1
|
Mantovani A, Marchesi F, Di Mitri D, Garlanda C. Macrophage diversity in cancer dissemination and metastasis. Cell Mol Immunol 2024; 21:1201-1214. [PMID: 39402303 PMCID: PMC11528009 DOI: 10.1038/s41423-024-01216-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/06/2024] [Indexed: 11/02/2024] Open
Abstract
Invasion and metastasis are hallmarks of cancer. In addition to the well-recognized hematogenous and lymphatic pathways of metastasis, cancer cell dissemination can occur via the transcoelomic and perineural routes, which are typical of ovarian and pancreatic cancer, respectively. Macrophages are a universal major component of the tumor microenvironment and, in established tumors, promote growth and dissemination to secondary sites. Here, we review the role of tumor-associated macrophages (TAMs) in cancer cell dissemination and metastasis, emphasizing the diversity of myeloid cells in different tissue contexts (lungs, liver, brain, bone, peritoneal cavity, nerves). The generally used models of lung metastasis fail to capture the diversity of pathways and tissue microenvironments. A better understanding of TAM diversity in different tissue contexts may pave the way for tailored diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Alberto Mantovani
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy.
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy.
- William Harvey Research Institute, Queen Mary University, London, UK.
| | - Federica Marchesi
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Diletta Di Mitri
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (Milan), Italy
| |
Collapse
|
2
|
The Molecular and Cellular Strategies of Glioblastoma and Non-Small-Cell Lung Cancer Cells Conferring Radioresistance. Int J Mol Sci 2022; 23:ijms232113577. [PMID: 36362359 PMCID: PMC9656305 DOI: 10.3390/ijms232113577] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Ionizing radiation (IR) has been shown to play a crucial role in the treatment of glioblastoma (GBM; grade IV) and non-small-cell lung cancer (NSCLC). Nevertheless, recent studies have indicated that radiotherapy can offer only palliation owing to the radioresistance of GBM and NSCLC. Therefore, delineating the major radioresistance mechanisms may provide novel therapeutic approaches to sensitize these diseases to IR and improve patient outcomes. This review provides insights into the molecular and cellular mechanisms underlying GBM and NSCLC radioresistance, where it sheds light on the role played by cancer stem cells (CSCs), as well as discusses comprehensively how the cellular dormancy/non-proliferating state and polyploidy impact on their survival and relapse post-IR exposure.
Collapse
|
3
|
Freeman AJ, Kearney CJ, Silke J, Oliaro J. Unleashing TNF cytotoxicity to enhance cancer immunotherapy. Trends Immunol 2021; 42:1128-1142. [PMID: 34750058 DOI: 10.1016/j.it.2021.10.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/08/2021] [Accepted: 10/10/2021] [Indexed: 01/02/2023]
Abstract
Tumor necrosis factor (TNF) is a proinflammatory cytokine that is produced and secreted by cytotoxic lymphocytes upon tumor target recognition. Depending on the context, TNF can mediate either pro-survival or pro-death signals. The potential cytotoxicity of T cell-produced TNF, particularly in the context of T cell-directed immunotherapies, has been largely overlooked. However, a spate of recent studies investigating tumor immune evasion through the application of CRISPR-based gene-editing screens have highlighted TNF-mediated killing as an important component of the mammalian T cell antitumor repertoire. In the context of the current understanding of the role of TNF in antitumor immunity, we discuss these studies and touch on their therapeutic implications. Collectively, we provide an enticing prospect to augment immunotherapy responses through TNF cytotoxicity.
Collapse
Affiliation(s)
- Andrew J Freeman
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Conor J Kearney
- Translational Haematology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - John Silke
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Jane Oliaro
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Immunology and Pathology, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|
4
|
The Role of Tumor-Associated Macrophages in the Progression and Chemoresistance of Ovarian Cancer. Cells 2020; 9:cells9051299. [PMID: 32456078 PMCID: PMC7290435 DOI: 10.3390/cells9051299] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/18/2020] [Accepted: 05/21/2020] [Indexed: 01/11/2023] Open
Abstract
Tumor-associated macrophages (TAMs) constitute the main population of immune cells present in the ovarian tumor microenvironment. These cells are characterized by high plasticity and can be easily polarized by colony-stimulating factor-1, which is released by tumor cells, into an immunosuppressive M2-like phenotype. These cells are strongly implicated in both the progression and chemoresistance of ovarian cancer. The main pro-tumoral function of M2-like TAMs is the secretion of a variety of cytokines, chemokines, enzymes and exosomes that reach microRNAs, directly inducing the invasion potential and chemoresistance of ovarian cancer cells by triggering their pro-survival signaling pathways. The M2-like TAMs are also important players in the metastasis of ovarian cancer cells in the peritoneum through their assistance in spheroid formation and attachment of cancer cells to the metastatic area—the omentum. Moreover, TAMs interplay with other immune cells, such as lymphocytes, natural killer cells, and dendritic cells, to inhibit their responsiveness, resulting in the development of immunosuppression. The detrimental character of the M2-like type of TAMs in ovarian tumors has been confirmed by a number of studies, demonstrating the positive correlation between their high level in tumors and low overall survival of patients.
Collapse
|
5
|
Worzfeld T, Pogge von Strandmann E, Huber M, Adhikary T, Wagner U, Reinartz S, Müller R. The Unique Molecular and Cellular Microenvironment of Ovarian Cancer. Front Oncol 2017; 7:24. [PMID: 28275576 PMCID: PMC5319992 DOI: 10.3389/fonc.2017.00024] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/07/2017] [Indexed: 12/13/2022] Open
Abstract
The reciprocal interplay of cancer cells and host cells is an indispensable prerequisite for tumor growth and progression. Cells of both the innate and adaptive immune system, in particular tumor-associated macrophages (TAMs) and T cells, as well as cancer-associated fibroblasts enter into a malicious liaison with tumor cells to create a tumor-promoting and immunosuppressive tumor microenvironment (TME). Ovarian cancer, the most lethal of all gynecological malignancies, is characterized by a unique TME that enables specific and efficient metastatic routes, impairs immune surveillance, and mediates therapy resistance. A characteristic feature of the ovarian cancer TME is the role of resident host cells, in particular activated mesothelial cells, which line the peritoneal cavity in huge numbers, as well as adipocytes of the omentum, the preferred site of metastatic lesions. Another crucial factor is the peritoneal fluid, which enables the transcoelomic spread of tumor cells to other pelvic and peritoneal organs, and occurs at more advanced stages as a malignancy-associated effusion. This ascites is rich in tumor-promoting soluble factors, extracellular vesicles and detached cancer cells as well as large numbers of T cells, TAMs, and other host cells, which cooperate with resident host cells to support tumor progression and immune evasion. In this review, we summarize and discuss our current knowledge of the cellular and molecular interactions that govern this interplay with a focus on signaling networks formed by cytokines, lipids, and extracellular vesicles; the pathophysiologial roles of TAMs and T cells; the mechanism of transcoelomic metastasis; and the cell type selective processing of signals from the TME.
Collapse
Affiliation(s)
- Thomas Worzfeld
- Institute of Pharmacology, Biochemical-Pharmacological Center (BPC), Philipps University, Marburg, Germany; Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Elke Pogge von Strandmann
- Experimental Tumor Research, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Philipps University , Marburg , Germany
| | - Magdalena Huber
- Institute of Medical Microbiology and Hygiene, Biomedical Research Center, Philipps University , Marburg , Germany
| | - Till Adhikary
- Institute of Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University , Marburg , Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital of Giessen and Marburg (UKGM) , Marburg , Germany
| | - Silke Reinartz
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University , Marburg , Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University , Marburg , Germany
| |
Collapse
|
6
|
Abstract
Deregulated inflammatory response plays a pivotal role in the initiation, development and progression of tumours. Potential molecular mechanism(s) that drive the establishment of an inflammatory-tumour microenvironment is not entirely understood owing to the complex cross-talk between pro-inflammatory and tumorigenic mediators such as cytokines, chemokines, oncogenes, enzymes, transcription factors and immune cells. These molecular mediators are critical linchpins between inflammation and cancer, and their activation and/or deactivation are influenced by both extrinsic (i.e. environmental and lifestyle) and intrinsic (i.e. hereditary) factors. At present, the research pertaining to inflammation-associated cancers is accumulating at an exponential rate. Interest stems from hope that new therapeutic strategies against molecular mediators can be identified to assist in cancer treatment and patient management. The present review outlines the various molecular and cellular inflammatory mediators responsible for tumour initiation, progression and development, and discusses the critical role of chronic inflammation in tumorigenesis.
Collapse
|
7
|
Shiozaki A, Bai XH, Shen-Tu G, Moodley S, Takeshita H, Fung SY, Wang Y, Keshavjee S, Liu M. Claudin 1 mediates TNFα-induced gene expression and cell migration in human lung carcinoma cells. PLoS One 2012; 7:e38049. [PMID: 22675434 PMCID: PMC3365005 DOI: 10.1371/journal.pone.0038049] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Accepted: 05/02/2012] [Indexed: 12/26/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is an important mechanism in carcinogenesis. To determine the mechanisms that are involved in the regulation of EMT, it is crucial to develop new biomarkers and therapeutic targets towards cancers. In this study, when TGFβ1 and TNFα were used to induce EMT in human lung carcinoma A549 cells, we found an increase in an epithelial cell tight junction marker, Claudin 1. We further identified that it was the TNFα and not the TGFβ1 that induced the fibroblast-like morphology changes. TNFα also caused the increase in Claudin-1 gene expression and protein levels in Triton X-100 soluble cytoplasm fraction. Down-regulation of Claudin-1, using small interfering RNA (siRNA), inhibited 75% of TNFα-induced gene expression changes. Claudin-1 siRNA effectively blocked TNFα-induced molecular functional networks related to inflammation and cell movement. Claudin-1 siRNA was able to significantly reduce TNF-enhanced cell migration and fibroblast-like morphology. Furthermore, over expression of Claudin 1 with a Claudin 1-pcDNA3.1/V5-His vector enhanced cell migration. In conclusion, these observations indicate that Claudin 1 acts as a critical signal mediator in TNFα-induced gene expression and cell migration in human lung cancer cells. Further analyses of these cellular processes may be helpful in developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Atsushi Shiozaki
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Xiao-hui Bai
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Grace Shen-Tu
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Serisha Moodley
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Hiroki Takeshita
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Shan-Yu Fung
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Yingchun Wang
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, University Health Network Toronto General Research Institute, Toronto, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Canada
- * E-mail:
| |
Collapse
|
8
|
Kamangar F, Neuhaus IM, Koo JYM. An evidence-based review of skin cancer rates on biologic therapies. J DERMATOL TREAT 2011; 23:305-15. [DOI: 10.3109/09546634.2011.652064] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Faranak Kamangar
- University of California, Davis School of Medicine, San Francisco Psoriasis Skin and Treatment Center,
San Francisco, CA, USA
| | | | - John Y. M. Koo
- UCSF Department of Dermatology, UCSF Psoriasis Skin and Treatment Center,
San Francisco, CA, USA
| |
Collapse
|
9
|
Souto JC, Vila L, Brú A. Polymorphonuclear neutrophils and cancer: intense and sustained neutrophilia as a treatment against solid tumors. Med Res Rev 2011; 31:311-63. [PMID: 19967776 DOI: 10.1002/med.20185] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Polymorphonuclear neutrophils (PMN) are the most abundant circulating immune cells and represent the first line of immune defense against infection. This review of the biomedical literature of the last 40 years shows that they also have a powerful antitumoral effect under certain circumstances. Typically, the microenvironment surrounding a solid tumor possesses many of the characteristics of chronic inflammation, a condition considered very favorable for tumor growth and spread. However, there are many circumstances that shift the chronic inflammatory state toward an acute inflammatory response around a tumor. This shift seems to convert PMN into very efficient anticancer effector cells. Clinical reports of unexpected antitumoral effects linked to the prolonged use of granulocyte colony-stimulating factor, which stimulates an intense and sustained neutrophilia, suggest that an easy way to fight solid tumors would be to encourage the development of intense peritumoral PMN infiltrates. Specifically designed clinical trials are urgently needed to evaluate the safety and efficacy of such drug-induced neutrophilia in patients with solid tumors. This antitumoral role of neutrophils may provide new avenues for the clinical treatment of cancer.
Collapse
Affiliation(s)
- Juan Carlos Souto
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.
| | | | | |
Collapse
|
10
|
Stoicov C, Li H, Cerny J, Houghton J. How the study of Helicobacter infection can contribute to the understanding of carcinoma development. Clin Microbiol Infect 2009; 15:813-22. [DOI: 10.1111/j.1469-0691.2009.02965.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
11
|
Stewart TJ, Christine Lutsiak ME, Abrams SI. Immune Consequences of Protracted Host-Tumor Interactions in a Transgenic Mouse Model of Mammary Carcinoma. Cancer Invest 2009; 26:237-49. [DOI: 10.1080/07357900701708419] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
12
|
Abstract
Tumour necrosis factor (TNF) is a major inflammatory cytokine that was first identified for its ability to induce rapid haemorrhagic necrosis of experimental cancers. When efforts to harness this anti-tumour activity in cancer treatments were underway, a paradoxical tumour-promoting role of TNF became apparent. Now that links between inflammation and cancer are appreciated, is TNF a target or a therapeutic in malignant disease -- or both?
Collapse
Affiliation(s)
- Frances Balkwill
- Centre for Cancer and Inflammation, Institute of Cancer, Barts, UK.
| |
Collapse
|
13
|
Abstract
The tumor microenvironment is created by the tumor and dominated by tumor-induced interactions. Although various immune effector cells are recruited to the tumor site, their anti-tumor functions are downregulated, largely in response to tumor-derived signals. Infiltrates of inflammatory cells present in human tumors are chronic in nature and are enriched in regulatory T cells (T(reg)) as well as myeloid suppressor cells (MSC). Immune cells in the tumor microenvironment not only fail to exercise antitumor effector functions, but they are co-opted to promote tumor growth. Sustained activation of the NF-kappaB pathway in the tumor milieu represents one mechanism that appears to favor tumor survival and drive abortive activation of immune cells. The result is tumor escape from the host immune system. Tumor escape is accomplished through the activation of one or several molecular mechanisms that lead to inhibition of immune cell functions or to apoptosis of anti-tumor effector cells. The ability to block tumor escape depends on a better understanding of cellular and molecular pathways operating in the tumor microenvironment. Novel therapeutic strategies that emerge are designed to change the pro-tumor microenvironment to one favoring acute responses and potent anti-tumor activity.
Collapse
|
14
|
Ariztia EV, Lee CJ, Gogoi R, Fishman DA. The Tumor Microenvironment: Key to Early Detection. Crit Rev Clin Lab Sci 2008; 43:393-425. [PMID: 17050079 DOI: 10.1080/10408360600778836] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The tumor microenvironment plays an important role equal to the tumor cell population in the progression of cancer. Consisting of stromal fibroblasts, inflammatory cells, components of the vasculature, normal epithelia, and extracellular matrix, the surrounding environment interacts or "cross-talks" with tumor cells through the release of growth factors, cytokines, proteases, and other bioactive molecules. Tumor growth, formation of new vascular networks, evasion of the host immune system, and invasion and metastasis are processes that co-evolve and become finely optimized and regulated within the tumor microenvironment. However, relatively recent reports on three areas of study have come together to add new levels of complexity to the tumor microenvironment. These include ectodomain shedding of proteins, shedding of membrane-derived vesicles, and novel roles for phospholipids. These dynamic changes that take place in the tumor microenvironment provide new avenues for study and for the early detection of cancer, whereas proteomic technologies provide the means to detect these unique proteins and lipids. Here we review the evolving concepts of the tumor microenvironment that, together with advances in proteomic technologies, hold the promise to facilitate the detection of early-stage cancer.
Collapse
Affiliation(s)
- Edgardo V Ariztia
- Department of Obstetrics and Gynecology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | |
Collapse
|
15
|
Manu KA, Kuttan G. Effect of Punarnavine, an alkaloid from Boerhaavia diffusa, on cell-mediated immune responses and TIMP-1 in B16F-10 metastatic melanoma-bearing mice. Immunopharmacol Immunotoxicol 2008; 29:569-86. [PMID: 18075866 DOI: 10.1080/08923970701692676] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Effect of Punarnavine on the cell-mediated immune (CMI) response in metastatic condition was studied using C57BL/6 mice model. Administration of Punarnavine enhanced Natural Killer (NK) cell activity, antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent complement mediated cytotoxicity (ACC) and the activity was observed in treated group much earlier compared to the metastatic tumor-bearing control. Production of cytokines such as IL-2 and IFN-gamma were significantly enhanced by the administration of Punarnavine compared to the untreated metastatic tumor-bearing control. Peaks of pro-inflammatory cytokines such as IL-1beta, IL-6 and TNF-alpha were significantly lowered by Punarnavine administration compared to metastatic control. The level and expression of TIMP-1 was also enhanced by the administration of Punarnavine compared to metastatic tumor bearing control. These results indicate Punarnavine could enhance the immune response against metastatic progression of B16F-10 melanoma cells in mice.
Collapse
Affiliation(s)
- K A Manu
- Amala Cancer Research Centre, Amala Nagar, Thrissur, Kerala State, India
| | | |
Collapse
|
16
|
Abstract
Death receptor ligands (FasL, TRAIL) activate apoptosis in cells expressing the cognate receptors. Evidence suggests that these ligands also deliver pro-inflammatory signals. In the tumor microenvironment, "Fas counterattack" mounted by tumors against immune cells is mediated by tumor-associated FasL. But death ligands crosslinking their receptors also induce inhibition of apoptosis and activation of the transcription factor, NFkappaB, with a subsequent burst of pro-inflammatory cytokine production and tumor growth promotion. NFkappaB, a key link between inflammation and cancer, regulates dual activities of death ligands, depending on molecular signals in the tumor microenvironment. This paper focuses on death ligands as an example of the extensive repertoire of strategies devised by tumors for escape from immune control.
Collapse
Affiliation(s)
- Theresa L Whiteside
- Department of Pathology, Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
17
|
Kulbe H, Thompson R, Wilson JL, Robinson S, Hagemann T, Fatah R, Gould D, Ayhan A, Balkwill F. The inflammatory cytokine tumor necrosis factor-alpha generates an autocrine tumor-promoting network in epithelial ovarian cancer cells. Cancer Res 2007; 67:585-92. [PMID: 17234767 PMCID: PMC2679985 DOI: 10.1158/0008-5472.can-06-2941] [Citation(s) in RCA: 296] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Constitutive expression of the inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) is characteristic of malignant ovarian surface epithelium. We investigated the hypothesis that this autocrine action of TNF-alpha generates and sustains a network of other mediators that promote peritoneal cancer growth and spread. When compared with two ovarian cancer cell lines that did not make TNF-alpha, constitutive production of TNF-alpha was associated with greater release of the chemokines CCL2 and CXCL12, the cytokines interleukin-6 (IL-6) and macrophage migration-inhibitory factor (MIF), and the angiogenic factor vascular endothelial growth factor (VEGF). TNF-alpha production was associated also with increased peritoneal dissemination when the ovarian cancer cells were xenografted. We next used RNA interference to generate stable knockdown of TNF-alpha in ovarian cancer cells. Production of CCL2, CXCL12, VEGF, IL-6, and MIF was decreased significantly in these cells compared with wild-type or mock-transfected cells, but in vitro growth rates were unaltered. Tumor growth and dissemination in vivo were significantly reduced when stable knockdown of TNF-alpha was achieved. Tumors derived from TNF-alpha knockdown cells were noninvasive and well circumscribed and showed high levels of apoptosis, even in the smallest deposits. This was reflected in reduced vascularization of TNF-alpha knockdown tumors. Furthermore, culture supernatants from such cells failed to stimulate endothelial cell growth in vitro. We conclude that autocrine production of TNF-alpha by ovarian cancer cells stimulates a constitutive network of other cytokines, angiogenic factors, and chemokines that may act in an autocrine/paracrine manner to promote colonization of the peritoneum and neovascularization of developing tumor deposits.
Collapse
Affiliation(s)
- Hagen Kulbe
- Centre for Translational Oncology, Institute of Cancer and the Cancer Research-UK Clinical Centre, London, United Kingdom
| | - Richard Thompson
- Centre for Translational Oncology, Institute of Cancer and the Cancer Research-UK Clinical Centre, London, United Kingdom
| | - Julia L. Wilson
- Centre for Translational Oncology, Institute of Cancer and the Cancer Research-UK Clinical Centre, London, United Kingdom
| | - Stephen Robinson
- Centre for Translational Oncology, Institute of Cancer and the Cancer Research-UK Clinical Centre, London, United Kingdom
| | - Thorsten Hagemann
- Centre for Translational Oncology, Institute of Cancer and the Cancer Research-UK Clinical Centre, London, United Kingdom
| | - Rewas Fatah
- Bone and Joint Research Unit, William Harvey Research Institute, Barts and The London, Queen Mary’s School of Medicine and Dentistry, London, United Kingdom
| | - David Gould
- Bone and Joint Research Unit, William Harvey Research Institute, Barts and The London, Queen Mary’s School of Medicine and Dentistry, London, United Kingdom
| | - Ayse Ayhan
- Department of Pathology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Frances Balkwill
- Centre for Translational Oncology, Institute of Cancer and the Cancer Research-UK Clinical Centre, London, United Kingdom
| |
Collapse
|
18
|
Abstract
Interactions between tumor infiltrating leukocytes and tumor cells have been of great interest because of the possibility that immune cells either interfere with tumor progression or actively promote tumor growth. The tumor microenvironment is shaped by cells entering it, and their functions reflect the local conditions. Successive changes occurring at the tumor site during tumor progression resemble chronic inflammation. This chronic inflammatory reaction seems to be largely orchestrated by the tumor, and it seems to promote tumor survival. Molecular and cellular mechanisms linking the inflammatory reaction and cancer are emerging, and this review summarizes the current understanding of interactions between inflammatory and cancer cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Theresa L Whiteside
- University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA 15213, USA
| |
Collapse
|
19
|
Ness RB, Modugno F. Endometriosis as a model for inflammation–hormone interactions in ovarian and breast cancers. Eur J Cancer 2006; 42:691-703. [PMID: 16531042 DOI: 10.1016/j.ejca.2006.01.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2006] [Accepted: 01/12/2006] [Indexed: 12/01/2022]
Abstract
Chronic inflammation has been implicated in a variety of cancers. In this review, we consider associations between endometriosis and cancers both local (ovarian) and distant (breast). We review the epidemiological data linking endometriosis to ovarian and breast cancers. We then consider evidence for a role for sex steroid hormones and for inflammation in the aetiology of each of these cancers. Finally, we consider that endometriosis may promote alterations in sex steroid hormones and inflammatory mediators. A possible explanation for the association between endometriosis and these reproductive cancers may then be local and systemic enhancement of aberrant inflammatory and hormonal mediators. If this hypothesis is true, endometriosis may need to be considered as a risk factor for ovarian and breast cancers, triggering increasingly intensive surveillance. Moreover, treatments for endometriosis may require consideration of the impact on long-term cancer risk.
Collapse
Affiliation(s)
- Roberta B Ness
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Room 513 Parran Hall, 130 DeSoto Street, Pittsburgh, PA 15261, USA.
| | | |
Collapse
|
20
|
Szlosarek P, Charles KA, Balkwill FR. Tumour necrosis factor-alpha as a tumour promoter. Eur J Cancer 2006; 42:745-50. [PMID: 16517151 DOI: 10.1016/j.ejca.2006.01.012] [Citation(s) in RCA: 287] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2006] [Accepted: 01/11/2006] [Indexed: 12/17/2022]
Abstract
It is becoming more evident that many aspects of tumour promotion arise from persistent and unresolving inflammation. One of the key molecules mediating the inflammatory processes in tumour promotion is the cytokine, tumour necrosis factor-alpha (TNF-alpha). Clinically, elevated serum concentrations and increased expression of TNF-alpha are present in various pre-neoplastic and malignant diseases, compared with serum and tissue from healthy individuals. Although over the last few decades high-dose administration of TNF-alpha has been used as a cytotoxic agent, recent pre-clinical cancer models have provided critical evidence to support the link between chronic, low level TNF-alpha exposure and the acquisition of pro-malignant phenotype (i.e., increased growth, invasion and metastasis). Furthermore, sophisticated cellular systems are being utilised to dissect the crucial role TNF-alpha plays in the communication of stromal/inflammatory cells and tumour cells. Understanding the intricate roles of TNF-alpha in the process of tumour promotion will assist in the development of novel cancer therapeutics.
Collapse
Affiliation(s)
- Peter Szlosarek
- Centre for Translational Oncology, Institute of Cancer and the CR-UK Clinical Centre, Barts and The London, Queen Mary's School of Medicine and Dentistry, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | | | | |
Collapse
|
21
|
Madhusudan S, Muthuramalingam SR, Braybrooke JP, Wilner S, Kaur K, Han C, Hoare S, Balkwill F, Ganesan TS. Study of etanercept, a tumor necrosis factor-alpha inhibitor, in recurrent ovarian cancer. J Clin Oncol 2005; 23:5950-9. [PMID: 16135466 DOI: 10.1200/jco.2005.04.127] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Convincing data support the link between inflammation and ovarian cancer. Tumor necrosis factor-alpha (TNF-alpha), a major mediator of inflammation, is chronically produced in the ovarian tumor microenvironment and may enhance tumor growth and invasion by inducing the secretion of cytokines, proangiogenic factors, and metalloproteinases. Etanercept is a recombinant human soluble p75 TNF receptor that binds to TNF-alpha and renders it biologically unavailable. In the current study, we sought to determine the toxicity, biologic activity, and therapeutic efficacy of etanercept in recurrent ovarian cancer. PATIENTS AND METHODS We initiated a phase I-B, nonrandomized, open-label study in patients with recurrent ovarian cancer. Etanercept was administered subcutaneously at a dose of 25 mg twice weekly (cohort one) and 25 mg thrice weekly (cohort two) until disease progression. RESULTS Thirty patients were recruited (cohort one, 17 patients; cohort two, 13 patients). Eighteen of the 30 patients (cohort one, 11 patients; cohort two, seven patients) completed > or = 12 weeks of treatment. Six patients achieved prolonged disease stabilization (cohort one, two patients [40 and 25 weeks]; cohort two, four patients [34, 24, 22, and 24 weeks]). A significant rise in immunoreactive TNF was seen in all patients (pretreatment compared with end of treatment). A phytohemagglutinin-stimulated whole-blood cytokine assay showed a significant fall in interleukin-6 (cohort one [11 of 17]) and CCL2 (cohort one [13 of 17]) levels. Common adverse effects were injection-site reactions and fatigue. CONCLUSION We provide evidence for the biologic activity and safety of etanercept in recurrent ovarian cancer. Our data suggest possible clinical activity that must be confirmed in future studies.
Collapse
Affiliation(s)
- Srinivasan Madhusudan
- Cancer Research UK Medical Oncology Unit, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Yang WL, Roland IH, Godwin AK, Xu XX. Loss of TNF-α-regulated COX-2 expression in ovarian cancer cells. Oncogene 2005; 24:7991-8002. [PMID: 16044148 DOI: 10.1038/sj.onc.1208943] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cyclooxygenase 2 (COX-2) is often found overexpressed in cancer and is thought to have a role in carcinogenic promotion, and thus is a target for therapeutic intervention. Here, we investigated the regulation of COX-2 expression in normal and cancer ovarian surface epithelial cells. Tumor necrosis factor alpha (TNF-alpha) is a potent inducer of COX-2 expression in the ovarian surface epithelium and this regulation is a critical step in ovulation. We observed that TNF-alpha stimulated COX-2 expression in human primary and immortalized epithelial (HIO) cell lines. The stimulation was suppressed by inhibitors of several signaling pathways, indicating the collaboration of TNF-alpha-activated signaling pathways mediates the regulation of COX-2 expression. In five ovarian cancer cell lines analysed, four did not express detectable COX-2 and TNF-alpha failed to elicit COX-2 expression. In NIH:OVCAR-5, the only ovarian cancer cell line expressing COX-2, signal pathway inhibitors no longer affected TNF-alpha-induced COX-2 expression. Thus, we conclude that TNF-alpha mediated signaling is uncoupled from the modulation of COX-2 expression in ovarian cancer. The loss of COX-2 expression was also observed to associate closely with epithelial neoplastic morphological transformation. The frequent loss of COX-2 expression suggests in ovarian cancer, unlike in other epithelial cancers, COX-2 expression does not contribute to ovarian cancer malignancy.
Collapse
Affiliation(s)
- Wan-Lin Yang
- Ovarian Cancer and Tumor Cell Biology Programs, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | | | | | | |
Collapse
|
23
|
Zarovni N, Monaco L, Corti A. Inhibition of tumor growth by intramuscular injection of cDNA encoding tumor necrosis factor alpha coupled to NGR and RGD tumor-homing peptides. Hum Gene Ther 2004; 15:373-82. [PMID: 15053862 DOI: 10.1089/104303404322959524] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The antitumor properties of tumor necrosis factor alpha (TNF) and its efficacy in selective destruction of tumor-associated vessels are well known. Besides the TNF protein, the TNF gene has been used for gene therapy of cancer and shown to induce antitumor responses both in animal models and in patients. We show here that the therapeutic properties of the TNF gene are improved by fusing the TNF sequence with those of peptides able to target tumor vessels, such as CNGRCG or ACDCRGDCFCG. Intramuscular administration of plasmid DNA encoding CNGRCG-TNF and ACDCRGDCFCG-TNF (pNGR-TNF and pRGD-TNF, respectively), but not plasmids encoding TNF (pTNF) or empty vector (pMock), inhibited the growth of subcutaneous murine B16F1 melanomas and RMA-T lymphomas implanted at sites distant from the site of plasmid injection. The combination of pNGR-TNF or pRGD-TNF with doxorubicin or melphalan induced stronger effects than single agents. These treatments induced antitumor effects without activating toxic or negative feedback mechanisms. In addition, pRGD-TNF increased the uptake of an antibody directed to a tumor-associated antigen. These results suggest that the therapeutic properties of NGR-TNF and RGD-TNF cDNAs are greater than those of TNF cDNA and provide the rationale for developing new gene therapy approaches based on vascular targeting with TNF coupled to tumor-homing peptides.
Collapse
Affiliation(s)
- Natasa Zarovni
- Department of Biological and Technological Research, and Cancer Immunotherapy and Gene Therapy Program, San Raffaele H. Scientific Institute, 20132 Milan, Italy
| | | | | |
Collapse
|
24
|
Mochizuki Y, Nakanishi H, Kodera Y, Ito S, Yamamura Y, Kato T, Hibi K, Akiyama S, Nakao A, Tatematsu M. TNF-alpha promotes progression of peritoneal metastasis as demonstrated using a green fluorescence protein (GFP)-tagged human gastric cancer cell line. Clin Exp Metastasis 2004; 21:39-47. [PMID: 15065601 DOI: 10.1023/b:clin.0000017181.01474.35] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The mechanisms underlying progression of peritoneal metastasis by gastric cancer after micrometastasis formation remain unclear. In the present study, we investigated metastasis to the abdominal wall peritoneum, one of the major features of peritoneal spread, using a human gastric cancer cell line (GCIY-EGFP) tagged with the green fluorescence protein gene (GFP). This model allows sensitive, specific and sequential observation of metastasis development from the initial deposits to peritoneal carcinomatosis at the end stage. In the initial phase, GCIY-EGFP cells could form micrometastasis selectively on the omentum and mesenterium in a milky spot-dependent manner, but not on abdominal wall peritoneum lacking milky spots until the late stages. In vitro analysis using primary mesothelial cells revealed addition of TNF-alpha to decrease their stress fibers, leading to morphological change followed by exposure of the submesothelial extracellular matrix (ECM) in intercellular gaps. Such TNF-alpha pretreatment was found to enhance attachment of tumor cells to the mesothelial monolayer. When tumor cells were injected into the peritoneal cavity of TNF-alpha pretreated mice, they could metastasize to the abdominal wall peritoneum from the very early stages, resulting in accelerated accumulation of ascites than in TNF-alpha non-pretreatment controls. RT-PCR analysis revealed that tumor cells express cytokines and chemokines, including TNF-alpha. Furthermore, TNF-alpha treatment results in up-regulation of expression of monocyte chemoattractant protein-1 (MCP-1) and IL-8 by mesothelial cells and of TNF-alpha itself by inflammatory leukocytes in the peritoneal cavity. These results suggest that metastasis to the abdominal wall peritoneum occurs as a second step from the first omental metastasis in a milky spot-independent manner and that TNF-alpha derived from tumor cells, mesothelial cells and inflammatory leukocytes in the peritoneal cavity may be involved in the progression of peritoneal metastasis.
Collapse
Affiliation(s)
- Yoshinari Mochizuki
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Azmy IAF, Balasubramanian SP, Wilson AG, Stephenson TJ, Cox A, Brown NJ, Reed MWR. Role of tumour necrosis factor gene polymorphisms (-308 and -238) in breast cancer susceptibility and severity. Breast Cancer Res 2004; 6:R395-400. [PMID: 15217507 PMCID: PMC468647 DOI: 10.1186/bcr802] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2003] [Revised: 03/21/2004] [Accepted: 04/26/2004] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Genetic polymorphisms in the promoter region of the tumour necrosis factor (TNF) gene can regulate gene expression and have been associated with inflammatory and malignant conditions. We have investigated two polymorphisms in the promoter of the TNF gene (-308 G>A and -238 G>A) for their role in breast cancer susceptibility and severity by means of an allelic association study. METHODS Using a case-control study design, breast cancer patients (n = 709) and appropriate age-matched and sex-matched controls obtained from the Breast Screening Unit (n = 498) were genotyped for these TNF polymorphisms, using a high-throughput allelic discrimination method. RESULTS Allele frequencies for both polymorphisms were similar in both breast cancer cases and controls. However, the -308 polymorphism was found to be associated with vascular invasion in breast tumours (P = 0.024). Comparison with other standard prognostic indices did not show any association for either genotype. CONCLUSIONS We demonstrated no association between the -308G>A polymorphism and the -238G>A polymorphism in the promoter region of TNF and susceptibility to breast cancer, in a large North European population. However, the -308 G>A polymorphism was found to be associated with the presence of vascular invasion in breast tumours.
Collapse
Affiliation(s)
- Iman A F Azmy
- Academic Surgical Oncology Unit, University of Sheffield, UK.
| | | | | | | | | | | | | |
Collapse
|
26
|
Mignogna MD, Fedele S, Lo Russo L, Lo Muzio L, Bucci E. Immune activation and chronic inflammation as the cause of malignancy in oral lichen planus: is there any evidence ? Oral Oncol 2004; 40:120-30. [PMID: 14693234 DOI: 10.1016/j.oraloncology.2003.08.001] [Citation(s) in RCA: 159] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The association of chronic inflammation with a variety of epithelial malignancies has been recognised for centuries. Well established examples include, among many others, oesophageal adenocarcinoma associated with chronic oesophagitis and bowel cancer associated with chronic inflammatory bowel diseases. By now no data, other than clinical observation, have been available in understanding the pathogenesis of these inflammation-related tumours. However, recent molecular studies on the relationship between solid malignancies and the surrounding stroma have given new insights. There is now enough evidence to accept that the chronic inflammatory process per se is able to provide a cytokine-based microenvironment which is able to influence cell survival, growth, proliferation, differentiation and movement, hence contributing to cancer initiation, progression, invasion and metastasis. Here it is discussed whether also oral lichen planus (OLP), being a chronic inflammatory autoimmune disease which has been clinically associated with development of oral squamous cell carcinoma, might be categorised among these disorders. With this aim, we critically reviewed and detailed the presence, in OLP subepithelial infiltrate, of inflammatory cells and cytokine networks that might act to promote squamous tumorigenesis.
Collapse
Affiliation(s)
- Michele D Mignogna
- Section of Oral Medicine, Department of Odontostomatological and Maxillofacial Sciences, University Federico II, Naples, Italy.
| | | | | | | | | |
Collapse
|
27
|
Szlosarek PW, Balkwill FR. Tumour necrosis factor alpha: a potential target for the therapy of solid tumours. Lancet Oncol 2003; 4:565-73. [PMID: 12965278 DOI: 10.1016/s1470-2045(03)01196-3] [Citation(s) in RCA: 267] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Tumour necrosis factor alpha (TNFalpha), named for its antitumour properties, was isolated almost 30 years ago. It is a vital member of the multifunctional TNF superfamily and has important roles in immunity and cellular remodelling as well as influencing apoptosis and cell survival. Its central role in inflammation has led to the development of TNFalpha antagonists as effective therapies for rheumatoid arthritis and inflammatory bowel disease. In this review, we discuss the evidence which has accumulated during the past decade that implicates TNFalpha in inflammatory pathways that increase tumorigenesis. There is convincing evidence that under specific conditions TNFalpha is a tumour promoter and helps to produce the toxic effects associated with conventional cancer therapy, such as the cytokine release syndrome and cisplatin-induced nephrotoxicity. Several trials have been set up to investigate the role of TNFalpha antagonists in cancer. It is hoped that these agents inhibit the neoplastic process either alone or in combination with other agents, and ameliorate the side effects of cancer therapy.
Collapse
Affiliation(s)
- Peter W Szlosarek
- Cancer Research UK, Translational Oncology Laboratory, Barts and The London, Queen Mary's School of Medicine and Dentistry, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | | |
Collapse
|
28
|
Abstract
BACKGROUND Endometriosis appears to predispose to ovarian cancer. How this may occur has been little discussed. STUDY DESIGN This article reviews the English language literature for in vitro, animal, clinical, and epidemiologic studies linking the two conditions. RESULTS Pathology case series consistently report endometrioid and clear cell types of ovarian cancer arising from endometriotic foci. Epidemiologic studies have been consistent with this association. There are also marked similarities between the proposed etiology of ovarian cancer and the observed pathophysiology of endometriosis. Specifically, both are characterized by immune alterations. Both conditions are promoted by estrogen excess and by progesterone deficit. Finally, steroid hormones interacting with the immune system may stimulate both endometriosis and ovarian cancer. I propose that the biology common to endometriosis and ovarian cancer represents not just a parallelism, but instead a causal pathway: aberrant immune function, fed by and feeding on estrogens, unbalanced by progesterone, may create a positive feed-forward loop that enhances the growth and invasiveness of endometriosis and promotes its malignant transformation. CONCLUSIONS The same pathophysiology may orchestrate the progression of endometriosis and its transformation to endometroid and clear cell ovarian neoplasias. This notion of a unifying biology suggests a directed approach to future research and identifies possible chemoprevention strategies for women with endometriosis.
Collapse
Affiliation(s)
- Roberta B Ness
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, the University of Pittsburgh Cancer Institute, Pittsburgh, PA 15261, USA.
| |
Collapse
|
29
|
Abstract
Nuclear factor-kappa B (NF-kappa B) has long been known to play a central role in the immune system by regulating the expression of key genes. Moreover, activation of this transcription factor helps a wide variety of cell types survive damage induced by pro-apoptotic stimuli. Because of its crucial role in the regulation of pro-inflammatory genes, NF-kappa B is a promising target for the discovery of anti-inflammatory drugs. More recently, NF-kappa B has also emerged as a major culprit in a variety of human cancers mainly because of its ability to protect transformed cells from apoptosis. The pharmaceutical industry should, therefore, seriously consider testing inhibitors of NF-kappa B, identified as part of their anti-inflammatory drug discovery programs, in combination with other chemotherapeutic drugs in models of cancer.
Collapse
Affiliation(s)
- Burkhard Haefner
- Department of Inflammatory Disease, Johnson & Johnson Pharmaceutical R&D, A Division of Janssen Pharmaceutica, Turnhoutseweg 30, 2340 Beerse, Belgium.
| |
Collapse
|
30
|
Affiliation(s)
- Fran Balkwill
- ICRF Translational Oncology Laboratory, Barts and the London, Queen Mary's School of Medicine and Dentistry, Science Building, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
31
|
Abstract
The epithelial tumour microenvironment is a complex tissue comprising variable numbers of tumour cells, fibroblasts, endothelial cells and infiltrating leucocytes. Cytokines are key molecules controlling autocrine or paracrine communications within and between these individual cell types. Under some circumstances, endogenous cytokines may orchestrate host responses against the tumour, but there is increasing evidence that the cytokine network contributes to tumour growth, progression and host immuno-suppression. In this review we outline some of the actions of endogenous cytokines in epithelial tumours with particular emphasis on tumour necrosis factor alpha, TNF, related inflammatory cytokines and the chemokine group of chemoattractant cytokines.
Collapse
Affiliation(s)
- Julia Wilson
- ICRF Translational Oncology Laboratory, Barts and The London, Queen Mary's School of Medicine and Dentistry, London EC1M 6BQ, UK
| | | |
Collapse
|
32
|
Abstract
The response of the body to a cancer is not a unique mechanism but has many parallels with inflammation and wound healing. This article reviews the links between cancer and inflammation and discusses the implications of these links for cancer prevention and treatment. We suggest that the inflammatory cells and cytokines found in tumours are more likely to contribute to tumour growth, progression, and immunosuppression than they are to mount an effective host antitumour response. Moreover cancer susceptibility and severity may be associated with functional polymorphisms of inflammatory cytokine genes, and deletion or inhibition of inflammatory cytokines inhibits development of experimental cancer. If genetic damage is the "match that lights the fire" of cancer, some types of inflammation may provide the "fuel that feeds the flames". Over the past ten years information about the cytokine and chemokine network has led to development of a range of cytokine/chemokine antagonists targeted at inflammatory and allergic diseases. The first of these to enter the clinic, tumour necrosis factor antagonists, have shown encouraging efficacy. In this article we have provided a rationale for the use of cytokine and chemokine blockade, and further investigation of non-steroidal anti-inflammatory drugs, in the chemoprevention and treatment of malignant diseases.
Collapse
Affiliation(s)
- F Balkwill
- ICRF Translational Oncology Laboratory, St Bartholomew's and Royal London School of Medicine and Dentistry, UK.
| | | |
Collapse
|
33
|
OH BONGR, SASAKI MASAHIRO, PERINCHERY GEETHA, RYU SOOB, PARK YANGI, CARROLL PETER, DAHIYA RAJVIR. FREQUENT GENOTYPE CHANGES AT −308, AND 488 REGIONS OF THE TUMOR NECROSIS FACTOR-α (TNF-α) GENE IN PATIENTS WITH PROSTATE CANCER. J Urol 2000. [DOI: 10.1016/s0022-5347(05)67683-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- BONG R. OH
- From the Department of Urology, Chonnam University Medical School, Kwangju, Korea, the Department of Obstetrics and Gynecology, School of Medicine, Hokkaido University, Sapporo, Japan, and the Department of Urology, University of California at San Francisco and Veterans Affairs Medical Center, San Francisco, California
| | - MASAHIRO SASAKI
- From the Department of Urology, Chonnam University Medical School, Kwangju, Korea, the Department of Obstetrics and Gynecology, School of Medicine, Hokkaido University, Sapporo, Japan, and the Department of Urology, University of California at San Francisco and Veterans Affairs Medical Center, San Francisco, California
| | - GEETHA PERINCHERY
- From the Department of Urology, Chonnam University Medical School, Kwangju, Korea, the Department of Obstetrics and Gynecology, School of Medicine, Hokkaido University, Sapporo, Japan, and the Department of Urology, University of California at San Francisco and Veterans Affairs Medical Center, San Francisco, California
| | - SOO B. RYU
- From the Department of Urology, Chonnam University Medical School, Kwangju, Korea, the Department of Obstetrics and Gynecology, School of Medicine, Hokkaido University, Sapporo, Japan, and the Department of Urology, University of California at San Francisco and Veterans Affairs Medical Center, San Francisco, California
| | - YANG I. PARK
- From the Department of Urology, Chonnam University Medical School, Kwangju, Korea, the Department of Obstetrics and Gynecology, School of Medicine, Hokkaido University, Sapporo, Japan, and the Department of Urology, University of California at San Francisco and Veterans Affairs Medical Center, San Francisco, California
| | - PETER CARROLL
- From the Department of Urology, Chonnam University Medical School, Kwangju, Korea, the Department of Obstetrics and Gynecology, School of Medicine, Hokkaido University, Sapporo, Japan, and the Department of Urology, University of California at San Francisco and Veterans Affairs Medical Center, San Francisco, California
| | - RAJVIR DAHIYA
- From the Department of Urology, Chonnam University Medical School, Kwangju, Korea, the Department of Obstetrics and Gynecology, School of Medicine, Hokkaido University, Sapporo, Japan, and the Department of Urology, University of California at San Francisco and Veterans Affairs Medical Center, San Francisco, California
| |
Collapse
|
34
|
Feldman AM, Combes A, Wagner D, Kadakomi T, Kubota T, Li YY, McTiernan C. The role of tumor necrosis factor in the pathophysiology of heart failure. J Am Coll Cardiol 2000; 35:537-44. [PMID: 10716453 DOI: 10.1016/s0735-1097(99)00600-2] [Citation(s) in RCA: 357] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Recent studies have focused their attention on the role of the proinflammatory cytokine tumor necrosis factor (TNF) in the development of heart failure. First recognized as an endotoxin-induced serum factor that caused necrosis of tumors and cachexia, it is now recognized that TNF participates in the pathophysiology of a group of inflammatory diseases including rheumatoid arthritis and Crohn's disease. The normal heart does not express TNF; however, the failing heart produces robust quantities. Furthermore, there is a direct relationship between the level of TNF expression and the severity of disease. In addition, both in vivo and in vitro studies demonstrate that TNF effects cellular and biochemical changes that mirror those seen in patients with congestive heart failure. Furthermore, in animal models, the development of the heart failure phenotype can be abrogated at least in part by anticytokine therapy. Based on information from experimental studies, investigators are now evaluating the clinical efficacy of novel anticytokine and anti-TNF strategies in patients with heart failure; one such strategy is the use of a recombinantly produced chimeric TNF alpha soluble receptor. Thus, in view of the emerging importance of proinflammatory cytokines in the pathogenesis of heart disease, we review the biology of TNF, its role in inflammatory diseases, the effects of TNF on the physiology of the heart and the development of clinical strategies that target the cytokine pathways.
Collapse
Affiliation(s)
- A M Feldman
- Cardiovascular Institute of the UPMC Health System, Pittsburgh, Pennsylvania 15213, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Gaiotti D, Chung J, Iglesias M, Nees M, Baker PD, Evans CH, Woodworth CD. Tumor necrosis factor-alpha promotes human papillomavirus (HPV) E6/E7 RNA expression and cyclin-dependent kinase activity in HPV-immortalized keratinocytes by a ras-dependent pathway. Mol Carcinog 2000; 27:97-109. [PMID: 10657902 DOI: 10.1002/(sici)1098-2744(200002)27:2<97::aid-mc5>3.0.co;2-v] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Tumor necrosis factor-alpha (TNF-alpha) inhibits growth of normal cervical keratinocytes but stimulates proliferation of human papillomavirus (HPV)-immortalized and cervical carcinoma-derived cell lines when mitogens such as epidermal growth factor (EGF) or serum are depleted. Current work identifies the mechanism of growth stimulation. TNF-alpha promoted cell cycle progression by increasing expression of HPV-16 E6/E7 RNAs and enhancing activity of cyclin-dependent kinase (cdk)2 and cdc2 after 3 d. Increased kinase activity was mediated by upregulation of cyclins A and B and decreases in cdk inhibitors p21(waf) and p27(kip). TNF-alpha stimulated these changes in part by increasing transcription and stabilization of RNA for amphiregulin, an EGF receptor ligand, and amphiregulin directly increased HPV-16 E6/E7 and cyclin A RNAs. To define which components of the EGF receptor signaling pathway were important, HPV-immortalized cells were transfected with activated or dominant negative mutants of Ha-ras, raf, or MAPKK. Expression of activated Ha-ras maintained HPV-16 and cyclin gene expression and promoted rapid growth in the absence of EGF. Furthermore, ras activation was necessary for TNF-alpha mitogenesis as transfection with a dominant negative ras mutant (Asn-17) strongly inhibited growth. Thus, activation of ras promotes expression of HPV-16 E6/E7 RNAs, induces cyclins A and B, and mediates growth stimulation of immortal keratinocytes by TNF-alpha. These studies define a pathway by which ras mutations, which occur in a subset of cervical cancers, may contribute to pathogenesis. Mol. Carcinog. 27:97-109, 2000. Published by Wiley-Liss, Inc.
Collapse
Affiliation(s)
- D Gaiotti
- Laboratory of Biology, National Cancer Institute, Bethesda, MD 20892-4255, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Bookman MA. Biological therapy for gynecologic malignancies. Cancer Treat Res 1998; 95:115-47. [PMID: 9619282 DOI: 10.1007/978-1-4615-5447-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- M A Bookman
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| |
Collapse
|
37
|
van Geene, Kehoe, Luesley, Ward. Growth promoting properties of cell free malignant ascites and benign peritoneal fluid on epithelial ovarian carcinoma cells in culture. Int J Gynecol Cancer 1998. [DOI: 10.1046/j.1525-1438.1998.09715.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
38
|
Gallagher G, Lindemann M, Oh HH, Ferencik S, Walz MK, Schmitz A, Richards S, Eskdale J, Field M, Grosse-Wilde H. Association of the TNFa2 microsatellite allele with the presence of colorectal cancer. TISSUE ANTIGENS 1997; 50:47-51. [PMID: 9243755 DOI: 10.1111/j.1399-0039.1997.tb02833.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Independently, our two groups collaborating in this report performed association studies to consider the influence of the TNF region within the human MHC on the presence of colorectal cancer. In the Glasgow Study, 84 colorectal cancer patients were compared with 91 controls at the TNFa microsatellite locus. Analysis of individual alleles by Fisher's exact test revealed a significant overrepresentation of the TNFa2 allele in the colorectal cancer group, after correcting for multiple comparisons. In the Essen Study, 47 colorectal cancer patients were compared with 117 matched controls, and the hypothesis of TNFa2 overrepresentation in colorectal cancer was confirmed. These data provide evidence for the involvement of the TNF locus in the pathogenetic etiology of colorectal cancer.
Collapse
Affiliation(s)
- G Gallagher
- Department of Surgery, University of Glasgow, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Burke F, East N, Upton C, Patel K, Balkwill FR. Interferon gamma induces cell cycle arrest and apoptosis in a model of ovarian cancer: enhancement of effect by batimastat. Eur J Cancer 1997; 33:1114-21. [PMID: 9376192 DOI: 10.1016/s0959-8049(97)88065-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Locoregional human IFN-gamma may have activity against refractory ovarian cancer. We investigated this further in an ovarian cancer xenograft model. Administered at clinically relevant doses, intraperitoneal IFN-gamma prolonged the survival of mice bearing multiple established peritoneal tumours, with optimal treatment giving a 3-6-fold increase in median survival time. Daily dosing, which was superior to intermittent treatment, decreased DNA synthesis and induced apoptosis in tumour cells with maximal effects after 7-21 days treatment. This was preceded by an increase in p53 protein at 48 h. The effect of IFN-gamma was not enhanced by sequential treatment with carboplatin. However, the matrix metalloprotease inhibitor, batimastat, further increased mouse survival when given after IFN-gamma. Thus IFN-gamma is cytotoxic to ovarian epithelial cells in vivo and intensive locoregional dosing over short periods is effective. Sequential administration of novel agents that perturb the host/tumour relationship may be of benefit.
Collapse
Affiliation(s)
- F Burke
- Biological Therapy Laboratory, Imperial Cancer Research Fund, London, U.K
| | | | | | | | | |
Collapse
|
40
|
Aderka D. The potential biological and clinical significance of the soluble tumor necrosis factor receptors. Cytokine Growth Factor Rev 1996; 7:231-40. [PMID: 8971478 DOI: 10.1016/s1359-6101(96)00026-3] [Citation(s) in RCA: 216] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The role of TNF receptors (TNF-Rs) is not limited to signal transduction but includes extracellular regulatory functions affecting systemic TNF bioavailability. This review summarizes the regulation of TNF-R shedding and its kinetics, the complex interaction between the soluble receptors and their ligand in vitro and in vivo, and the potential diagnostic, prognostic and therapeutic value of the soluble receptors in malignant, inflammatory, infectious and autoimmune disorders.
Collapse
Affiliation(s)
- D Aderka
- Sackler Faculty of Medicine, Tel Aviv University, Dept of Oncology, Tel-Aviv Medical Center, Israel
| |
Collapse
|
41
|
Burke F, Balkwill FR. Cytokines in animal models of cancer. BIOTHERAPY (DORDRECHT, NETHERLANDS) 1996; 8:229-41. [PMID: 8813335 DOI: 10.1007/bf01877209] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cytokines are a complex family of mediators that play a wide role in development, immunity, inflammation and tissue repair. Their use in therapy is still in its infancy and animal models have a key role to play in optimizing doses and schedules. Whilst xenogeneic and syngeneic transplantable systems have traditionally been used to look at the effects of cytokines in tumour models, oncogene transgenic mice prone to develop cancer, may now have a role to play. Moreover, gene therapy has allowed the investigation of ectopically expressed high and continuous levels of cytokines. We will attempt to review the literature on the effect of cytokines and their combinations in these models of cancer.
Collapse
Affiliation(s)
- F Burke
- Biological Therapies Laboratory, Imperial Cancer Research Fund, London, UK
| | | |
Collapse
|
42
|
Behammer W, Kluge M, Rüschoff J, Männel DN. Tumor necrosis factor effects on ascites formation in an experimental tumor model. J Interferon Cytokine Res 1996; 16:403-8. [PMID: 8727081 DOI: 10.1089/jir.1996.16.403] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
In humans, treatment of malignant ascites with bolus TNF leads to resolution of the ascites. In an experimental model NMRI nude mice were inoculated intraperitoneally with human NIH-OVCAR3 adenocarcinoma cells, resulting in production of ascites and intraperitoneal tumor growth. Ascites formation and tumor growth after IP injection of recombinant human TNF was determined. Depending on the treatment schedule, a dual effect of TNF on the development of ascites was seen. Doses of TNF (1-10 micrograms/g) given once per week completely prevented ascites production, whereas the same doses of TNF given on a daily schedule induced enhanced ascites formation in an inverse TNF dose relationship. The area of tumor cell-covered peritoneal lining corresponded to these findings, indicating a correlation of tumor mass with ascites production. In an attempt to prevent renewal of ascites after drainage, neither inhibition nor enhancement in ascites production was seen when TNF was given five times per week. However, doses of 10 micrograms/g of TNF once per week led to almost complete inhibition of ascites reappearance. Histological examination of animals that received repeated TNF treatment demonstrated chronic peritonitis with strong stromal proliferation, angiogenesis, and increased adhesion of tumor cells to the peritoneum.
Collapse
Affiliation(s)
- W Behammer
- Institute of Pathology/Tumorimmunology, University of Regensburg, Germany
| | | | | | | |
Collapse
|
43
|
Giavazzi R. Cytokine-mediated tumor-endothelial cell interaction in metastasis. Curr Top Microbiol Immunol 1996; 213 ( Pt 2):13-30. [PMID: 9053288 DOI: 10.1007/978-3-642-61109-4_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- R Giavazzi
- Mario Negri Institute for Pharmacological Research, Bergamo, Italy
| |
Collapse
|
44
|
Walker W, Gallagher G. The development of a novel immunotherapy model of human ovarian cancer in human PBL-severe combined immunodeficient (SCID) mice. Clin Exp Immunol 1995; 101:494-501. [PMID: 7664496 PMCID: PMC1553225 DOI: 10.1111/j.1365-2249.1995.tb03140.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The reported ability of SCID mice to accept xenografts of both human tumors and peripheral blood lymphocytes (PBL) provides the potential for the development of novel immunotherapy models in these animals. This study describes the development of a novel small animal model of human ovarian cancer. This was achieved by engrafting a human ovarian cancer cell line (Ovan-4) into the peritoneal cavity of immunodeficient SCID and immune reconstituted human PBL-SCID mice. When transplanted to SCID mice this cell line exhibited growth characteristics similar to the clinical disease observed in patients with implantation of metastatic nodules onto the interior surface of the peritoneal wall. Reconstituted human PBL-SCID mice challenged with identical numbers of Ovan-4 cells exhibited a significant increase in survival time, suggesting a role for cells of the human immune system in preventing the development of this type of malignancy in vivo. Furthermore, vaccination of human PBL-SCID mice against Ovan-4 produced tumour-specific human antibodies in the serum of these animals. Animals reconstituted with CD8-depleted PBL exhibited increased serum immunoglobulin levels and produced enhanced anti-Ovan-4 activity after vaccination. Subsequent challenge of these animals with Ovan-4 revealed a further increase in survival time. These results suggest that human antibodies may have a role in immunity against ovarian cancer and could be of therapeutic value in this type of disease.
Collapse
Affiliation(s)
- W Walker
- Department of Immunology, University of Strathclyde, Glasgow, UK
| | | |
Collapse
|
45
|
Negus RP, Stamp GW, Relf MG, Burke F, Malik ST, Bernasconi S, Allavena P, Sozzani S, Mantovani A, Balkwill FR. The detection and localization of monocyte chemoattractant protein-1 (MCP-1) in human ovarian cancer. J Clin Invest 1995; 95:2391-6. [PMID: 7738202 PMCID: PMC295866 DOI: 10.1172/jci117933] [Citation(s) in RCA: 240] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Chemokines may control the macrophage infiltrate found in many solid tumors. In human ovarian cancer, in situ hybridization detected mRNA for the macrophage chemokine monocyte chemoattractant protein-1 (MCP-1) in 16/17 serous carcinomas, 4/4 mucinous carcinomas, 2/2 endometrioid carcinomas, and 1/3 borderline tumors. In serous tumors, mRNA expression mainly localized to the epithelial areas, as did immunoreactive MCP-1 protein. In the other tumors, both stromal and epithelial expression were seen. All tumors contained variable numbers of cells positive for the macrophage marker CD68. MCP-1 mRNA was also detected in the stroma of 5/5 normal ovaries. RT-PCR demonstrated mRNA for MCP-1 in 7/7 serous carcinomas and 6/6 ovarian cancer cell lines. MCP-1 protein was detected by ELISA in ascites from patients with ovarian cancer (mean 4.28 ng/ml) and was produced primarily by the cancer cells. Human MCP-1 protein was also detected in culture supernatants from cell lines and in ascites from human ovarian tumor xenografts which induce a peritoneal monocytosis in nude mice. We conclude that the macrophage chemoattractant MCP-1 is produced by epithelial ovarian cancer and that the tumor cells themselves are probably a major source. MCP-1 may contribute to the accumulation of tumor-associated macrophages, which may subsequently influence tumor behavior.
Collapse
Affiliation(s)
- R P Negus
- Biological Therapies Laboratory, Imperial Cancer Research Fund, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Burke F, Knowles RG, East N, Balkwill FR. The role of indoleamine 2,3-dioxygenase in the anti-tumour activity of human interferon-gamma in vivo. Int J Cancer 1995; 60:115-22. [PMID: 7814143 DOI: 10.1002/ijc.2910600117] [Citation(s) in RCA: 80] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We have studied the relationship between L-tryptophan metabolism and the response to human IFN-gamma in 3 human ovarian cancer xenografts growing in nude mice. During IFN-gamma therapy all 3 tumours showed a profound depletion in L-tryptophan and a corresponding rise in L-kynurenine. The microenvironment surrounding the tumours was also depleted of L-tryptophan. The IFN-gamma-inducible enzyme indoleamine dioxygenase, IDO, was induced in treated tumours. While there was a variability in IDO mRNA expression in the different xenografts tested, in situ hybridization showed that the gene was induced at all levels of the tumour, and not just the periphery. These results show that induction of IDO by IFN-gamma in vivo can metabolize L-tryptophan rapidly enough for it to become depleted, despite a continued supply of L-tryptophan from the host. The IDO mRNA and protein remained induced after the L-tryptophan levels had returned to normal, suggesting that the gene may be post-transcriptionally regulated and/or the IDO co-factor supply may be limited. Another IFN-gamma-inducible gene, tryptophanyl tRNA synthetase, was also induced in the tumour. It is possible that this enzyme, which is responsible for synthesizing tryptophanyl tRNA, acts in a compensatory manner by allowing protein synthesis to continue despite low free L-tryptophan concentrations. There was no correlation of the above parameters with the anti-tumour response to IFN-gamma, suggesting that other mechanisms must play a role. L-tryptophan depletion may be a contributor to a multifactorial growth inhibition of tumour cells following IFN-gamma treatment, but cannot on its own explain their growth inhibition.
Collapse
Affiliation(s)
- F Burke
- Biological Therapy Laboratory, Imperial Cancer Research Fund, London, UK
| | | | | | | |
Collapse
|
47
|
Jäckel M, Köpf-Maier P, Tausch-Treml R. Lack of cell-cycle-specific effects of recombinant tumor necrosis factor in vivo. Cancer Immunol Immunother 1994; 39:337-41. [PMID: 7987866 PMCID: PMC11038530 DOI: 10.1007/bf01519988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/1994] [Accepted: 08/17/1994] [Indexed: 01/28/2023]
Abstract
Several in vitro studies have demonstrated that tumor cells arrested in the G2 and M phases of the cell cycle expressed an increased sensitivity to the tumor necrosis factor (TNF). The scope of the present study was to investigate whether this cycle dependence of TNF effects also exists in vivo. The experiments were performed by using the Lewis lung carcinoma (LLC), which had been allotransplanted to nude mice. In order to induce delays of the tumor cell cycle in G2, the animals were treated with etoposide (40 mg/kg body weight i.p.) or with local radiation (15 Gy), each increasing the G2 fraction of the LLC from 10% to 35% and 50% respectively. For combination therapy with recombinant (r)TNF, the tumor was transplanted to four groups of six mice each, one of them serving as a control group the others being treated either with a G2 inductor alone, with rTNF alone, or with rTNF and a G2 inductor combined. Administration of rTNF (125 or 250 micrograms/kg body weight i.v.) was always carried out 24 h after therapy with etoposide or radiation when the maximum of G2 accumulation had developed. The growth behavior of the treated tumors revealed that the response of the LLC to rTNF in vivo was not improved by pretreatment with a G2 inductor and, thus, obviously lacked cell-cycle specificity. It is supposed that direct interactions of TNF with the tumor cells, which are a basic requirement for cell-cycle-linked phenomena, play a minor role in the therapeutic outcome of the LLC under in vivo conditions.
Collapse
Affiliation(s)
- M Jäckel
- Department of Otorhinolaryngology, Klinikum Benjamin-Franklin, Free University of Berlin, Germany
| | | | | |
Collapse
|
48
|
Brown PD. Clinical trials of a low molecular weight matrix metalloproteinase inhibitor in cancer. Ann N Y Acad Sci 1994; 732:217-21. [PMID: 7978793 DOI: 10.1111/j.1749-6632.1994.tb24737.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- P D Brown
- British Bio-technology Ltd., Cowley, Oxford, United Kingdom
| |
Collapse
|
49
|
Christen RD, Isonishi S, Jones JA, Jekunen AP, Hom DK, Kröning R, Gately DP, Thiebaut FB, Los G, Howell SB. Signaling and drug sensitivity. Cancer Metastasis Rev 1994; 13:175-89. [PMID: 7923549 DOI: 10.1007/bf00689635] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Even though alterations in receptor and nonreceptor kinases are involved in the development of human cancer, many cancer cell lines still retain their responsiveness to growth factors. We have investigated the hypothesis that cellular signaling events regulate the sensitivity of cancer cells to chemotherapeutic agents. In 2008 human ovarian carcinoma cells, activation of a number of different transduction pathways resulted in a 2 to 4-fold increase in the sensitivity to cisplatin. These signaling events include pathways activated by the epidermal growth factor (EGF) receptor, tumor necrosis factor alpha (TNF alpha) receptor, bombesin receptor, protein kinase A (PKA), and protein kinase C (PKC). Enhanced sensitivity to chemotherapeutic agents is presumed to be mediated by phosphorylation of critical target protein(s). beta-tubulin has been identified as one such target for the protein kinase signaling cascade. For other signal transduction pathways the key substrates that regulate drug sensitivity have not yet been identified. Recent work has shown that DNA damaging agents activate signaling cascades one of which involves the Src, Ras, and Raf proteins as intermediates and results in induction of a number of genes, including c-fos, c-jun, and the growth arrest and DNA damage-inducible (gadd) genes. This signaling cascade has been shown to involve activation of protein kinase C and to have a protective function. With the growing understanding of how signaling events relate to damage response and drug sensitivity, new and potentially useful strategies for modulating drug sensitivity are evolving.
Collapse
Affiliation(s)
- R D Christen
- Department of Medicine, University of California San Diego, La Jolla, CA 92093-0812
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Miles DW, Happerfield LC, Naylor MS, Bobrow LG, Rubens RD, Balkwill FR. Expression of tumour necrosis factor (TNF alpha) and its receptors in benign and malignant breast tissue. Int J Cancer 1994; 56:777-82. [PMID: 8119765 DOI: 10.1002/ijc.2910560603] [Citation(s) in RCA: 98] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
As well as having anti-tumour activity in animal models. TNF alpha has been implicated in the promotion of tumour invasion and metastasis. Expression and localization of TNF alpha mRNA and protein has been investigated in a series of benign and malignant breast tissues. TNF alpha mRNA was expressed in a minority of cells (< 0.5%) in 4/11 cases of benign breast tissue and in a higher proportion of invasive carcinomas (43/49 cases). Expression of TNF alpha mRNA was focal and confined to the tumour stroma. The presence of TNF alpha was confirmed by immunohistochemistry and again was predominantly stromal. Expression of mRNA and protein localized to infiltrating macrophages defined by the antibody EBM/11 in adjacent sections and using 2-colour immunofluorescence. Although the average number of macrophages per high-power field did not vary among invasive carcinomas of different histological type or grade, the number of cells expressing TNF alpha increased with increasing tumour grade. Immunodetectable p75 TNF receptor and to a lesser extent p55 receptor was present on cells of the mononuclear infiltrate and on the endothelium of invasive carcinomas, but not on the tumour cells themselves.
Collapse
Affiliation(s)
- D W Miles
- Imperial Cancer Research Fund Clinical Oncology Unit, Guy's Hospital, London, UK
| | | | | | | | | | | |
Collapse
|