201
|
Fujimura T, Aiba S. Significance of Immunosuppressive Cells as a Target for Immunotherapies in Melanoma and Non-Melanoma Skin Cancers. Biomolecules 2020; 10:E1087. [PMID: 32707850 PMCID: PMC7464513 DOI: 10.3390/biom10081087] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
Tumor-associated macrophages (TAMs) have been detected in most skin cancers. TAMs produce various chemokines and angiogenic factors that promote tumor development, along with other immunosuppressive cells such as myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs) and tumor-associated neutrophils. TAMs generated from monocytes develop into functional, fully activated macrophages, and TAMs obtain various immunosuppressive functions to maintain the tumor microenvironment. Since TAMs express PD1 to maintain the immunosuppressive M2 phenotype by PD1/PD-L1 signaling from tumor cells, and the blockade of PD1/PD-L1 signaling by anti-PD1 antibodies (Abs) activate and re-polarize TAMs into immunoreactive M1 phenotypes, TAMs represent a potential target for anti-PD1 Abs. The main population of TAMs comprises CD163+ M2 macrophages, and CD163+ TAMs release soluble (s)CD163 and several proinflammatory chemokines (CXCL5, CXCL10, CCL19, etc.) as a result of TAM activation to induce an immunosuppressive tumor microenvironment together with other immunosuppressive cells. Since direct blockade of PD1/PD-L1 signaling between tumor cells and tumor-infiltrating T cells (both effector T cells and Tregs) is mandatory for inducing an anti-immune response by anti-PD1 Abs, anti-PD1 Abs need to reach the tumor microenvironment to induce anti-immune responses in the tumor-bearing host. Taken together, TAM-related factors could offer a biomarker for anti-PD1 Ab-based immunotherapy. Understanding the crosstalk between TAMs and immunosuppressive cells is important for optimizing PD1 Ab-based immunotherapy.
Collapse
Affiliation(s)
- Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan;
| | | |
Collapse
|
202
|
Capucetti A, Albano F, Bonecchi R. Multiple Roles for Chemokines in Neutrophil Biology. Front Immunol 2020; 11:1259. [PMID: 32733442 PMCID: PMC7363767 DOI: 10.3389/fimmu.2020.01259] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/18/2020] [Indexed: 11/23/2022] Open
Abstract
Chemokines are recognized as the most critical mediators for selective neutrophil recruitment during inflammatory conditions. Furthermore, they are considered fundamental regulators of neutrophil mobilization from the bone marrow (BM) to the bloodstream and for their homing back at the end of their life for apoptosis and clearance. However, chemokines are also important mediators of neutrophil effector functions including oxidative burst, degranulation, neutrophil extracellular trap (NET)osis, and production of inflammatory mediators. Neutrophils have been historically considered as a homogeneous population. In recent years, several maturation stages and subsets with different phenotypic profiles and effector functions were described both in physiological and pathological conditions such as infections, autoimmunity, and cancer. The aim of this review is to give an overview of the current evidence regarding the role of chemokines and chemokine receptors in neutrophil biology, including their possible role in neutrophil maturation, differentiation, and in defining emerging neutrophil subsets.
Collapse
Affiliation(s)
- Arianna Capucetti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Francesca Albano
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy.,Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Raffaella Bonecchi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy.,Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| |
Collapse
|
203
|
Rébé C, Ghiringhelli F. Interleukin-1β and Cancer. Cancers (Basel) 2020; 12:E1791. [PMID: 32635472 PMCID: PMC7408158 DOI: 10.3390/cancers12071791] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Within a tumor, IL-1β is produced and secreted by various cell types, such as immune cells, fibroblasts, or cancer cells. The IL1B gene is induced after "priming" of the cells and a second signal is required to allow IL-1β maturation by inflammasome-activated caspase-1. IL-1β is then released and leads to transcription of target genes through its ligation with IL-1R1 on target cells. IL-1β expression and maturation are guided by gene polymorphisms and by the cellular context. In cancer, IL-1β has pleiotropic effects on immune cells, angiogenesis, cancer cell proliferation, migration, and metastasis. Moreover, anti-cancer treatments are able to promote IL-1β production by cancer or immune cells, with opposite effects on cancer progression. This raises the question of whether or not to use IL-1β inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Cédric Rébé
- Platform of Transfer in Cancer Biology, Centre Georges François Leclerc, INSERM LNC UMR1231, University of Bourgogne Franche-Comté, F-21000 Dijon, France
| | - François Ghiringhelli
- Platform of Transfer in Cancer Biology, Centre Georges François Leclerc, INSERM LNC UMR1231, University of Bourgogne Franche-Comté, F-21000 Dijon, France
| |
Collapse
|
204
|
Chillà A, Margheri F, Biagioni A, Del Rosso T, Fibbi G, Del Rosso M, Laurenzana A. Cell-Mediated Release of Nanoparticles as a Preferential Option for Future Treatment of Melanoma. Cancers (Basel) 2020; 12:cancers12071771. [PMID: 32630815 PMCID: PMC7408438 DOI: 10.3390/cancers12071771] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 01/15/2023] Open
Abstract
Targeted and immune therapies have unquestionably improved the prognosis of melanoma patients. However the treatment of this neoplasm still requires approaches with a higher therapeutic index, in order to reduce shortcomings related to toxic effects and aspecific targeting. This means developing therapeutic tools derived with high affinity molecules for tumor components differentially expressed in melanoma cells with respect to their normal counterpart. Nanomedicine has sought to address this problem owing to the high modulability of nanoparticles. This approach exploits not only the enhanced permeability and retention effect typical of the tumor microenvironment (passive targeting), but also the use of specific "molecular antennas" that recognize some tumor-overexpressed molecules (active targeting). This line of research has given rise to the so-called "smart nanoparticles," some of which have already passed the preclinical phase and are under clinical trials in melanoma patients. To further improve nanoparticles partition within tumors, for some years now a line of thought is exploiting the molecular systems that regulate the innate tumor-homing activity of platelets, granulocytes, monocytes/macrophages, stem cells, endothelial-colony-forming cells, and red blood cells loaded with nanoparticles. This new vision springs from the results obtained with some of these cells in regenerative medicine, an approach called "cell therapy." This review takes into consideration the advantages of cell therapy as the only one capable of overcoming the limits of targeting imposed by the increased interstitial pressure of tumors.
Collapse
Affiliation(s)
- Anastasia Chillà
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
| | - Tommaso Del Rosso
- Department of Physics, Pontifical Catholic University of Rio de Janeiro, 22451-900 Rio de Janeiro-RJ, Brazil;
| | - Gabriella Fibbi
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
- Correspondence: (M.D.R.); (A.L.)
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences School of Health Sciences, University of Florence-Viale G.B. Morgagni, 50–50134 Florence, Italy; (A.C.); (F.M.); (A.B.); (G.F.)
- Correspondence: (M.D.R.); (A.L.)
| |
Collapse
|
205
|
Zhang WH, Wang WQ, Gao HL, Xu SS, Li S, Li TJ, Han X, Xu HX, Li H, Jiang W, Ye LY, Lin X, Wu CT, Yu XJ, Liu L. Tumor-Infiltrating Neutrophils Predict Poor Survival of Non-Functional Pancreatic Neuroendocrine Tumor. J Clin Endocrinol Metab 2020; 105:dgaa196. [PMID: 32285127 DOI: 10.1210/clinem/dgaa196] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/11/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVE This study retrospectively characterized the immune infiltrating profile in nonfunctional pancreatic neuroendocrine tumors (NF-PanNETs). METHODS Tumor tissues from the 109-patient Fudan cohort and a 73-patient external validation set were evaluated by immunohistochemistry for 9 immune cell types: tumor-infiltrating neutrophils (TINs), tumor-associated macrophages (TAMs), CD11c+ dendritic cells, anti-NCR1+ natural killer (NK) cells, CD4+ and CD8+ T cells, CD45RO+ memory T cells, FOXP3+ regulatory T cells (Tregs), and CD20+ B cells. RESULTS TINs were primarily distributed in the intratumoral area, dendritic cells and NK cells were scattered evenly in intratumoral and stromal areas, and Tregs were rarely detected. The remaining 5 cell types were primarily present in peritumoral stroma. Total TINs (P < .001) and TAMs (P = .002) increased as NF-PanNET grade rose. Kaplan-Meier analyses showed that high intratumoral TINs, total TAMs, and stromal CD4+ T-cell infiltration correlated with shorter recurrence-free survival (RFS, P = .010, P = .027, and P = .035, respectively) and overall survival (OS, P = .017, P = .029, and P = .045, respectively). Additionally, high intratumoral CD8+ T cell infiltration correlated with prolonged RFS (P = .039). Multivariate Cox regression demonstrated that intratumoral TINs, World Health Organization (WHO) classification, and eighth edition of the American Joint Committee on Cancer tumor-node-metastasis staging system (AJCC8th TNM) were independent factors for RFS (P = .043, P = .023, and P = .029, respectively), whereas intratumoral TINs and WHO classification were independent factors for OS (P = .010 and P = .007, respectively). Furthermore, the combination of TINs, WHO classification, and AJCC8th TNM remarkably improved prognostic accuracy for RFS. These results have been verified in the external validation set. CONCLUSION Intratumoral TINs are an independent and unfavorable predictor of postoperative NF-PanNETs. A combination of TINs, WHO classification, and AJCC8th TNM could improve prognostic accuracy for RFS.
Collapse
Affiliation(s)
- Wu-Hu Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wen-Quan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - He-Li Gao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shuai-Shuai Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shuo Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Tian-Jiao Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xuan Han
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Hua-Xiang Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Hao Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wang Jiang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Long-Yun Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xuan Lin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Chun-Tao Wu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xian-Jun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Liang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| |
Collapse
|
206
|
Patel A, Ravaud A, Motzer RJ, Pantuck AJ, Staehler M, Escudier B, Martini JF, Lechuga M, Lin X, George DJ. Neutrophil-to-Lymphocyte Ratio as a Prognostic Factor of Disease-free Survival in Postnephrectomy High-risk Locoregional Renal Cell Carcinoma: Analysis of the S-TRAC Trial. Clin Cancer Res 2020; 26:4863-4868. [PMID: 32546645 DOI: 10.1158/1078-0432.ccr-20-0704] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/28/2020] [Accepted: 06/09/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE In the S-TRAC trial, adjuvant sunitinib improved disease-free survival (DFS) compared with placebo in patients with locoregional renal cell carcinoma (RCC) at high risk of recurrence. This post hoc exploratory analysis investigated the neutrophil-to-lymphocyte ratio (NLR) for predictive and prognostic significance in the RCC adjuvant setting. EXPERIMENTAL DESIGN Kaplan-Meier estimates and Cox proportional analyses were performed on baseline NLR and change from baseline at week 4 to assess their association with DFS. Univariate P values were two-sided and based on an unstratified log-rank test. RESULTS 609 of 615 patients had baseline NLR values; 574 patients had baseline and week 4 values. Sunitinib-treated patients with baseline NLR <3 had longer DFS versus placebo (7.1 vs. 4.7; HR, 0.71; P = 0.02). For baseline NLR ≥3, DFS was similar regardless of treatment (sunitinib 6.8 vs. placebo not reached; HR, 1.03; P = 0.91). A ≥25% NLR decrease at week 4 was associated with longer DFS versus no change (6.8 vs. 5.3 years; HR, 0.71; P = 0.01). A greater proportion of sunitinib-treated patients had ≥25% NLR decrease at week 4 (71.2%) versus placebo (17.4%). Patients with ≥25% NLR decrease at week 4 received a higher median cumulative sunitinib dose (10,137.5 mg) versus no change (8,168.8 mg) or ≥25% increase (6,712.5 mg). CONCLUSIONS In the postnephrectomy high-risk RCC patient cohort, low baseline NLR may help identify those most suitable for adjuvant sunitinib. A ≥25% NLR decrease at week 4 may be an early indicator of those most likely to tolerate treatment and derive DFS benefit.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xun Lin
- Pfizer Inc, La Jolla, California
| | | |
Collapse
|
207
|
Strakhova R, Cadassou O, Cros-Perrial E, Jordheim LP. Regulation of tumor infiltrated innate immune cells by adenosine. Purinergic Signal 2020; 16:289-295. [PMID: 32529478 DOI: 10.1007/s11302-020-09701-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/28/2020] [Indexed: 01/04/2023] Open
Abstract
Cancer has the ability to escape the immune system using different molecular actors. Adenosine is known to be involved in mechanisms which control inflammatory reactions and prevent excessive immune response. This purine nucleoside can be translocated from the cell or produced in the extracellular space by 5'-ectonucleotidases. Once bound to its receptors on the surface of immune effector cells, adenosine activates various molecular pathways, which lead to functional inhibition of the cell or its death. Some tumors are infiltrated by the different cells of immune system but are able to use adenosine as an immunosuppressive molecule and thus inhibit immune anticancer response. This mechanism is well described on adaptive cells, but much less on innate cells. This review outlines major effects of adenosine on innate immune cells, its consequences on cancer progression, and possible ways to block the adenosine-dependent immunosuppressive effect.
Collapse
Affiliation(s)
- Regina Strakhova
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France
| | - Octavia Cadassou
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France
| | - Emeline Cros-Perrial
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France
| | - Lars Petter Jordheim
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, 69008, Lyon, France.
| |
Collapse
|
208
|
Martins-Cardoso K, Almeida VH, Bagri KM, Rossi MID, Mermelstein CS, König S, Monteiro RQ. Neutrophil Extracellular Traps (NETs) Promote Pro-Metastatic Phenotype in Human Breast Cancer Cells through Epithelial-Mesenchymal Transition. Cancers (Basel) 2020; 12:E1542. [PMID: 32545405 PMCID: PMC7352979 DOI: 10.3390/cancers12061542] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/24/2022] Open
Abstract
Neutrophil extracellular traps (NETs) have been associated with several steps of tumor progression, including primary growth and metastasis. One of the key features for the acquisition of the metastatic ability is the epithelial-mesenchymal transition (EMT), a complex cellular program. In this study, we evaluated the ability of isolated NETs in modulating the pro-metastatic phenotype of human breast cancer cells. Tumor cells were treated with isolated NETs and then samples were generated for cell migration, quantitative RT-PCR, western blotting, immunofluorescence, and flow cytometry assays. RNA-seq data from The Cancer Genome Atlas (TCGA) database were assessed. NETs changed the typical epithelial morphology of MCF7 cells into a mesenchymal phenotype, a process that was accompanied by enhanced migratory properties. Additional EMT traits were observed: increased expression of N-cadherin and fibronectin, while the E-cadherin expression was repressed. Notably, NETs positively regulated the gene expression of several factors linked to the pro-inflammatory and pro-metastatic properties. Analyses of TCGA data showed that samples from breast cancer patients exhibit a significant correlation between pro-tumoral and neutrophil signature gene expression, including several EMT and pro-metastatic factors. Therefore, NETs drive pro-metastatic phenotype in human breast cancer cells through the activation of the EMT program.
Collapse
Affiliation(s)
- Karina Martins-Cardoso
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941 590, Brazil; (K.M.-C.); (V.H.A.)
| | - Vitor H. Almeida
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941 590, Brazil; (K.M.-C.); (V.H.A.)
| | - Kayo M. Bagri
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941 590, Brazil; (K.M.B.); (M.I.D.R.); (C.S.M.); (S.K.)
| | - Maria Isabel Doria Rossi
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941 590, Brazil; (K.M.B.); (M.I.D.R.); (C.S.M.); (S.K.)
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro 21941 913, Brazil
| | - Claudia S. Mermelstein
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941 590, Brazil; (K.M.B.); (M.I.D.R.); (C.S.M.); (S.K.)
| | - Sandra König
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941 590, Brazil; (K.M.B.); (M.I.D.R.); (C.S.M.); (S.K.)
| | - Robson Q. Monteiro
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941 590, Brazil; (K.M.-C.); (V.H.A.)
| |
Collapse
|
209
|
Costanzo-Garvey DL, Keeley T, Case AJ, Watson GF, Alsamraae M, Yu Y, Su K, Heim CE, Kielian T, Morrissey C, Frieling JS, Cook LM. Neutrophils are mediators of metastatic prostate cancer progression in bone. Cancer Immunol Immunother 2020; 69:1113-1130. [PMID: 32114681 PMCID: PMC7230043 DOI: 10.1007/s00262-020-02527-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 02/17/2020] [Indexed: 12/13/2022]
Abstract
Bone metastatic prostate cancer (BM-PCa) significantly reduces overall patient survival and is currently incurable. Current standard immunotherapy showed promising results for PCa patients with metastatic, but less advanced, disease (i.e., fewer than 20 bone lesions) suggesting that PCa growth in bone contributes to response to immunotherapy. We found that: (1) PCa stimulates recruitment of neutrophils, the most abundant immune cell in bone, and (2) that neutrophils heavily infiltrate regions of prostate tumor in bone of BM-PCa patients. Based on these findings, we examined the impact of direct neutrophil-prostate cancer interactions on prostate cancer growth. Bone marrow neutrophils directly induced apoptosis of PCa in vitro and in vivo, such that neutrophil depletion in bone metastasis models enhanced BM-PCa growth. Neutrophil-mediated PCa killing was found to be mediated by suppression of STAT5, a transcription factor shown to promote PCa progression. However, as the tumor progressed in bone over time, neutrophils from late-stage bone tumors failed to elicit cytotoxic effector responses to PCa. These findings are the first to demonstrate that bone-resident neutrophils inhibit PCa and that BM-PCa are able to progress via evasion of neutrophil-mediated killing. Enhancing neutrophil cytotoxicity in bone may present a novel therapeutic option for bone metastatic prostate cancer.
Collapse
Affiliation(s)
- Diane L Costanzo-Garvey
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Med Center, Omaha, NE, 68192, USA
| | - Tyler Keeley
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Med Center, Omaha, NE, 68192, USA
| | - Adam J Case
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Gabrielle F Watson
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Massar Alsamraae
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Med Center, Omaha, NE, 68192, USA
| | - Yangsheng Yu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Med Center, Omaha, NE, 68192, USA
| | - Kaihong Su
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Med Center, Omaha, NE, 68192, USA
- Department of Medical Education, California University of Science and Medicine, San Bernadino, CA, USA
| | - Cortney E Heim
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Med Center, Omaha, NE, 68192, USA
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Med Center, Omaha, NE, 68192, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Jeremy S Frieling
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Leah M Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Med Center, Omaha, NE, 68192, USA.
| |
Collapse
|
210
|
Labani-Motlagh A, Ashja-Mahdavi M, Loskog A. The Tumor Microenvironment: A Milieu Hindering and Obstructing Antitumor Immune Responses. Front Immunol 2020; 11:940. [PMID: 32499786 PMCID: PMC7243284 DOI: 10.3389/fimmu.2020.00940] [Citation(s) in RCA: 480] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/22/2020] [Indexed: 12/20/2022] Open
Abstract
The success of cancer immunotherapy relies on the knowledge of the tumor microenvironment and the immune evasion mechanisms in which the tumor, stroma, and infiltrating immune cells function in a complex network. The potential barriers that profoundly challenge the overall clinical outcome of promising therapies need to be fully identified and counteracted. Although cancer immunotherapy has increasingly been applied, we are far from understanding how to utilize different strategies in the best way and how to combine therapeutic options to optimize clinical benefit. This review intends to give a contemporary and detailed overview of the different roles of immune cells, exosomes, and molecules acting in the tumor microenvironment and how they relate to immune activation and escape. Further, current and novel immunotherapeutic options will be discussed.
Collapse
Affiliation(s)
| | | | - Angelica Loskog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
211
|
Onesti CE, Josse C, Boulet D, Thiry J, Beaumecker B, Bours V, Jerusalem G. Blood eosinophilic relative count is prognostic for breast cancer and associated with the presence of tumor at diagnosis and at time of relapse. Oncoimmunology 2020; 9:1761176. [PMID: 32923121 PMCID: PMC7458605 DOI: 10.1080/2162402x.2020.1761176] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Cancer outcome is associated with circulating immune cells, including eosinophils. Here we analyze the relative eosinophil count (REC) in different breast cancer subtypes. Methods Stage I–III breast cancer patients were included in the study and classified as REC-high vs low (cutoff 1.5%) or relative lymphocyte count (RLC)-high vs low (cutoff 17.5%). The co-primary endpoints were the breast cancer-specific survival (BCSS) or the time to treatment failure (TTF) in the REC groups. Results Overall 930 patients were included in the study. We observed a benefit for REC-high vs REC-low in TTF (HR 0.610, 95% CI 0.458–0.812), and in BCSS (HR 0.632, 95% CI 0.433–0.923). Similarly, we observed a better TTF (HR 0.421, 95% CI 0.262–0.677) and BCSS (HR 0.350, 95% CI 0.200–0.614) in RLC-high vs low. A lower relapse rate was observed in the REC-high vs REC-low group (17.1% vs 24.7%, p = 0.005), not confirmed in the multivariate analysis. A lower median REC at baseline and at relapse was observed compared to REC after surgery and during cancer-free follow-up (p < .0001). Conclusions REC could be a new promising, affordable and accessible predictive and prognostic biomarker in all breast cancer subtypes.
Collapse
Affiliation(s)
- Concetta Elisa Onesti
- Medical Oncology Department, Centre Hospitalier Universitaire Sart-Tilman, Liège, Belgium.,Laboratory of Human Genetics, GIGA Institute, Liège, Belgium
| | - Claire Josse
- Medical Oncology Department, Centre Hospitalier Universitaire Sart-Tilman, Liège, Belgium.,Laboratory of Human Genetics, GIGA Institute, Liège, Belgium
| | - Delphine Boulet
- Laboratory of Human Genetics, GIGA Institute, Liège, Belgium
| | - Jérôme Thiry
- Laboratory of Human Genetics, GIGA Institute, Liège, Belgium
| | | | - Vincent Bours
- Laboratory of Human Genetics, GIGA Institute, Liège, Belgium.,Department of Human Genetics, Centre Hospitalier Universitaire Sart-Tilman, Liège, Belgium
| | - Guy Jerusalem
- Medical Oncology Department, Centre Hospitalier Universitaire Sart-Tilman, Liège, Belgium.,Faculty of Medicine, Liège University, Liège, Belgium
| |
Collapse
|
212
|
Carneiro-Goetten JOL, Rodrigues BS, Nogoceke RA, do Nascimento TG, Moreno-Amaral AN, Stuelp-Campelo PM, Elifio-Esposito S. Neutrophils activated by BJcuL, a C-type lectin isolated from Bothrops jararacussu venom, decrease the invasion potential of neuroblastoma SK-N-SH cells in vitro. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20190073. [PMID: 32425992 PMCID: PMC7216824 DOI: 10.1590/1678-9199-jvatitd-2019-0073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 04/01/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Neuroblastoma is a pediatric tumor with a mortality rate of 40% in the most aggressive cases. Tumor microenvironment components as immune cells contribute to the tumor progression; thereby, the modulation of immune cells to a pro-inflammatory and antitumoral profile could potentialize the immunotherapy, a suggested approach for high-risk patients. Preview studies showed the antitumoral potential of BJcuL, a C- type lectin isolated from Bothrops jararacussu venom. It was able to induce immunomodulatory responses, promoting the rolling and adhesion of leukocytes and the activation of neutrophils. METHODS SK-N-SH cells were incubated with conditioned media (CM) obtained during the treatment of neutrophils with BJcuL and fMLP, a bacteria-derived peptide highly effective for activating neutrophil functions. Then we evaluated the effect of the same stimulation on the co-cultivation of neutrophils and SK-N-SH cells. Tumor cells were tested for viability, migration, and invasion potential. RESULTS In the viability assay, only neutrophils treated with BJcuL (24 h) and cultivated with SK-N-SH were cytotoxic. Migration of tumor cells decreased when incubated directly (p < 0.001) or indirectly (p < 0.005) with untreated neutrophils. When invasion potential was evaluated, neutrophils incubated with BJcuL reduced the total number of colonies of SK-N-SH cells following co-cultivation for 24 h (p < 0.005). Treatment with CM resulted in decreased anchorage-free survival following 24 h of treatment (p < 0.001). CONCLUSION Data demonstrated that SK-N-SH cells maintain their migratory potential in the face of neutrophil modulation by BJcuL, but their invasive capacity was significantly reduced.
Collapse
Affiliation(s)
| | - Bruna Santos Rodrigues
- Escola de Ciências da Vida, Pontifícia Universidade Católica do
Paraná (PUCPR), Curitiba, PR, Brazil
| | - Rodrigo Amauri Nogoceke
- Escola de Ciências da Vida, Pontifícia Universidade Católica do
Paraná (PUCPR), Curitiba, PR, Brazil
| | | | - Andrea Novais Moreno-Amaral
- Programa de Pós-graduação em Ciências da Saúde, Pontifícia
Universidade Católica do Paraná (PUCPR), Curitiba, PR, Brazil
| | | | - Selene Elifio-Esposito
- Escola de Ciências da Vida, Pontifícia Universidade Católica do
Paraná (PUCPR), Curitiba, PR, Brazil
| |
Collapse
|
213
|
Ishiguro S, Upreti D, Robben N, Burghart R, Loyd M, Ogun D, Le T, Delzeit J, Nakashima A, Thakkar R, Nakashima A, Suzuki K, Comer J, Tamura M. Water extract from Euglena gracilis prevents lung carcinoma growth in mice by attenuation of the myeloid-derived cell population. Biomed Pharmacother 2020; 127:110166. [PMID: 32361165 DOI: 10.1016/j.biopha.2020.110166] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 12/22/2022] Open
Abstract
The partially purified water extract from Euglena gracilis (EWE) was evaluated for its antitumor and immunomodulatory effects in cell cultures and in a mouse orthotopic lung carcinoma allograft model. In two-dimensional cell culture, the EWE treatment inhibited cell growth of both murine Lewis lung carcinoma (LLC) and human lung carcinoma cells (A549 and H1299) in a dose- and time-dependent manner. In contrast, the growth of mouse bone marrow cells (BMCs), but not mouse splenocytes (SPLs), was stimulated by the treatment with EWE. In three-dimensional spheroid culture, spheroid growth of LLC cells was significantly attenuated by EWE treatment. In a mouse LLC orthotopic allograft model, pretreatment with EWE (150-200 mg/kg/day, via drinking water) three weeks prior to the LLC cell inoculation, but not post-treatment after LLC cell inoculation, significantly attenuated the growth of LLC tumors in immunocompetent syngeneic mouse lung. This tumor growth attenuation coincided with a significant decrease in the population of myeloid-derived cells, primarily neutrophils. Flow cytometric analysis revealed that the EWE treatment significantly attenuated growth of granulocytic myeloid-derived suppressor cells (gMDSC) in BMCs and that this decrease was due to induction of gMDSC-specific apoptosis and differentiation of monocytic MDSCs (mMDSC) to macrophages. The present study provides evidence that EWE pretreatment inhibits lung carcinoma growth mainly by stimulating host antitumor immunity through attenuation of growth of gMDSCs and decreasing the number of peripheral granulocytes. This study suggests that the partially purified extract derived from Euglena gracilis contains significant bioactive materials that prevent lung carcinoma growth.
Collapse
Affiliation(s)
- Susumu Ishiguro
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Deepa Upreti
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Nicole Robben
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Riley Burghart
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Mayme Loyd
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Damilola Ogun
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Tran Le
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Jennifer Delzeit
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Arashi Nakashima
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Ravindra Thakkar
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | | | - Kengo Suzuki
- Euglena Co., Ltd., Minato-ku, Tokyo 108-0014, Japan.
| | - Jeffrey Comer
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Masaaki Tamura
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| |
Collapse
|
214
|
Elliot A, Myllymäki H, Feng Y. Inflammatory Responses during Tumour Initiation: From Zebrafish Transgenic Models of Cancer to Evidence from Mouse and Man. Cells 2020; 9:cells9041018. [PMID: 32325966 PMCID: PMC7226149 DOI: 10.3390/cells9041018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
The zebrafish is now an important model organism for cancer biology studies and provides unique and complementary opportunities in comparison to the mammalian equivalent. The translucency of zebrafish has allowed in vivo live imaging studies of tumour initiation and progression at the cellular level, providing novel insights into our understanding of cancer. Here we summarise the available transgenic zebrafish tumour models and discuss what we have gleaned from them with respect to cancer inflammation. In particular, we focus on the host inflammatory response towards transformed cells during the pre-neoplastic stage of tumour development. We discuss features of tumour-associated macrophages and neutrophils in mammalian models and present evidence that supports the idea that these inflammatory cells promote early stage tumour development and progression. Direct live imaging of tumour initiation in zebrafish models has shown that the intrinsic inflammation induced by pre-neoplastic cells is tumour promoting. Signals mediating leukocyte recruitment to pre-neoplastic cells in zebrafish correspond to the signals that mediate leukocyte recruitment in mammalian tumours. The activation state of macrophages and neutrophils recruited to pre-neoplastic cells in zebrafish appears to be heterogenous, as seen in mammalian models, which provides an opportunity to study the plasticity of innate immune cells during tumour initiation. Although several potential mechanisms are described that might mediate the trophic function of innate immune cells during tumour initiation in zebrafish, there are several unknowns that are yet to be resolved. Rapid advancement of genetic tools and imaging technologies for zebrafish will facilitate research into the mechanisms that modulate leukocyte function during tumour initiation and identify targets for cancer prevention.
Collapse
Affiliation(s)
| | | | - Yi Feng
- Correspondence: ; Tel.: +44-(0)131-242-6685
| |
Collapse
|
215
|
Zhang H, Lv H, Weng M, Wang H, Cata JP, Chen W, Miao C. Preoperative leukocytosis is associated with increased tumor-infiltrating neutrophil extracellular traps and worse outcomes in esophageal cancer. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:441. [PMID: 32395485 PMCID: PMC7210211 DOI: 10.21037/atm.2020.03.190] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background We evaluated the correlation between preoperative white blood cell (WBC) count and the prognosis in esophageal cancer (EC) patients who underwent esophagectomy, and explored the potential link between preoperative WBC count and tumor-infiltrating neutrophil extracellular traps (NETs) in EC. Methods From January 2013 to December 2017, 3,096 patients at Fudan University Shanghai Cancer Center (FUSCC) undergoing esophagectomy for EC were enrolled in this retrospective cohort. The prognostic value of preoperative WBC count together with tumor-infiltrating NETs was investigated. Results Leukocytosis (≥10,000/µL) was significantly associated with decreased overall survival (OS) and disease-free survival (DFS) (P<0.05). Further, moderate leukocytosis (≥7,000/µL) were also identified as an independent prognostic factor for survival. Additionally, moderate leukocytosis was correlated with male sex (P=0.006), advanced T stage (P<0.001), TNM stage (P<0.001) and ineffective postoperative chemotherapy (P<0.001), and moderate leukocytosis even predicted increased relapse postoperatively (P<0.001). Importantly, patients with moderate leukocytosis had a significantly higher level of intra-tumoral NETs infiltration (P<0.001), and the higher level of NETs infiltration were associated with worse OS and DFS (P<0.001). Conclusions Our data indicated that preoperative moderate leukocytosis is associated with increased tumor-infiltrating NETs and is an independent prognostic factor for survival in EC after surgery.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hu Lv
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Meilin Weng
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Huihui Wang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Juan P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Anaesthesiology and Surgical Oncology Research Group, Houston, TX, USA
| | - Wankun Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
216
|
Morgan MM, Schuler LA, Ciciliano JC, Johnson BP, Alarid ET, Beebe DJ. Modeling chemical effects on breast cancer: the importance of the microenvironment in vitro. Integr Biol (Camb) 2020; 12:21-33. [PMID: 32118264 PMCID: PMC7060306 DOI: 10.1093/intbio/zyaa002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/18/2019] [Accepted: 02/01/2020] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that our ability to predict chemical effects on breast cancer is limited by a lack of physiologically relevant in vitro models; the typical in vitro breast cancer model consists of the cancer cell and excludes the mammary microenvironment. As the effects of the microenvironment on cancer cell behavior becomes more understood, researchers have called for the integration of the microenvironment into in vitro chemical testing systems. However, given the complexity of the microenvironment and the variety of platforms to choose from, identifying the essential parameters to include in a chemical testing platform is challenging. This review discusses the need for more complex in vitro breast cancer models and outlines different approaches used to model breast cancer in vitro. We provide examples of the microenvironment modulating breast cancer cell responses to chemicals and discuss strategies to help pinpoint what components should be included in a model.
Collapse
Affiliation(s)
- Molly M Morgan
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Jordan C Ciciliano
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Brian P Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Elaine T Alarid
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Oncology, University of Wisconsin-Madison, Madison, WI, USA
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
217
|
Wu L, Saxena S, Singh RK. Neutrophils in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1224:1-20. [PMID: 32036601 DOI: 10.1007/978-3-030-35723-8_1] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neutrophils are the first responders to inflammation, infection, and injury. As one of the most abundant leukocytes in the immune system, neutrophils play an essential role in cancer progression, through multiple mechanisms, including promoting angiogenesis, immunosuppression, and cancer metastasis. Recent studies demonstrating elevated neutrophil to lymphocyte ratios suggest neutrophil as a potential therapeutic target and biomarker for disease status in cancer. This chapter will discuss the phenotypic and functional changes in the neutrophil in the tumor microenvironment, the underlying mechanism(s) of neutrophil facilitated cancer metastasis, and clinical potential of neutrophils as a prognostic/diagnostic marker and therapeutic target.
Collapse
Affiliation(s)
- Lingyun Wu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sugandha Saxena
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rakesh K Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
218
|
Vokalova L, Balogh A, Toth E, Van Breda SV, Schäfer G, Hoesli I, Lapaire O, Hahn S, Than NG, Rossi SW. Placental Protein 13 (Galectin-13) Polarizes Neutrophils Toward an Immune Regulatory Phenotype. Front Immunol 2020; 11:145. [PMID: 32117288 PMCID: PMC7028707 DOI: 10.3389/fimmu.2020.00145] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 01/20/2020] [Indexed: 12/17/2022] Open
Abstract
Termed as galectin-13, placental protein 13 (PP13) is exclusively expressed in the placenta of anthropoid primates. Research on PP13 in normal and pathologic pregnancies show alteration of PP13 concentrations in pregnancy affected by preeclampsia or gestational diabetes. Galectins are also described as potent immunomodulators, and PP13 regulates T cell function in the placenta. Therefore, this study aims to investigate the effects of PP13 on neutrophils; a cell type often ignored in pregnancy, but present in the uterus and placenta from the early stages of pregnancy. Since neutrophil function is dysregulated during pathologic pregnancies, a link between PP13 and neutrophil activity is possible. We determined that PP13 reduces the apoptosis rate in neutrophils. Also, PP13 increases the expression of PD-L1 and production of HGF, TNF-α, reactive oxygen species (ROS), and MMP-9 in these cells. This phenotype resembles one observed in permissive tumor neutrophils; able to sustain tissue and vessel growth, and inhibit T cell activation. At the same time, PP13 does not alter all neutrophil functions, i.e., extrusion of neutrophil extracellular traps, degranulation, phagocytosis, and ROS production following bacterial exposure. PP13 seems to play an essential role in regulating the activity of neutrophils in the placenta by polarizing them toward a placental-growth-permissive phenotype.
Collapse
Affiliation(s)
- Lenka Vokalova
- Prenatal Medicine, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Andrea Balogh
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Eszter Toth
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Shane V Van Breda
- Prenatal Medicine, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Günther Schäfer
- Prenatal Medicine, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Irene Hoesli
- Department of Antenatal Care, University Women's Hospital Basel, Basel, Switzerland
| | - Olav Lapaire
- Department of Antenatal Care, University Women's Hospital Basel, Basel, Switzerland
| | - Sinuhe Hahn
- Prenatal Medicine, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Nandor Gabor Than
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary.,First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary.,Maternity Private Clinic of Obstetrics and Gynecology, Budapest, Hungary
| | - Simona W Rossi
- Prenatal Medicine, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| |
Collapse
|
219
|
Shaul ME, Eyal O, Guglietta S, Aloni P, Zlotnik A, Forkosh E, Levy L, Weber LM, Levin Y, Pomerantz A, Nechushtan H, Eruslanov E, Singhal S, Robinson MD, Krieg C, Fridlender ZG. Circulating neutrophil subsets in advanced lung cancer patients exhibit unique immune signature and relate to prognosis. FASEB J 2020; 34:4204-4218. [PMID: 31957112 DOI: 10.1096/fj.201902467r] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/19/2019] [Accepted: 01/06/2020] [Indexed: 12/31/2022]
Abstract
The accumulation of circulating low-density neutrophils (LDN) has been described in cancer patients and associated with tumor-supportive properties, as opposed to the high-density neutrophils (HDN). Here we aimed to evaluate the clinical significance of circulating LDN in lung cancer patients, and further assessed its diagnostic vs prognostic value. Using mass cytometry (CyTOF), we identified major subpopulations within the circulating LDN/HDN subsets and determined phenotypic modulations of these subsets along tumor progression. LDN were highly enriched in the low-density (LD) fraction of advanced lung cancer patients (median 7.0%; range 0.2%-80%, n = 64), but not in early stage patients (0.7%; 0.05%-6%; n = 35), healthy individuals (0.8%; 0%-3.5%; n = 15), or stable chronic obstructive pulmonary disease (COPD) patients (1.2%; 0.3%-7.4%, n = 13). Elevated LDN (>10%) remarkably related with poorer prognosis in late stage patients. We identified three main neutrophil subsets which proportions are markedly modified in cancer patients, with CD66b+ /CD10low /CXCR4+ /PDL1inter subset almost exclusively found in advanced lung cancer patients. We found substantial variability in subsets between patients, and demonstrated that HDN and LDN retain a degree of inherent spontaneous plasticity. Deep phenotypic characterization of cancer-related circulating neutrophils and their modulation along tumor progression is an important advancement in understanding the role of myeloid cells in lung cancer.
Collapse
Affiliation(s)
- Merav E Shaul
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Ophir Eyal
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Silvia Guglietta
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Dermatology, Medical University of South Carolina, Charleston, SC, USA
| | - Pazzit Aloni
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Asaf Zlotnik
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Ester Forkosh
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Liran Levy
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Lukas M Weber
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Yonathan Levin
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Alon Pomerantz
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Hovav Nechushtan
- Sharrett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Evgeniy Eruslanov
- Thoracic Oncology Research Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Sunil Singhal
- Thoracic Oncology Research Laboratory, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark D Robinson
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Carsten Krieg
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.,Department of Dermatology, Medical University of South Carolina, Charleston, SC, USA
| | - Zvi G Fridlender
- Institute of Pulmonary Medicine, Hadassah Hebrew University Hospital, Jerusalem, Israel
| |
Collapse
|
220
|
Krishnamurthy A, Mittal S, Kothandaraman S, Dhanushkodi M, John A. Exploring the prognostic significance of the pretreatment inflammatory markers in hypopharyngeal cancers: A retrospective analysis. CANCER RESEARCH, STATISTICS, AND TREATMENT 2020. [DOI: 10.4103/crst.crst_152_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
221
|
Huang H, Zhang H, Onuma AE, Tsung A. Neutrophil Elastase and Neutrophil Extracellular Traps in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1263:13-23. [PMID: 32588320 PMCID: PMC11770835 DOI: 10.1007/978-3-030-44518-8_2] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tumor-associated neutrophils (TANs) play a major role during cancer development and progression in the tumor microenvironment. Neutrophil elastase (NE) is a serine protease normally expressed in neutrophil primary granules. Formation of neutrophil extracellular traps (NETs), a mechanism used by neutrophils, has been traditionally associated with the capture and killing of bacteria. However, there are recent discoveries suggesting that NE secretion and NETs formation are also involved in the tumor microenvironment. Here, we focus on how NE and NETs play a key regulatory function in the tumor microenvironment, such as tumor proliferation, distant metastasis, tumor-associated thrombosis, and antitumor activity. Additionally, the potential use of NETs, NE, or associated molecules as potential disease activity biomarkers or therapeutic targets will be introduced.
Collapse
Affiliation(s)
- Hai Huang
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
| | - Hongji Zhang
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
- Department of Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Amblessed E Onuma
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH, USA
| | - Allan Tsung
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
222
|
Wu L, Awaji M, Saxena S, Varney ML, Sharma B, Singh RK. IL-17-CXC Chemokine Receptor 2 Axis Facilitates Breast Cancer Progression by Up-Regulating Neutrophil Recruitment. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:222-233. [PMID: 31654638 PMCID: PMC6943375 DOI: 10.1016/j.ajpath.2019.09.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 07/31/2019] [Accepted: 09/04/2019] [Indexed: 12/20/2022]
Abstract
Recent evidence suggests that interactions among proinflammatory cytokines, chemokines, and cancer cell-recruited neutrophils result in enhanced metastasis and chemotherapy resistance. Nonetheless, the detailed mechanism remains unclear. Our aim was to discover the role of IL-17, CXC chemokine receptor 2 (CXCR2) ligands, and cancer-associated neutrophils in chemotherapy resistance and metastasis in breast cancer. Mice were injected with Cl66 murine mammary tumor cells, Cl66 cells resistant to doxorubicin (Cl66-Dox), or Cl66 cells resistant to paclitaxel (Cl66-Pac). Higher levels of IL-17 receptor, CXCR2 chemokines, and CXCR2 were observed in tumors generated from Cl66-Dox and Cl66-Pac cells in comparison with tumors generated from Cl66 cells. Tumors generated from Cl66-Dox and Cl66-Pac cells had higher infiltration of neutrophils and T helper 17 cells. In comparison with primary tumor sites, there were increased levels of CXCR2, CXCR2 ligands, and IL-17 receptor within the metastatic lesions. Moreover, IL-17 increased the expression of CXCR2 ligands and cell proliferation of Cl66 cells. The supernatant of Cl66-Dox and Cl66-Pac cells enhanced neutrophil chemotaxis. In addition, IL-17-induced neutrophil chemotaxis was dependent on CXCR2 signaling. Collectively, these data demonstrate that the IL-17-CXCR2 axis facilitates the recruitment of neutrophils to the tumor sites, thus allowing them to play a cancer-promoting role in cancer progression.
Collapse
Affiliation(s)
- Lingyun Wu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Mohammad Awaji
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sugandha Saxena
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Michelle L Varney
- Internal Medicine-Oncology/Hematology, University of Nebraska Medical Center, Omaha, Nebraska
| | | | - Rakesh K Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska.
| |
Collapse
|
223
|
Tang F, Tie Y, Tu C, Wei X. Surgical trauma-induced immunosuppression in cancer: Recent advances and the potential therapies. Clin Transl Med 2020; 10:199-223. [PMID: 32508035 PMCID: PMC7240866 DOI: 10.1002/ctm2.24] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 02/05/2023] Open
Abstract
Surgical resection remains the mainstay treatment for solid cancers, especially for localized disease. However, the postoperative immunosuppression provides a window for cancer cell proliferation and awakening dormant cancer cells, leading to rapid recurrences or metastases. This immunosuppressive status after surgery is associated with the severity of surgical trauma since immunosuppression induced by minimally invasive surgery is less than that of an extensive open surgery. The systemic response to tissue damages caused by surgical operations and the subsequent wound healing induced a cascade alteration in cellular immunity. After surgery, patients have a high level of circulating damage-associated molecular patterns (DAMPs), triggering a local and systemic inflammation. The inflammatory metrics in the immediate postoperative period was associated with the prognosis of cancer patients. Neutrophils provide the first response to surgical trauma, and the production of neutrophil extracellular traps (NETs) promotes cancer progression. Activated macrophage during wound healing presents a tumor-associated phenotype that cancers can exploit for their survival advantage. In addition, the amplification and activation of myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs) or the elevated programmed death ligand-1 and vascular endothelial growth factor expression under surgical trauma, exacerbate the immunosuppression and favor of the formation of the premetastatic niche. Therapeutic strategies to reduce the cellular immunity impairment after surgery include anti-DAMPs, anti-postoperative inflammation or inflammatory/pyroptosis signal, combined immunotherapy with surgery, antiangiogenesis and targeted therapies for neutrophils, macrophages, MDSCs, and Tregs. Further, the application of enhanced recovery after surgery also has a feasible outcome for postoperative immunity restoration. Overall, current therapies to improve the cellular immunity under the special condition after surgery are relatively lacking. Further understanding the underlying mechanisms of surgical trauma-related immunity dysfunction, phenotyping the immunosuppressive cells, and developing the related therapeutic intervention should be explored.
Collapse
Affiliation(s)
- Fan Tang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
- Department of OrthopeadicsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Yan Tie
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuanPeople's Republic of China
| | - Chongqi Tu
- Department of OrthopeadicsWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Xiawei Wei
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| |
Collapse
|
224
|
Unmasking the Many Faces of Tumor-Associated Neutrophils and Macrophages: Considerations for Targeting Innate Immune Cells in Cancer. Trends Cancer 2019; 5:789-798. [PMID: 31813456 DOI: 10.1016/j.trecan.2019.10.013] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 10/27/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023]
Abstract
Immunotherapy has emerged at the forefront of cancer therapy; however, patient survival remains low for many cancer types. In consideration of this, non-T cell immune populations, such as innate immune cells, have been identified as potential immunotherapeutic targets. In noncancerous settings, neutrophils are first responders to injury and infection, and work in a partnership with macrophages to regulate inflammation. However, the diversity of tumor-associated neutrophils (TANs) remains elusive. Furthermore, it is likely that TANs and tumor-associated macrophages (TAMs) act in tandem within tumors and contribute both contrasting and synergistic roles in tumor progression. In this Opinion, we discuss the complexity of TAN and TAM functions, the interplay between TANs and TAMs, and major considerations required for implementing TAN/TAM-based therapies.
Collapse
|
225
|
Polak KL, Chernosky NM, Smigiel JM, Tamagno I, Jackson MW. Balancing STAT Activity as a Therapeutic Strategy. Cancers (Basel) 2019; 11:cancers11111716. [PMID: 31684144 PMCID: PMC6895889 DOI: 10.3390/cancers11111716] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/23/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022] Open
Abstract
Driven by dysregulated IL-6 family member cytokine signaling in the tumor microenvironment (TME), aberrant signal transducer and activator of transcription (STAT3) and (STAT5) activation have been identified as key contributors to tumorigenesis. Following transformation, persistent STAT3 activation drives the emergence of mesenchymal/cancer-stem cell (CSC) properties, important determinants of metastatic potential and therapy failure. Moreover, STAT3 signaling within tumor-associated macrophages and neutrophils drives secretion of factors that facilitate metastasis and suppress immune cell function. Persistent STAT5 activation is responsible for cancer cell maintenance through suppression of apoptosis and tumor suppressor signaling. Furthermore, STAT5-mediated CD4+/CD25+ regulatory T cells (Tregs) have been implicated in suppression of immunosurveillance. We discuss these roles for STAT3 and STAT5, and weigh the attractiveness of different modes of targeting each cancer therapy. Moreover, we discuss how anti-tumorigenic STATs, including STAT1 and STAT2, may be leveraged to suppress the pro-tumorigenic functions of STAT3/STAT5 signaling.
Collapse
Affiliation(s)
- Kelsey L Polak
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| | - Noah M Chernosky
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| | - Jacob M Smigiel
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| | - Ilaria Tamagno
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| | - Mark W Jackson
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
226
|
Birch GP, Campbell T, Bradley M, Dhaliwal K. Optical Molecular Imaging of Inflammatory Cells in Interventional Medicine-An Emerging Strategy. Front Oncol 2019; 9:882. [PMID: 31572676 PMCID: PMC6751259 DOI: 10.3389/fonc.2019.00882] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022] Open
Abstract
The optical molecular imaging of inflammation is an emerging strategy for interventional medicine and diagnostics. The host's inflammatory response and adaptation to acute and chronic diseases provides unique signatures that have the potential to guide interventions. Thus, there are emerging a suite of molecular imaging and sensing approaches for a variety of targets in this area. This review will focus on two key cellular orchestrators that dominate this area, neutrophils and macrophages, with recent developments in molecular probes and approaches discussed.
Collapse
Affiliation(s)
- Gavin P Birch
- EaStChem School of Chemistry, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Thane Campbell
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark Bradley
- EaStChem School of Chemistry, University of Edinburgh, Edinburgh, United Kingdom
| | - Kevin Dhaliwal
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
227
|
An Autocrine Wnt5a Loop Promotes NF-κB Pathway Activation and Cytokine/Chemokine Secretion in Melanoma. Cells 2019; 8:cells8091060. [PMID: 31510045 PMCID: PMC6770184 DOI: 10.3390/cells8091060] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/29/2019] [Accepted: 09/05/2019] [Indexed: 12/19/2022] Open
Abstract
Wnt5a signaling has been implicated in the progression of cancer by regulating multiple cellular processes, largely migration and invasion, epithelial-mesenchymal transition (EMT), and metastasis. Since Wnt5a signaling has also been involved in inflammatory processes in infectious and inflammatory diseases, we addressed the role of Wnt5a in regulating NF-κB, a pivotal mediator of inflammatory responses, in the context of cancer. The treatment of melanoma cells with Wnt5a induced phosphorylation of the NF-κB subunit p65 as well as IKK phosphorylation and IκB degradation. By using cDNA overexpression, RNA interference, and dominant negative mutants we determined that ROR1, Dvl2, and Akt (from the Wnt5a pathway) and TRAF2 and RIP (from the NF-κB pathway) are required for the Wnt5a/NF-κB crosstalk. Wnt5a also induced p65 nuclear translocation and increased NF-κB activity as evidenced by reporter assays and a NF-κB-specific upregulation of RelB, Bcl-2, and Cyclin D1. Further, stimulation of melanoma cells with Wnt5a increased the secretion of cytokines and chemokines, including IL-6, IL-8, IL-11, and IL-6 soluble receptor, MCP-1, and TNF soluble receptor I. The inhibition of endogenous Wnt5a demonstrated that an autocrine Wnt5a loop is a major regulator of the NF-κB pathway in melanoma. Taken together, these results indicate that Wnt5a activates the NF-κB pathway and has an immunomodulatory effect on melanoma through the secretion of cytokines and chemokines.
Collapse
|
228
|
Jiang C, Cao S, Li N, Jiang L, Sun T. PD-1 and PD-L1 correlated gene expression profiles and their association with clinical outcomes of breast cancer. Cancer Cell Int 2019; 19:233. [PMID: 31516390 PMCID: PMC6734479 DOI: 10.1186/s12935-019-0955-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 09/02/2019] [Indexed: 12/16/2022] Open
Abstract
Background Immunotherapies that targeting programmed cell death 1 (PD-1) and programmed death-ligand 1 (PD-L1) have obtained prominent success in breast cancer (BC). However, not all the patients benefit from the antibody therapy. This study aimed to identify PD-1/PD-L1 correlated genes and pathways as well as investigate their potential as prognostic marker in BC. Materials and methods By analysing transcriptional data of BC from TCGA, we identified PD-1 and PD-L1 correlated genes by WGCNA analysis and explored the biological process as well as pathways they enriched. Co-expression analysis were performed for PD-1/PD-L1 with immune infiltration and checkpoints. The prognostic value of PD-1 and PD-L1 were also investigated. Results PD-1 and PD-L1 expression showed significant difference in different molecular subtypes and stages. PD-1 correlated genes enriched in T cell activation, lymphocyte activation, leukocyte migration while PD-L1 correlated genes demonstrated enrichment including T cell apoptotic process, tolerance induction and cytolysis. Immune infiltration analysis suggested that PD-1 and PD-L1 were related with Neutrophils (r = 0.65, r = 0.48) and Fibroblasts (r = 0.59, r = 0.47). For immune checkpoints analysis, PD-1 was associated with HLA-A (r = 0.804) and INPP5D (r = 0.782) while PD-L1 correlated with CTLA4 (r = 0.843) and CD27 (r = 0.823). PD-1 was associated favorable survival of BC (HR = 0.67, P = 0.012) while PD-L1 did not demonstrate significant association with BC prognosis (HR = 0.85, P = 0.313). Conclusion PD-1 and PD-L1 correlated genes participated in biological process including T cell activation, lymphocyte activation, leukocyte migration, T cell apoptotic process, tolerance induction and cytolysis. PD-1/PD-L1 expression also demonstrated relation with immune infiltration and immune checkpoints. High PD-1 expression predicted better survival of breast cancer patients.
Collapse
Affiliation(s)
- Cui Jiang
- 1Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and & Institute, 44 Xiaoheyan Road, Dadong District, Shenyang, 110042 Liaoning People's Republic of China
| | - SunRun Cao
- 2Institute of Translational Medicine, China Medical University, No.77 Puhe Road, Shenyang, 110001 Liaoning People's Republic of China
| | - Na Li
- 2Institute of Translational Medicine, China Medical University, No.77 Puhe Road, Shenyang, 110001 Liaoning People's Republic of China
| | - Lei Jiang
- 1Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and & Institute, 44 Xiaoheyan Road, Dadong District, Shenyang, 110042 Liaoning People's Republic of China
| | - Tao Sun
- 1Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and & Institute, 44 Xiaoheyan Road, Dadong District, Shenyang, 110042 Liaoning People's Republic of China
| |
Collapse
|
229
|
Cretella D, Digiacomo G, Giovannetti E, Cavazzoni A. PTEN Alterations as a Potential Mechanism for Tumor Cell Escape from PD-1/PD-L1 Inhibition. Cancers (Basel) 2019; 11:1318. [PMID: 31500143 PMCID: PMC6770107 DOI: 10.3390/cancers11091318] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 12/24/2022] Open
Abstract
The recent approval of immune checkpoint inhibitors drastically changed the standard treatments in many advanced cancer patients, but molecular changes within the tumor can prevent the activity of immunotherapy drugs. Thus, the introduction of the inhibitors of the immune checkpoint programmed death-1/programmed death ligand-1 (PD-1/PD-L1), should prompt deeper studies on resistance mechanisms, which can be caused by oncogenic mutations detected in cancer cells. PTEN, a tumor suppressor gene, dephosphorylates the lipid signaling intermediate PIP3 with inhibition of AKT activity, one of the main effectors of the PI3K signaling axis. As a consequence of genetic or epigenetic aberrations, PTEN expression is often altered, with increased activation of PI3K axis. Interestingly, some data confirmed that loss of PTEN expression modified the pattern of cytokine secretion creating an immune-suppressive microenvironment with increase of immune cell populations that can promote tumor progression. Moreover, PTEN loss may be ascribed to reduction of tumor infiltrating lymphocytes (TILs), which can explain the absence of activity of immune checkpoint inhibitors. This review describes the role of PTEN loss as a mechanism responsible for resistance to anti PD-1/PD-L1 treatment. Moreover, combinatorial strategies between PD-1/PD-L1 inhibitors and PI3K/AKT targeting drugs are proposed as a new strategy to overcome resistance to immune checkpoint inhibition.
Collapse
Affiliation(s)
- Daniele Cretella
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Graziana Digiacomo
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081HV Amsterdam, The Netherlands.
- Fondazione Pisana per la Scienza, 56017 Pisa, Italy.
| | - Andrea Cavazzoni
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
230
|
Buoncervello M, Gabriele L, Toschi E. The Janus Face of Tumor Microenvironment Targeted by Immunotherapy. Int J Mol Sci 2019; 20:E4320. [PMID: 31484464 PMCID: PMC6747403 DOI: 10.3390/ijms20174320] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 08/28/2019] [Accepted: 09/03/2019] [Indexed: 02/08/2023] Open
Abstract
The tumor microenvironment (TME) is a complex entity where host immune and non-immune cells establish a dynamic crosstalk with cancer cells. Through cell-cell interactions, which are mediated by key signals, such as the PD-1/PD-L1 axis, as well as the release of soluble mediators, this articulated process defines the nature of TME determining tumor development, prognosis, and response to therapy. Specifically, tumors are characterized by cellular plasticity that allows for the microenvironment to polarize towards inflammation or immunosuppression. Thus, the dynamic crosstalk among cancer, stromal, and immune components crucially favors the dominance of one of the Janus-faced contexture of TME crucial to the outcome of tumor development and therapeutic response. However, mostly, TME is dominated by an immunosuppressive landscape that blocks antitumor immunity and sustain tumor progression. Hence, in most cases, the immunosuppressive components of TME are highly competent in suppressing tumor-specific CD8+ T lymphocytes, the effectors of cancer destruction. In this complex context, immunotherapy aims to arm the hidden Janus face of TME disclosing and potentiating antitumor immune signals. Herein, we discuss recent knowledge on the immunosuppressive crosstalk within TME, and share perspectives on how immunotherapeutic approaches may exploit tumor immune signals to generate antitumor immunity.
Collapse
Affiliation(s)
- Maria Buoncervello
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Lucia Gabriele
- Tumor Immunology Section, Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Elena Toschi
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy.
| |
Collapse
|
231
|
Heeke S, Mograbi B, Alix-Panabières C, Hofman P. Never Travel Alone: The Crosstalk of Circulating Tumor Cells and the Blood Microenvironment. Cells 2019; 8:cells8070714. [PMID: 31337010 PMCID: PMC6678604 DOI: 10.3390/cells8070714] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/10/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022] Open
Abstract
Commonly, circulating tumor cells (CTCs) are described as source of metastasis in cancer patients. However, in this process cancer cells of the primary tumor site need to survive the physical and biological challenges in the blood stream before leaving the circulation to become the seed of a new metastatic site in distant parenchyma. Most of the CTCs released in the blood stream will not resist those challenges and will consequently fail to induce metastasis. A few of them, however, interact closely with other blood cells, such as neutrophils, platelets, and/or macrophages to survive in the blood stream. Recent studies demonstrated that the interaction and modulation of the blood microenvironment by CTCs is pivotal for the development of new metastasis, making it an interesting target for potential novel treatment strategies. This review will discuss the recent research on the processes in the blood microenvironment with CTCs and will outline currently investigated treatment strategies.
Collapse
Affiliation(s)
- Simon Heeke
- Université Côte d'Azur, CHU Nice, FHU OncoAge, 06000 Nice, France
- Université Côte d'Azur, CNRS UMR7284, Inserm U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), FHU OncoAge, 06000 Nice, France
| | - Baharia Mograbi
- Université Côte d'Azur, CHU Nice, FHU OncoAge, 06000 Nice, France
- Université Côte d'Azur, CNRS UMR7284, Inserm U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), FHU OncoAge, 06000 Nice, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre, EA2415, Montpellier University, 34093 Montpellier, France
| | - Paul Hofman
- Université Côte d'Azur, CHU Nice, FHU OncoAge, 06000 Nice, France.
- Université Côte d'Azur, CNRS UMR7284, Inserm U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), FHU OncoAge, 06000 Nice, France.
- Laboratory of Clinical and Experimental Pathology and Biobank BB-0033-00025, Pasteur Hospital, FHU OncoAge, 06000 Nice, France.
| |
Collapse
|
232
|
Otsuka I. Cutaneous Metastasis after Surgery, Injury, Lymphadenopathy, and Peritonitis: Possible Mechanisms. Int J Mol Sci 2019; 20:E3286. [PMID: 31277406 PMCID: PMC6651228 DOI: 10.3390/ijms20133286] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/27/2019] [Accepted: 07/01/2019] [Indexed: 12/12/2022] Open
Abstract
Cutaneous metastases from internal malignancies are uncommon. Umbilical metastasis, also known as Sister Joseph nodule (SJN), develops in patients with carcinomatous peritonitis or superficial lymphadenopathy, while non-SJN skin metastases develop after surgery, injury, and lymphadenopathy. In this review, the possible mechanisms of skin metastases are discussed. SJNs develop by the contiguous or lymphatic spread of tumor cells. After surgery and injury, tumor cells spread by direct implantation or hematogenous metastasis, and after lymphadenopathy, they spread by extranodal extension. The inflammatory response occurring during wound healing is exploited by tumor cells and facilitates tumor growth. Macrophages are crucial drivers of tumor-promoting inflammation, which is a source of survival, growth and angiogenic factors. Angiogenesis is promoted by the vascular endothelial growth factor (VEGF), which also mediates tumor-associated immunodeficiency. In the subcutaneous tissues that surround metastatic lymph nodes, adipocytes promote tumor growth. In the elderly, age-associated immunosuppression may facilitate hematogenous metastasis. Anti-VEGF therapy affects recurrence patterns but at the same time, may increase the risk of skin metastases. Immune suppression associated with inflammation may play a key role in skin metastasis development. Thus, immune therapies, including immune checkpoint inhibitors reactivating cytotoxic T-cell function and inhibiting tumor-associated macrophage function, appear promising.
Collapse
Affiliation(s)
- Isao Otsuka
- Department of Obstetrics and Gynecology, Kameda Medical Center, Kamogawa 296-8602, Japan.
| |
Collapse
|
233
|
Yang W, Shi J, Zhou Y, Liu T, Zhan F, Zhang K, Liu N. Integrating proteomics and transcriptomics for the identification of potential targets in early colorectal cancer. Int J Oncol 2019; 55:439-450. [PMID: 31268166 PMCID: PMC6615923 DOI: 10.3892/ijo.2019.4833] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/20/2019] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies worldwide. At present, CRC can often be treated upon diagnosis at stage I or II, or when dysplasia is detected; however, 60-70% of cases are not diagnosed until they have developed into late stages of the disease or until the malignancy is identified. Diagnosis of CRC at an early stage remains a challenge due to the absence of early-stage-specific biomarkers. To identify potential targets of early stage CRC, label-free proteomics analysis was applied to paired tumor-benign tissue samples from patients with stage II CRC (n=21). A total of 2,968 proteins were identified; corresponding RNA-Sequencing data were retrieved from The Cancer Genome Atlas-colon adenocarcinoma. Numerous bioinformatics methods, including differential expression analysis, weighted correlation network analysis, Gene Ontology and protein-protein interaction analyses, were applied to the proteomics and transcriptomics data. A total of 111 key proteins, which appeared as both differentially expressed proteins and mRNAs in the hub module, were identified as key candidates. Among these, three potential targets [protein-arginine deiminase type-2 (PADI2), Fc fragment of IgG binding protein (FCGBP) and phosphoserine aminotransferase 1] were identified from the pathological data. Furthermore, the survival analysis indicated that PADI2 and FCGBP were associated with the prognosis of CRC. The findings of the present study suggested potential targets for the identification of early stage CRC, and may improve understanding of the mechanism underlying the occurrence of CRC.
Collapse
Affiliation(s)
- Wang Yang
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Jian Shi
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yan Zhou
- Department of Gastrointestinal Surgery, The Second Hospital of Shandong University, Shandong 250000, P.R. China
| | - Tongjun Liu
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Fangling Zhan
- Central Laboratory, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Kai Zhang
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Ning Liu
- Central Laboratory, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
234
|
Wen L, Guo L, Zhang W, Li Y, Jiang W, Di X, Ma J, Feng L, Zhang K, Shou J. Cooperation Between the Inflammation and Coagulation Systems Promotes the Survival of Circulating Tumor Cells in Renal Cell Carcinoma Patients. Front Oncol 2019; 9:504. [PMID: 31263677 PMCID: PMC6590108 DOI: 10.3389/fonc.2019.00504] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 05/28/2019] [Indexed: 12/24/2022] Open
Abstract
Most renal cell carcinoma (RCC) patients die from metastasis or recurrence after the spread of cancer to another organ, but the mechanisms underlying the intravascular survival of circulating tumor cells (CTCs) have not been completely deciphered. Additionally, although elevated plasma C-reactive protein (CRP) levels and thrombocytosis are strongly correlated and both indicate a poor prognosis for RCC patients, the bridge connecting inflammation and coagulation remains poorly understood. To explore the complicated relationship among inflammation, the coagulation system and CTC survival, we obtained viable CTC counts and clinical information from 106 treatment-naïve patients. In addition, we performed RNA sequencing on peripheral blood leukocytes from 21 of these patients. Patients with elevated CRP and fibrinogen (FIB) levels had higher CTC counts than patients with normal levels of these indexes. Each pair of the three variables (CTC count, CRP level and FIB level) was positively correlated. According to transcriptomic analysis of blood leukocytes, the functions of the 257 genes identified as being positively correlated with the CTC count indicated neutrophil extracellular trap (NET) formation. Indeed, gene set enrichment analysis (GSEA) suggested that NET formation or increased levels of NET markers would promote CTC viability. Additionally, the calculated NET score was positively correlated with the plasma FIB concentration, and both of these values were increased in patients with elevated CRP levels. Moreover, immunofluorescence staining showed that NETs were entangled with viable renal cancer cells and that the NET frameworks were decorated with NET-derived tissue factor (TF). Finally, analysis of 533 RCC samples from The Cancer Genome Atlas (TCGA) indicated that the NET score and TF value are independent prognostic factors for RCC patients. Collectively, NETs formed by intravascular neutrophils further activate the coagulation system. Both the DNA scaffold sprouted and fibrin net triggered by NETs anchor and shield CTCs from attack. Thus, degrading this framework maybe could destroy the double shelter of CTCs, the pioneers of metastasis.
Collapse
Affiliation(s)
- Li Wen
- Department of Urinary Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liping Guo
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen Zhang
- Department of Immunology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yajian Li
- Department of Urinary Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weixing Jiang
- Department of Urinary Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xuebing Di
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - JianHui Ma
- Department of Urinary Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kaitai Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianzhong Shou
- Department of Urinary Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|