW, Australia.
| |
Collapse
|
221
|
Abstract
Human IL6 is a cytokine produced by many cell types that has pleiotropic effects. In agreement, anti-IL6 therapy reduces inflammation, hepatic acute phase proteins, and anemia and has antiangiogenic effects. Blocking IL6 has demonstrated therapeutic efficacy with drug registration in Castleman disease and inflammatory diseases (rheumatoid arthritis) without major toxicity. Interestingly, the inhibition of C-reactive protein (CRP) production is a trustworthy surrogate marker of anti-IL6 therapy efficacy. Clinically registered IL6 inhibitors include siltuximab, an anti-IL6 mAb, and tocilizumab, an anti-IL6R mAb. In various cancers, in particular plasma cell cancers, large randomized trials showed no efficacy of IL6 inhibitors, despite a full inhibition of CRP production in treated patients in vivo, the numerous data showing an involvement of IL6 in these diseases, and initial short-term treatments demonstrating a dramatic inhibition of cancer cell proliferation in vivo. A likely explanation is the plasticity of cancer cells, with the presence of various subclones, making the outgrowth of cancer subclones possible using growth factors other than IL6. In addition, current therapeutic strategies used in these cancers already target IL6 activity. Thus, anti-IL6 therapeutics are able to neutralize IL6 production in vivo and are safe and useful in inflammatory diseases and Castleman disease.
Collapse
Affiliation(s)
- Jean-François Rossi
- Department of Hematology, CHU de Montpellier, Montpellier, France. INSERM U1040, Montpellier, France. Université Montpellier I, Montpellier, France.
| | - Zhao-Yang Lu
- Unité de Thérapie Cellulaire, CHU de Montpellier, Montpellier, France
| | | | - Bernard Klein
- INSERM U1040, Montpellier, France. Université Montpellier I, Montpellier, France
| |
Collapse
|
222
|
Wouters M, Dijkgraaf EM, Kuijjer ML, Jordanova ES, Hollema H, Welters M, van der Hoeven J, Daemen T, Kroep JR, Nijman HW, van der Burg SH. Interleukin-6 receptor and its ligand interleukin-6 are opposite markers for survival and infiltration with mature myeloid cells in ovarian cancer. Oncoimmunology 2015; 3:e962397. [PMID: 25964862 PMCID: PMC4353164 DOI: 10.4161/21624011.2014.962397] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 10/30/2014] [Indexed: 11/19/2022] Open
Abstract
An increased level of interleukin-6 (IL-6) in epithelial ovarian cancer (EOC) is correlated with a worse prognosis. IL-6 stimulates tumor-growth and inflammation. We investigated the intricate interaction between the IL-6 signaling pathway and tumor-infiltrating myeloid cells (TIMs) to determine their prognostic impact in EOC. 160 EOC samples were analyzed for the expression of IL-6, its receptor (IL-6R) and downstream signaling via pSTAT3 by immunohistochemistry. Triple color immunofluorescence confocal microscopy was used to identify myeloid cell populations by CD14, CD33, and CD163. The relationship between these markers, tumor-infiltrating immune cells, clinical-pathological characteristics and survival was investigated. EOC displayed a dense infiltration with myeloid cells, in particular of the CD163+ type. The distribution pattern of all myeloid subtypes was comparable among the different histological subtypes. Analysis of the tumor cells revealed a high expression of IL-6R in 15% and of IL-6 in 23% of patients. Interestingly, tumors expressing IL-6 or IL-6R formed two different groups. Tumors with a high expression of IL-6R displayed low mature myeloid cell infiltration and a longer disease-specific survival (DSS), especially in late stage tumors. High expression of IL-6R was an independent prognostic factor for survival by multivariate analyses (hazard ratio = 0.474, p = 0.011). In contrast, tumors with high epithelial IL-6 expression displayed a dense infiltration of mature myeloid cells and were correlated with a shorter DSS. Furthermore, in densely CD8+ T-cell infiltrated tumors, the ratio between these lymphoid cells and CD163+ myeloid cells was predictive for survival. Thus, IL-6 and IL-6R are opposite markers for myeloid cell infiltration and survival.
Collapse
Key Words
- DSS, disease-specific survival
- EOC, epithelial ovarian cancer
- FIGO, International Federation of Gynecology and Obstetrics
- IL-6, interleukin-6; IL-6R, interleukin-6 receptor
- IL-6R, interleukin-6, IL-6, interleukin-6 receptor
- MDSC, myeloid-derived suppressor cell
- T reg, regulatory T cell
- TAM, tumor-associated macrophage
- TIL, tumor-infiltrating lymphocytes
- TIM, tumor-infiltrating myeloid cell
- TMA, tissue microarray
- epithelial ovarian cancer
- pSTAT3
- pSTAT3, phosphorylated signal transducer and activator of transcription 3
- tumor-infiltrating myeloid cells
Collapse
Affiliation(s)
- McA Wouters
- Department of Gynecologic Oncology; University of Groningen; University Medical Center Groningen ; Hanzeplein ; Groningen, The Netherlands ; Department of Medical Microbiology; University of Groningen; University Medical Center Groningen ; Hanzeplein , Groningen, The Netherlands
| | - E M Dijkgraaf
- Department of ClinicalOncology; Leiden University Medical Center ; Albinusdreef , Leiden, The Netherlands
| | - M L Kuijjer
- Department of Biostatistics and Computational Biology; Dana-Farber Cancer Institute ; Boston, MA USA ; Department of Biostatistics; Harvard School of Public Health ; Boston, MA USA
| | - E S Jordanova
- Center for Gynaecological Oncology Amsterdam ; VUMC, De Boelelaan , Amsterdam, The Netherlands
| | - H Hollema
- Department of Pathology; University of Groningen; University Medical Center Groningen ; Hanzeplein , Groningen, The Netherlands
| | - Mjp Welters
- Department of ClinicalOncology; Leiden University Medical Center ; Albinusdreef , Leiden, The Netherlands
| | - Jjm van der Hoeven
- Department of ClinicalOncology; Leiden University Medical Center ; Albinusdreef , Leiden, The Netherlands
| | - T Daemen
- Department of Medical Microbiology; University of Groningen; University Medical Center Groningen ; Hanzeplein , Groningen, The Netherlands
| | - J R Kroep
- Department of ClinicalOncology; Leiden University Medical Center ; Albinusdreef , Leiden, The Netherlands
| | - H W Nijman
- Department of Gynecologic Oncology; University of Groningen; University Medical Center Groningen ; Hanzeplein ; Groningen, The Netherlands
| | - S H van der Burg
- Department of ClinicalOncology; Leiden University Medical Center ; Albinusdreef , Leiden, The Netherlands
| |
Collapse
|
223
|
Fujiwara A, Higashiyama M, Kanou T, Okami J, Tokunaga T, Tomita Y, Kodama K. Granulocyte-colony stimulating factor (G-CSF) producing malignant pleural mesothelioma: Report of a case. Thorac Cancer 2015; 6:105-9. [PMID: 26273344 PMCID: PMC4448476 DOI: 10.1111/1759-7714.12140] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/12/2014] [Indexed: 11/26/2022] Open
Abstract
This report presents a case of malignant pleural mesothelioma (MPM) producing granulocyte colony-stimulating factor (G-CSF) that was treated by tumor resection. A 76-year-old male presented with a huge right-side chest wall tumor, along with a slight fever and chest wall pain. Laboratory findings showed an increased white blood cell count (64600 cells/μL) and C-reactive protein level (20.57 mg/dL). The patient underwent surgical removal of the tumor along with tissue from the chest wall and histopathological analysis led to a diagnosis of sarcomatous type of MPM. Immunohistochemical findings for both anti-human G-CSF and interleukin-6 monoclonal antibodies were positive. Although the general condition of the patient quickly improved after surgery, local recurrence occurred two months later and he died of respiratory failure seven months after the operation, though surgery provided symptom relief. G-CSF-producing MPMs usually show a poor prognosis, though less-invasive surgery may be considered for relief of symptoms.
Collapse
Affiliation(s)
- Ayako Fujiwara
- Department of General Thoracic Surgery, Osaka Medical Center for Cancer and Cardiovascular Diseases Osaka, Japan
| | - Masahiko Higashiyama
- Department of General Thoracic Surgery, Osaka Medical Center for Cancer and Cardiovascular Diseases Osaka, Japan
| | - Takashi Kanou
- Department of General Thoracic Surgery, Osaka Medical Center for Cancer and Cardiovascular Diseases Osaka, Japan
| | - Jiro Okami
- Department of General Thoracic Surgery, Osaka Medical Center for Cancer and Cardiovascular Diseases Osaka, Japan
| | - Toshiteru Tokunaga
- Department of General Thoracic Surgery, Osaka Medical Center for Cancer and Cardiovascular Diseases Osaka, Japan
| | - Yasuhiko Tomita
- Department of Pathology, Osaka Medical Center for Cancer and Cardiovascular Diseases Osaka, Japan
| | - Ken Kodama
- Department of General Thoracic Surgery, Yao Municipal Hospital Yao, Japan
| |
Collapse
|
224
|
Mao Y, Zhang Y, Qu Q, Zhao M, Lou Y, Liu J, huang O, Chen X, Wu J, Shen K. Cancer-associated fibroblasts induce trastuzumab resistance in HER2 positive breast cancer cells. MOLECULAR BIOSYSTEMS 2015; 11:1029-40. [PMID: 25648538 DOI: 10.1039/c4mb00710g] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
CAFs isolated from HER2+ patients secreted higher levels of IL6 which expanded cancer stem cells and activated multiple pathways, then induced trastuzumab resistance in HER2 positive breast cancer cells.
Collapse
|
225
|
Klemm F, Joyce JA. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol 2014; 25:198-213. [PMID: 25540894 DOI: 10.1016/j.tcb.2014.11.006] [Citation(s) in RCA: 552] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 11/20/2014] [Accepted: 11/21/2014] [Indexed: 02/08/2023]
Abstract
The tumor microenvironment (TME) not only plays a pivotal role during cancer progression and metastasis but also has profound effects on therapeutic efficacy. In the case of microenvironment-mediated resistance this can involve an intrinsic response, including the co-option of pre-existing structural elements and signaling networks, or an acquired response of the tumor stroma following the therapeutic insult. Alternatively, in other contexts, the TME has a multifaceted ability to enhance therapeutic efficacy. This review examines recent advances in our understanding of the contribution of the TME during cancer therapy and discusses key concepts that may be amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Florian Klemm
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Johanna A Joyce
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
226
|
Engineered microenvironments provide new insights into ovarian and prostate cancer progression and drug responses. Adv Drug Deliv Rev 2014; 79-80:193-213. [PMID: 24969478 DOI: 10.1016/j.addr.2014.06.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 05/30/2014] [Accepted: 06/16/2014] [Indexed: 02/06/2023]
Abstract
Tissue engineering technologies, which have originally been designed to reconstitute damaged tissue structure and function, can mimic not only tissue regeneration processes but also cancer development and progression. Bioengineered approaches allow cell biologists to develop sophisticated experimentally and physiologically relevant cancer models to recapitulate the complexity of the disease seen in patients. Tissue engineering tools enable three-dimensionality based on the design of biomaterials and scaffolds that re-create the geometry, chemistry, function and signalling milieu of the native tumour microenvironment. Three-dimensional (3D) microenvironments, including cell-derived matrices, biomaterial-based cell culture models and integrated co-cultures with engineered stromal components, are powerful tools to study dynamic processes like proteolytic functions associated with cancer progression, metastasis and resistance to therapeutics. In this review, we discuss how biomimetic strategies can reproduce a humanised niche for human cancer cells, such as peritoneal or bone-like microenvironments, addressing specific aspects of ovarian and prostate cancer progression and therapy response.
Collapse
|
227
|
Zhang P, Goodrich C, Fu C, Dong C. Melanoma upregulates ICAM-1 expression on endothelial cells through engagement of tumor CD44 with endothelial E-selectin and activation of a PKCα-p38-SP-1 pathway. FASEB J 2014; 28:4591-609. [PMID: 25138157 DOI: 10.1096/fj.11-202747] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer metastasis involves multistep adhesive interactions between tumor cells (TCs) and endothelial cells (ECs), but the molecular mechanisms of intercellular communication in the tumor microenvironment remain elusive. Using static and flow coculture systems in conjunction with flow cytometry, we discovered that certain receptors on the ECs are upregulated on melanoma cell adhesion. Direct contact but not separate coculture between human umbilical endothelial cells (HUVECs) and a human melanoma cell line (Lu1205) increased intercellular adhesion molecule 1 (ICAM-1) and E-selectin expression on HUVECs by 3- and 1.5-fold, respectively, compared with HUVECs alone. The nonmetastatic cell line WM35 failed to promote ICAM-1 expression changes in HUVECs on contact. Enzyme-linked immunosorbent assay (ELISA) revealed that EC-TC contact has a synergistic effect on the expression of the cytokines interleukin (IL)-8, IL-6, and growth-related oncogene α (Gro-α). By using E-selectin cross-linking and beads coated with CD44 immunopurified from Lu1205 cells, we showed that CD44/selectin ligation was responsible for the ICAM-1 up-regulation on HUVECs. Protein kinase Cα (PKC-α) activation was found to be the downstream target of the CD44/selectin-initiated signaling, as ICAM-1 elevation was inhibited by siRNA targeting PKCα or a dominant negative form of PKCα (PKCα DN). Western blot analysis and electrophoretic mobility shift assays (EMSAs) showed that TC-EC contact mediated p38 phosphorylation and binding of the transcription factor SP-1 to its regulation site. In conclusion, CD44/selectin binding signals ICAM-1 up-regulation on the EC surface through a PKCα-p38-SP-1 pathway, which further enhances melanoma cell adhesion to ECs during metastasis.
Collapse
Affiliation(s)
- Pu Zhang
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Chris Goodrich
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Changliang Fu
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Cheng Dong
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
228
|
Ataie-Kachoie P, Pourgholami MH, Richardson DR, Morris DL. Gene of the month: Interleukin 6 (IL-6). J Clin Pathol 2014; 67:932-7. [PMID: 25031389 DOI: 10.1136/jclinpath-2014-202493] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The Interleukin 6 (IL-6) gene encodes the classic proinflammatory cytokine IL-6. It is also known as interferon-β2 (IFN-β2), B cell stimulatory factor-2 and hybridoma/plasmacytoma growth factor. IL-6 is a multifunctional cytokine with a central role in many physiological inflammatory and immunological processes. Due to its major role in initiation as well as resolving inflammation, deregulation of IL-6 is a mainstay of chronic inflammatory and autoimmune diseases. Additionally, IL-6 has been shown to be implicated in pathogenesis of many human malignancies. Thus, a better understanding of IL-6 and its role in various pathological conditions could enable the development of strategies to use it as a therapeutic target. This short review focuses on the structure, regulation and biological activities of IL-6. In addition we discuss the role of IL-6 in diseases with inflammatory background and cancer and also the therapeutic applications of anti-IL-6 agents.
Collapse
|
229
|
Lopez J, Banerjee S, Kaye SB. New developments in the treatment of ovarian cancer--future perspectives. Ann Oncol 2014; 24 Suppl 10:x69-x76. [PMID: 24265409 DOI: 10.1093/annonc/mdt475] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Over the past 40 years, the treatment of ovarian cancer has undoubtedly improved as a result of better multi-modality care and platinum-based chemotherapy. More recently, the introduction of anti-angiogenic therapy, PARP inhibitors and a weekly regimen for paclitaxel indicate that results are likely to improve further. However, major challenges remain and these will be reviewed in this article. We assess key issues in anti-angiogenic treatment including potential ways for addressing resistance; we review the current studies of PARP inhibitor treatment, which shows most promise in patients with germline BRCA mutations; we describe the potential for folate-receptor-directed therapy, given the high level of FR expression in ovarian cancer and we highlight the potential for molecular targeted therapy, focusing on specific subgroups of the disease with targets such as the PI3 K/AKT and RAS/RAF/MEK pathways and the ErbB family of oncogenes. We anticipate that progress will accelerate with a better understanding of the molecular pathogenesis of the various subtypes of ovarian cancer, leading to an increasingly personalized approach to treating women with this disease.
Collapse
Affiliation(s)
- J Lopez
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, Sutton
| | | | | |
Collapse
|
230
|
McMurray JS, Mandal PK, Liao WS, Klostergaard J, Robertson FM. The consequences of selective inhibition of signal transducer and activator of transcription 3 (STAT3) tyrosine705 phosphorylation by phosphopeptide mimetic prodrugs targeting the Src homology 2 (SH2) domain. JAKSTAT 2014; 1:263-347. [PMID: 24058783 PMCID: PMC3670284 DOI: 10.4161/jkst.22682] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Herein we review our progress on the development of phosphopeptide-based prodrugs targeting the SH2 domain of STAT3 to prevent recruitment to cytokine and growth factor receptors, activation, nuclear translocation and transcription of genes involved in cancer. We developed high affinity phosphopeptides (KI = 46–200 nM). Corresponding prodrugs inhibited constitutive and IL-6 induced Tyr705 phosphorylation at 0.5–1 μM in a variety of human cancer cell lines. They were not cytotoxic at 5 μM in vitro but they inhibited tumor growth in a human xenograft breast cancer model in mice, accompanied by reduced VEGF expression and angiogenesis.
Collapse
Affiliation(s)
- John S McMurray
- The Department of Experimental Therapeutics; The University of Texas MD Anderson Cancer Center; Houston, TX USA
| | | | | | | | | |
Collapse
|
231
|
Momčilović M, Mangano K, Jevtić B, Mammana S, Stošić-Grujičić S, Nicoletti F, Miljković D. Saquinavir-NO Inhibits IL-6 Production in Macrophages. Basic Clin Pharmacol Toxicol 2014; 115:499-506. [DOI: 10.1111/bcpt.12268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 05/05/2014] [Indexed: 01/05/2023]
Affiliation(s)
- Miljana Momčilović
- Department of Immunology; Institute for Biological Research “Siniša Stanković”; University of Belgrade; Belgrade Serbia
| | - Katia Mangano
- Department of Biomedical Sciences; University of Catania; Catania Italy
| | - Bojan Jevtić
- Department of Immunology; Institute for Biological Research “Siniša Stanković”; University of Belgrade; Belgrade Serbia
| | - Santa Mammana
- Department of Biomedical Sciences; University of Catania; Catania Italy
| | - Stanislava Stošić-Grujičić
- Department of Immunology; Institute for Biological Research “Siniša Stanković”; University of Belgrade; Belgrade Serbia
| | - Ferdinando Nicoletti
- Department of Biomedical Sciences; University of Catania; Catania Italy
- OncoNOx Aps; Copenhagen Denmark
| | - Djordje Miljković
- Department of Immunology; Institute for Biological Research “Siniša Stanković”; University of Belgrade; Belgrade Serbia
| |
Collapse
|
232
|
Abstract
Paraneoplastic thrombocytosis is associated with many solid tumors and often correlates with reduced survival. Recent studies suggest that a pathogenic feed back loop may be operative between platelets and tumor cells, with reciprocal interactions between tumor growth/metastasis and thrombocytosis/platelet activation. Specific molecular pathways have been identified in which tumors can stimulate platelet production and activation; activated platelets can, in turn, promote tumor growth and metastasis. Taken together, these findings provide exciting new potential targets for therapeutic intervention.
Collapse
|
233
|
Immunosuppression through constitutively activated NF-κB signalling in human ovarian cancer and its reversal by an NF-κB inhibitor. Br J Cancer 2014; 110:2965-74. [PMID: 24867687 PMCID: PMC4056060 DOI: 10.1038/bjc.2014.251] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Revised: 04/07/2014] [Accepted: 04/14/2014] [Indexed: 12/29/2022] Open
Abstract
Background: Although T-cell immunity is thought to be involved in the prognosis of epithelial ovarian cancer (EOC) patients, immunosuppressive conditions hamper antitumour immune responses. Thus, their mechanisms and overcoming strategies need to be investigated. Methods: The role of NF-κB in human EOC cells and macrophages was evaluated by in vitro production of immunosuppressive IL-6 and IL-8 by EOC cells and in vivo analysis of immune responses in nude mice implanted with human EOC cells using an NF-κB inhibitor DHMEQ. Results: In EOC patients, increased plasma IL-6, IL-8, and arginase were observed. The NF-κB inhibitor DHMEQ inhibited the production of IL-6 and IL-8 by EOC cell lines. Immunosuppression of human DCs and macrophages by culture supernatant of EOC cells was reversed with the pretreatment of DHMEQ. Administration of DHMEQ to nude mice implanted with human EOC resulted in the restoration of T-cell stimulatory activity of murine DCs along with the reduction of tumour accumulation and arginase expression of MDSCs. Nuclear factor-κB inhibition in tumour-bearing mice also enhanced antitumour effects of transferred murine naive T cells. Conclusions: NF-κB is involved in the immunosuppression induced by human EOC, and its inhibitor may restore antitumour immune responses, indicating that NF-κB is an attractive target for EOC treatment.
Collapse
|
234
|
Landskron G, De la Fuente M, Thuwajit P, Thuwajit C, Hermoso MA. Chronic inflammation and cytokines in the tumor microenvironment. J Immunol Res 2014; 2014:149185. [PMID: 24901008 PMCID: PMC4036716 DOI: 10.1155/2014/149185] [Citation(s) in RCA: 1215] [Impact Index Per Article: 110.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/15/2014] [Indexed: 12/14/2022] Open
Abstract
Acute inflammation is a response to an alteration induced by a pathogen or a physical or chemical insult, which functions to eliminate the source of the damage and restore homeostasis to the affected tissue. However, chronic inflammation triggers cellular events that can promote malignant transformation of cells and carcinogenesis. Several inflammatory mediators, such as TNF-α, IL-6, TGF-β, and IL-10, have been shown to participate in both the initiation and progression of cancer. In this review, we explore the role of these cytokines in important events of carcinogenesis, such as their capacity to generate reactive oxygen and nitrogen species, their potential mutagenic effect, and their involvement in mechanisms for epithelial mesenchymal transition, angiogenesis, and metastasis. Finally, we will provide an in-depth analysis of the participation of these cytokines in two types of cancer attributable to chronic inflammatory disease: colitis-associated colorectal cancer and cholangiocarcinoma.
Collapse
Affiliation(s)
- Glauben Landskron
- Disciplinary Program, Institute of Biomedical Sciences, School of Medicine, University of Chile, Independencia 1027, 8380453 Santiago, Chile
| | - Marjorie De la Fuente
- Disciplinary Program, Institute of Biomedical Sciences, School of Medicine, University of Chile, Independencia 1027, 8380453 Santiago, Chile
| | - Peti Thuwajit
- Department of Immunology, School of Medicine, Siriraj Hospital, Mahidol University, 2 Prannok Road, Bangkok Noi, Bangkok 10700, Thailand
| | - Chanitra Thuwajit
- Department of Immunology, School of Medicine, Siriraj Hospital, Mahidol University, 2 Prannok Road, Bangkok Noi, Bangkok 10700, Thailand
| | - Marcela A. Hermoso
- Disciplinary Program, Institute of Biomedical Sciences, School of Medicine, University of Chile, Independencia 1027, 8380453 Santiago, Chile
| |
Collapse
|
235
|
Abstract
Clear cell carcinomas of the female genital tract are rare tumours with a fearsome reputation for having poor responses to conventional platinum-based chemotherapy and poor prognosis. However, it is now clear that early-stage ovarian clear cell carcinoma has an excellent prognosis and may not require any adjuvant therapy. In addition, radiotherapy may also have a key role to play in adjuvant management of clear cell tumours. Identification of patients who truly do not need adjuvant chemotherapy is important. The past 3 years has seen a significant improvement in our understanding of clear cell carcinoma biology-in particular, the role of mutations in the chromatin remodelling gene ARID1A as key drivers that are common to clear cell carcinomas of ovarian and endometrial origin. Moreover, gynaecological clear cell carcinomas appear to share many features with renal clear cell tumours, suggesting a common pathogenesis. This raises the possibility of clinical trials that include patients with clear cell tumours from different organs of origin. Dissecting the role of disordered chromatin organisation in clear cell carcinoma pathogenesis is a key priority. Finally, the role of endometriosis and the attendant chronic inflammation are recognised. The inflammatory cytokine interleukin-6 appears to play a key role in clear cell carcinoma biology and is an excellent potential therapeutic target.
Collapse
|
236
|
Abstract
Growing understanding of the role of thrombocytosis, high platelet turnover, and the presence of activated platelets in the circulation in cancer progression and metastasis has brought megakaryocytes into focus. Platelet biology is essential to hemostasis, vascular integrity, angiogenesis, inflammation, innate immunity, wound healing, and cancer biology. However, before megakaryocyte/platelet-directed therapies can be considered for clinical use, understanding of the mechanism and biology of paraneoplastic thrombocytosis in malignancy is required. Here, we provide an overview of the clinical implications, biological significance, and mechanisms of paraneoplastic thrombocytosis in the context of ovarian cancer.
Collapse
Affiliation(s)
- Ashley N Davis
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vahid Afshar-Kharghan
- Department of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TTX; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
237
|
Rangwala F, Bendell JC, Kozloff MF, Arrowood CC, Dellinger A, Meadows J, Tourt-Uhlig S, Murphy J, Meadows KL, Starr A, Broderick S, Brady JC, Cushman SM, Morse MA, Uronis HE, Hsu SD, Zafar SY, Wallace J, Starodub AN, Strickler JH, Pang H, Nixon AB, Hurwitz HI. Phase I study of capecitabine, oxaliplatin, bevacizumab, and everolimus in advanced solid tumors. Invest New Drugs 2014; 32:700-9. [PMID: 24711126 DOI: 10.1007/s10637-014-0089-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/13/2014] [Indexed: 01/25/2023]
Abstract
PURPOSE To define maximum tolerated dose (MTD), toxicities, and pharmacodynamics of capecitabine, oxaliplatin, bevacizumab, and everolimus in advanced solid tumor patients. DESIGN This was a standard "3 + 3" dose-escalation trial. All subjects received bevacizumab 7.5 mg/kg on day 1 of each cycle. Doses for capecitabine, oxaliplatin and everolimus were modified per dose limiting toxicity (DLT). Baseline and on-treatment plasma biomarkers were analyzed. Archived tumor mRNA levels were evaluated for NRP1, NRP2 and VEGF-A isoforms. RESULTS Twenty-nine patients were evaluable for toxicity and 30 for efficacy. Two DLTs were observed in cohort 1 and one DLT each was observed in cohort -1 and -1b. Grade ≥3 toxicities included neutropenia, hypertension, perforation/fistula/hemorrhage, hypertriglyceridemia, diarrhea, and thromboembolism. Twelve subjects experienced partial response (PR); 12 had stable disease as best response. Three of seven chemorefractory metastatic colorectal cancer (mCRC) subjects experienced PR; 8 of 15 chemonaive mCRC subjects experienced PR. Plasma TβRIII and IL-6 increased on treatment but without correlation to outcome. Increased VEGF165 levels significantly correlated with longer progression free survival. CONCLUSIONS Everolimus with full dose capecitabine, oxaliplatin, and bevacizumab had unacceptable toxicity. MTD was: everolimus 5 mg daily; capecitabine 680 mg/m(2) BID days 1-14; oxaliplatin 100 mg/m(2) and bevacizumab 7.5 mg/kg, day 1. Activity was noted in mCRC.
Collapse
MESH Headings
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antimetabolites, Antineoplastic/administration & dosage
- Antimetabolites, Antineoplastic/adverse effects
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Bevacizumab
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Capecitabine
- Deoxycytidine/administration & dosage
- Deoxycytidine/adverse effects
- Deoxycytidine/analogs & derivatives
- Everolimus
- Female
- Fluorouracil/administration & dosage
- Fluorouracil/adverse effects
- Fluorouracil/analogs & derivatives
- Humans
- Immunosuppressive Agents/administration & dosage
- Immunosuppressive Agents/adverse effects
- Male
- Maximum Tolerated Dose
- Middle Aged
- Neoplasms/drug therapy
- Neuropilin-1/genetics
- Neuropilin-1/metabolism
- Neuropilin-2/genetics
- Neuropilin-2/metabolism
- Organoplatinum Compounds/administration & dosage
- Organoplatinum Compounds/adverse effects
- Oxaliplatin
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Sirolimus/administration & dosage
- Sirolimus/adverse effects
- Sirolimus/analogs & derivatives
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Fatima Rangwala
- Duke University Medical Center, Seeley G. Mudd Bldg 10 Bryan Searle Drive, Box 3052, Durham, NC, 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Dai L, Xia P, Di W. Sphingosine 1-phosphate: a potential molecular target for ovarian cancer therapy? Cancer Invest 2014; 32:71-80. [PMID: 24499107 DOI: 10.3109/07357907.2013.876646] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Sphingosine 1-phosphate (S1P) is an important signaling regulator involved in tumor progression in multiple neoplasms. However, the role of S1P in the pathogenesis of ovarian cancer remains unclear. Herein, we summarize recent advances in understanding the impact of S1P signaling in ovarian cancer progression. S1P, aberrantly produced in ovarian cancer patients, is involved in the regulation of key cellular processes that contribute to ovarian cancer initiation and progression. Moreover, agents that block the S1P signaling pathway inhibit ovarian cancer cell growth or induce apoptosis. Hence, current evidence suggests that S1P may become a potential molecular target for ovarian cancer therapy.
Collapse
Affiliation(s)
- Lan Dai
- Department of Obstetrics and Gynecology, Renji Hospital, Shanghai Jiaotong University School of Medicine , Shanghai , People's Republic of China1
| | | | | |
Collapse
|
239
|
Pahne-Zeppenfeld J, Schröer N, Walch-Rückheim B, Oldak M, Gorter A, Hegde S, Smola S. Cervical cancer cell-derived interleukin-6 impairs CCR7-dependent migration of MMP-9-expressing dendritic cells. Int J Cancer 2014; 134:2061-73. [PMID: 24136650 DOI: 10.1002/ijc.28549] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 09/09/2013] [Accepted: 09/27/2013] [Indexed: 12/14/2022]
Abstract
Cervical carcinogenesis is a consequence of persistent infection with high-risk human papillomaviruses (HPVs). Recent studies indicate that HPV-transformed cells actively instruct their microenvironment to promote carcinogenesis. Here, we demonstrate that cervical cancer cells activate monocytes to produce their own CCL2 for further monocyte recruitment and reprogram their function during differentiation and maturation to dendritic cells (DCs). Our data show that cervical cancer cells suppress the induction of the chemokine receptor CCR7 in phenotypically mature DCs and impair their migration toward a lymph node homing chemokine, required to initiate adaptive immune responses. We confirmed the presence of CD83(+)CCR7(low) DCs in cancer biopsies. The second factor essential for DC migration, matrix-metalloproteinase MMP-9, which also has vasculogenic and protumorigenic properties, is not suppressed but upregulated in immature as well as mature DCs. We identified interleukin-6 (IL-6) as a crucial cervical cancer cell-derived mediator and nuclear factor kappaB (NF-jB) as the central signaling pathway targeted in DCs. Anti-IL-6 antibodies reverted not only NF-jB inhibition and restored CCR7-dependent migration but also blocked MMP-9 induction. This is the first report demonstrating the dissociation of CCR7 and MMP-9 expression in phenotypically mature CD83(+) DCs by cancer cells. Our results show that cervical cancer cells actively shape the local microenvironment. They induce the accumulation of myeloid cells and skew their function from immune activation to local production of protumorigenic MMP-9. Neutralizing anti-IL-6 antibodies can counteract this functional dysbalance and should therefore be considered for adjuvant cervical cancer therapy.
Collapse
|
240
|
Kode J, Taur P, Gulia A, Jambhekar N, Agarwal M, Puri A. Pasteurization of bone for tumour eradication prior to reimplantation - an in vitro & pre-clinical efficacy study. Indian J Med Res 2014; 139:585-597. [PMID: 24927346 PMCID: PMC4078498 DOI: pmid/24927346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND & OBJECTIVES In current era of limb-salvage therapy, pasteurization of bone sarcomas is receiving growing attention as a potential extracorporeal treatment and cost-effective alternative to allografts and radiation before surgical reimplantation. Detailed in vitro and in vivo pre-clinical study to evaluate efficacy of pasteurization to eradicate malignant cells has not been reported yet. The present study was carried out to assess the efficacy of pasteurization to kill tumour cells both in vitro and in vivo. METHODS Surgically resected specimens of osteosarcomas (n=4) were cut into equal halves and one section was pasteurized by heating at 60°C to 65°C for 40 min. Paired samples before and after pasteurization were studied in vitro for DNA ploidy, evaluation of histological change and elimination of mitotic activity. These tissues were transplanted in immune-deficient NOD-SCID mice to evaluate effect on tumour-generating ability, presence of human nuclei, osteopontin and cytokine/chemokines released in tumour-transplanted mice. RESULTS Non-pasteurized tumour samples had viable tumour cells which exhibited significant growth in culture, increased proliferative ability and clonogenic potential while respective pasteurized tumour tissues did not grow in culture and did not exhibit clonogenicity. Flow cytometry revealed that propidium iodide positive dead cells increased significantly (P< 0.01) post pasteurization. Seven of 12 non-pasteurized tumour transplanted mice demonstrated tumour-forming ability as against 0 of 12 in pasteurized tumour transplanted mice. Solid tumour xenografts exhibited strong expression of anti-human nuclei and osteopontin by immunohistochemistry as well as secretary human interluekin-6 (IL-6) while pasteurized mice failed to express these markers. INTERPRETATION & CONCLUSIONS This study has provided a basis to establish pasteurization as being efficacious in ensuring tumour eradication from resected bone tumour specimens. Pasteurized tumour bearing bone can thus safely be used to reconstruct large defects after tumour resection.
Collapse
Affiliation(s)
- Jyoti Kode
- Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Prasad Taur
- Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Ashish Gulia
- Department of Orthopedic Oncology, Tata Memorial Hospital, Mumbai, India
| | | | - Manish Agarwal
- Department of Orthopedic Oncology, Tata Memorial Hospital, Mumbai, India
| | - Ajay Puri
- Department of Orthopedic Oncology, Tata Memorial Hospital, Mumbai, India
| |
Collapse
|
241
|
Chang Q, Daly L, Bromberg J. The IL-6 feed-forward loop: a driver of tumorigenesis. Semin Immunol 2014; 26:48-53. [PMID: 24613573 DOI: 10.1016/j.smim.2014.01.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 01/23/2014] [Indexed: 12/19/2022]
Abstract
IL-6 signaling plays a prominent role in tumorigenesis and metastasis. In this review we discuss the recent evidence describing the tumor intrinsic and extrinsic functions of this signaling pathway. Although blockade of this pathway in pre-clinical models leads to a reduction in tumor growth and metastasis, its clinical success is less evident. Thus, identifying the features of tumors/patients that predict response to anti-IL6 therapy are needed.
Collapse
Affiliation(s)
- Qing Chang
- Department of Medicine, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Laura Daly
- Department of Medicine, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, NY, USA
| | - Jacqueline Bromberg
- Department of Medicine, Memorial Sloan-Kettering Cancer Center (MSKCC), New York, NY, USA; Weill Cornell Medical College (WCMC), New York, NY, USA.
| |
Collapse
|
242
|
Rockall AG, Avril N, Lam R, Iannone R, Mozley PD, Parkinson C, Bergstrom D, Sala E, Sarker SJ, McNeish IA, Brenton JD. Repeatability of quantitative FDG-PET/CT and contrast-enhanced CT in recurrent ovarian carcinoma: test-retest measurements for tumor FDG uptake, diameter, and volume. Clin Cancer Res 2014; 20:2751-60. [PMID: 24573555 DOI: 10.1158/1078-0432.ccr-13-2634] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Repeatability of baseline FDG-PET/CT measurements has not been tested in ovarian cancer. This dual-center, prospective study assessed variation in tumor 2[18F]fluoro-2-deoxy-D-glucose (FDG) uptake, tumor diameter, and tumor volume from sequential FDG-PET/CT and contrast-enhanced computed tomography (CECT) in patients with recurrent platinum-sensitive ovarian cancer. EXPERIMENTAL DESIGN Patients underwent two pretreatment baseline FDG-PET/CT (n = 21) and CECT (n = 20) at two clinical sites with different PET/CT instruments. Patients were included if they had at least one target lesion in the abdomen with a standardized uptake value (SUV) maximum (SUVmax) of ≥ 2.5 and a long axis diameter of ≥ 15 mm. Two independent reading methods were used to evaluate repeatability of tumor diameter and SUV uptake: on site and at an imaging clinical research organization (CRO). Tumor volume reads were only performed by CRO. In each reading set, target lesions were independently measured on sequential imaging. RESULTS Median time between FDG-PET/CT was two days (range 1-7). For site reads, concordance correlation coefficients (CCC) for SUVmean, SUVmax, and tumor diameter were 0.95, 0.94, and 0.99, respectively. Repeatability coefficients were 16.3%, 17.3%, and 8.8% for SUVmean, SUVmax, and tumor diameter, respectively. Similar results were observed for CRO reads. Tumor volume CCC was 0.99 with a repeatability coefficient of 28.1%. CONCLUSIONS There was excellent test-retest repeatability for FDG-PET/CT quantitative measurements across two sites and two independent reading methods. Cutoff values for determining change in SUVmean, SUVmax, and tumor volume establish limits to determine metabolic and/or volumetric response to treatment in platinum-sensitive relapsed ovarian cancer.
Collapse
Affiliation(s)
- Andrea G Rockall
- Authors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, Pennsylvania
| | - Norbert Avril
- Authors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, Pennsylvania
| | - Raymond Lam
- Authors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, Pennsylvania
| | - Robert Iannone
- Authors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, Pennsylvania
| | - P David Mozley
- Authors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, Pennsylvania
| | - Christine Parkinson
- Authors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, PennsylvaniaAuthors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, Pennsylvania
| | - Donald Bergstrom
- Authors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, Pennsylvania
| | - Evis Sala
- Authors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, Pennsylvania
| | - Shah-Jalal Sarker
- Authors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, Pennsylvania
| | - Iain A McNeish
- Authors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, PennsylvaniaAuthors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, Pennsylvania
| | - James D Brenton
- Authors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, PennsylvaniaAuthors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge Biomedical Research Centre; and Cambridge Experimental Cancer Medicine Centre, Cambridge, United Kingdom; Merck and Co, Whitehouse Station; Sanofi, Bridgewater, New Jersey; and Merck Imaging, West Point, PennsylvaniaAuthors' Affiliations: Department of Radiology, St. Bartholomew's Hospital/Barts Health NHS Trust; Department of Medical Oncology, St Bartholomew's Hospital/Barts Health NHS Trust; Department of Nuclear Medicine, Barts Cancer Institute; Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London; Departments of Oncology and Radiology, Cambridge University Hospitals Foundation Trust; Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre; NIHR Cambridge
| |
Collapse
|
243
|
Angevin E, Tabernero J, Elez E, Cohen SJ, Bahleda R, van Laethem JL, Ottensmeier C, Lopez-Martin JA, Clive S, Joly F, Ray-Coquard I, Dirix L, Machiels JP, Steven N, Reddy M, Hall B, Puchalski TA, Bandekar R, van de Velde H, Tromp B, Vermeulen J, Kurzrock R. A phase I/II, multiple-dose, dose-escalation study of siltuximab, an anti-interleukin-6 monoclonal antibody, in patients with advanced solid tumors. Clin Cancer Res 2014; 20:2192-204. [PMID: 24563479 DOI: 10.1158/1078-0432.ccr-13-2200] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE This phase I/II study evaluated safety, efficacy, and pharmacokinetics of escalating, multiple doses of siltuximab, a chimeric anti-interleukin (IL)-6 monoclonal antibody derived from a new Chinese hamster ovary (CHO) cell line in patients with advanced/refractory solid tumors. EXPERIMENTAL DESIGN In the phase I dose-escalation cohorts, 20 patients with advanced/refractory solid tumors received siltuximab 2.8 or 5.5 mg/kg every 2 weeks or 11 or 15 mg/kg every 3 weeks intravenously (i.v.). In the phase I expansion (n = 24) and phase II cohorts (n = 40), patients with Kirsten rat sarcoma-2 (KRAS)-mutant tumors, ovarian, pancreatic, or anti-EGF receptor (EGFR) refractory/resistant non-small cell lung cancer (NSCLC), colorectal, or H&N cancer received 15 mg/kg every 3 weeks. The phase II primary efficacy endpoint was complete response, partial response, or stable disease >6 weeks. RESULTS Eighty-four patients (35 colorectal, 29 ovarian, 9 pancreatic, and 11 other) received a median of three (range, 1-45) cycles. One dose-limiting toxicity occurred at 5.5 mg/kg. Common grade ≥3 adverse events were hepatic function abnormalities (15%), physical health deterioration (12%), and fatigue (11%). Ten percent of patients had siltuximab-related grade ≥3 adverse events. Neutropenia (4%) was the only possibly related adverse event grade ≥3 reported in >1 patient. Serious adverse events were reported in 42%; most were related to underlying disease. The pharmacokinetic profile of CHO-derived siltuximab appears similar to the previous cell line. No objective responses occurred; 5 of 84 patients had stable disease >6 weeks. Hemoglobin increased ≥1.5 g/dL in 33 of 47 patients. At 11 and 15 mg/kg, completely sustained C-reactive protein suppression was observed. CONCLUSIONS Siltuximab monotherapy appears to be well tolerated but without clinical activity in solid tumors, including ovarian and KRAS-mutant cancers. The recommended phase II doses were 11 and 15 mg/kg every 3 weeks.
Collapse
Affiliation(s)
- Eric Angevin
- Authors' Affiliations: Institut de Cancérologie Gustave Roussy, Villejuif; Centre Francois Baclesse, CHU Côte de Nacre, Caen; Centre Léon Bérard Lyon, Lyon, France; Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona; 12 de Octubre University Hospital, Madrid, Spain; Fox Chase Cancer Center, Philadelphia; Janssen Research & Development, Spring House, Pennsylvania; formerly University of Texas, MD Anderson Cancer Center, Houston, Texas; currently UC San Diego Moores Cancer Center, San Diego, California; Erasme University Hospital; Cliniques Universitaires Saint-Luc and Institut de Recherche Clinique et Expérimentale (Pole MIRO), Université Catholique de Louvain, Brussels; AZ Sint-Augustinus, Antwerp; Janssen Research & Development, Beerse, Belgium; Southampton University Hospitals NHS Trust, Southampton; Western General Hospital, Edinburgh; University Hospital Birmingham NHS Foundation Trust, Birmingham, United Kingdom; and Janssen Research & Development, Leiden, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Taniguchi K, Karin M. IL-6 and related cytokines as the critical lynchpins between inflammation and cancer. Semin Immunol 2014; 26:54-74. [PMID: 24552665 DOI: 10.1016/j.smim.2014.01.001] [Citation(s) in RCA: 526] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/06/2014] [Indexed: 11/17/2022]
Abstract
Inflammatory responses play pivotal roles in cancer development, including tumor initiation, promotion, progression, and metastasis. Cytokines are now recognized as important mediators linking inflammation and cancer, and are therefore potential therapeutic and preventive targets as well as prognostic factors. The interleukin (IL)-6 family of cytokines, especially IL-6 and IL-11, is highly up-regulated in many cancers and considered as one of the most important cytokine families during tumorigenesis and metastasis. This review discusses molecular mechanisms linking the IL-6 cytokine family to solid malignancies and their treatment.
Collapse
Affiliation(s)
- Koji Taniguchi
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; UC San Diego Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
245
|
Domcke S, Sinha R, Levine DA, Sander C, Schultz N. Evaluating cell lines as tumour models by comparison of genomic profiles. Nat Commun 2014; 4:2126. [PMID: 23839242 PMCID: PMC3715866 DOI: 10.1038/ncomms3126] [Citation(s) in RCA: 1094] [Impact Index Per Article: 99.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 06/07/2013] [Indexed: 02/06/2023] Open
Abstract
Cancer cell lines are frequently used as in vitro tumour models. Recent molecular profiles of hundreds of cell lines from The Cancer Cell Line Encyclopedia and thousands of tumour samples from the Cancer Genome Atlas now allow a systematic genomic comparison of cell lines and tumours. Here we analyse a panel of 47 ovarian cancer cell lines and identify those that have the highest genetic similarity to ovarian tumours. Our comparison of copy-number changes, mutations and mRNA expression profiles reveals pronounced differences in molecular profiles between commonly used ovarian cancer cell lines and high-grade serous ovarian cancer tumour samples. We identify several rarely used cell lines that more closely resemble cognate tumour profiles than commonly used cell lines, and we propose these lines as the most suitable models of ovarian cancer. Our results indicate that the gap between cell lines and tumours can be bridged by genomically informed choices of cell line models for all tumour types. Cell lines are widely used in cancer research to study tumour biology. Here Domcke et al. compare genomic data from ovarian cancer cell lines with those from clinical ovarian tumour samples and identify cell lines that most closely resemble the genomic features of high-grade serous ovarian cancer.
Collapse
Affiliation(s)
- Silvia Domcke
- Computational Biology Center, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box 460, New York, New York 10065, USA
| | | | | | | | | |
Collapse
|
246
|
Son D, Na YR, Hwang ES, Seok SH. Platelet-derived growth factor-C (PDGF-C) induces anti-apoptotic effects on macrophages through Akt and Bad phosphorylation. J Biol Chem 2014; 289:6225-35. [PMID: 24421315 DOI: 10.1074/jbc.m113.508994] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
PDGF-C, which is abundant in the malignant breast tumor microenvironment, plays an important role in cell growth and survival. Because tumor-associated macrophages (TAMs) contribute to cancer malignancy, macrophage survival mechanisms are an attractive area of research into controlling tumor progression. In this study, we investigated PDGF-C-mediated signaling pathways involved in anti-apoptotic effects in macrophages. We found that the human malignant breast cancer cell line MDA-MB-231 produced high quantities of PDGF-C, whereas benign MCF-7 cells did not. Recombinant PDGF-C induced PDGF receptor α chain phosphorylation, followed by Akt and Bad phosphorylation in THP-1-derived macrophages. MDA-MB-231 culture supernatants also activated macrophage PDGF-Rα. PDGF-C prevented staurosporine-induced macrophage apoptosis by inhibiting the activation of caspase-3, -7, -8, and -9 and cleavage of poly(ADP-ribose) polymerase. Finally, TAMs isolated from the PDGF-C knockdown murine breast cancer cell line 4T1 and PDGF-C knockdown MDA-MB-231-derived tumor mass showed higher rates of apoptosis than the respective WT controls. Collectively, our results suggest that tumor cell-derived PDGF-C enhances TAM survival, promoting tumor malignancy.
Collapse
Affiliation(s)
- Dain Son
- From the Department of Microbiology and Immunology, and Institute of Endemic Disease, College of Medicine, Seoul National University, Seoul 110-799, Korea
| | | | | | | |
Collapse
|
247
|
Kumar J, Ward AC. Role of the interleukin 6 receptor family in epithelial ovarian cancer and its clinical implications. Biochim Biophys Acta Rev Cancer 2014; 1845:117-25. [PMID: 24388871 DOI: 10.1016/j.bbcan.2013.12.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 12/08/2013] [Accepted: 12/24/2013] [Indexed: 01/10/2023]
Abstract
Ovarian cancer is the most lethal gynecological malignancy, with few effective treatment options in most cases. Therefore, understanding the biology of ovarian cancer remains an important area of research in order to improve clinical outcomes. Cytokine receptor signaling through the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway is an essential component of normal development and homeostasis. However, numerous studies have implicated perturbation of this pathway in a range of cancers. In particular, members of the IL-6R family acting via the downstream STAT3 transcription factor play an important role in a number of solid tumors - including ovarian cancer - by altering the expression of target genes that impact on key phenotypes. This has led to the development of specific inhibitors of this pathway which are being used in combination with standard chemotherapeutic agents. This review focuses on the role of IL-6R family members in the etiology of epithelial ovarian cancer, and the application of therapies specifically targeting IL-6R signaling in this disease setting.
Collapse
Affiliation(s)
- Janani Kumar
- School of Medicine, Deakin University, Geelong, Victoria, Australia; Strategic Research Centre in Molecular and Medical Research, Deakin University, Geelong, Victoria, Australia
| | - Alister C Ward
- School of Medicine, Deakin University, Geelong, Victoria, Australia; Strategic Research Centre in Molecular and Medical Research, Deakin University, Geelong, Victoria, Australia.
| |
Collapse
|
248
|
Singha B, Phyo SA, Gatla HR, Vancurova I. Quantitative analysis of bortezomib-induced IL-8 gene expression in ovarian cancer cells. Methods Mol Biol 2014; 1172:295-304. [PMID: 24908316 DOI: 10.1007/978-1-4939-0928-5_27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Interleukin-8 (IL-8), originally discovered as the neutrophil chemoattractant and inducer of leukocyte-mediated inflammation, contributes to cancer progression through its induction of tumor cell proliferation, survival, and migration. IL-8 expression is increased in many types of advanced cancers, including ovarian cancer, and correlates with poor prognosis. Bortezomib (BZ) is the first FDA-approved proteasome inhibitor that has shown remarkable antitumor activity in multiple myeloma and other hematological malignancies. In solid tumors, including ovarian carcinoma, BZ has been less effective as a single agent; however, the mechanisms remain unknown. We have recently shown that in ovarian cancer cells, BZ greatly increases IL-8 expression, while expression of other NFκB-regulated cytokines, IL-6 and TNF, is unchanged. In this chapter, we describe a protocol that uses real-time qRT-PCR to quantitatively analyze mRNA levels of IL-8 and IL-6 in BZ-treated ovarian cancer cells. The protocol can be easily modified and used for analysis of other cytokines in different cell types.
Collapse
Affiliation(s)
- Bipradeb Singha
- Department of Biology, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | | | | | | |
Collapse
|
249
|
Nagasaki T, Hara M, Nakanishi H, Takahashi H, Sato M, Takeyama H. Interleukin-6 released by colon cancer-associated fibroblasts is critical for tumour angiogenesis: anti-interleukin-6 receptor antibody suppressed angiogenesis and inhibited tumour-stroma interaction. Br J Cancer 2013; 110:469-78. [PMID: 24346288 PMCID: PMC3899773 DOI: 10.1038/bjc.2013.748] [Citation(s) in RCA: 314] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 09/12/2013] [Accepted: 10/31/2013] [Indexed: 02/08/2023] Open
Abstract
Background: Interleukin-6 (IL-6) has an important role in cancer progression, and high levels of plasma IL-6 are correlated with a poor prognosis in a variety of cancers. It has also been reported that tumour stromal fibroblasts are necessary for steps in cancer progression, such as angiogenesis. There have been few reports of a correlation between fibroblast actions and IL-6 levels. In this study, we examined the correlation between cancer stromal fibroblasts and IL-6 and the utility of IL-6 as a therapeutic target in human colon cancer. Methods: The expression levels of IL-6 and VEGF of fibroblasts and cancer cell lines were evaluated using real-time PCR and ELISA. The anti-angiogenic effect of inhibiting IL-6 signalling was measured in an angiogenesis model and animal experiment. Results: We demonstrate that stromal fibroblasts isolated from colon cancer produced significant amounts of IL-6 and that colon cancer cells enhanced IL-6 production by stromal fibroblasts. Moreover, IL-6 enhanced VEGF production by fibroblasts, thereby inducing angiogenesis. In vivo, anti-IL6 receptor antibody targeting stromal tissue showed greater anti-tumour activity than did anti-IL6 receptor antibody targeting xenografted cancer cells. Conclusion: Cancer stromal fibroblasts were an important source of IL-6 in colon cancer. IL-6 produced by activated fibroblasts induced tumour angiogenesis by stimulating adjacent stromal fibroblasts. The relationship between IL-6 and stromal fibroblasts offers new approaches to cancer therapy.
Collapse
Affiliation(s)
- T Nagasaki
- Department of Gastroenterological Surgery, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - M Hara
- Department of Gastroenterological Surgery, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - H Nakanishi
- Division of Oncological Pathology, Aichi Cancer Center Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan
| | - H Takahashi
- Department of Gastroenterological Surgery, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - M Sato
- Department of Gastroenterological Surgery, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - H Takeyama
- Department of Gastroenterological Surgery, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| |
Collapse
|
250
|
Konstantinopoulos PA, Matulonis UA. Current status and evolution of preclinical drug development models of epithelial ovarian cancer. Front Oncol 2013; 3:296. [PMID: 24377084 PMCID: PMC3858677 DOI: 10.3389/fonc.2013.00296] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 11/23/2013] [Indexed: 01/06/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy and the fifth most common cause of female cancer death in the United States. Although important advances in surgical and chemotherapeutic strategies over the last three decades have significantly improved the median survival of EOC patients, the plateau of the survival curve has not changed appreciably. Given that EOC is a genetically and biologically heterogeneous disease, identification of specific molecular abnormalities that can be targeted in each individual ovarian cancer on the basis of predictive biomarkers promises to be an effective strategy to improve outcome in this disease. However, for this promise to materialize, appropriate preclinical experimental platforms that recapitulate the complexity of these neoplasms and reliably predict antitumor activity in the clinic are critically important. In this review, we will present the current status and evolution of preclinical models of EOC, including cell lines, immortalized normal cells, xenograft models, patient-derived xenografts, and animal models, and will discuss their potential for oncology drug development.
Collapse
Affiliation(s)
- Panagiotis A Konstantinopoulos
- Medical Gynecologic Oncology Program, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School , Boston, MA , USA
| | - Ursula A Matulonis
- Medical Gynecologic Oncology Program, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School , Boston, MA , USA
| |
Collapse
|