151
|
The absolute lymphocyte to monocyte ratio is associated with poor prognosis in classical Hodgkin lymphoma patients younger than 60 years of age. Hematol Oncol 2014; 33:133-40. [DOI: 10.1002/hon.2155] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 05/28/2014] [Accepted: 06/25/2014] [Indexed: 11/07/2022]
|
152
|
Tumour-infiltrating Gr-1+ myeloid cells antagonize senescence in cancer. Nature 2014; 515:134-7. [DOI: 10.1038/nature13638] [Citation(s) in RCA: 224] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 07/01/2014] [Indexed: 12/19/2022]
|
153
|
Cheng J, Tian L, Ma J, Gong Y, Zhang Z, Chen Z, Xu B, Xiong H, Li C, Huang Q. Dying tumor cells stimulate proliferation of living tumor cells via caspase-dependent protein kinase Cδ activation in pancreatic ductal adenocarcinoma. Mol Oncol 2014; 9:105-14. [PMID: 25156550 DOI: 10.1016/j.molonc.2014.07.024] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 07/30/2014] [Accepted: 07/30/2014] [Indexed: 12/30/2022] Open
Abstract
Pancreatic cancer is one of the most lethal human cancers, and radiotherapy is often implemented for locally advanced pancreatic ductal adenocarcinoma. Tumor cell repopulation is a major challenge in treating cancers after radiotherapy. In order to address the problem of tumor repopulation, our previous studies have demonstrated that dying cells stimulate the proliferation of living tumor cells after radiotherapy. In particular, dying cells undergoing apoptosis also activate survival or proliferation signals and release growth factors to surrounding living cells. In the present study, we used an in vitro model to examine the possible mechanisms for dying cell stimulated tumor repopulation in pancreatic cancer. In this model, a small number of living, luciferase-labeled pancreatic cancer cells (reporter) were seeded onto a layer of a much larger number of irradiated, unlabeled pancreatic cancer cells and the growth of the living cells was measured over time as a gage of tumor repopulation. Our results indicate that irradiated, dying Panc1 feeder cells significantly stimulated the proliferation of living Panc1 reporter cells. Importantly, we identified that the percentage of apoptotic cells and the cleavage of caspases 3 and 7 and protein kinase Cδ (PKCδ) were increased in irradiated Panc1 cells. We presumed that caspases 3 and 7 and PKCδ as integral mediators in the process of dying pancreatic cancer cell stimulation of living tumor cell growth. In order to demonstrate the importance of caspases 3, 7 and PKCδ, we introduced dominant-negative mutants of caspase 3 (DN_C3), caspase 7 (DN_C7), or PKCδ (DN_PKCδ) into Panc1 cells using lentiviral vectors. The stably transduced Panc1 cells were irradiated and used as feeders and we found a significant decrease in the growth of living Panc1 reporter cells when compared with irradiated wild-type Panc1 cells as feeders. Moreover, the role of PKCδ in the growth stimulation of living tumor cells was further confirmed using a pan PKC inhibitor GF109203x and a specific PKCδ inhibitor, rottlerin. Additionally, we found significantly increased phosphorylation of Akt, p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase/stress-activated protein kinase (JNK1/2) in the irradiated Panc1 cells. Mechanistically, PKCδ cleavage was attenuated in both DN_C3 and DN_C7 transduced Panc1 cells, and both Akt and p38 MAPK phosphorylation were attenuated in DN_PKCδ transduced Panc1 cells following radiation. Thus, this report suggests a novel finding that cellular signaling caspase 3/7-PKCδ-Akt/p38 MAPK is crucial to the repopulation in Panc1 cells after radiotherapy.
Collapse
Affiliation(s)
- Jin Cheng
- The Comprehensive Cancer Center & Shanghai Key Laboratory for Pancreatic Diseases, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201620, China
| | - Ling Tian
- Experimental Research Center, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201620, China
| | - Jingjing Ma
- The Comprehensive Cancer Center & Shanghai Key Laboratory for Pancreatic Diseases, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201620, China
| | - Yanping Gong
- Experimental Research Center, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201620, China
| | - Zhengxiang Zhang
- The Comprehensive Cancer Center & Shanghai Key Laboratory for Pancreatic Diseases, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201620, China
| | - Zhiwei Chen
- The Comprehensive Cancer Center & Shanghai Key Laboratory for Pancreatic Diseases, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201620, China
| | - Bing Xu
- The Comprehensive Cancer Center & Shanghai Key Laboratory for Pancreatic Diseases, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201620, China
| | - Hui Xiong
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center, Shanghai 201203, China
| | - Chuanyuan Li
- The Department of Dermatology, Duke University Medical Center, Durham, NC 27710, USA.
| | - Qian Huang
- The Comprehensive Cancer Center & Shanghai Key Laboratory for Pancreatic Diseases, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201620, China.
| |
Collapse
|
154
|
Head and neck cancer relapse after chemoradiotherapy correlates with CD163+ macrophages in primary tumour and CD11b+ myeloid cells in recurrences. Br J Cancer 2014; 111:1509-18. [PMID: 25093488 PMCID: PMC4200089 DOI: 10.1038/bjc.2014.446] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 06/27/2014] [Accepted: 07/13/2014] [Indexed: 12/22/2022] Open
Abstract
Background: We investigated the prognostic role of tumour-associated macrophages (TAMs) in patients with head and neck squamous cell carcinoma (HNSCC) treated with definitive chemoradiotherapy (CRT). Methods: The expression of CD68+, CD163+ and CD11b+ cells was assessed using immunohistochemistry in n=106 pre-treatment tumour biopsy samples and was correlated with clinicopathological characteristics, including T-stage, N-stage, grading, tumour localisation, age and sex as well as local failure-free survival (LFFS), distant metastases-free survival (DMFS), progression-free (PFS), and overall survival (OS). Finally, TAMs expression and vessel density (CD31) were examined in n=12 available early local recurrence samples and compared with their matched primary tumours . The diagnostic images and radiotherapy plans of these 12 patients were also analysed. All local recurrences occurred in the high radiation dose region (⩾70 Gy). Results: With a median follow-up of 40 months, OS at 2 years was 60.5%. High CD163 expression in primary tumours was associated with decreased OS (P=0.010), PFS (P=0.033), LFFS (P=0.036) and DMFS (P=0.038) in multivariate analysis. CD163 demonstrated a strong prognostic value only in human papillomavirus (p16INK4)-negative patients. Early local recurrence specimens demonstrated a significantly increased infiltration of CD11b+ myeloid cells (P=0.0097) but decreased CD31-positive vessel density (P=0.0004) compared with their matched primary samples. Conclusions: Altogether, baseline CD163 expression predicts for an unfavourable clinical outcome in HNSCC after definitive CRT. Early local recurrences showed increased infiltration by CD11b+ cells. These data provide important insight on the role of TAMs in mediating response to CRT in patients with HNSCC.
Collapse
|
155
|
Pastushenko I, Vermeulen PB, Van den Eynden GG, Rutten A, Carapeto FJ, Dirix LY, Van Laere S. Mechanisms of tumour vascularization in cutaneous malignant melanoma: clinical implications. Br J Dermatol 2014; 171:220-33. [PMID: 24641095 DOI: 10.1111/bjd.12973] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2014] [Indexed: 01/02/2023]
Abstract
Malignant melanoma represents < 10% of all skin cancers but is responsible for the majority of skin-cancer-related deaths. Metastatic melanoma has historically been considered as one of the most therapeutically challenging malignancies. Fortunately, for the first time after decades of basic research and clinical investigation, new drugs have produced major clinical responses. Angiogenesis has been considered an important target for cancer treatment. Initial efforts have focused primarily on targeting endothelial and tumour-related vascular endothelial growth factor signalling. Here, we review different mechanisms of tumour vascularization described in melanoma and discuss the potential clinical implications.
Collapse
Affiliation(s)
- I Pastushenko
- Department of Dermatology, Hospital Clínico Universitario 'Lozano Blesa', Zaragoza, 50009, Spain
| | | | | | | | | | | | | |
Collapse
|
156
|
Dexamethasone inhibits in vivo tumor growth by the alteration of bone marrow CD11b+ myeloid cells. Int Immunopharmacol 2014; 21:494-500. [DOI: 10.1016/j.intimp.2014.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 05/28/2014] [Accepted: 06/04/2014] [Indexed: 01/28/2023]
|
157
|
Shen CJ, Sharma A, Vuong DV, Erler JT, Pruschy M, Broggini-Tenzer A. Ionizing radiation induces tumor cell lysyl oxidase secretion. BMC Cancer 2014; 14:532. [PMID: 25052686 PMCID: PMC4223762 DOI: 10.1186/1471-2407-14-532] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 07/16/2014] [Indexed: 11/15/2022] Open
Abstract
Background Ionizing radiation (IR) is a mainstay of cancer therapy, but irradiation can at times also lead to stress responses, which counteract IR-induced cytotoxicity. IR also triggers cellular secretion of vascular endothelial growth factor, transforming growth factor β and matrix metalloproteinases, among others, to promote tumor progression. Lysyl oxidase is known to play an important role in hypoxia-dependent cancer cell dissemination and metastasis. Here, we investigated the effects of IR on the expression and secretion of lysyl oxidase (LOX) from tumor cells. Methods LOX-secretion along with enzymatic activity was investigated in multiple tumor cell lines in response to irradiation. Transwell migration assays were performed to evaluate invasive capacity of naïve tumor cells in response to IR-induced LOX. In vivo studies for confirming IR-enhanced LOX were performed employing immunohistochemistry of tumor tissues and ex vivo analysis of murine blood serum derived from locally irradiated A549-derived tumor xenografts. Results LOX was secreted in a dose dependent way from several tumor cell lines in response to irradiation. IR did not increase LOX-transcription but induced LOX-secretion. LOX-secretion could not be prevented by the microtubule stabilizing agent patupilone. In contrast, hypoxia induced LOX-transcription, and interestingly, hypoxia-dependent LOX-secretion could be counteracted by patupilone. Conditioned media from irradiated tumor cells promoted invasiveness of naïve tumor cells, while conditioned media from irradiated, LOX- siRNA-silenced cells did not stimulate their invasive capacity. Locally applied irradiation to tumor xenografts also increased LOX-secretion in vivo and resulted in enhanced LOX-levels in the murine blood serum. Conclusions These results indicate a differential regulation of LOX-expression and secretion in response to IR and hypoxia, and suggest that LOX may contribute towards an IR-induced migratory phenotype in sublethally-irradiated tumor cells and tumor progression.
Collapse
Affiliation(s)
| | | | | | | | - Martin Pruschy
- Laboratory for Molecular Radiobiology, University Hospital Zurich, 8091 Zürich, Switzerland.
| | | |
Collapse
|
158
|
Reisz JA, Bansal N, Qian J, Zhao W, Furdui CM. Effects of ionizing radiation on biological molecules--mechanisms of damage and emerging methods of detection. Antioxid Redox Signal 2014; 21:260-92. [PMID: 24382094 PMCID: PMC4060780 DOI: 10.1089/ars.2013.5489] [Citation(s) in RCA: 486] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 12/07/2013] [Accepted: 01/01/2014] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE The detrimental effects of ionizing radiation (IR) involve a highly orchestrated series of events that are amplified by endogenous signaling and culminating in oxidative damage to DNA, lipids, proteins, and many metabolites. Despite the global impact of IR, the molecular mechanisms underlying tissue damage reveal that many biomolecules are chemoselectively modified by IR. RECENT ADVANCES The development of high-throughput "omics" technologies for mapping DNA and protein modifications have revolutionized the study of IR effects on biological systems. Studies in cells, tissues, and biological fluids are used to identify molecular features or biomarkers of IR exposure and response and the molecular mechanisms that regulate their expression or synthesis. CRITICAL ISSUES In this review, chemical mechanisms are described for IR-induced modifications of biomolecules along with methods for their detection. Included with the detection methods are crucial experimental considerations and caveats for their use. Additional factors critical to the cellular response to radiation, including alterations in protein expression, metabolomics, and epigenetic factors, are also discussed. FUTURE DIRECTIONS Throughout the review, the synergy of combined "omics" technologies such as genomics and epigenomics, proteomics, and metabolomics is highlighted. These are anticipated to lead to new hypotheses to understand IR effects on biological systems and improve IR-based therapies.
Collapse
Affiliation(s)
- Julie A Reisz
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine , Winston-Salem, North Carolina
| | | | | | | | | |
Collapse
|
159
|
Filatenkov A, Baker J, Müller AM, Ahn GO, Kohrt H, Dutt S, Jensen K, Dejbakhsh-Jones S, Negrin RS, Shizuru JA, Engleman EG, Strober S. Treatment of 4T1 metastatic breast cancer with combined hypofractionated irradiation and autologous T-cell infusion. Radiat Res 2014; 182:163-9. [PMID: 24992165 DOI: 10.1667/rr13471.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The goal of this study was to determine whether a combination of local tumor irradiation and autologous T-cell transplantation can effectively treat metastatic 4T1 breast cancer in mice. BALB/c mice were injected subcutaneously with luciferase-labeled 4T1 breast tumor cells and allowed to grow for 21 days, at which time metastases appeared in the lungs. Primary tumors were treated at that time with 3 daily fractions of 20 Gy of radiation each. Although this approach could eradicate primary tumors, tumors in the lungs grew progressively. We attempted to improve efficacy of the radiation by adding autologous T-cell infusions. Accordingly, T cells were purified from the spleens of tumor-bearing mice after completion of irradiation and cryopreserved. Cyclophosphamide was administered thereafter to induce lymphodepletion, followed by T-cell infusion. Although the addition of cyclophosphamide to irradiation did not improve survival or reduce tumor progression, the combination of radiation, cyclophosphamide and autologous T-cell infusion induced durable remissions and markedly improved survival. We conclude that the combination of radiation and autologous T-cell infusion is an effective treatment for metastatic 4T1 breast cancer.
Collapse
Affiliation(s)
- Alexander Filatenkov
- a Division of Immunology and Rheumatology, Department of Medicine, Stanford University, School of Medicine, Stanford, California 94305
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Al-Batran SE, Werner D. Recent advances and future trends in the targeted therapy of metastatic gastric cancer. Expert Rev Gastroenterol Hepatol 2014; 8:555-69. [PMID: 24665840 DOI: 10.1586/17474124.2014.902304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The better understanding of the molecular mechanisms behind gastric cancer has led to the development of new therapeutic strategies that are likely to improve patient outcomes in the near future. Recently, targeting the HER2 and the VEGF pathways with trastuzumab and ramucirumab, respectively, have been found to improve survival, while directed therapies against a number of other pathways are under clinical evaluation. These include the hepatocyte growth factor and its receptor c-MET, the insulin-like growth factor 1, the fibroblast growth factor, the mammalian target of rapamycin (mTOR), the epidermal growth factor receptor, and other pathways, as well as relevant immunotherapeutic strategies. This article reviews recent advances and future trends of these concepts for gastric cancer and adenocarcinoma of the gastroesophageal junction.
Collapse
Affiliation(s)
- Salah-Eddin Al-Batran
- Krankenhaus Nordwest, UCT-University Cancer Center Frankfurt, Frankfurt am Main, Germany
| | | |
Collapse
|
161
|
Radiobiologie des doses ablatives en radiothérapie stéréotaxique : mise au point des données récentes. Cancer Radiother 2014; 18:264-9. [DOI: 10.1016/j.canrad.2014.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 04/15/2014] [Indexed: 11/24/2022]
|
162
|
Abstract
Low oxygen tension, hypoxia, is a characteristic of many tumors and associated with the poor prognosis. Hypoxia invites bone marrow derived cells (BMDCs) from bone marrow to the site of tumor. These recruited CXCR4+ BMDCs provide favorable environment for the tumor growth by acquiring pro-angiogenic phenotype such as CD45+VEGFR2+ Endothelial Progenitor Cells (EPC), or CD45+Tie2+ myeloid cells. CD11b+CD13+ myeloid population of the BMDCs modulate tumor progression. These myeloid populations retain immunosuppressive characteristics, for example, myeloid derived suppressor cells (MDSCs), and regulates immune- suppression by inhibiting cytotoxic T cell function. In addition, MDSCs were observed at the premetastatic niche of the distant organs in other tumors. Protumorigenic and prometastatic role of the myeloid cells provides a basis for therapeutic targeting of immunosuppression and thus inhibiting tumor development and metastasis.
Collapse
Affiliation(s)
- B R Achyut
- Tumor Angiogenesis Lab, Biochemistry and Molecular Biology Department, Cancer Center, Georgia Regents University, USA
| | - Ali S Arbab
- Tumor Angiogenesis Lab, Biochemistry and Molecular Biology Department, Cancer Center, Georgia Regents University, USA
| |
Collapse
|
163
|
Yu CF, Lin CM, Wang SC, Chen FH, Hong JH, Tsai CS, Yang YC, Chiang CS. Effects of pre-irradiation and SDF-1 suppression on the progression of murine astrocytoma cells grown in different stromal beds. Int J Radiat Biol 2014; 90:1162-8. [PMID: 24937369 DOI: 10.3109/09553002.2014.930539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE To examine whether brain tumors grown in pre-irradiated (PreIR) thigh have a similar tumor bed effect (TBE) as in PreIR brain tissue. MATERIAL AND METHODS Tumor growth delay and immunohistochemical (IHC) staining for CD31, an endothelial surface marker, and PIMO, a hypoxia marker, were used to study the TBE of a murine astrocytoma, ALTS1C1, or a stromal-derived factor-1 (SDF-1) gene-silenced astrocytoma, ALTS1C1-SDFkd, growing in different PreIR stroma beds. RESULTS ALTS1C1 tumors growing in both PreIR brain and PreIR thigh had reduced microvascular density (MVD) and more chronic hypoxia, but tumor growth delay was only seen in PreIR brain tissue. In contrast, ALTS1C1-SDFkd tumors showed tumor growth delay in PreIR thigh, with little effect in PreIR brain tissue. CONCLUSIONS This study cautions that both the tumor and the nature of the PreIR stromal bed are important when using pre-irradiation as a model of recurrent brain tumors after radiation therapy.
Collapse
Affiliation(s)
- Ching-Fang Yu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing-Hua University , Hsinchu , Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
164
|
Brown JM. Vasculogenesis: a crucial player in the resistance of solid tumours to radiotherapy. Br J Radiol 2014; 87:20130686. [PMID: 24338942 DOI: 10.1259/bjr.20130686] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Tumours have two main ways to develop a vasculature: by angiogenesis, the sprouting of endothelial cells from nearby blood vessels, and vasculogenesis, the formation of blood vessels from circulating cells. Because tumour irradiation abrogates local angiogenesis, the tumour must rely on the vasculogenesis pathway for regrowth after irradiation. Tumour irradiation produces a marked influx of CD11b(+) myeloid cells (macrophages) into the tumours, and these are crucial to the formation of blood vessels in the tumours after irradiation and for the recurrence of the tumours. This process is driven by increased tumour hypoxia, which increases levels of HIF-1 (hypoxia-inducible factor 1), which in turn upregulates SDF-1 (stromal cell-derived factor 1 or CXCL12), the main driver of the vasculogenesis pathway. Inhibition of HIF-1 or of its downstream target SDF-1 prevents the radiation-induced influx of the CD11b(+) myeloid cells and delays or prevents the tumours from recurring following irradiation. Others and we have shown that with a variety of tumours in both mice and rats, the inhibition of the SDF-1/CXCR4 pathway delays or prevents the recurrence of implanted or autochthonous tumours following irradiation or following treatment with vascular disrupting agents or some chemotherapeutic drugs such as paclitaxel. In addition to the recruited macrophages, endothelial progenitor cells (EPCs) are also recruited to the irradiated tumours, a process also driven by SDF-1. Together, the recruited proangiogenic macrophages and the EPCs reform the tumour vasculature and allow the tumour to regrow following irradiation. This is a new paradigm with major implications for the treatment of solid tumours by radiotherapy.
Collapse
Affiliation(s)
- J M Brown
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| |
Collapse
|
165
|
Koh YW, Park CS, Yoon DH, Suh C, Huh J. Should the cut-off values of the lymphocyte to monocyte ratio for prediction of prognosis in diffuse large B-cell lymphoma be changed in elderly patients? Eur J Haematol 2014; 93:340-8. [PMID: 24766257 DOI: 10.1111/ejh.12354] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2014] [Indexed: 01/20/2023]
Abstract
OBJECTIVES A recent study suggested a prognostic role for the peripheral blood absolute lymphocyte/monocyte ratio (LMR) at diagnosis of diffuse large B-cell lymphoma (DLBCL). Here, we investigated the significance of LMR in DLBCL patients in relation to advanced age. METHODS We examined the prognostic impact of LMR in 603 DLBCL treated with rituximab plus CHOP, using the receiver operating characteristic curve analysis for optimal cut-off values, and performed a subgroup analysis according to age. RESULTS In elderly groups (age ≥ 70), absolute monocyte count was significantly increased, whereas LMR was significantly decreased compared to younger groups. Patients under 70 yr of age with LMR < 3.04 had significantly lower overall survival (OS) and progression-free survival (PFS) compared to those with LMR ≥ 3.04 (P < 0.001 for both). However, in elderly patients (age ≥ 70), there was no significant difference in OS between patients' LMR levels using the 3.04 cut-off value (P = 0.059). Therefore, a new LMR cut-off value of 2.36 was selected in elderly patients, having observed that elderly patients with LMR < 2.36 had significantly lower OS compared to those with LMR ≥ 2.36 (P = 0.021). In multivariate analysis, LMR remained a significant prognostic factor for OS (P = 0.004) or PFS (P < 0.001). CONCLUSIONS We suggest the use of a different cut-off value of LMR in elderly patients to distinguish high-risk from low-risk groups.
Collapse
Affiliation(s)
- Young Wha Koh
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | | | | | | | | |
Collapse
|
166
|
Boer JC, Walenkamp AME, den Dunnen WFA. Recruitment of bone marrow derived cells during anti-angiogenic therapy in GBM: the potential of combination strategies. Crit Rev Oncol Hematol 2014; 92:38-48. [PMID: 24933160 DOI: 10.1016/j.critrevonc.2014.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 04/10/2014] [Accepted: 05/02/2014] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma (GBM) is a highly vascular tumor characterized by rapid and invasive tumor growth, followed by oxygen depletion, hypoxia and neovascularization, which generate a network of disorganized, tortuous and permeable vessels. Recruitment of bone marrow derived cells (BMDC) is crucial for vasculogenesis. These cells may act as vascular progenitors by integrating into the newly formed blood vessels or as vascular modulators by releasing pro-angiogenic factors. In patients with recurrent GBM, anti-vascular endothelial growth factor (VEGF) therapy has been evaluated in combination with chemotherapy, yielding improvements in progression-free survival (PFS). However, benefits are temporary as vascular tumors acquire angiogenic pathways independently of VEGF. Specifically, acute hypoxia following prolonged VEGF depletion induces the recruitment of certain myeloid cell subpopulations, which highly contribute to treatment refractoriness. Here we review the molecular mechanisms of neovascularization in relation to bevacizumab therapy with special emphasis on the recruitment of BMDCs and possible combination therapies for GBM patients.
Collapse
Affiliation(s)
- Jennifer C Boer
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Annemiek M E Walenkamp
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Wilfred F A den Dunnen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
167
|
Liu T, Xie C, Ma H, Zhang S, Liang Y, Shi L, Yu D, Feng Y, Zhang T, Wu G. Gr-1+CD11b+ cells facilitate Lewis lung cancer recurrence by enhancing neovasculature after local irradiation. Sci Rep 2014; 4:4833. [PMID: 24776637 PMCID: PMC4003474 DOI: 10.1038/srep04833] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 04/11/2014] [Indexed: 12/15/2022] Open
Abstract
Studies have shown that bone marrow-derived cells play an important role in tumor recurrence after chemotherapy and radiotherapy. In this study, we examined the relationship between the accumulation of Gr-1+CD11b+ cells and tumor recurrence after irradiation in tumor-bearing mice. By transplanting bone marrow cells into whole body-irradiated mice depleted of bone marrow, we assessed the role of Gr-1+CD11b+ cells in lung carcinoma models after local irradiation (LI). 20 Gy local irradiation could recruit CD11b+CXCR4+ cells into the irradiated tissues, and the recruited CD11b+CXCR4+ cells could promote tumor recurrence. Further 6 Gy whole body irradiation (WBI6Gy) could decrease tumor recurrence by inhibiting the accumulation of Gr-1+CD11b+ cells and then suppressing tumor vasculogenesis and angiogenesis. Our results suggest that the accumulation of CD11b+Gr-1+ cells promote tumor re-growth after local irradiation by enhancing tumor neovascularization, and low dose of whole body irradiation or irradiation of enlarged spleen may provide a new alternative for anti-angiogenesis therapies.
Collapse
Affiliation(s)
- Tao Liu
- 1] Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, P.R. China [2]
| | - Congying Xie
- 1] Departments of Radiation Oncology and Chemotherapy, The First Affiliated Hospital of Wenzhou Medical College, Wenzhou, Zhejiang, 325002, P.R. China [2]
| | - Hong Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, P.R. China
| | - Sheng Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, P.R. China
| | - Yicheng Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, P.R. China
| | - Liangliang Shi
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, P.R. China
| | - Dandan Yu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, P.R. China
| | - Yiming Feng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, P.R. China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, P.R. China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, P.R. China
| |
Collapse
|
168
|
Tumors as organs: biologically augmenting radiation therapy by inhibiting transforming growth factor β activity in carcinomas. Semin Radiat Oncol 2014; 23:242-51. [PMID: 24012338 DOI: 10.1016/j.semradonc.2013.05.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Transforming growth factor β (TGFβ) plays critical roles in regulating a plethora of physiological processes in normal organs, including morphogenesis, embryonic development, stem cell differentiation, immune regulation, and wound healing. Though considered a tumor suppressor, TGFβ is a critical mediator of tumor microenvironment, in which it likewise mediates tumor and stromal cell phenotype, recruitment, inflammation, immune function, and angiogenesis. The fact that activation of TGFβ is an early and persistent event in irradiated tissues and that TGFβ signaling controls effective DNA damage response provides a new means to manipulate tumor response to radiation. Here we discuss preclinical studies unraveling TGFβ effects in cancer treatment and review TGFβ biology in lung cancer as an example of the opportunities for TGFβ pathway inhibition as a pharmaceutical approach to augment radiation therapy.
Collapse
|
169
|
Martin BJ. Inhibiting vasculogenesis after radiation: a new paradigm to improve local control by radiotherapy. Semin Radiat Oncol 2014; 23:281-7. [PMID: 24012342 DOI: 10.1016/j.semradonc.2013.05.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Tumors are supported by blood vessels, and it has long been debated whether their response to irradiation is affected by radiation damage to the vasculature. We have shown in preclinical models that, indeed, radiation is damaging to the tumor vasculature and strongly inhibits tumor angiogenesis. However, the vasculature can recover by colonization from circulating cells, primarily proangiogenenic CD11b+ monocytes or macrophages from the bone marrow. This secondary pathway of blood vessel formation, known as vasculogenesis, thus acts to restore the tumor vasculature and allows the tumor to recur following radiation. The stimulus for the influx of these CD11b+ cells into tumors following irradiation is the increased levels of hypoxia-inducible factor-1 in the tumor due to induced tumor hypoxia secondary to blood vessel loss. This increases tumor levels of the chemokine stromal cell-derived factor-1, which has chemokine receptors CXCR4 and CXCR7 on monocytes and endothelial cells thereby capturing these cells in the tumors. The increase in CD11b+ monocytes in tumors following irradiation can be prevented using antibodies or small molecules that inhibit hypoxia-inducible factor-1 or the interaction of stromal cell-derived factor-1 with its receptors. We show that the effect of inhibiting these chemokine-chemokine receptor interactions is a marked increase in the radiation response of transplanted or chemically induced tumors in mice and rats. This strategy of inhibiting vasculogenesis following tumor irradiation is a new paradigm in radiotherapy and suggests that higher levels of local control of tumors in several sites would be achievable with this strategy.
Collapse
Affiliation(s)
- Brown J Martin
- Department of Radiation Oncology, Stanford University, Stanford, CA.
| |
Collapse
|
170
|
Brown JM, Carlson DJ, Brenner DJ. The tumor radiobiology of SRS and SBRT: are more than the 5 Rs involved? Int J Radiat Oncol Biol Phys 2014; 88:254-62. [PMID: 24411596 DOI: 10.1016/j.ijrobp.2013.07.022] [Citation(s) in RCA: 403] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/14/2013] [Accepted: 07/17/2013] [Indexed: 12/26/2022]
Abstract
Stereotactic radiosurgery (SRS) and stereotactic body radiation therapy (SBRT), also known as stereotactic ablative radiation therapy (SABR), are rapidly becoming accepted practice for the radiation therapy of certain tumors. Typically, SRS and SBRT involve the delivery of 1 or a few large-dose fractions of 8 to 30 Gy per fraction: a major paradigm shift from radiation therapy practice over the past 90 years, when, with relatively large amounts of normal tissues receiving high doses, the goal was to maximize tumor response for an acceptable level of normal tissue injury. The development of SRS and SBRT have come about because of technologic advances in image guidance and treatment delivery techniques that enable the delivery of large doses to tumors with reduced margins and high gradients outside the target, thereby minimizing doses to surrounding normal tissues. Because the results obtained with SRS and SBRT have been impressive, they have raised the question whether classic radiobiological modeling, and the linear-quadratic (LQ) model, are appropriate for large doses per fraction. In addition to objections to the LQ model, the possibility of additional biological effects resulting from endothelial cell damage, enhanced tumor immunity, or both have been raised to account for the success of SRS and SBRT. In this review, we conclude that the available preclinical and clinical data do not support a need to change the LQ model or to invoke phenomena over and above the classic 5 Rs of radiobiology and radiation therapy, with the likely exception that for some tumors high doses of irradiation may produce enhanced antitumor immunity. Thus, we suggest that for most tumors, the standard radiobiology concepts of the 5 Rs are sufficient to explain the clinical data, and the excellent results obtained from clinical studies are the result of the much larger biologically effective doses that are delivered with SRS and SBRT.
Collapse
Affiliation(s)
- J Martin Brown
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.
| | - David J Carlson
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - David J Brenner
- Center for Radiological Research, Columbia University Medical Center, New York, New York
| |
Collapse
|
171
|
Liu Z, Liu Z, Zhang X, Xue P, Zhang H. RY10-4 suppressed metastasis of MDA-MB-231 by stabilizing ECM and E-cadherin. Biomed Pharmacother 2014; 68:439-45. [PMID: 24721328 DOI: 10.1016/j.biopha.2014.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/04/2014] [Indexed: 12/21/2022] Open
Abstract
In the article, we investigated the anti-metastasis mechanism of RY10-4, an anti-tumor compound derived from protoapigenone, in breast tumor cells MB-MDA-231. The analog of protoapigenone with an unaromatic B-ring was verified to suppress the proliferation of several tumor cells by previous research that also showed that several tumor progression such as inducing apoptosis and anti-angiogenesis could be acted on by RY10-4. In the article, we investigated the mechanism about how RY10-4 suppressed the invasion of MDA-MB-231. Firstly, the transwells assays with and without matrigel were adapted to evaluate the anti-metastasis and anti-invasion activity. Much research had demonstrated that the ECM and E-cadherin/β-catenin complex play an important role in cell adhesion and the formation of the cell skeleton, and as we knew the abnormal and absent expression of ECM and E-cadherin/β-catenin complex are found in many malignant cells. The result demonstrated that the amount and distribution of E-cadherin/β-catenin complex were backed on track by RY10-4, and the expression of MMP-2/9 in MDA-MB-231, which functions as a major negative factor of ECM, was down-regulated after co-cultured with RY10-4. Furthermore the pathway related to MMP-2/9 and E-cadherin was assessed by the western blot. As the results showed, the MAPK pathway and the spread of β-catenin were affected by RY10-4 to exert the anti-metastasis on MDA-MB-231. Collectively, the research revealed a novel anti-tumor ability of RY10-4 by inhibiting migration and invasion in MDA-MB-231.
Collapse
Affiliation(s)
- Ziwei Liu
- School of Chemical Engineering & Pharmacy, Wuhan Institute of Technology, Xiongchu Avenue, Wuhan, China
| | - Zhimei Liu
- Humanwell Healthcare (group) Co. Ltd, Gaoxing Avenue, Wuhan, China
| | - Xiulan Zhang
- School of Chemical Engineering & Pharmacy, Wuhan Institute of Technology, Xiongchu Avenue, Wuhan, China
| | - Pingping Xue
- Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation of Hubei Province, College of Pharmacy, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Zhang
- School of Chemical Engineering & Pharmacy, Wuhan Institute of Technology, Xiongchu Avenue, Wuhan, China.
| |
Collapse
|
172
|
Diaz-Montero CM, Finke J, Montero AJ. Myeloid-derived suppressor cells in cancer: therapeutic, predictive, and prognostic implications. Semin Oncol 2014; 41:174-84. [PMID: 24787291 DOI: 10.1053/j.seminoncol.2014.02.003] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Immune evasion is a hallmark of cancer. While there are multiple different mechanisms that cancer cells employ, myeloid-derived suppressor cells (MDSCs) are one of the key drivers of tumor-mediated immune evasion. MDSCs begin as myeloid cells recruited to the tumor microenvironment, where they are transformed into potent immunosuppressive cells. However, our understanding of the clinical relevance of MDSCs in cancer patients has significantly lagged behind the preclinical literature in part due to the absence of a cognate molecule present in mice, as well as to the considerable heterogeneity of MDSCs. However, if one evaluates the clinical literature through the filter of clinically robust endpoints, such as overall survival, three important phenotypes emerge: promyelocytic, monocytic, and granulocytic. Based on these studies, MDSCs have clear prognostic importance in multiple solid tumors, and emerging data support the utility of circulating MDSCs as a predictive marker for cancer immunotherapy, and even as an early leading marker for predicting clinical response to systemic chemotherapy in patients with advanced solid tumors. More recent preclinical data in immunosuppressed murine models suggest that MDSCs play an important role in tumor progression and the metastatic process that is independent of their immunosuppressive properties. Consequently, targeting MDSCs either in combination with cancer immunotherapy or independently as part of an approach to inhibit the metastatic process appears to be a very clinically promising strategy. We review different approaches to target MDSCs that could potentially be tested in future clinical trials in cancer patients.
Collapse
Affiliation(s)
- C Marcela Diaz-Montero
- Lerner Research Institute Department of Immunology; Cleveland Clinic Foundation, Cleveland, OH
| | - Jim Finke
- Lerner Research Institute Department of Immunology; Cleveland Clinic Foundation, Cleveland, OH
| | - Alberto J Montero
- Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH.
| |
Collapse
|
173
|
Wang H, Melton DW, Porter L, Sarwar ZU, McManus LM, Shireman PK. Altered macrophage phenotype transition impairs skeletal muscle regeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1167-1184. [PMID: 24525152 DOI: 10.1016/j.ajpath.2013.12.020] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 11/05/2013] [Accepted: 12/26/2013] [Indexed: 12/31/2022]
Abstract
Monocyte/macrophage polarization in skeletal muscle regeneration is ill defined. We used CD11b-diphtheria toxin receptor transgenic mice to transiently deplete monocytes/macrophages at multiple stages before and after muscle injury induced by cardiotoxin. Fat accumulation within regenerated muscle was maximal when ablation occurred at the same time as cardiotoxin-induced injury. Early ablation (day 1 after cardiotoxin) resulted in the smallest regenerated myofiber size together with increased residual necrotic myofibers and fat accumulation. However, muscle regeneration after late (day 4) ablation was similar to controls. Levels of inflammatory cells in injured muscle following early ablation and associated with impaired muscle regeneration were determined by flow cytometry. Delayed, but exaggerated, monocyte [CD11b(+)(CD90/B220/CD49b/NK1.1/Ly6G)(-)(F4/80/I-Ab/CD11c)(-)Ly6C(+/-)] accumulation occurred; interestingly, Ly6C(+) and Ly6C(-) monocytes were present concurrently in ablated animals and control mice. In addition to monocytes, proinflammatory, Ly6C(+) macrophage accumulation following early ablation was delayed compared to controls. In both groups, CD11b(+)F4/80(+) cells exhibited minimal expression of the M2 markers CD206 and CD301. Nevertheless, early ablation delayed and decreased the transient accumulation of CD11b(+)F4/80(+)Ly6C(-)CD301(-) macrophages; in control animals, the later tissue accumulation of these cells appeared to correspond to that of anti-inflammatory macrophages, determined by cytokine production and arginase activity. In summary, impairments in muscle regeneration were associated with exaggerated monocyte recruitment and reduced Ly6C(-) macrophages; the switch of macrophage/monocyte subsets is critical to muscle regeneration.
Collapse
Affiliation(s)
- Hanzhou Wang
- Department of Surgery, University of Texas Health Science Center, San Antonio, Texas
| | - David W Melton
- Department of Surgery, University of Texas Health Science Center, San Antonio, Texas; Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, Texas; Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, Texas
| | - Laurel Porter
- Department of Surgery, University of Texas Health Science Center, San Antonio, Texas
| | - Zaheer U Sarwar
- Department of Surgery, University of Texas Health Science Center, San Antonio, Texas
| | - Linda M McManus
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas; Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, Texas
| | - Paula K Shireman
- Department of Surgery, University of Texas Health Science Center, San Antonio, Texas; Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, Texas; The South Texas Veterans Health Care System, San Antonio, Texas.
| |
Collapse
|
174
|
Fang M, Peng CW, Yuan JP, Zhang ZL, Pang DW, Li Y. Coevolution of the tumor microenvironment revealed by quantum dot-based multiplexed imaging of hepatocellular carcinoma. Future Oncol 2014; 9:1029-37. [PMID: 23837765 DOI: 10.2217/fon.13.63] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
AIM This study aimed to provide new insights into the mechanisms of hepatocellular carcinoma (HCC) invasion by simultaneously imaging tumor cells and major components of the tumor microenvironment. MATERIALS & METHODS Formalin-fixed paraffin-embedded human HCC tissues were studied by conventional immunohistochemistry and quantum dot-based multiplexed imaging to reveal type IV collagen, LOX and tumor angiogenesis. RESULTS Type IV collagen degradation and repatterning in the extracellular matrix (ECM) was a continuous process, making the ECM harder, although more fragile and less resistant to cancer invasion. The distribution of LOX among cancer nests was heterogeneous, with higher expression in small cancer nests and lower expression in large cancer nests. LOX expression in cancer cells was associated with rigid stroma and tumor angiogenesis. Tumor angiogenesis occurred with type IV collagen presence. At the cancer invasion front, the ECM was hydrolyzed, with the prominent linear reorientation of type IV collagen surrounding cancer nests adjacent to neovessels. CONCLUSION The visualization of the temporal-spatial relationship between type IV collagen, LOX and tumor angiogenesis revealed the coevolution process of HCC cells and their microenvironment, emphasizing an active role of the ECM during cancer invasion.
Collapse
Affiliation(s)
- Min Fang
- Department of Oncology, Zhongnan Hospital of Wuhan University & Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Cancer Clinical Study Center, Number 169 Donghu Road, Wuchang District, Wuhan, 430071, PR China
| | | | | | | | | | | |
Collapse
|
175
|
Farina AR, Mackay AR. Gelatinase B/MMP-9 in Tumour Pathogenesis and Progression. Cancers (Basel) 2014; 6:240-96. [PMID: 24473089 PMCID: PMC3980597 DOI: 10.3390/cancers6010240] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/20/2014] [Accepted: 01/21/2014] [Indexed: 12/14/2022] Open
Abstract
Since its original identification as a leukocyte gelatinase/type V collagenase and tumour type IV collagenase, gelatinase B/matrix metalloproteinase (MMP)-9 is now recognised as playing a central role in many aspects of tumour progression. In this review, we relate current concepts concerning the many ways in which gelatinase B/MMP-9 influences tumour biology. Following a brief outline of the gelatinase B/MMP-9 gene and protein, we analyse the role(s) of gelatinase B/MMP-9 in different phases of the tumorigenic process, and compare the importance of gelatinase B/MMP-9 source in the carcinogenic process. What becomes apparent is the importance of inflammatory cell-derived gelatinase B/MMP-9 in tumour promotion, early progression and triggering of the "angiogenic switch", the integral relationship between inflammatory, stromal and tumour components with respect to gelatinase B/MMP-9 production and activation, and the fundamental role for gelatinase B/MMP-9 in the formation and maintenance of tumour stem cell and metastatic niches. It is also apparent that gelatinase B/MMP-9 plays important tumour suppressing functions, producing endogenous angiogenesis inhibitors, promoting inflammatory anti-tumour activity, and inducing apoptosis. The fundamental roles of gelatinase B/MMP-9 in cancer biology underpins the need for specific therapeutic inhibitors of gelatinase B/MMP-9 function, the use of which must take into account and substitute for tumour-suppressing gelatinase B/MMP-9 activity and also limit inhibition of physiological gelatinase B/MMP-9 function.
Collapse
Affiliation(s)
- Antonietta Rosella Farina
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, Via Vetoio, Coppito 2, L'Aquila 67100, Italy.
| | - Andrew Reay Mackay
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, Via Vetoio, Coppito 2, L'Aquila 67100, Italy.
| |
Collapse
|
176
|
Hellevik T, Martinez-Zubiaurre I. Radiotherapy and the tumor stroma: the importance of dose and fractionation. Front Oncol 2014; 4:1. [PMID: 24478982 PMCID: PMC3896881 DOI: 10.3389/fonc.2014.00001] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/03/2014] [Indexed: 01/04/2023] Open
Abstract
Ionizing radiation is a non-specific but highly effective way to kill malignant cells. However, tumor recurrence sustained by a minor fraction of surviving tumor cells is a commonplace phenomenon caused by the activation of both cancer cell intrinsic resistance mechanisms, and also extrinsic intermediaries of therapy resistance, represented by non-malignant cells and structural components of the tumor stroma. The improved accuracy offered by advanced radiotherapy (RT)-technology permits reduced volume of healthy tissue in the irradiated field, and has been triggering an increase in the prescription of high-dose oligo-fractionated regimens in the clinics. Given the remarkable clinical success of high-dose RT and the current therapeutic shift occurring in the field, in this review we revise the existing knowledge on the effects that different radiation regimens exert on the different compartments of the tumor microenvironment, and highlight the importance of anti-tumor immunity and other tumor cell extrinsic mechanisms influencing therapeutic responses to high-dose radiation.
Collapse
Affiliation(s)
- Turid Hellevik
- Department of Oncology, University Hospital of Northern-Norway , Tromsø , Norway ; Translational Cancer Research Group, Department of Clinical Medicine, University of Tromsø , Tromsø , Norway
| | - Iñigo Martinez-Zubiaurre
- Translational Cancer Research Group, Department of Clinical Medicine, University of Tromsø , Tromsø , Norway
| |
Collapse
|
177
|
Tsai YD, Wang CP, Chen CY, Lin LW, Hwang TZ, Lu LF, Hsu HF, Chung FM, Lee YJ, Houng JY. Pretreatment circulating monocyte count associated with poor prognosis in patients with oral cavity cancer. Head Neck 2014; 36:947-53. [DOI: 10.1002/hed.23400] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 03/06/2013] [Accepted: 05/23/2013] [Indexed: 11/11/2022] Open
Affiliation(s)
- Yu-Duan Tsai
- Division of Neurology, E-Da Hospital; I-Shou University; Kaohsiung Taiwan
- Department of Medical Nutrition; Institute of Biotechnology and Chemical Engineering and I-Shou University; Kaohsiung Taiwan
| | - Chao-Ping Wang
- Division of Cardiology, Department of Internal Medicine, E-Da Hospital; I-Shou University; Kaohsiung Taiwan
| | - Chih-Yu Chen
- Department of Dentistry, E-Da Hospital; I-Shou University; Kaohsiung Taiwan
| | - Li-Wen Lin
- Department of Dentistry, E-Da Hospital; I-Shou University; Kaohsiung Taiwan
| | - Tzer-Zen Hwang
- Department of Otolaryngology, E-Da Hospital; I-Shou University; Kaohsiung Taiwan
| | - Li-Fen Lu
- Division of Cardiac Surgery, Department of Surgery, E-Da Hospital; I-Shou University; Kaohsiung Taiwan
| | - Hsia-Fen Hsu
- Department of Medical Nutrition; Institute of Biotechnology and Chemical Engineering and I-Shou University; Kaohsiung Taiwan
| | - Fu-Mei Chung
- Division of Cardiology, Department of Internal Medicine, E-Da Hospital; I-Shou University; Kaohsiung Taiwan
| | | | - Jer-Yiing Houng
- Department of Medical Nutrition; Institute of Biotechnology and Chemical Engineering and I-Shou University; Kaohsiung Taiwan
| |
Collapse
|
178
|
Walters MJ, Ebsworth K, Berahovich RD, Penfold MET, Liu SC, Al Omran R, Kioi M, Chernikova SB, Tseng D, Mulkearns-Hubert EE, Sinyuk M, Ransohoff RM, Lathia JD, Karamchandani J, Kohrt HEK, Zhang P, Powers JP, Jaen JC, Schall TJ, Merchant M, Recht L, Brown JM. Inhibition of CXCR7 extends survival following irradiation of brain tumours in mice and rats. Br J Cancer 2014; 110:1179-88. [PMID: 24423923 PMCID: PMC3950859 DOI: 10.1038/bjc.2013.830] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/12/2013] [Accepted: 12/18/2013] [Indexed: 12/26/2022] Open
Abstract
Background: In experimental models of glioblastoma multiforme (GBM), irradiation (IR) induces local expression of the chemokine CXCL12/SDF-1, which promotes tumour recurrence. The role of CXCR7, the high-affinity receptor for CXCL12, in the tumour's response to IR has not been addressed. Methods: We tested CXCR7 inhibitors for their effects on tumour growth and/or animal survival post IR in three rodent GBM models. We used immunohistochemistry to determine where CXCR7 protein is expressed in the tumours and in human GBM samples. We used neurosphere formation assays with human GBM xenografts to determine whether CXCR7 is required for cancer stem cell (CSC) activity in vitro. Results: CXCR7 was detected on tumour cells and/or tumour-associated vasculature in the rodent models and in human GBM. In human GBM, CXCR7 expression increased with glioma grade and was spatially associated with CXCL12 and CXCL11/I-TAC. In the rodent GBM models, pharmacological inhibition of CXCR7 post IR caused tumour regression, blocked tumour recurrence, and/or substantially prolonged survival. CXCR7 expression levels on human GBM xenograft cells correlated with neurosphere-forming activity, and a CXCR7 inhibitor blocked sphere formation by sorted CSCs. Conclusions: These results indicate that CXCR7 inhibitors could block GBM tumour recurrence after IR, perhaps by interfering with CSCs.
Collapse
Affiliation(s)
- M J Walters
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA 94043, USA
| | - K Ebsworth
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA 94043, USA
| | - R D Berahovich
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA 94043, USA
| | - M E T Penfold
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA 94043, USA
| | - S-C Liu
- Department of Radiation Oncology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - R Al Omran
- Department of Radiation Oncology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - M Kioi
- Department of Radiation Oncology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - S B Chernikova
- Department of Radiation Oncology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - D Tseng
- Department of Radiation Oncology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - E E Mulkearns-Hubert
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - M Sinyuk
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - R M Ransohoff
- 1] Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA [2] Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - J D Lathia
- 1] Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA [2] Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - J Karamchandani
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - H E K Kohrt
- Department of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - P Zhang
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA 94043, USA
| | - J P Powers
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA 94043, USA
| | - J C Jaen
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA 94043, USA
| | - T J Schall
- ChemoCentryx Inc., 850 Maude Ave, Mountain View, CA 94043, USA
| | - M Merchant
- Department of Neurology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - L Recht
- Department of Neurology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| | - J M Brown
- Department of Radiation Oncology, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA
| |
Collapse
|
179
|
Liu T, Du X, Sheng X. Gene expression changes after ionizing radiation in endothelial cells derived from human endometrial cancer-preliminary outcomes. Arch Gynecol Obstet 2014; 289:1315-23. [PMID: 24385285 DOI: 10.1007/s00404-013-3136-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 12/16/2013] [Indexed: 01/09/2023]
Abstract
BACKGROUND Accumulating evidence has demonstrated that death of microvascular endothelial cells plays a decisive role in the tumor response against radiotherapy. Nevertheless, radiation-induced gene alterations on cancer-associated endothelial cells of human endometrial carcinoma remain poorly understood. The purpose of this study was to elucidate the gene expression changes after X-ray radiation in human endometrial carcinoma vascular endothelial cells and to provide new targets for combined treatment of radiation and anti-angiogenesis in human endometrial carcinoma. MATERIALS AND METHODS Endometrial cancer-derived endothelial cells, which obtained before and 4 h after 400 cGy X-ray radiation from four endometrial carcinomas, were analyzed by gene expression profile. The selected meaningful genes from gene microarray experiments were validated by real-time quantitative PCR. RESULTS Microarray analyses showed 49 significantly changed genes which were common to all the microarray experiments. There into, 14 genes were found to be in persistent up-regulation and 14 in persistent down-regulation 4 h after X-ray radiation when compared with the control group. These genes were involved in cell cycle and growth regulation, cell-apoptosis, chemokine, cell signaling, cellular stress response, angiogenesis, DNA synthesis and repair and cell adhesion. Eight randomly selected genes were validated by real-time PCR. DISCUSSION The genes of cancer-derived endothelial cells regulated by X-ray radiation as well as their related signal pathways, which obtained from gene expression profiling data, were relevant to radiosensitivity of endometrial cancer. This study shows that the identified genes and their related signaling pathways are candidated biomarkers for radiation and anti-angiogenesis of human endometrial carcinoma.
Collapse
Affiliation(s)
- Ting Liu
- Department of Gynecologic Oncology, Shandong Cancer Hospital, Jinan, 250117, Shandong, People's Republic of China
| | | | | |
Collapse
|
180
|
Abu El-Asrar AM, Mohammad G, Nawaz MI, Siddiquei MM, Van den Eynde K, Mousa A, De Hertogh G, Opdenakker G. Relationship between vitreous levels of matrix metalloproteinases and vascular endothelial growth factor in proliferative diabetic retinopathy. PLoS One 2013; 8:e85857. [PMID: 24392031 PMCID: PMC3877391 DOI: 10.1371/journal.pone.0085857] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 12/03/2013] [Indexed: 11/17/2022] Open
Abstract
To investigate which matrix metalloproteinases (MMPs) are more likely to be involved in the angiogenic process in proliferative diabetic retinopathy (PDR), we measured the levels of MMPs in the vitreous fluid from patients with PDR and controls and correlated these levels with the levels of vascular endothelial growth factor (VEGF). Vitreous samples from 32 PDR and 24 nondiabetic patients were studied by mosaic multiplex MMPs enzyme-linked immunosorbent assay (ELISA), single ELISA, Western blot and zymography analysis. Epiretinal membranes from 11 patients with PDR were studied by immunohistochemistry. MMP-8 and MMP-13 were not detected. ELISA, Western blot and gelatin ymography assays revealed significant increases in the expression levels of MMP-1, MMP-7, MMP-9 and VEGF in vitreous samples from PDR patients compared to nondiabetic controls, whereas MMP-2 and MMP-3 were not upregulated in vitreous samples from PDR patients. Significant correlations existed between ELISA and zymography assays for the quantitation of MMP-2 (r=0.407; p=0.039) and MMP-9 (r=0.711; p<0.001). Significant correlations were observed between levels of VEGF and levels of MMP-1 (r=0.845; P<0.001) and MMP-9 (r=0.775; p<0.001), and between levels of MMP-1 and MMP-9 (r=0.857; p<0.001). In epiretinal membranes, cytoplasmic immunoreactivity for MMP-9 was present in vascular endothelial cells and stromal monocytes/macrophages and neutrophils. Our findings suggest that among the MMPs measured, MMP-1 and MMP-9 may contribute to the angiogenic switch in PDR.
Collapse
Affiliation(s)
- Ahmed M. Abu El-Asrar
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ghulam Mohammad
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mohd. Imtiaz Nawaz
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | | | - Kathleen Van den Eynde
- Laboratory of Histochemistry and Cytochemistry, University of Leuven, KU Leuven, Belgium
| | - Ahmed Mousa
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Gert De Hertogh
- Laboratory of Histochemistry and Cytochemistry, University of Leuven, KU Leuven, Belgium
| | - Ghislain Opdenakker
- Rega Institute for Medical Research, Department of Microbiology and Immunology, University of Leuven, KU Leuven, Belgium
| |
Collapse
|
181
|
Heger M, van Golen RF, Broekgaarden M, Michel MC. The molecular basis for the pharmacokinetics and pharmacodynamics of curcumin and its metabolites in relation to cancer. Pharmacol Rev 2013; 66:222-307. [PMID: 24368738 DOI: 10.1124/pr.110.004044] [Citation(s) in RCA: 376] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This review addresses the oncopharmacological properties of curcumin at the molecular level. First, the interactions between curcumin and its molecular targets are addressed on the basis of curcumin's distinct chemical properties, which include H-bond donating and accepting capacity of the β-dicarbonyl moiety and the phenylic hydroxyl groups, H-bond accepting capacity of the methoxy ethers, multivalent metal and nonmetal cation binding properties, high partition coefficient, rotamerization around multiple C-C bonds, and the ability to act as a Michael acceptor. Next, the in vitro chemical stability of curcumin is elaborated in the context of its susceptibility to photochemical and chemical modification and degradation (e.g., alkaline hydrolysis). Specific modification and degradatory pathways are provided, which mainly entail radical-based intermediates, and the in vitro catabolites are identified. The implications of curcumin's (photo)chemical instability are addressed in light of pharmaceutical curcumin preparations, the use of curcumin analogues, and implementation of nanoparticulate drug delivery systems. Furthermore, the pharmacokinetics of curcumin and its most important degradation products are detailed in light of curcumin's poor bioavailability. Particular emphasis is placed on xenobiotic phase I and II metabolism as well as excretion of curcumin in the intestines (first pass), the liver (second pass), and other organs in addition to the pharmacokinetics of curcumin metabolites and their systemic clearance. Lastly, a summary is provided of the clinical pharmacodynamics of curcumin followed by a detailed account of curcumin's direct molecular targets, whereby the phenotypical/biological changes induced in cancer cells upon completion of the curcumin-triggered signaling cascade(s) are addressed in the framework of the hallmarks of cancer. The direct molecular targets include the ErbB family of receptors, protein kinase C, enzymes involved in prostaglandin synthesis, vitamin D receptor, and DNA.
Collapse
Affiliation(s)
- Michal Heger
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
182
|
Signal transduction in tumor angiogenesis. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
183
|
Liu SC, Alomran R, Chernikova SB, Lartey F, Stafford J, Jang T, Merchant M, Zboralski D, Zöllner S, Kruschinski A, Klussmann S, Recht L, Brown JM. Blockade of SDF-1 after irradiation inhibits tumor recurrences of autochthonous brain tumors in rats. Neuro Oncol 2013; 16:21-8. [PMID: 24335554 PMCID: PMC3870826 DOI: 10.1093/neuonc/not149] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Tumor irradiation blocks local angiogenesis, forcing any recurrent tumor to form new vessels from circulating cells. We have previously demonstrated that the post-irradiation recurrence of human glioblastomas in the brains of nude mice can be delayed or prevented by inhibiting circulating blood vessel–forming cells by blocking the interaction of CXCR4 with its ligand stromal cell-derived factor (SDF)–1 (CXCL12). In the present study we test this strategy by directly neutralizing SDF-1 in a clinically relevant model using autochthonous brain tumors in immune competent hosts. Methods We used NOX-A12, an l-enantiomeric RNA oligonucleotide that binds and inhibits SDF-1 with high affinity. We tested the effect of this inhibitor on the response to irradiation of brain tumors in rat induced by n-ethyl-N-nitrosourea. Results Rats treated in utero with N-ethyl-N-nitrosourea began to die of brain tumors from approximately 120 days of age. We delivered a single dose of whole brain irradiation (20 Gy) on day 115 of age, began treatment with NOX-A12 immediately following irradiation, and continued with either 5 or 20 mg/kg for 4 or 8 weeks, doses and times equivalent to well-tolerated human exposures. We found a marked prolongation of rat life span that was dependent on both drug dose and duration of treatment. In addition we treated tumors only when they were visible by MRI and demonstrated complete regression of the tumors that was not achieved by irradiation alone or with the addition of temozolomide. Conclusions Inhibition of SDF-1 following tumor irradiation is a powerful way of improving tumor response of glioblastoma multiforme.
Collapse
Affiliation(s)
- Shie-Chau Liu
- Corresponding author: J. Martin Brown, PhD, Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, A246, 1050A Arastradero Rd, Palo Alto, CA 94304-1334.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Guo C, Buranych A, Sarkar D, Fisher PB, Wang XY. The role of tumor-associated macrophages in tumor vascularization. Vasc Cell 2013; 5:20. [PMID: 24314323 PMCID: PMC3913793 DOI: 10.1186/2045-824x-5-20] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/25/2013] [Indexed: 12/15/2022] Open
Abstract
Tumor vascularization is a highly complex process that involves the interaction between tumors and their surrounding stroma, as well as many distinct angiogenesis-regulating factors. Tumor associated macrophages (TAMs) represent one of the most abundant cell components in the tumor environment and key contributors to cancer-related inflammation. A large body of evidence supports the notion that TAMs play a critical role in promoting the formation of an abnormal tumor vascular network and subsequent tumor progression and invasion. Clinical and experimental evidence has shown that high levels of infiltrating TAMs are associated with poor patient prognosis and tumor resistance to therapies. In addition to stimulating angiogenesis during tumor growth, TAMs enhance tumor revascularization in response to cytotoxic therapy (e.g., radiotherapy), thereby causing cancer relapse. In this review, we highlight the emerging data related to the phenotype and polarization of TAMs in the tumor microenvironment, as well as the underlying mechanisms of macrophage function in the regulation of the angiogenic switch and tumor vascularization. Additionally, we discuss the potential of targeting pro-angiogenic TAMs, or reprograming TAMs toward a tumoricidal and angiostatic phenotype, to promote normalization of the tumor vasculature to enhance the outcome of cancer therapies.
Collapse
Affiliation(s)
- Chunqing Guo
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, PO BOX 980033, Richmond VA23298, USA
| | - Annicole Buranych
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, PO BOX 980033, Richmond VA23298, USA
| | - Devanand Sarkar
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, PO BOX 980033, Richmond VA23298, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond VA23298, USA
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond VA23298, USA
| | - Paul B Fisher
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, PO BOX 980033, Richmond VA23298, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond VA23298, USA
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond VA23298, USA
| | - Xiang-Yang Wang
- Department of Human & Molecular Genetics, Virginia Commonwealth University School of Medicine, PO BOX 980033, Richmond VA23298, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond VA23298, USA
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond VA23298, USA
| |
Collapse
|
185
|
Wu CY, Yang LH, Yang HY, Knoff J, Peng S, Lin YH, Wang C, Alvarez RD, Pai SI, Roden RBS, Hung CF, Wu TC. Enhanced cancer radiotherapy through immunosuppressive stromal cell destruction in tumors. Clin Cancer Res 2013; 20:644-57. [PMID: 24300786 DOI: 10.1158/1078-0432.ccr-13-1334] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE Radiotherapy kills cancer cells by causing DNA damage, and stimulates a systemic antitumor immune response by releasing tumor antigen and endogenous adjuvant within the tumor microenvironment. However, radiotherapy also induces the recruitment of immunosuppressive myeloid cells, which can interfere with the antitumor immune responses elicited by apoptotic tumor cells. We hypothesized that local delivery of vaccine following radiotherapy will lead to the priming of antigen-specific CTL immune responses and render immunosuppressive myeloid cells susceptible to killing by the activated CTLs. EXPERIMENTAL DESIGN Using several antigenic systems, we tested whether intratumoral injection of antigenic peptide/protein in irradiated tumors would be able to prime CTLs as well as load myeloid cells with antigen, rendering them susceptible to antigen-specific CTL killing. RESULTS We show that by combining radiotherapy and targeted antigenic peptide delivery to the tumor, the adjuvant effect generated by radiotherapy itself was sufficient to elicit the priming and expansion of antigen-specific CTLs, through the type I IFN-dependent pathway, leading to synergistic therapeutic antitumor effects compared with either treatment alone. In addition, using two different types of transgenic mice, we demonstrated that CTL-mediated killing of stromal cells in tumors by our approach is important for tumor control. Finally, we confirmed the efficacy of this approach in our preclinical model using two clinically tested therapeutic human papilloma virus (HPV) vaccines. CONCLUSIONS These data serve as an important foundation for the future clinical translation of radiotherapy combined with a clinically tested therapeutic HPV vaccine for the control of HPV-associated cancers.
Collapse
Affiliation(s)
- Chao-Yi Wu
- Authors' Affiliations: Departments of Pathology, Obstetrics and Gynecology, Molecular Microbiology and Immunology, Oncology, and Otolaryngology-Head and Neck Surgery, Johns Hopkins Medical Institutions; Department of Biostatistics and Bioinformatics, Sidney Kimmel Cancer Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland; Departments of Obstetrics and Gynecology, Kunming Medical University, Yunnan, China; Nephrology, Chang-Gung Memorial Hospital, Gueishan; Department of Obstetrics and Gynecology, Tri-Service General Hospital, Taipei, Taiwan; and Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
Yoshida N, Oda M, Kuroda Y, Katayama Y, Okikawa Y, Masunari T, Fujiwara M, Nishisaka T, Sasaki N, Sadahira Y, Mihara K, Asaoku H, Matsui H, Seto M, Kimura A, Arihiro K, Sakai A. Clinical significance of sIL-2R levels in B-cell lymphomas. PLoS One 2013; 8:e78730. [PMID: 24236041 PMCID: PMC3827264 DOI: 10.1371/journal.pone.0078730] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 09/16/2013] [Indexed: 02/03/2023] Open
Abstract
Elevated soluble interleukin-2 receptor (sIL-2R) in sera is observed in patients with malignant lymphoma (ML). Therefore, sIL-2R is commonly used as a diagnostic and prognostic marker for ML, but the mechanisms responsible for the increase in sIL-2R levels in patients with B-cell lymphomas have not yet been elucidated. We first hypothesized that lymphoma cells expressing IL-2R and some proteinases such as matrix metalloproteinases (MMPs) in the tumor microenvironment can give rise to increased sIL-2R in sera. However, flow cytometric studies revealed that few lymphoma cells expressed IL-2R α chain (CD25) in diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL), and most CD25-expressing cells in the tumor were T-cells. Distinct correlations between CD25 expression on B-lymphoma cells and sIL-2R levels were not observed. We then confirmed that MMP-9 plays an important role in producing sIL-2R in functional studies. Immunohistochemical (IHC) analysis also revealed that MMP-9 is mainly derived from tumor-associated macrophages (TAMs). We therefore evaluated the number of CD68 and CD163 positive macrophages in the tumor microenvironment using IHC analysis. A positive correlation between the levels of sIL-2R in sera and the numbers of CD68 positive macrophages in the tumor microenvironment was confirmed in FL and extranodal DLBCL. These results may be useful in understanding the pathophysiology of B-cell lymphomas.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Biomarkers, Tumor/blood
- Cell Line, Tumor
- Humans
- Interleukin-2 Receptor alpha Subunit/metabolism
- Lymphoma, B-Cell/blood
- Lymphoma, B-Cell/mortality
- Lymphoma, B-Cell/pathology
- Macrophages/enzymology
- Matrix Metalloproteinase 9/metabolism
- Middle Aged
- Prognosis
- Receptors, Cell Surface/metabolism
- Receptors, Interleukin-2/blood
- Statistics, Nonparametric
- Survival Analysis
- Tumor Microenvironment
Collapse
Affiliation(s)
- Noriaki Yoshida
- Department of Hematology and Oncology, Research Institute Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
- Division of Molecular Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Cancer Genetics, Nagoya University Graduate School of Medicine at Aichi Cancer Center Research Institute, Nagoya, Japan
- * E-mail: (NY); (AS)
| | - Miyo Oda
- Department of Anatomical Pathology, Hiroshima University, Hiroshima, Japan
| | - Yoshiaki Kuroda
- Department of Hematology and Oncology, Research Institute Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yuta Katayama
- Department of Internal Medicine, Hiroshima Red Cross Hospital & Atomic-Bomb Survivors Hospital, Hiroshima, Japan
| | - Yoshiko Okikawa
- Department of Hematology, National Hospital Organization Kure Medical Center, Kure, Japan
| | - Taro Masunari
- Department of Hematology, Chugoku Central Hospital, Fukuyama, Japan
| | - Megumu Fujiwara
- Department of Pathology, Hiroshima Red Cross Hospital & Atomic-Bomb Survivors Hospital, Hiroshima, Japan
| | - Takashi Nishisaka
- Department of Laboratory Pathology, Hiroshima Prefecture Hospital, Hiroshima, Japan
| | - Naomi Sasaki
- Department of Pathology, Kure Kyosai Hospital, Kure, Japan
| | - Yoshito Sadahira
- Department of Pathology, Kawasaki Medical School, Kurashiki, Japan
| | - Keichiro Mihara
- Department of Hematology and Oncology, Research Institute Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Hideki Asaoku
- Department of Internal Medicine, Hiroshima Red Cross Hospital & Atomic-Bomb Survivors Hospital, Hiroshima, Japan
| | - Hirotaka Matsui
- Department of Molecular Oncology & Leukemia Program Project, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Masao Seto
- Division of Molecular Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Cancer Genetics, Nagoya University Graduate School of Medicine at Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Akiro Kimura
- Department of Hematology and Oncology, Research Institute Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
- Department of Laboratory medicine, Kure Kyosai Hospital, Kure, Japan
| | - Koji Arihiro
- Department of Anatomical Pathology, Hiroshima University, Hiroshima, Japan
| | - Akira Sakai
- Department of Hematology and Oncology, Research Institute Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
- Department of Radiation Life Science, Fukushima Medical University, Fukushima, Japan
- * E-mail: (NY); (AS)
| |
Collapse
|
187
|
Angiogenic capacity of M1- and M2-polarized macrophages is determined by the levels of TIMP-1 complexed with their secreted proMMP-9. Blood 2013; 122:4054-67. [PMID: 24174628 DOI: 10.1182/blood-2013-05-501494] [Citation(s) in RCA: 193] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A proangiogenic function of tissue-infiltrating monocytes/macrophages has long been attributed to their matrix metalloproteinase-9 zymogen (proMMP-9). Herein, we evaluated the capacity of human monocytes, mature M0 macrophages, and M1- and M2-polarized macrophages to induce proMMP-9-mediated angiogenesis. Only M2 macrophages induced angiogenesis at levels comparable with highly angiogenic neutrophils previously shown to release their proMMP-9 in a unique form, free of tissue inhibitor of metalloproteinases-1 (TIMP-1). Macrophage differentiation was accompanied by induction of low-angiogenic, TIMP-1-encumbered proMMP-9. However, polarization toward the M2, but not the M1 phenotype, caused a substantial downregulation of TIMP-1 expression, resulting in production of angiogenic, TIMP-deficient proMMP-9. Correspondingly, the angiogenic potency of M2 proMMP-9 was lost after its complexing with TIMP-1, whereas TIMP-1 silencing in M0/M1 macrophages rendered them both angiogenic. Similar to human cells, murine bone marrow-derived M2 macrophages also shut down their TIMP-1 expression and produced proMMP-9 unencumbered by TIMP-1. Providing proof that angiogenic capacity of murine M2 macrophages depended on their TIMP-free proMMP-9, Mmp9-null M2 macrophages were nonangiogenic, although their TIMP-1 was severely downregulated. Our study provides a unifying molecular mechanism for high angiogenic capacity of TIMP-free proMMP-9 that would be uniquely produced in a pathophysiological microenvironment by influxing neutrophils and/or M2 polarized macrophages.
Collapse
|
188
|
Braun J, Strittmatter K, Nübel T, Komljenovic D, Sator-Schmitt M, Bäuerle T, Angel P, Schorpp-Kistner M. Loss of stromal JUNB does not affect tumor growth and angiogenesis. Int J Cancer 2013; 134:1511-6. [DOI: 10.1002/ijc.28477] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 07/29/2013] [Accepted: 08/01/2013] [Indexed: 11/08/2022]
Affiliation(s)
- Jennifer Braun
- Division of Signal Transduction and Growth Control DKFZ DKFZ-ZMBH Alliance; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Karin Strittmatter
- Division of Signal Transduction and Growth Control DKFZ DKFZ-ZMBH Alliance; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Tobias Nübel
- Division of Signal Transduction and Growth Control DKFZ DKFZ-ZMBH Alliance; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Dorde Komljenovic
- Department of Medical Physics in Radiology DKFZ; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Melanie Sator-Schmitt
- Division of Signal Transduction and Growth Control DKFZ DKFZ-ZMBH Alliance; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Tobias Bäuerle
- Department of Medical Physics in Radiology DKFZ; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Peter Angel
- Division of Signal Transduction and Growth Control DKFZ DKFZ-ZMBH Alliance; German Cancer Research Center (DKFZ); Heidelberg Germany
| | - Marina Schorpp-Kistner
- Division of Signal Transduction and Growth Control DKFZ DKFZ-ZMBH Alliance; German Cancer Research Center (DKFZ); Heidelberg Germany
| |
Collapse
|
189
|
Hematopoietic stem and progenitor cell migration after hypofractionated radiation therapy in a murine model. Int J Radiat Oncol Biol Phys 2013; 87:1162-70. [PMID: 24113056 DOI: 10.1016/j.ijrobp.2013.08.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 08/26/2013] [Accepted: 08/27/2013] [Indexed: 12/16/2022]
Abstract
PURPOSE To characterize the recruitment of bone marrow (BM)-derived hematopoietic stem and progenitor cells (HSPCs) within tumor microenvironment after radiation therapy (RT) in a murine, heterotopic tumor model. METHODS AND MATERIALS Lewis lung carcinoma tumors were established in C57BL/6 mice and irradiated with 30 Gy given as 2 fractions over 2 days. Tumors were imaged with positron emission tomography/computed tomography (PET/CT) and measured daily with digital calipers. The HSPC and myelomonocytic cell content was assessed via immunofluorescent staining and flow cytometry. Functionality of tumor-associated HSPCs was verified in vitro using colony-forming cell assays and in vivo by rescuing lethally irradiated C57BL/6 recipients. RESULTS Irradiation significantly reduced tumor volumes and tumor regrowth rates compared with nonirradiated controls. The number of CD133(+) HSPCs present in irradiated tumors was higher than in nonirradiated tumors during all stages of regrowth. CD11b(+) counts were similar. PET/CT imaging and growth rate analysis based on standardized uptake value indicated that HSPC recruitment directly correlated to the extent of regrowth and intratumor cell activity after irradiation. The BM-derived tumor-associated HSPCs successfully formed hematopoietic colonies and engrafted irradiated mice. Finally, targeted treatment with a small animal radiation research platform demonstrated localized HSPC recruitment to defined tumor subsites exposed to radiation. CONCLUSIONS Hypofractionated irradiation resulted in a pronounced and targeted recruitment of BM-derived HSPCs, possibly as a mechanism to promote tumor regrowth. These data indicate for the first time that radiation therapy regulates HSPC content within regrowing tumors.
Collapse
|
190
|
Park SI, Lee C, Sadler WD, Koh AJ, Jones J, Seo JW, Soki FN, Cho SW, Daignault SD, McCauley LK. Parathyroid hormone-related protein drives a CD11b+Gr1+ cell-mediated positive feedback loop to support prostate cancer growth. Cancer Res 2013; 73:6574-83. [PMID: 24072746 DOI: 10.1158/0008-5472.can-12-4692] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the tumor microenvironment, CD11b(+)Gr1(+) bone marrow-derived cells are a predominant source of protumorigenic factors such as matrix metalloproteinases (MMP), but how distal tumors regulate these cells in the bone marrow is unclear. Here we addressed the hypothesis that the parathyroid hormone-related protein (PTHrP) potentiates CD11b(+)Gr1(+) cells in the bone marrow of prostate tumor hosts. In two xenograft models of prostate cancer, levels of tumor-derived PTHrP correlated with CD11b(+)Gr1(+) cell recruitment and microvessel density in the tumor tissue, with evidence for mediation of CD11b(+)Gr1(+) cell-derived MMP-9 but not tumor-derived VEGF-A. CD11b(+)Gr1(+) cells isolated from mice with PTHrP-overexpressing tumors exhibited relatively increased proangiogenic potential, suggesting that prostate tumor-derived PTHrP potentiates this activity of CD11b(+)Gr1(+) cells. Administration of neutralizing PTHrP monoclonal antibody reduced CD11b(+)Gr1(+) cells and MMP-9 in the tumors. Mechanistic investigations in vivo revealed that PTHrP elevated Y418 phosphorylation levels in Src family kinases in CD11b(+)Gr1(+) cells via osteoblast-derived interleukin-6 and VEGF-A, thereby upregulating MMP-9. Taken together, our results showed that prostate cancer-derived PTHrP acts in the bone marrow to potentiate CD11b(+)Gr1(+) cells, which are recruited to tumor tissue where they contribute to tumor angiogenesis and growth.
Collapse
Affiliation(s)
- Serk In Park
- Authors' Affiliations: Departments of Medicine and Cancer Biology; Center for Bone Biology; Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry; Comprehensive Cancer Center Biostatistics Core; and Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Lee DY, Chunta JL, Park SS, Huang J, Martinez AA, Grills IS, Krueger SA, Wilson GD, Marples B. Pulsed versus conventional radiation therapy in combination with temozolomide in a murine orthotopic model of glioblastoma multiforme. Int J Radiat Oncol Biol Phys 2013; 86:978-85. [PMID: 23845846 DOI: 10.1016/j.ijrobp.2013.04.034] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 03/29/2013] [Accepted: 04/16/2013] [Indexed: 11/28/2022]
Abstract
PURPOSE To evaluate the efficacy of pulsed low-dose radiation therapy (PLRT) combined with temozolomide (TMZ) as a novel treatment approach for radioresistant glioblastoma multiforme (GBM) in a murine model. METHODS AND MATERIALS Orthotopic U87MG hGBM tumors were established in Nu-Foxn1(nu) mice and imaged weekly using a small-animal micropositron emission tomography (PET)/computed tomography (CT) system. Tumor volume was determined from contrast-enhanced microCT images and tumor metabolic activity (SUVmax) from the F18-FDG microPET scan. Tumors were irradiated 7 to 10 days after implantation with a total dose of 14 Gy in 7 consecutive days. The daily treatment was given as a single continuous 2-Gy dose (RT) or 10 pulses of 0.2 Gy using an interpulse interval of 3 minutes (PLRT). TMZ (10 mg/kg) was given daily by oral gavage 1 hour before RT. Tumor vascularity and normal brain damage were assessed by immunohistochemistry. RESULTS Radiation therapy with TMZ resulted in a significant 3- to 4-week tumor growth delay compared with controls, with PLRT+TMZ the most effective. PLRT+TMZ resulted in a larger decline in SUVmax than RT+TMZ. Significant differences in survival were evident. Treatment after PLRT+TMZ was associated with increased vascularization compared with RT+TMZ. Significantly fewer degenerating neurons were seen in normal brain after PLRT+TMZ compared with RT+TMZ. CONCLUSIONS PLRT+TMZ produced superior tumor growth delay and less normal brain damage when compared with RT+TMZ. The differential effect of PLRT on vascularization may confirm new treatment avenues for GBM.
Collapse
Affiliation(s)
- David Y Lee
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
192
|
Strategies for optimizing the response of cancer and normal tissues to radiation. Nat Rev Drug Discov 2013; 12:526-42. [PMID: 23812271 DOI: 10.1038/nrd4003] [Citation(s) in RCA: 327] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Approximately 50% of all patients with cancer receive radiation therapy at some point during the course of their treatment, and the majority of these patients are treated with curative intent. Despite recent advances in the planning of radiation treatment and the delivery of image-guided radiation therapy, acute toxicity and potential long-term side effects often limit the ability to deliver a sufficient dose of radiation to control tumours locally. In the past two decades, a better understanding of the hallmarks of cancer and the discovery of specific signalling pathways by which cells respond to radiation have provided new opportunities to design molecularly targeted therapies to increase the therapeutic window of radiation therapy. Here, we review efforts to develop approaches that could improve outcomes with radiation therapy by increasing the probability of tumour cure or by decreasing normal tissue toxicity.
Collapse
|
193
|
Abstract
The vascular endothelium is a dynamic cellular "organ" that controls passage of nutrients into tissues, maintains the flow of blood, and regulates the trafficking of leukocytes. In tumors, factors such as hypoxia and chronic growth factor stimulation result in endothelial dysfunction. For example, tumor blood vessels have irregular diameters; they are fragile, leaky, and blood flow is abnormal. There is now good evidence that these abnormalities in the tumor endothelium contribute to tumor growth and metastasis. Thus, determining the biological basis underlying these abnormalities is critical for understanding the pathophysiology of tumor progression and facilitating the design and delivery of effective antiangiogenic therapies.
Collapse
Affiliation(s)
- Andrew C Dudley
- Department of Cellular and Molecular Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA; Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina 27599, USA; and McAllister Heart Institute, Chapel Hill, North Carolina 27599, USA.
| |
Collapse
|
194
|
Russell JS, Brown JM. The irradiated tumor microenvironment: role of tumor-associated macrophages in vascular recovery. Front Physiol 2013; 4:157. [PMID: 23882218 PMCID: PMC3713331 DOI: 10.3389/fphys.2013.00157] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 06/11/2013] [Indexed: 12/12/2022] Open
Abstract
Radiotherapy is an important modality used in the treatment of more than 50% of cancer patients in the US. However, despite sophisticated techniques for radiation delivery as well as the combination of radiation with chemotherapy, tumors can recur. Thus, any method of improving the local control of the primary tumor by radiotherapy would produce a major improvement in the curability of cancer patients. One of the challenges in the field is to understand how the tumor vasculature can regrow after radiation in order to support tumor recurrence, as it is unlikely that any of the endothelial cells within the tumor could survive the doses given in a typical radiotherapy regimen. There is now considerable evidence from both preclinical and clinical studies that the tumor vasculature can be restored following radiotherapy from an influx of circulating cells consisting primarily of bone marrow derived monocytes and macrophages. The radiation-induced influx of bone marrow derived cells (BMDCs) into tumors can be prevented through the blockade of various cytokine pathways and such strategies can inhibit tumor recurrence. However, the post-radiation interactions between surviving tumor cells, recruited immune cells, and the remaining stroma remain poorly defined. While prior studies have described the monocyte/macrophage inflammatory response within normal tissues and in the tumor microenvironment, less is known about this response with respect to a tumor after radiation therapy. The goal of this review is to summarize existing research studies to provide an understanding of how the myelomonocytic lineage may influence vascular recovery within the irradiated tumor microenvironment.
Collapse
Affiliation(s)
- Jeffery S Russell
- Department of Medical Oncology, Stanford University School of Medicine Stanford, CA, USA
| | | |
Collapse
|
195
|
Oncology scan--How radiation effects on cells in the stromal microenvironment influence tumor development, proliferation, and recovery. Int J Radiat Oncol Biol Phys 2013; 86:593-5. [PMID: 23773382 DOI: 10.1016/j.ijrobp.2013.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
196
|
Sessa C, Lorusso P, Tolcher A, Farace F, Lassau N, Delmonte A, Braghetti A, Bahleda R, Cohen P, Hospitel M, Veyrat-Follet C, Soria JC. Phase I Safety, Pharmacokinetic and Pharmacodynamic Evaluation of the Vascular Disrupting Agent Ombrabulin (AVE8062) in Patients with Advanced Solid Tumors. Clin Cancer Res 2013; 19:4832-42. [DOI: 10.1158/1078-0432.ccr-13-0427] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
197
|
Matrix metalloproteinases: inflammatory regulators of cell behaviors in vascular formation and remodeling. Mediators Inflamm 2013; 2013:928315. [PMID: 23840100 PMCID: PMC3694547 DOI: 10.1155/2013/928315] [Citation(s) in RCA: 301] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 05/15/2013] [Indexed: 12/21/2022] Open
Abstract
Abnormal angiogenesis and vascular remodeling contribute to pathogenesis of a number of disorders such as tumor, arthritis, atherosclerosis, restenosis, hypertension, and neurodegeneration. During angiogenesis and vascular remodeling, behaviors of stem/progenitor cells, endothelial cells (ECs), and vascular smooth muscle cells (VSMCs) and its interaction with extracellular matrix (ECM) play a critical role in the processes. Matrix metalloproteinases (MMPs), well-known inflammatory mediators are a family of zinc-dependent proteolytic enzymes that degrade various components of ECM and non-ECM molecules mediating tissue remodeling in both physiological and pathological processes. MMPs including MMP-1, MMP-2, MMP-3, MMP-7, MMP-8, MMP-9, MMP-12, and MT1-MMP, are stimulated and activated by various stimuli in vascular tissues. Once activated, MMPs degrade ECM proteins or other related signal molecules to promote recruitment of stem/progenitor cells and facilitate migration and invasion of ECs and VSMCs. Moreover, vascular cell proliferation and apoptosis can also be regulated by MMPs via proteolytically cleaving and modulating bioactive molecules and relevant signaling pathways. Regarding the importance of vascular cells in abnormal angiogenesis and vascular remodeling, regulation of vascular cell behaviors through modulating expression and activation of MMPs shows therapeutic potential.
Collapse
|
198
|
Notch1 regulates angio-supportive bone marrow-derived cells in mice: relevance to chemoresistance. Blood 2013; 122:143-53. [PMID: 23690447 DOI: 10.1182/blood-2012-11-459347] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Host responses to chemotherapy can induce resistance mechanisms that facilitate tumor regrowth. To determine the contribution of bone marrow-derived cells (BMDCs), we exposed tumor-bearing mice to chemotherapeutic agents and evaluated the influx and contribution of a genetically traceable subpopulation of BMDCs (vascular endothelial-cadherin-Cre-enhanced yellow fluorescent protein [VE-Cad-Cre-EYFP]). Treatment of tumor-bearing mice with different chemotherapeutics resulted in a three- to 10-fold increase in the influx of VE-Cad-Cre-EYFP. This enhanced influx was accompanied by a significant increase in angiogenesis. Expression profile analysis revealed a progressive change in the EYFP population with loss of endothelial markers and an increase in mononuclear markers. In the tumor, 2 specific populations of VE-Cad-Cre-EYFP BMDCs were identified: Gr1⁺/CD11b⁺ and Tie2high/platelet endothelial cell adhesion moleculelow cells, both located in perivascular areas. A common signature of the EYFP population that exits the bone marrow is an increase in Notch. Inducible inactivation of Notch in the EYFP⁺ BMDCs impaired homing of these BMDCs to the tumor. Importantly, Notch deletion reduced therapy-enhanced angiogenesis, and was associated with an increased antitumor effect of the chemotherapy. These findings revealed the functional significance of a specific population of supportive BMDCs in response to chemotherapeutics and uncovered a new potential strategy to enhance anticancer therapy.
Collapse
|
199
|
Lin CH, Lee HT, Lee SD, Lee W, Cho CWC, Lin SZ, Wang HJ, Okano H, Su CY, Yu YL, Hsu CY, Shyu WC. Role of HIF-1α-activated Epac1 on HSC-mediated neuroplasticity in stroke model. Neurobiol Dis 2013; 58:76-91. [PMID: 23702312 DOI: 10.1016/j.nbd.2013.05.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/02/2013] [Accepted: 05/10/2013] [Indexed: 12/27/2022] Open
Abstract
Exchange protein activated by cAMP-1 (Epac1) plays an important role in cell proliferation, cell survival and neuronal signaling, and activation of Epac1 in endothelial progenitor cells increases their homing to ischemic muscles and promotes neovascularization in a model of hind limb ischemia. Moreover, upregulation of Epac1 occurs during organ development and in diseases such as myocardial hypertrophy, diabetes, and Alzheimer's disease. We report here that hypoxia upregulated Epac1 through HIF-1α induction in the CD34-immunosorted human umbilical cord blood hematopoietic stem cells (hUCB(34)). Importantly, implantation of hUCB(34) subjected to hypoxia-preconditioning (HP-hUCB(34)) improved stroke outcome, more than did implantation of untreated hUCB(34), in rodents subjected to cerebral ischemia, and this required Epac1-to-matrix metalloprotease (MMP) signaling. This improved therapeutic efficacy correlated with better engraftment and differentiation of these cells in the ischemic host brain. In addition, more than did implantation of untreated HP-hUCB(34), implantation of HP-hUCB(34) improved cerebral blood flow into the ischemic brain via induction of angiogenesis, facilitated proliferation/recruitment of endogenous neural progenitor cells in the ischemic brain, and promoted neurite outgrowth following cerebral ischemia. Consistent with our proposed role of Epac1-to-MMP signaling in hypoxia-preconditioning, the above mentioned effects of implanting HP-hUCB(34) could be abolished by pharmacological inhibition and genetic disruption/deletion of Epac1 or MMPs. We have discovered a HIF-1α-to-Epac1-to-MMP signaling pathway that is required for the improved therapeutic efficacy resulting from hypoxia preconditioning of hUCB(34) in vitro prior to their implantation into the host brain in vivo.
Collapse
Affiliation(s)
- Chen-Huan Lin
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Yu F, Jia X, Du F, Wang J, Wang Y, Ai W, Fan D. miR-155-deficient bone marrow promotes tumor metastasis. Mol Cancer Res 2013; 11:923-36. [PMID: 23666369 DOI: 10.1158/1541-7786.mcr-12-0686] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Infiltration of immune cells in primary tumors and metastatic sites is known to influence tumor progression and metastasis. Macrophages represent the most abundant immune cells in the tumor microenvironment, and evidence has shown that macrophages promote seeding, extravasation, and persistent growth of tumor cells at metastatic sites. miR-155 plays an essential role in immune cell development/function, and its aberrant expression is associated with lymphomas and several solid tumor types. However, it is unknown how miR-155 expression in immune cells affects solid tumor growth and metastasis. To this end, bone marrow transplantation was performed using miR-155-deficient mice as bone marrow donors and wild-type (WT) mice as recipients, and the chimeric mice were inoculated with tumor cells. We demonstrate that bone marrow lacking miR-155 significantly enhanced lung metastasis without a substantial effect on primary tumor growth. Relative to mice with WT bone marrow, miR-155-deficient bone marrow accumulated more macrophages in the spleen and lungs. Further analysis revealed that miR-155-deficient macrophages in metastatic sites exhibited a tumor-promoting M2 phenotype. In vitro study suggested that miR-155-null macrophages were prone to M2 polarization upon incubation with tumor cell-conditioned medium, due to elevated expression of C/EBPβ, an identified miR-155 target. These data, for the first time, demonstrate that miR-155 in host immune cells plays a vital role in modulating solid tumor metastasis by affecting the recruitment and polarization of bone marrow-derived macrophages. IMPLICATIONS Targeted inhibition of miR-155 delays tumor development but inhibition in host immune cells may encourage metastasis.
Collapse
Affiliation(s)
- Fang Yu
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, 6439 Garners Ferry Road, Columbia, SC 29208, USA.
| | | | | | | | | | | | | |
Collapse
|