151
|
Bockorny B, Semenisty V, Macarulla T, Borazanci E, Wolpin BM, Stemmer SM, Golan T, Geva R, Borad MJ, Pedersen KS, Park JO, Ramirez RA, Abad DG, Feliu J, Muñoz A, Ponz-Sarvise M, Peled A, Lustig TM, Bohana-Kashtan O, Shaw SM, Sorani E, Chaney M, Kadosh S, Vainstein Haras A, Von Hoff DD, Hidalgo M. BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: the COMBAT trial. Nat Med 2020; 26:878-885. [PMID: 32451495 DOI: 10.1038/s41591-020-0880-x] [Citation(s) in RCA: 344] [Impact Index Per Article: 68.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 04/09/2020] [Indexed: 12/16/2022]
Abstract
Programmed cell death 1 (PD-1) inhibitors have limited effect in pancreatic ductal adenocarcinoma (PDAC), underscoring the need to co-target alternative pathways. CXC chemokine receptor 4 (CXCR4) blockade promotes T cell tumor infiltration and is synergistic with anti-PD-1 therapy in PDAC mouse models. We conducted a phase IIa, open-label, two-cohort study to assess the safety, efficacy and immunobiological effects of the CXCR4 antagonist BL-8040 (motixafortide) with pembrolizumab and chemotherapy in metastatic PDAC (NCT02826486). The primary outcome was objective response rate (ORR). Secondary outcomes were overall survival (OS), disease control rate (DCR) and safety. In cohort 1, 37 patients with chemotherapy-resistant disease received BL-8040 and pembrolizumab. The DCR was 34.5% in the evaluable population (modified intention to treat, mITT; N = 29), including nine patients (31%) with stable disease and one patient (3.4%) with partial response. Median OS (mOS) was 3.3 months in the ITT population. Notably, in patients receiving study drugs as second-line therapy, the mOS was 7.5 months. BL-8040 increased CD8+ effector T cell tumor infiltration, decreased myeloid-derived suppressor cells (MDSCs) and further decreased circulating regulatory T cells. In cohort 2, 22 patients received BL-8040 and pembrolizumab with chemotherapy, with an ORR, DCR and median duration of response of 32%, 77% and 7.8 months, respectively. These data suggest that combined CXCR4 and PD-1 blockade may expand the benefit of chemotherapy in PDAC and warrants confirmation in subsequent randomized trials.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antineoplastic Agents, Immunological
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- CD8-Positive T-Lymphocytes/pathology
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/secondary
- Female
- Fluorouracil/administration & dosage
- Humans
- Irinotecan/administration & dosage
- Leucovorin/administration & dosage
- Liver Neoplasms/drug therapy
- Liver Neoplasms/secondary
- Lung Neoplasms/drug therapy
- Lung Neoplasms/secondary
- Lymph Nodes/pathology
- Lymphatic Metastasis
- Lymphocytes, Tumor-Infiltrating/pathology
- Male
- Middle Aged
- Myeloid-Derived Suppressor Cells/pathology
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/pathology
- Peptides/administration & dosage
- Peritoneal Neoplasms/drug therapy
- Peritoneal Neoplasms/secondary
- Receptors, CXCR4/antagonists & inhibitors
- Retroperitoneal Neoplasms/drug therapy
- Retroperitoneal Neoplasms/secondary
- Survival Rate
- T-Lymphocytes, Regulatory/pathology
- Treatment Outcome
Collapse
Affiliation(s)
- Bruno Bockorny
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Teresa Macarulla
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, IOB Quirón, Barcelona, Spain
| | - Erkut Borazanci
- HonorHealth Research Institute, Scottsdale, AZ, USA
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Brian M Wolpin
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Salomon M Stemmer
- Institute of Oncology, Davidoff Center, Rabin Medical Center, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Talia Golan
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Oncology, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Ravit Geva
- Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Tel-Aviv University, Tel Aviv, Israel
| | - Mitesh J Borad
- Oncology, Mayo Clinic Cancer Center, Scottsdale, AZ, USA
| | | | - Joon Oh Park
- Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | - Jaime Feliu
- Instituto de Investigación Hospital Universitario La Paz (IdIPAZ), Cátedra UAM-AMGEN, CIBERONC, Madrid, Spain
| | - Andres Muñoz
- Medical Oncology Service, Instituto de Investigación Sanitaria Hospital Gregorio Marañón, Universidad Complutense, Madrid, Spain
| | - Mariano Ponz-Sarvise
- Clinica Universidad de Navarra and Program in Solid Tumors (CIMA), Universidad de Navarra, IDISNA, Pamplona, Spain
| | - Amnon Peled
- Goldyne Savad Institute of Gene Therapy, Hebrew University of Jerusalem, Jerusalem, Israel
| | | | | | | | | | - Marya Chaney
- Early Oncology Development, Merck & Co., Inc, Kenilworth, NJ, USA
| | | | | | - Daniel D Von Hoff
- HonorHealth Research Institute, Scottsdale, AZ, USA
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Manuel Hidalgo
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
- New York Presbyterian Hospital, New York, NY, USA.
| |
Collapse
|
152
|
Kell DB, Heyden EL, Pretorius E. The Biology of Lactoferrin, an Iron-Binding Protein That Can Help Defend Against Viruses and Bacteria. Front Immunol 2020; 11:1221. [PMID: 32574271 PMCID: PMC7271924 DOI: 10.3389/fimmu.2020.01221] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022] Open
Abstract
Lactoferrin is a nutrient classically found in mammalian milk. It binds iron and is transferred via a variety of receptors into and between cells, serum, bile, and cerebrospinal fluid. It has important immunological properties, and is both antibacterial and antiviral. In particular, there is evidence that it can bind to at least some of the receptors used by coronaviruses and thereby block their entry. Of importance are Heparan Sulfate Proteoglycans (HSPGs) and the host receptor angiotensin-converting enzyme 2 (ACE2), as based on other activities lactoferrin might prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from attaching to the host cells. Lactoferrin (and more specifically enteric-coated LF because of increased bioavailability) may consequently be of preventive and therapeutic value during the present COVID-19 pandemic.
Collapse
Affiliation(s)
- Douglas B Kell
- Department of Biochemistry, Faculty of Health and Life Sciences, Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom.,The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark.,Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | | | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
153
|
Horowitz LF, Rodriguez AD, Dereli-Korkut Z, Lin R, Castro K, Mikheev AM, Monnat RJ, Folch A, Rostomily RC. Multiplexed drug testing of tumor slices using a microfluidic platform. NPJ Precis Oncol 2020; 4:12. [PMID: 32435696 PMCID: PMC7237421 DOI: 10.1038/s41698-020-0117-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 03/25/2020] [Indexed: 12/11/2022] Open
Abstract
Current methods to assess the drug response of individual human cancers are often inaccurate, costly, or slow. Functional approaches that rapidly and directly assess the response of patient cancer tissue to drugs or small molecules offer a promising way to improve drug testing, and have the potential to identify the best therapy for individual patients. We developed a digitally manufactured microfluidic platform for multiplexed drug testing of intact cancer slice cultures, and demonstrate the use of this platform to evaluate drug responses in slice cultures from human glioma xenografts and patient tumor biopsies. This approach retains much of the tissue microenvironment and can provide results rapidly enough, within days of surgery, to guide the choice of effective initial therapies. Our results establish a useful preclinical platform for cancer drug testing and development with the potential to improve cancer personalized medicine.
Collapse
Affiliation(s)
- L. F. Horowitz
- Department of Bioengineering, University of Washington, Seattle, WA 98195 USA
- Department of Neurosurgery, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195 USA
- Department of Pathology, University of Washington, Seattle, WA 98195 USA
| | - A. D. Rodriguez
- Department of Bioengineering, University of Washington, Seattle, WA 98195 USA
| | - Z. Dereli-Korkut
- Department of Neurosurgery, Houston Methodist Hospital and Research Institute, Houston, TX USA
| | - R. Lin
- Department of Bioengineering, University of Washington, Seattle, WA 98195 USA
| | - K. Castro
- Department of Bioengineering, University of Washington, Seattle, WA 98195 USA
| | - A. M. Mikheev
- Department of Neurosurgery, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195 USA
- Department of Neurosurgery, Houston Methodist Hospital and Research Institute, Houston, TX USA
| | - R. J. Monnat
- Department of Pathology, University of Washington, Seattle, WA 98195 USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195 USA
| | - A. Folch
- Department of Bioengineering, University of Washington, Seattle, WA 98195 USA
| | - R. C. Rostomily
- Department of Neurosurgery, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195 USA
- Department of Neurosurgery, Houston Methodist Hospital and Research Institute, Houston, TX USA
- Weill Cornell School of Medicine, Department of Neurosurgery, New York, NY USA
| |
Collapse
|
154
|
Mehta A, Hwang WL, Weekes C. The present and future of systemic and microenvironment-targeted therapy for pancreatic adenocarcinoma. ANNALS OF PANCREATIC CANCER 2020; 3:3. [PMID: 33294843 PMCID: PMC7720884 DOI: 10.21037/apc-2020-pda-05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Metastatic pancreatic adenocarcinoma remains one of the deadliest cancer diagnoses with 5-year survival rates as low as 3%. For decades, gemcitabine remained the mainstay of systemic therapy before the approvals of FOLFIRINOX and gemcitabine with nab-paclitaxel. Despite these advances in the early 2010s, almost all patients progress on systemic chemotherapy and significant effort is needed to identify novel therapeutic targets. A promising array of approaches is currently under investigation, enabled by deeper understanding of the immune system within the tumor microenvironment (TME) and of the key vulnerabilities in pathways essential for tumor survival. In this review, we will explore the different approaches to boost tumor immunity and to target tumor metabolic pathways that are currently under clinical investigation for systemic treatment, and highlight the promising therapeutic areas that may give rise to the next generation of therapies for pancreatic cancer.
Collapse
Affiliation(s)
- Arnav Mehta
- Department of Hematology/Oncology, Massachusetts General Hospital, Boston, MA, USA
- Dana Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - William L. Hwang
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Colin Weekes
- Department of Hematology/Oncology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
155
|
Combined antitumor effects of anti-EGFR variant III CAR-T cell therapy and PD-1 checkpoint blockade on glioblastoma in mouse model. Cell Immunol 2020; 352:104112. [PMID: 32305131 DOI: 10.1016/j.cellimm.2020.104112] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 12/15/2022]
Abstract
Glioblastoma is one of the deadliest cancers. Chimeric antigen receptor (CAR)-T cell therapy against solid tumors has been far from satisfactory largely due to the immunosuppressive tumor microenvironment, such as PD-1 mediated T cell exhaustion. In the present study, we investigated the combined antitumor effects of anti-EGFR variant III CAR-T cell therapy and PD-1 checkpoint blockade on glioblastoma in mouse model. The results demonstrated that CAR-T cells with PD-1 blockade exhibit higher killing efficiency in vitro. Additionally, CAR-T cells with PD-1 blockade showed more effective and persistent therapeutic effects on glioblastoma and led to significantly increased number of tumor infiltrating lymphocytes (TILs) in the mouse model. In conclusion, PD-1 checkpoint blockade significantly enhanced the antitumor activity of anti-human EGFRvIII CAR-T cells by overcoming TILs exhaustion. The outcomes of the present study provide a novel strategy for improving the potency of CAR-T cell therapies in solid tumors.
Collapse
|
156
|
Deleuze A, Saout J, Dugay F, Peyronnet B, Mathieu R, Verhoest G, Bensalah K, Crouzet L, Laguerre B, Belaud-Rotureau MA, Rioux-Leclercq N, Kammerer-Jacquet SF. Immunotherapy in Renal Cell Carcinoma: The Future Is Now. Int J Mol Sci 2020; 21:ijms21072532. [PMID: 32260578 PMCID: PMC7177761 DOI: 10.3390/ijms21072532] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022] Open
Abstract
Renal cell carcinoma is the third type of urologic cancer and has a poor prognosis with 30% of metastatic patients at diagnosis. The antiangiogenics and targeted immunotherapies led to treatment remodeling emphasizing the role of the tumour microenvironment. However, long-term responses are rare with a high rate of resistance. New strategies are emerging to improve the efficacy and the emerging drugs are under evaluation in ongoing trials. With the different treatment options, there is an urgent need to identify biomarkers in order to predict the efficacy of drugs and to better stratify patients. Owing to the limitations of programmed death-ligand 1 (PD-L1), the most studied immunohistochemistry biomarkers, and of the tumor mutational burden, the identification of more reliable markers is an unmet need. New technologies could help in this purpose.
Collapse
Affiliation(s)
- Antoine Deleuze
- Université Rennes, Inserm, EHESP (Ecole des Hautes Etudes en Santé Publique), IRSET (Institut de recherche en santé, environnement et travail), UMR 1085, 35000 Rennes, France; (A.D.); (J.S.); (F.D.); (R.M.); (M.-A.B.-R.); (N.R.-L.)
- Department of Medical Oncology, Centre Eugene Marquis, 35000 Rennes, France; (L.C.); (B.L.)
| | - Judikaël Saout
- Université Rennes, Inserm, EHESP (Ecole des Hautes Etudes en Santé Publique), IRSET (Institut de recherche en santé, environnement et travail), UMR 1085, 35000 Rennes, France; (A.D.); (J.S.); (F.D.); (R.M.); (M.-A.B.-R.); (N.R.-L.)
| | - Frédéric Dugay
- Université Rennes, Inserm, EHESP (Ecole des Hautes Etudes en Santé Publique), IRSET (Institut de recherche en santé, environnement et travail), UMR 1085, 35000 Rennes, France; (A.D.); (J.S.); (F.D.); (R.M.); (M.-A.B.-R.); (N.R.-L.)
- Department of Cytogenetics, University Hospital, 35000 Rennes, France
| | - Benoit Peyronnet
- Department of Urology, University Hospital, 35000 Rennes, France; (B.P.); (G.V.); (K.B.)
| | - Romain Mathieu
- Université Rennes, Inserm, EHESP (Ecole des Hautes Etudes en Santé Publique), IRSET (Institut de recherche en santé, environnement et travail), UMR 1085, 35000 Rennes, France; (A.D.); (J.S.); (F.D.); (R.M.); (M.-A.B.-R.); (N.R.-L.)
- Department of Urology, University Hospital, 35000 Rennes, France; (B.P.); (G.V.); (K.B.)
| | - Gregory Verhoest
- Department of Urology, University Hospital, 35000 Rennes, France; (B.P.); (G.V.); (K.B.)
| | - Karim Bensalah
- Department of Urology, University Hospital, 35000 Rennes, France; (B.P.); (G.V.); (K.B.)
| | - Laurence Crouzet
- Department of Medical Oncology, Centre Eugene Marquis, 35000 Rennes, France; (L.C.); (B.L.)
| | - Brigitte Laguerre
- Department of Medical Oncology, Centre Eugene Marquis, 35000 Rennes, France; (L.C.); (B.L.)
| | - Marc-Antoine Belaud-Rotureau
- Université Rennes, Inserm, EHESP (Ecole des Hautes Etudes en Santé Publique), IRSET (Institut de recherche en santé, environnement et travail), UMR 1085, 35000 Rennes, France; (A.D.); (J.S.); (F.D.); (R.M.); (M.-A.B.-R.); (N.R.-L.)
- Department of Cytogenetics, University Hospital, 35000 Rennes, France
| | - Nathalie Rioux-Leclercq
- Université Rennes, Inserm, EHESP (Ecole des Hautes Etudes en Santé Publique), IRSET (Institut de recherche en santé, environnement et travail), UMR 1085, 35000 Rennes, France; (A.D.); (J.S.); (F.D.); (R.M.); (M.-A.B.-R.); (N.R.-L.)
- Department of Pathology, University Hospital, 35000 Rennes, France
| | - Solène-Florence Kammerer-Jacquet
- Université Rennes, Inserm, EHESP (Ecole des Hautes Etudes en Santé Publique), IRSET (Institut de recherche en santé, environnement et travail), UMR 1085, 35000 Rennes, France; (A.D.); (J.S.); (F.D.); (R.M.); (M.-A.B.-R.); (N.R.-L.)
- Department of Pathology, University Hospital, 35000 Rennes, France
- Correspondence: ; Tel.: +33-2-99-28-42-79; Fax: +33-2-99-28-42-84
| |
Collapse
|
157
|
Kenerson HL, Sullivan KM, Seo YD, Stadeli KM, Ussakli C, Yan X, Lausted C, Pillarisetty VG, Park JO, Riehle KJ, Yeh M, Tian Q, Yeung RS. Tumor slice culture as a biologic surrogate of human cancer. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:114. [PMID: 32175407 DOI: 10.21037/atm.2019.12.88] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background The tumor microenvironment (TME) is critical to every aspect of cancer biology. Organotypic tumor slice cultures (TSCs) preserve the original TME and have demonstrated utility in predicting drug sensitivity, but the association between clinicopathologic parameters and in vitro TSC behavior has not been well-defined. Methods One hundred and eight fresh tumor specimens from liver resections at a tertiary academic center were procured and precisely cut with a Vibratome to create 250 μm × 6 mm slices. These fixed-dimension TSCs were grown on polytetrafluoroethylene inserts, and their metabolic activities were determined by a colorimetric assay. Correlation between baseline activities and clinicopathologic parameters was assessed. Tissue CEA mRNA expression was determined by RNAseq. Results By standardizing the dimensions of a slice, we found that adjacent tumor slices have equivalent metabolic activities, while those derived from different tumors exhibit >30-fold range in baseline MTS absorbances, which correlated significantly with the percentage of tumor necrosis based on histologic assessment. Extending this to individual cancers, we were able to detect intra-tumoral heterogeneity over a span of a few millimeters, which reflects differences in tumor cell density and Ki-67 positivity. For colorectal cancers, tissue CEA expression based on RNAseq of tumor slices was found to correlate with clinical response to chemotherapies. Conclusions We report a standardized method to assess and compare human cancer growth ex vivo across a wide spectrum of tumor samples. TSC reflects the state of tumor behavior and heterogeneity, thus providing a simple approach to study of human cancers with an intact TME.
Collapse
Affiliation(s)
- Heidi L Kenerson
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Kevin M Sullivan
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Yongwoo D Seo
- Department of Surgery, University of Washington, Seattle, WA, USA
| | | | - Cigdem Ussakli
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Xiaowei Yan
- Institute for Systems Biology, Seattle, WA, USA
| | | | - Venu G Pillarisetty
- Department of Surgery, University of Washington, Seattle, WA, USA.,Northwest Liver Research Program, University of Washington, Seattle, WA, USA
| | - James O Park
- Department of Surgery, University of Washington, Seattle, WA, USA.,Northwest Liver Research Program, University of Washington, Seattle, WA, USA
| | - Kimberly J Riehle
- Department of Surgery, University of Washington, Seattle, WA, USA.,Northwest Liver Research Program, University of Washington, Seattle, WA, USA
| | - Matthew Yeh
- Department of Pathology, University of Washington, Seattle, WA, USA.,Northwest Liver Research Program, University of Washington, Seattle, WA, USA
| | - Qiang Tian
- Institute for Systems Biology, Seattle, WA, USA
| | - Raymond S Yeung
- Department of Surgery, University of Washington, Seattle, WA, USA.,Northwest Liver Research Program, University of Washington, Seattle, WA, USA
| |
Collapse
|
158
|
Chuan T, Li T, Yi C. Identification of CXCR4 and CXCL10 as Potential Predictive Biomarkers in Triple Negative Breast Cancer (TNBC). Med Sci Monit 2020; 26:e918281. [PMID: 31924747 PMCID: PMC6977636 DOI: 10.12659/msm.918281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Efficacious therapy for triple negative breast cancer (TNBC) continues to be a profound clinical challenge, but the key driven genes and convoluted signaling pathways are still unknown. MATERIAL AND METHODS A total of 223 samples (163 TNBC and 60 healthy breast tissues) were taken and deeply integrated analyzed by R software from 4 expression profiles in the study, including GSE53752, GSE45827, GSE65194, and GSE38959. We examined differentially expressed genes (DEGs) and screen for critical genes and pathways enrichment. The protein‑protein interaction (PPI) network of DEGs-associated was built through the STRING Version: 11.0 database and Cytoscape software to filter the hub gene. Then, we verified hug gene expression levels through the Oncomine database. Also, we analyzed the prognostic value of TNBC patient's hub genes using the Kaplan-Meier plotter database. RESULTS In our study, we filter out 365 DEGs, including 212 upregulated genes and 153 downregulated genes. Then, 10 hub genes were picked out by the intersection of 12 algorithms. At the same time, we discovered that CXCR4 and CXCL10 overexpression are favorable prognostic factors for recurrence-free survival of TNBC through the Kaplan-Meier plotter database. CONCLUSIONS Our research found that CXCR4 and CXCL10 overexpressed, and they were a favorable prognostic factor in patients with TNBC. CXCR4 and CXCL10 might be effective targets for TNBC therapy.
Collapse
Affiliation(s)
- Tian Chuan
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China (mainland)
| | - Tian Li
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China (mainland)
| | - Cui Yi
- Clinical Research Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China (mainland)
| |
Collapse
|
159
|
Chifu I, Heinze B, Fuss CT, Lang K, Kroiss M, Kircher S, Ronchi CL, Altieri B, Schirbel A, Fassnacht M, Hahner S. Impact of the Chemokine Receptors CXCR4 and CXCR7 on Clinical Outcome in Adrenocortical Carcinoma. Front Endocrinol (Lausanne) 2020; 11:597878. [PMID: 33281749 PMCID: PMC7691376 DOI: 10.3389/fendo.2020.597878] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 10/20/2020] [Indexed: 12/22/2022] Open
Abstract
Chemokine receptors have a negative impact on tumor progression in several human cancers and have therefore been of interest for molecular imaging and targeted therapy. However, their clinical and prognostic significance in adrenocortical carcinoma (ACC) is unknown. The aim of this study was to evaluate the chemokine receptor profile in ACC and to analyse its association with clinicopathological characteristics and clinical outcome. A chemokine receptor profile was initially evaluated by quantitative PCR in 4 normal adrenals, 18 ACC samples and human ACC cell line NCI-H295. High expression of CXCR4 and CXCR7 in both healthy and malignant adrenal tissue and ACC cells was confirmed. In the next step, we analyzed the expression and cellular localization of CXCR4 and CXCR7 in ACC by immunohistochemistry in 187 and 84 samples, respectively. These results were correlated with clinicopathological parameters and survival outcome. We detected strong membrane expression of CXCR4 and CXCR7 in 50% of ACC samples. Strong cytoplasmic CXCR4 staining was more frequent among samples derived from metastases compared to primaries (p=0.01) and local recurrences (p=0.04). CXCR4 membrane staining positively correlated with proliferation index Ki67 (r=0.17, p=0.028). CXCR7 membrane staining negatively correlated with Ki67 (r=-0.254, p=0.03) but positively with tumor size (r=0.3, p=0.02). No differences in progression-free or overall survival were observed between patients with strong and weak staining intensities for CXCR4 or CXCR7. Taken together, high expression of CXCR4 and CXCR7 in both local tumors and metastases suggests that some ACC patients might benefit from CXCR4/CXCR7-targeted therapy.
Collapse
Affiliation(s)
- Irina Chifu
- Division of Endocrinology and Diabetes, Department of Medicine I, University Hospital of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
| | - Britta Heinze
- Division of Endocrinology and Diabetes, Department of Medicine I, University Hospital of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
- *Correspondence: Britta Heinze,
| | - Carmina T. Fuss
- Division of Endocrinology and Diabetes, Department of Medicine I, University Hospital of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
| | - Katharina Lang
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Matthias Kroiss
- Division of Endocrinology and Diabetes, Department of Medicine I, University Hospital of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Wuerzburg, Wuerzburg, Germany
| | - Stefan Kircher
- Institute of Pathology, Interdisciplinary Bank of Biomaterials and Data (ibdw), University of Wuerzburg, Wuerzburg, Germany
| | - Cristina L. Ronchi
- Division of Endocrinology and Diabetes, Department of Medicine I, University Hospital of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, United Kingdom
| | - Barbara Altieri
- Division of Endocrinology and Diabetes, Department of Medicine I, University Hospital of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
| | - Andreas Schirbel
- Comprehensive Cancer Center Mainfranken, University of Wuerzburg, Wuerzburg, Germany
- Department of Nuclear Medicine, University Hospital of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
| | - Martin Fassnacht
- Division of Endocrinology and Diabetes, Department of Medicine I, University Hospital of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Wuerzburg, Wuerzburg, Germany
| | - Stefanie Hahner
- Division of Endocrinology and Diabetes, Department of Medicine I, University Hospital of Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
160
|
Abstract
Renal fibrosis is the final pathological process common to any ongoing, chronic kidney injury or maladaptive repair. Renal fibrosis is considered to be closely related to various cell types, such as fibroblasts, myofibroblasts, T cells, and other inflammatory cells. Multiple types of cells regulate renal fibrosis through the recruitment, proliferation, and activation of fibroblasts, and the production of the extracellular matrix. Cell trafficking is orchestrated by a family of small proteins called chemokines. Chemokines are cytokines with chemotactic properties, which are classified into 4 groups: CXCL, CCL, CX3CL, and XCL. Similarly, chemokine receptors are G protein-coupled seven-transmembrane receptors classified into 4 groups: XCR, CCR, CXCR, and CX3CR. Chemokine receptors are also implicated in the infiltration, differentiation, and survival of functional cells, triggering inflammation that leads to fibrosis development. In this review, we summarize the different chemokine receptors involved in the processes of fibrosis in different cell types. Further studies are required to identify the molecular mechanisms of chemokine signaling that contribute to renal fibrosis.
Collapse
|
161
|
Xun Y, Yang H, Li J, Wu F, Liu F. CXC Chemokine Receptors in the Tumor Microenvironment and an Update of Antagonist Development. Rev Physiol Biochem Pharmacol 2020; 178:1-40. [PMID: 32816229 DOI: 10.1007/112_2020_35] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chemokine receptors, a diverse group within the seven-transmembrane G protein-coupled receptor superfamily, are frequently overexpressed in malignant tumors. Ligand binding activates multiple downstream signal transduction cascades that drive tumor growth and metastasis, resulting in poor clinical outcome. These receptors are thus considered promising targets for anti-tumor therapy. This article reviews recent studies on the expression and function of CXC chemokine receptors in various tumor microenvironments and recent developments in cancer therapy using CXC chemokine receptor antagonists.
Collapse
Affiliation(s)
- Yang Xun
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Hua Yang
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Jiekai Li
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Fuling Wu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Fang Liu
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, China.
| |
Collapse
|
162
|
Daniel SK, Seo YD, Pillarisetty VG. The CXCL12-CXCR4/CXCR7 axis as a mechanism of immune resistance in gastrointestinal malignancies. Semin Cancer Biol 2019; 65:176-188. [PMID: 31874281 DOI: 10.1016/j.semcancer.2019.12.007] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/03/2019] [Accepted: 12/11/2019] [Indexed: 02/07/2023]
Abstract
Single agent checkpoint inhibitor therapy has not been effective for most gastrointestinal solid tumors, but combination therapy with drugs targeting additional immunosuppressive pathways is being attempted. One such pathway, the CXCL12-CXCR4/CXCR7 chemokine axis, has attracted attention due to its effects on tumor cell survival and metastasis as well as immune cell migration. CXCL12 is a small protein that functions in normal hematopoietic stem cell homing in addition to repair of damaged tissue. Binding of CXCL12 to CXCR4 leads to activation of G protein signaling kinases such as P13K/mTOR and MEK/ERK while binding to CXCR7 leads to β-arrestin mediated signaling. While some gastric and colorectal carcinoma cells have been shown to make CXCL12, the primary source in pancreatic cancer and peritoneal metastases is cancer-associated fibroblasts. Binding of CXCL12 to CXCR4 and CXCR7 on tumor cells leads to anti-apoptotic signaling through Bcl-2 and survivin upregulation, as well as promotion of the epithelial-to-mesechymal transition through the Rho-ROCK pathway and alterations in cell adhesion molecules. High levels of CXCL12 seen in the bone marrow, liver, and spleen could partially explain why these are popular sites of metastases for many tumors. CXCL12 is a chemoattractant for lymphocytes at lower levels, but becomes chemorepellant at higher levels; it is unclear exactly what gradient exists in the tumor microenvironment and how this influences tumor-infiltrating lymphocytes. AMD3100 (Plerixafor or Mozobil) is a small molecule CXCR4 antagonist and is the most frequently used drug targeting the CXCL12-CXCR4/CXCR7 axis in clinical trials for gastrointestinal solid tumors currently. Other small molecules and monoclonal antibodies against CXCR4 are being trialed. Further understanding of the CXCL12- CXCR4/CXCR7 chemokine axis in the tumor microenvironment will allow more effective targeting of this pathway in combination immunotherapy.
Collapse
Affiliation(s)
- Sara K Daniel
- University of Washington, Dept. of Surgery, Seattle, WA, USA
| | - Y David Seo
- University of Washington, Dept. of Surgery, Seattle, WA, USA
| | | |
Collapse
|
163
|
Zhang PF, Pei X, Li KS, Jin LN, Wang F, Wu J, Zhang XM. Circular RNA circFGFR1 promotes progression and anti-PD-1 resistance by sponging miR-381-3p in non-small cell lung cancer cells. Mol Cancer 2019; 18:179. [PMID: 31815619 PMCID: PMC6900862 DOI: 10.1186/s12943-019-1111-2] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 11/25/2019] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Immune system evasion, distance tumor metastases, and increased cell proliferation are the main reasons for the progression of non-small cell lung cancer (NSCLC) and the death of NSCLC patients. Dysregulation of circular RNAs plays a critical role in the progression of NSCLC; therefore, further understanding the biological mechanisms of abnormally expressed circRNAs is critical to discovering novel, promising therapeutic targets for NSCLC treatment. METHODS The expression of circular RNA fibroblast growth factor receptor 1 (circFGFR1) in NSCLC tissues, paired nontumor tissues, and cell lines was detected by RT-qPCR. The role of circFGFR1 in NSCLC progression was assessed both in vitro by CCK-8, clonal formation, wound healing, and Matrigel Transwell assays and in vivo by a subcutaneous tumor mouse assay. In vivo circRNA precipitation, RNA immunoprecipitation, and luciferase reporter assays were performed to explore the interaction between circFGFR1 and miR-381-3p. RESULTS Here, we report that circFGFR1 is upregulated in NSCLC tissues, and circFGFR1 expression is associated with deleterious clinicopathological characteristics and poor prognoses for NSCLC patients. Forced circFGFR1 expression promoted the migration, invasion, proliferation, and immune evasion of NSCLC cells. Mechanistically, circFGFR1 could directly interact with miR-381-3p and subsequently act as a miRNA sponge to upregulate the expression of the miR-381-3p target gene C-X-C motif chemokine receptor 4 (CXCR4), which promoted NSCLC progression and resistance to anti-programmed cell death 1 (PD-1)- based therapy. CONCLUSION Taken together, our results suggest the critical role of circFGFR1 in the proliferation, migration, invasion, and immune evasion abilities of NSCLC cells and provide a new perspective on circRNAs during NSCLC progression.
Collapse
Affiliation(s)
- Peng-Fei Zhang
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xu Pei
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ke-Sang Li
- Department of Hematology and Oncology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Zrhejiang, Ningbo, China
| | - Li-Na Jin
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fei Wang
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jing Wu
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xue-Mei Zhang
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
164
|
The Gastrointestinal Tumor Microenvironment: An Updated Biological and Clinical Perspective. JOURNAL OF ONCOLOGY 2019; 2019:6240505. [PMID: 31885581 PMCID: PMC6893275 DOI: 10.1155/2019/6240505] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/30/2019] [Indexed: 12/24/2022]
Abstract
Gastrointestinal cancers are still responsible for high numbers of cancer-related deaths despite advances in therapy. Tumor-associated cells play a key role in tumor biology, by supporting or halting tumor development through the production of extracellular matrix, growth factors, cytokines, and extracellular vesicles. Here, we review the roles of these tumor-associated cells in the initiation, angiogenesis, immune modulation, and resistance to therapy of gastrointestinal cancers. We also discuss novel diagnostic and therapeutic strategies directed at tumor-associated cells and their potential benefits for the survival of these patients.
Collapse
|
165
|
Xing Y, Zha WJ, Li XM, Li H, Gao F, Ye T, Du WQ, Liu YC. Circular RNA circ-Foxo3 inhibits esophageal squamous cell cancer progression via the miR-23a/PTEN axis. J Cell Biochem 2019; 121:2595-2605. [PMID: 31680314 DOI: 10.1002/jcb.29481] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 10/10/2019] [Indexed: 12/17/2022]
Abstract
Circ-Foxo3 is a circRNA encoded by the human FOXO3 gene and works as a sponge for potential microRNAs (miRNAs) to regulate cancer progression. However, the role of circ-Foxo3 in esophageal squamous cell cancer (ESCC) is not clear. In this study, circ-Foxo3 was lowly expressed in cell lines and ESCC tissues. Meanwhile, overexpression of circ-Foxo3 inhibited cell growth, migration, and invasion, whether in vivo or in vitro. Mechanically, we found a potential miRNA target, miR-23a, which negatively correlated with circ-Foxo3 in ESCC. Then, a luciferase assay confirmed the relationship between the circ-Foxo3 and miRNA. Moreover, circ-Foxo3 upregulation of PTEN occurred through "sponging" miR-23a. Taken together, these results indicated that the circ-Foxo3/miR-23a/PTEN pathway was critical for inhibiting the ESCC progression. This may provide a promising target for treat ESCC.
Collapse
Affiliation(s)
- Yao Xing
- Department of Radiotherapy, Taixing People's Hospital Affiliated to Bengbu Medical College, Bengbu, China
| | - Wen-Juan Zha
- Department of Radiotherapy, Taixing People's Hospital Affiliated to Bengbu Medical College, Bengbu, China
| | - Xiao-Min Li
- Department of Radiotherapy, Taixing People's Hospital Affiliated to Bengbu Medical College, Bengbu, China
| | - Hao Li
- Department of Clinical Laboratory, Taixing People's Hospital, Taixing, China
| | - Fei Gao
- Department of Radiotherapy, Taixing People's Hospital, Taixing, China
| | - Ting Ye
- Department of Clinical Laboratory, Taixing People's Hospital, Taixing, China
| | - Wang-Qi Du
- Department of Clinical Laboratory, Taixing People's Hospital, Taixing, China
| | - Yang-Chen Liu
- Department of Radiotherapy, Taixing People's Hospital, Taixing, China
| |
Collapse
|
166
|
Zhao R, Ni J, Lu S, Jiang S, You L, Liu H, Shou J, Zhai C, Zhang W, Shao S, Yang X, Pan H, Han W. CircUBAP2-mediated competing endogenous RNA network modulates tumorigenesis in pancreatic adenocarcinoma. Aging (Albany NY) 2019; 11:8484-8501. [PMID: 31584877 PMCID: PMC6814619 DOI: 10.18632/aging.102334] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 09/22/2019] [Indexed: 12/24/2022]
Abstract
We investigated the role of the competing endogenous RNA (ceRNA) network in the development and progression of pancreatic adenocarcinoma (PAAD). We analyzed the expression profiles of PAAD and normal pancreatic tissues from multiple GEO databases and identified 457 differentially expressed circular RNAs (DEcircRNAs), 19 microRNAs (DEmiRNAs) and 1993 mRNAs (DEmRNAs). We constructed a ceRNA network consisting of 4 DEcircRNAs, 3 DEmiRNAs and 149 DEmRNAs that regulates the NF-kappa B, PI3K-Akt, and Wnt signaling pathways. We then identified and validated five hub genes, CXCR4, HIF1A, ZEB1, SDC1 and TWIST1, which are overexpressed in PAAD tissues. The expression of CXCR4, HIF1A, ZEB1, and SDC1 in PAAD was regulated by circ-UBAP2 and hsa-miR-494. The expression of CXCR4 and ZEB1 correlated with the levels of M2 macrophages, T-regulatory cells (Tregs) and exhausted T cells in the PAAD tissues. The expression of CXCR4 and ZEB1 positively correlated with the expression of CTLA-4 and PD-1. This suggests that CXCR4 and ZEB1 proteins inhibit antigen presentation and promote immune escape mechanisms in PAAD cells. In summary, our data suggest that the circUBAP2-mediated ceRNA network modulates PAAD by regulating the infiltration and function of immune cells.
Collapse
Affiliation(s)
- Rongjie Zhao
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| | - Junjie Ni
- The First Clinical College, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Si Lu
- The Fourth Clinical College, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Sujing Jiang
- Department of Radiation and Medical Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Liangkun You
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| | - Hao Liu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| | - Jiawei Shou
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| | - Chongya Zhai
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| | - Wei Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| | - Shengpeng Shao
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Xinmei Yang
- Department of Oncology, The First Affiliated Hospital of Jiaxing University, Jiaxing 314000, Zhejiang, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang, China
| |
Collapse
|
167
|
Ma Y, Feng XF, Yang WX, You CG. Exploring the Pathological Mechanism of Bladder Cancer Based on Tumor Mutational Burden Analysis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1093815. [PMID: 31534952 PMCID: PMC6732589 DOI: 10.1155/2019/1093815] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/28/2019] [Indexed: 01/10/2023]
Abstract
Although immunotherapy has progressed in the treatment of bladder cancer, some patients still have poor prognosis. New therapeutic targets are eager to be discovered to improve the outcomes of bladder cancer. With the development of high-throughput sequencing and tumor profiling, potential tumor biomarkers were identified. Through the interpretation of related data from the Cancer Genome Atlas database (TCGA), some key genes have been discovered to drive the development and prognosis of urinary bladder neoplasm. On account of the success of immunotherapy in many cancer types, we established the relationship between tumor mutation burden and immune microenvironment of bladder cancer and found the changes of several immune cells in this disease. Based on the understanding of the bladder tumor genome and immune environment, this study is supposed to provide new therapies for the treatment of bladder neoplasm.
Collapse
Affiliation(s)
- Yao Ma
- Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Xiao-Fei Feng
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730030, China
- Key Laboratory of Osteoarthritis of Gansu Province, Lanzhou University Second Hospital, No. 82 Cuiyingmen, Lanzhou, Gansu 730030, China
| | - Wan-Xia Yang
- Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Chong-Ge You
- Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou 730030, China
| |
Collapse
|
168
|
Medina BD, Miller G. Rethinking T Cells in Pancreas Cancer. Clin Cancer Res 2019; 25:3747-3749. [PMID: 31072844 DOI: 10.1158/1078-0432.ccr-19-1073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 11/16/2022]
Abstract
Patients with pancreatic ductal adenocarcinoma do not benefit from checkpoint blockade. However, human tumors harbor evidence of adaptive immunity in clonally expanded T-cell populations. Immune intact modeling of human tumors identifies stromal sequestration as a mechanism of immune escape. Targeting the stroma combined with checkpoint blockade unleashes antitumor immunity.See related article by Seo et al., p. 3934.
Collapse
Affiliation(s)
- Benjamin D Medina
- Department of Surgery, NYU Langone Medical Center, New York, New York
| | - George Miller
- Department of Surgery, NYU Langone Medical Center, New York, New York.
| |
Collapse
|